Bio

Bio


Dr. Ingelsson obtained his MD (2000) and PhD (2005) at Uppsala University, Sweden. After internship, he did a residency in general medicine (2003-2006) and took up a postdoctoral research fellowship at the Framingham Heart Study (2006-2007). He moved to Karolinska Institutet (Stockholm, Sweden) in 2007 and was appointed Professor of Cardiovascular Epidemiology in 2010. From 2013-2016, he was a Professor of Molecular Epidemiology at Uppsala University. He was also a Visiting Professor at the Wellcome Trust Centre for Human Genetics at University of Oxford in 2012-2015. Since May 2016, he is Professor of Medicine at Stanford University.

For the past fifteen years, Dr. Ingelsson has been doing cardiovascular research with a special focus on the role of obesity and insulin resistance in development of subclinical and clinical cardiovascular disease. His research is translational and interdisciplinary, combining big data approaches, such as -omics in population-based cohorts, with gene editing in functional model systems to reach new insights into the pathophysiology of cardiovascular disease and related conditions, identification of new biomarkers for improved risk prediction, and discovery of novel targets for drug development.

He has had a leading role in many of the large efforts identifying new genetic loci associated with cardiovascular and metabolic traits, and has extensive experience from research on biomarkers and -omics methods, including development and application of prediction metrics and Mendelian randomization. He has served as PI of numerous –omics efforts in several Swedish cohort studies, including ULSAM, PIVUS, TwinGene and EpiHealth. Since 2013, his laboratory has refocused much of their research efforts towards characterization of genes discovered in genome-wide association studies using a combination of in-depth studies in human (including various -omics methods), in vitro studies (primarily adipocytes, hepatocytes and skeletal myocytes) and in vivo models (mice and zebrafish).

He has published over 300 peer-reviewed original articles (>110 as lead author), and >60 in journals with impact factor over 30. Before relocating to the U.S, he received many large European research grants, and after joining the Stanford faculty in May 2016, he has built a strong and well-funded research program. He has won several prestigious awards, such as the AHA Trudy Bush Fellowship for Cardiovascular Research in Women’s Health, ERC starting grant, Wallenberg Academy Fellow and the Göran Gustafsson Prize in Medicine in 2015 (to the most successful medical researcher in Sweden under age 45).

Academic Appointments


Administrative Appointments


  • Committee Member, Department of Medicine’s Professoriate Appointments and Promotions Committee (2018 - Present)
  • Affinity Group Leader, Stanford Diabetes Research Center (2017 - Present)
  • National Director, EATRIS.se (Swedish node of the European Infrastructure for Translational Medicine) (2015 - 2016)
  • Board Member, Faculty Board of the Disciplinary Domain of Medicine and Pharmacy, Uppsala University (2014 - 2015)
  • Executive Group Member, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet (2010 - 2012)

Honors & Awards


  • Göran Gustafsson Prize in Medicine, Göran Gustafsson Foundation (2015)
  • Wallenberg Academy Fellow, Knut och Alice Wallenberg Foundation (2013)
  • ERC Starting Grant Award, European Research Council (2013)
  • Fellow of the American Heart Association (FAHA), American Heart Association (2010)
  • Ingvar Carlsson Award, Swedish Foundation for Strategic Research (2009)
  • Trudy Bush Fellowship for Cardiovascular Research in Women’s Health, American Heart Association (2009)
  • Young Investigator Award, EuroPRevent 2008, European Association of Cardiovascular Prevention and Rehabilitation (2008)

Boards, Advisory Committees, Professional Organizations


  • Scientific advisor, Precision Wellness (2016 - Present)
  • Scientific advisor, Olink Proteomics (2017 - Present)

Professional Education


  • PhD, Uppsala University, Epidemiology (2005)
  • MD, Uppsala University, Medicine (2000)

Patents


  • Lindholm D, Fukaya E, Leeper NJ, Ingelsson E. "United States Patent 62/522,601 Systems and Methods for Predicting Heart Failure Using Leg Bioimpedance", Leland Stanford Junior University, Jun 20, 2017

Research & Scholarship

Current Research and Scholarly Interests


Our research area is cardiovascular medicine with a special focus on metabolic disturbances, such as obesity and insulin resistance and their role in the development of subclinical and clinical cardiovascular disease. The methods used are from the molecular epidemiology field where we use -omics studies of how cardiovascular disease and related conditions varies with DNA variation, RNA expression, and circulating biomarkers such as proteins and metabolites; but also functional characterization of candidate genes using CRISPR-Cas9 gene editing in cell and animal models. Our research is translational, trying to bridge population studies with molecular biology to reach new important insights into the pathophysiology of cardiovascular diseases, identification of new biomarkers for improved risk prediction, and discovery of novel targets for drug development.

Population-based studies
We are performing many population-based projects in the UK Biobank, which is an excellent example of science in the new era of open science initiatives and big data analytics. In 2006-2010, the UK Biobank recruited 502,650 participants aged 37-73 years to undergo physical measurements, detailed assessments about risk factors and future disease events, and sampling of blood, urine and saliva. Genome-wide genotyping on the UK Biobank Axiom Array and imputation to ~80 million variants has been performed in all participants. They have also been extensively examined, and outcome events are recorded in a longitudinal fashion.

We are working on a wide range of projects using this excellent cohort, including traditional epidemiological studies and GWAS addressing important, but understudied conditions, such as peripheral vascular disease and heart failure – including risk prediction studies to improve patient stratification, as well as studies of environmental risk factors, genetic determinants and their interactions; but also more novel approaches which aims at addressing causality of risk factors and biomarkers and importantly, at finding druggable targets using genomic approaches. The statistical power, as well as the opportunities to study new research questions, are unprecedented given the very large sample size (ten- to hundred-fold larger than all previous studies) and the extreme richness of the data.

In addition to UK Biobank, we are also working with several other datasets. I am still the PI for a range of –omics projects in several Swedish cohorts – ULSAM, PIVUS, TwinGene and EpiHealth. These include genomics, transcriptomics, epigenomics, proteomics and metabolomics, often used in combination - aiming at increasing the biological knowledge of obesity, insulin resistance and CVD, and to identify new biomarkers for risk prediction and novel drug targets.

Wet-lab approaches
To further characterize gene function after various –omics studies and use of in silico data on gene regulation and transcription from public resources, we proceed to studies of gene function in model systems. We use CRISPR-Cas9 techniques for gene editing in human SGBS adipocytes, HepG2 hepatocytes, HMCL-7304 skeletal myocytes, and murine 3T3-L1 adipocytes to study phenotypes related to obesity and insulin resistance. We transfect cells using our custom-built lentivirus CRISPR-Cas9 constructs, and assess the effect of knockdown or overexpression of candidate genes on basal and insulin-stimulated glucose uptake (using 14C-labeled deoxyglucose) and lipolysis (measuring glycerol after insulin and isoprenaline exposure), as well as insulin signaling proteins and adipogenesis. We address downstream effects of gene knockdown using transcriptomic and metabolomic profiling on cell lysates.

Teaching

2018-19 Courses


Publications

All Publications


  • Identification of metabolic profiles associated with human exposure to perfluoroalkyl substances JOURNAL OF EXPOSURE SCIENCE AND ENVIRONMENTAL EPIDEMIOLOGY Salihovic, S., Fall, T., Ganna, A., Broeckling, C. D., Prenni, J. E., Hyotylainen, T., Karrman, A., Lind, P., Ingelsson, E., Lind, L. 2019; 29 (2): 196–205

    Abstract

    Recent epidemiological studies suggest that human exposure to perfluoroalkyl substances (PFASs) may be associated with type 2 diabetes and other metabolic phenotypes. To gain further insights regarding PFASs exposure in humans, we here aimed to characterize the associations between different PFASs and the metabolome. In this cross-sectional study, we investigated 965 individuals from Sweden (all aged 70 years, 50% women) sampled in 2001-2004. PFASs were analyzed in plasma using isotope-dilution ultra-pressure liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS). Non-target metabolomics profiling was performed in plasma using UPLC coupled to time-of-flight mass spectrometry (UPLC-QTOFMS) operated in positive electrospray mode. Multivariate linear regression analysis was used to investigate associations between circulating levels of PFASs and metabolites. In total, 15 metabolites, predominantly from lipid pathways, were associated with levels of PFASs following adjustment for sex, smoking, exercise habits, education, energy, and alcohol intake, after correction for multiple testing. Perfluorononanoic acid (PFNA) and perfluoroundecanoic acid (PFUnDA) were strongly associated with multiple glycerophosphocholines and fatty acids including docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA). We also found that the different PFASs evaluated were associated with distinctive metabolic profiles, suggesting potentially different biochemical pathways in humans.

    View details for DOI 10.1038/s41370-018-0060-y

    View details for Web of Science ID 000459048700007

    View details for PubMedID 30185940

  • Substantial Cardiovascular Morbidity in Adults with Lower-Complexity Congenital Heart Disease. Circulation Saha, P., Potiny, P., Rigdon, J., Morello, M., Tcheandjieu, C., Romfh, A., Fernandes, S. M., McElhinney, D. B., Bernstein, D., Lui, G. K., Shaw, G. M., Ingelsson, E., Priest, J. R. 2019

    Abstract

    BACKGROUND: Although lower-complexity cardiac malformations constitute the majority of adult congenital heart disease (ACHD), the long-term risks of adverse cardiovascular events and relationship with conventional risk factors in this population are poorly understood. We aimed to quantify the risk of adverse cardiovascular events associated with lower-complexity ACHD that is unmeasured by conventional risk factors.METHODS: A multi-tiered classification algorithm was used to select individuals with lower-complexity ACHD and individuals without ACHD for comparison amongst >500,000 British adults in the UK Biobank (UKB). ACHD diagnoses were sub-classified as "isolated aortic valve (AoV)" and "non-complex" defects. Time-to-event analyses were conducted for primary endpoints of fatal or non-fatal acute coronary syndrome (ACS), ischemic stroke, heart failure (HF), and atrial fibrillation, and a secondary combined endpoint for major adverse cardiovascular event (MACE). Maximum follow-up time for the study period was 22 years using retrospectively and prospectively collected data from the UKB.RESULTS: We identified 2,006 individuals with lower-complexity ACHD and 497,983 unexposed individuals in the UKB (median [IQR] age at enrollment 58 [51,63]). Of the ACHD-exposed group, 59% were male; 51% were current or former smokers; 30% were obese; 69%, 41%, and 7% were diagnosed or treated for hypertension, hyperlipidemia, and diabetes respectively. After adjustment for 12 measured cardiovascular risk factors, ACHD remained strongly associated with the primary endpoints, with hazard ratios (HR) ranging from 2.0 (95% confidence interval [CI] 1.5-2.8, p<0.001) for ACS to 13.0 (95% CI 9.4-18.1, p<0.001) for HF. ACHD-exposed individuals with ≤2 cardiovascular risk factors had a 29% age-adjusted incidence rate of MACE in contrast to 13% in non-ACHD individuals with ≥5 risk factors.CONCLUSIONS: Individuals with lower-complexity ACHD had higher burden of adverse cardiovascular events relative to the general population that was unaccounted for by conventional cardiovascular risk factors. These findings highlight the need for closer surveillance of patients with mild to moderate ACHD and further investigation into management and mechanisms of cardiovascular risk unique to this growing population of high-risk adults.

    View details for DOI 10.1161/CIRCULATIONAHA.118.037064

    View details for PubMedID 30813762

  • No evidence of a causal association of type 2 diabetes and glucose metabolism with atrial fibrillation. Diabetologia Harati, H., Zanetti, D., Rao, A., Gustafsson, S., Perez, M., Ingelsson, E., Knowles, J. W. 2019

    Abstract

    AIMS/HYPOTHESIS: Several epidemiological studies have shown an increased risk of atrial fibrillation in individuals with type 2 diabetes or milder forms of dysglycaemia. We aimed to assess whether this relation is causal using a Mendelian randomisation approach.METHODS: Two-sample Mendelian randomisation was used to obtain estimates of the influence of type 2 diabetes, fasting blood glucose (FBG), and HbA1c on the risk of atrial fibrillation. Instrumental variables were constructed using available summary statistics from meta-analyses of genome-wide association studies (GWAS) for type 2 diabetes and associated phenotypes. Pleiotropic SNPs were excluded from the analyses. The most recent GWAS meta-analysis summary statistics for atrial fibrillation, which included over 1 million individuals (approximately 60,000 individuals with atrial fibrillation) was used for outcome analysis.RESULTS: Neither type 2 diabetes (OR 1.01 [95% CI 0.98, 1.03]; p=0.37), nor FBG (OR 0.95 [95% CI 0.82, 1.09] per mmol/l; p=0.49) or HbA1c (OR 1.01 [95% CI, 0.85, 1.17] per mmol/mol [%]; p=0.88) were associated with atrial fibrillation in Mendelian randomisation analyses. We had >80% statistical power to detect ORs of 1.08, 1.06 and 1.09 or larger for type 2 diabetes, FBG and HbA1c, respectively, for associations with atrial fibrillation.CONCLUSIONS/INTERPRETATION: This Mendelian randomisation analysis does not support a causal role of clinical significance between genetically programmed type 2 diabetes, FBG or HbA1c and development of atrial fibrillation. These data suggest that drug treatment to reduce dysglycaemia is unlikely to be an effective strategy for atrial fibrillation prevention.DATA AVAILABILITY: The datasets analysed during the current study are available from the following repository: Nielsen JB, Thorolfsdottir RB, Fritsche LG, et al (2018) GWAS summary statistics for AF (N=60,620 AF cases and 970,216 controls). Center for Statistical Genetics: http://csg.sph.umich.edu/willer/public/afib2018/nielsen-thorolfsdottir-willer-NG2018-AFib-gwas-summary-statistics.tbl.gz.

    View details for DOI 10.1007/s00125-019-4836-y

    View details for PubMedID 30810766

  • Protein-coding variants implicate novel genes related to lipid homeostasis contributing to body-fat distribution. Nature genetics Justice, A. E., Karaderi, T., Highland, H. M., Young, K. L., Graff, M., Lu, Y., Turcot, V., Auer, P. L., Fine, R. S., Guo, X., Schurmann, C., Lempradl, A., Marouli, E., Mahajan, A., Winkler, T. W., Locke, A. E., Medina-Gomez, C., Esko, T., Vedantam, S., Giri, A., Lo, K. S., Alfred, T., Mudgal, P., Ng, M. C., Heard-Costa, N. L., Feitosa, M. F., Manning, A. K., Willems, S. M., Sivapalaratnam, S., Abecasis, G., Alam, D. S., Allison, M., Amouyel, P., Arzumanyan, Z., Balkau, B., Bastarache, L., Bergmann, S., Bielak, L. F., Bluher, M., Boehnke, M., Boeing, H., Boerwinkle, E., Boger, C. A., Bork-Jensen, J., Bottinger, E. P., Bowden, D. W., Brandslund, I., Broer, L., Burt, A. A., Butterworth, A. S., Caulfield, M. J., Cesana, G., Chambers, J. C., Chasman, D. I., Chen, Y. I., Chowdhury, R., Christensen, C., Chu, A. Y., Collins, F. S., Cook, J. P., Cox, A. J., Crosslin, D. S., Danesh, J., de Bakker, P. I., Denus, S. d., Mutsert, R. d., Dedoussis, G., Demerath, E. W., Dennis, J. G., Denny, J. C., Angelantonio, E. D., Dorr, M., Drenos, F., Dube, M., Dunning, A. M., Easton, D. F., Elliott, P., Evangelou, E., Farmaki, A., Feng, S., Ferrannini, E., Ferrieres, J., Florez, J. C., Fornage, M., Fox, C. S., Franks, P. W., Friedrich, N., Gan, W., Gandin, I., Gasparini, P., Giedraitis, V., Girotto, G., Gorski, M., Grallert, H., Grarup, N., Grove, M. L., Gustafsson, S., Haessler, J., Hansen, T., Hattersley, A. T., Hayward, C., Heid, I. M., Holmen, O. L., Hovingh, G. K., Howson, J. M., Hu, Y., Hung, Y., Hveem, K., Ikram, M. A., Ingelsson, E., Jackson, A. U., Jarvik, G. P., Jia, Y., Jorgensen, T., Jousilahti, P., Justesen, J. M., Kahali, B., Karaleftheri, M., Kardia, S. L., Karpe, F., Kee, F., Kitajima, H., Komulainen, P., Kooner, J. S., Kovacs, P., Kramer, B. K., Kuulasmaa, K., Kuusisto, J., Laakso, M., Lakka, T. A., Lamparter, D., Lange, L. A., Langenberg, C., Larson, E. B., Lee, N. R., Lee, W., Lehtimaki, T., Lewis, C. E., Li, H., Li, J., Li-Gao, R., Lin, L., Lin, X., Lind, L., Lindstrom, J., Linneberg, A., Liu, C., Liu, D. J., Luan, J., Lyytikainen, L., MacGregor, S., Magi, R., Mannisto, S., Marenne, G., Marten, J., Masca, N. G., McCarthy, M. I., Meidtner, K., Mihailov, E., Moilanen, L., Moitry, M., Mook-Kanamori, D. O., Morgan, A., Morris, A. P., Muller-Nurasyid, M., Munroe, P. B., Narisu, N., Nelson, C. P., Neville, M., Ntalla, I., O'Connell, J. R., Owen, K. R., Pedersen, O., Peloso, G. M., Pennell, C. E., Perola, M., Perry, J. A., Perry, J. R., Pers, T. H., Ewing, A., Polasek, O., Raitakari, O. T., Rasheed, A., Raulerson, C. K., Rauramaa, R., Reilly, D. F., Reiner, A. P., Ridker, P. M., Rivas, M. A., Robertson, N. R., Robino, A., Rudan, I., Ruth, K. S., Saleheen, D., Salomaa, V., Samani, N. J., Schreiner, P. J., Schulze, M. B., Scott, R. A., Segura-Lepe, M., Sim, X., Slater, A. J., Small, K. S., Smith, B. H., Smith, J. A., Southam, L., Spector, T. D., Speliotes, E. K., Stefansson, K., Steinthorsdottir, V., Stirrups, K. E., Strauch, K., Stringham, H. M., Stumvoll, M., Sun, L., Surendran, P., Swart, K. M., Tardif, J., Taylor, K. D., Teumer, A., Thompson, D. J., Thorleifsson, G., Thorsteinsdottir, U., Thuesen, B. H., Tonjes, A., Torres, M., Tsafantakis, E., Tuomilehto, J., Uitterlinden, A. G., Uusitupa, M., van Duijn, C. M., Vanhala, M., Varma, R., Vermeulen, S. H., Vestergaard, H., Vitart, V., Vogt, T. F., Vuckovic, D., Wagenknecht, L. E., Walker, M., Wallentin, L., Wang, F., Wang, C. A., Wang, S., Wareham, N. J., Warren, H. R., Waterworth, D. M., Wessel, J., White, H. D., Willer, C. J., Wilson, J. G., Wood, A. R., Wu, Y., Yaghootkar, H., Yao, J., Yerges-Armstrong, L. M., Young, R., Zeggini, E., Zhan, X., Zhang, W., Zhao, J. H., Zhao, W., Zheng, H., Zhou, W., Zillikens, M. C., CHD Exome+ Consortium, Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium, EPIC-CVD Consortium, ExomeBP Consortium, Global Lipids Genetic Consortium, GoT2D Genes Consortium, InterAct, ReproGen Consortium, T2D-Genes Consortium, MAGIC Investigators, Rivadeneira, F., Borecki, I. B., Pospisilik, J. A., Deloukas, P., Frayling, T. M., Lettre, G., Mohlke, K. L., Rotter, J. I., Kutalik, Z., Hirschhorn, J. N., Cupples, L. A., Loos, R. J., North, K. E., Lindgren, C. M. 2019

    Abstract

    Body-fat distribution is a risk factor for adverse cardiovascular health consequences. We analyzed the association of body-fat distribution, assessed by waist-to-hip ratio adjusted for body mass index, with 228,985 predicted coding and splice site variants available on exome arrays in up to 344,369 individuals from five major ancestries (discovery) and 132,177 European-ancestry individuals (validation). We identified 15 common (minor allele frequency, MAF ≥5%) and nine low-frequency or rare (MAF <5%) coding novel variants. Pathway/gene set enrichment analyses identified lipid particle, adiponectin, abnormal white adipose tissue physiology and bone development and morphology as important contributors to fat distribution, while cross-trait associations highlight cardiometabolic traits. In functional follow-up analyses, specifically in Drosophila RNAi-knockdowns, we observed a significant increase in the total body triglyceride levels for two genes (DNAH10 and PLXND1). We implicate novel genes in fat distribution, stressing the importance of interrogating low-frequency and protein-coding variants.

    View details for DOI 10.1038/s41588-018-0334-2

    View details for PubMedID 30778226

  • Body composition and atrial fibrillation: a Mendelian randomization study. European heart journal Tikkanen, E., Gustafsson, S., Knowles, J. W., Perez, M., Burgess, S., Ingelsson, E. 2019

    Abstract

    Aims: Increases in fat-free mass and fat mass have been associated with higher risk of atrial fibrillation (AF) in observational studies. It is not known whether these associations reflect independent causal processes. Our aim was to evaluate independent causal roles of fat-free mass and fat mass on AF.Methods and results: We conducted a large observational study to estimate the associations between fat-free mass and fat mass on incident AF in the UK Biobank (N=487404, N events=10365). Genome-wide association analysis was performed to obtain genetic instruments for Mendelian randomization (MR). We evaluated the causal effects of fat-free mass and fat mass on AF with two-sample method by using genetic associations from AFGen consortium as outcome. Finally, we evaluated independent causal effects of fat-free mass and fat mass with multivariate MR. Both fat-free mass and fat mass had observational associations with incident AF [hazard ratio (HR)=1.77, 95% confidence interval (CI) 1.72-1.83; HR=1.40, 95% CI 1.37-1.43 per standard deviation increase in fat-free and fat mass, respectively]. The causal effects using the inverse-variance weighted method were 1.55 (95% CI 1.38-1.75) for fat-free mass and 1.30 (95% CI 1.17-1.45) for fat mass. Weighted median, Egger regression, and penalized methods showed similar estimates. The multivariate MR analysis suggested that the causal effects of fat-free and fat mass were independent of each other (causal risk ratios: 1.37, 95% CI 1.06-1.75; 1.28, 95% CI 1.03-1.58).Conclusion: Genetically programmed increases in fat-free mass and fat mass independently cause an increased risk of AF.

    View details for DOI 10.1093/eurheartj/ehz003

    View details for PubMedID 30721963

  • Disentangling the genetics of lean mass. The American journal of clinical nutrition Karasik, D., Zillikens, M. C., Hsu, Y., Aghdassi, A., Akesson, K., Amin, N., Barroso, I., Bennett, D. A., Bertram, L., Bochud, M., Borecki, I. B., Broer, L., Buchman, A. S., Byberg, L., Campbell, H., Campos-Obando, N., Cauley, J. A., Cawthon, P. M., Chambers, J. C., Chen, Z., Cho, N. H., Choi, H. J., Chou, W., Cummings, S. R., de Groot, L. C., De Jager, P. L., Demuth, I., Diatchenko, L., Econs, M. J., Eiriksdottir, G., Enneman, A. W., Eriksson, J., Eriksson, J. G., Estrada, K., Evans, D. S., Feitosa, M. F., Fu, M., Gieger, C., Grallert, H., Gudnason, V., Lenore, L. J., Hayward, C., Hofman, A., Homuth, G., Huffman, K. M., Husted, L. B., Illig, T., Ingelsson, E., Ittermann, T., Jansson, J., Johnson, T., Biffar, R., Jordan, J. M., Jula, A., Karlsson, M., Khaw, K., Kilpelainen, T. O., Klopp, N., Kloth, J. S., Koller, D. L., Kooner, J. S., Kraus, W. E., Kritchevsky, S., Kutalik, Z., Kuulasmaa, T., Kuusisto, J., Laakso, M., Lahti, J., Lang, T., Langdahl, B. L., Lerch, M. M., Lewis, J. R., Lill, C., Lind, L., Lindgren, C., Liu, Y., Livshits, G., Ljunggren, O., Loos, R. J., Lorentzon, M., Luan, J., Luben, R. N., Malkin, I., McGuigan, F. E., Medina-Gomez, C., Meitinger, T., Melhus, H., Mellstrom, D., Michaelsson, K., Mitchell, B. D., Morris, A. P., Mosekilde, L., Nethander, M., Newman, A. B., O'Connell, J. R., Oostra, B. A., Orwoll, E. S., Palotie, A., Peacock, M., Perola, M., Peters, A., Prince, R. L., Psaty, B. M., Raikkonen, K., Ralston, S. H., Ripatti, S., Rivadeneira, F., Robbins, J. A., Rotter, J. I., Rudan, I., Salomaa, V., Satterfield, S., Schipf, S., Shin, C. S., Smith, A. V., Smith, S. B., Soranzo, N., Spector, T. D., Stancakova, A., Stefansson, K., Steinhagen-Thiessen, E., Stolk, L., Streeten, E. A., Styrkarsdottir, U., Swart, K. M., Thompson, P., Thomson, C. A., Thorleifsson, G., Thorsteinsdottir, U., Tikkanen, E., Tranah, G. J., Uitterlinden, A. G., van Duijn, C. M., van Schoor, N. M., Vandenput, L., Vollenweider, P., Volzke, H., Wactawski-Wende, J., Walker, M., J Wareham, N., Waterworth, D., Weedon, M. N., Wichmann, H., Widen, E., Williams, F. M., Wilson, J. F., Wright, N. C., Yerges-Armstrong, L. M., Yu, L., Zhang, W., Zhao, J. H., Zhou, Y., Nielson, C. M., Harris, T. B., Demissie, S., Kiel, D. P., Ohlsson, C. 2019

    Abstract

    Background: Lean body mass (LM) plays an important role in mobility and metabolic function. We previously identified five loci associated with LM adjusted for fat mass in kilograms. Such an adjustment may reduce the power to identify genetic signals having an association with both lean mass and fat mass.Objectives: To determine the impact of different fat mass adjustments on genetic architecture of LM and identify additional LM loci.Methods: We performed genome-wide association analyses for whole-body LM (20 cohorts of European ancestry with n=38,292) measured using dual-energy X-ray absorptiometry) or bioelectrical impedance analysis, adjusted for sex, age, age2, and height with or without fat mass adjustments (Model 1 no fat adjustment; Model 2 adjustment for fat mass as a percentage of body mass; Model 3 adjustment for fat mass in kilograms).Results: Seven single-nucleotide polymorphisms (SNPs) in separate loci, including one novel LM locus (TNRC6B), were successfully replicated in an additional 47,227 individuals from 29 cohorts. Based on the strengths of the associations in Model 1 vs Model 3, we divided the LM loci into those with an effect on both lean mass and fat mass in the same direction and refer to those as "sumo wrestler" loci (FTO and MC4R). In contrast, loci with an impact specifically on LM were termed "body builder" loci (VCAN and ADAMTSL3). Using existing available genome-wide association study databases, LM increasing alleles of SNPs in sumo wrestler loci were associated with an adverse metabolic profile, whereas LM increasing alleles of SNPs in "body builder" loci were associated with metabolic protection.Conclusions: In conclusion, we identified one novel LM locus (TNRC6B). Our results suggest that a genetically determined increase in lean mass might exert either harmful or protective effects on metabolic traits, depending on its relation to fat mass.

    View details for DOI 10.1093/ajcn/nqy272

    View details for PubMedID 30721968

  • Common Genetic Variation in Relation to Brachial Vascular Dimensions and Flow-Mediated Vasodilation. Circulation. Genomic and precision medicine Dorr, M., Hamburg, N. M., Muller, C., Smith, N. L., Gustafsson, S., Lehtimaki, T., Teumer, A., Zeller, T., Li, X., Lind, L., Raitakari, O. T., Volker, U., Blankenberg, S., McKnight, B., Morris, A. P., Kahonen, M., Lemaitre, R. N., Wild, P. S., Nauck, M., Volzke, H., Munzel, T., Mitchell, G. F., Psaty, B. M., Lindgren, C. M., Larson, M. G., Felix, S. B., Ingelsson, E., Lyytikainen, L., Herrington, D., Benjamin, E. J., Schnabel, R. B. 2019; 12 (2): e002409

    View details for DOI 10.1161/CIRCGEN.118.002409

    View details for PubMedID 30779634

  • Trans-ethnic kidney function association study reveals putative causal genes and effects on kidney-specific disease aetiologies. Nature communications Morris, A. P., Le, T. H., Wu, H., Akbarov, A., van der Most, P. J., Hemani, G., Smith, G. D., Mahajan, A., Gaulton, K. J., Nadkarni, G. N., Valladares-Salgado, A., Wacher-Rodarte, N., Mychaleckyj, J. C., Dueker, N. D., Guo, X., Hai, Y., Haessler, J., Kamatani, Y., Stilp, A. M., Zhu, G., Cook, J. P., Arnlov, J., Blanton, S. H., de Borst, M. H., Bottinger, E. P., Buchanan, T. A., Cechova, S., Charchar, F. J., Chu, P., Damman, J., Eales, J., Gharavi, A. G., Giedraitis, V., Heath, A. C., Ipp, E., Kiryluk, K., Kramer, H. J., Kubo, M., Larsson, A., Lindgren, C. M., Lu, Y., Madden, P. A., Montgomery, G. W., Papanicolaou, G. J., Raffel, L. J., Sacco, R. L., Sanchez, E., Stark, H., Sundstrom, J., Taylor, K. D., Xiang, A. H., Zivkovic, A., Lind, L., Ingelsson, E., Martin, N. G., Whitfield, J. B., Cai, J., Laurie, C. C., Okada, Y., Matsuda, K., Kooperberg, C., Chen, Y. I., Rundek, T., Rich, S. S., Loos, R. J., Parra, E. J., Cruz, M., Rotter, J. I., Snieder, H., Tomaszewski, M., Humphreys, B. D., Franceschini, N. 2019; 10 (1): 29

    Abstract

    Chronic kidney disease (CKD) affects ~10% of the global population, with considerable ethnic differences in prevalence and aetiology. We assemble genome-wide association studies of estimated glomerular filtration rate (eGFR), a measure of kidney function that defines CKD, in 312,468 individuals of diverse ancestry. We identify 127 distinct association signals with homogeneous effects on eGFR across ancestries and enrichment in genomic annotations including kidney-specific histone modifications. Fine-mapping reveals 40 high-confidence variants driving eGFR associations and highlights putative causal genes with cell-type specific expression in glomerulus, and inproximal and distal nephron. Mendelian randomisation supports causal effects of eGFR on overall and cause-specific CKD, kidney stone formation, diastolic blood pressure and hypertension. These results define novel molecular mechanisms and putative causal genes for eGFR, offering insight into clinical outcomes and routes to CKD treatment development.

    View details for DOI 10.1038/s41467-018-07867-7

    View details for PubMedID 30604766

  • Longitudinal effects of aging on plasma proteins levels in older adults - associations with kidney function and hemoglobin levels. PloS one Lind, L., Sundstrom, J., Larsson, A., Lampa, E., Arnlov, J., Ingelsson, E. 2019; 14 (2): e0212060

    Abstract

    BACKGROUND: A targeted proteomics chip has been shown to be useful to discover novel associations of proteins with cardiovascular disease. We investigated how these proteins change with aging, and whether this change is related to a decline in kidney function, or to a change in hemoglobin levels.MATERIAL AND METHODS: In the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study, including 1,016 participants from the general population aged 70 at baseline, 84 proteins were measured at ages 70, 75, 80. At these occasions, glomerular filtration rate (eGFR) was estimated and the hemoglobin levels were measured.RESULTS: Sixty-one of the 84 evaluated proteins changed significantly during the 10-year follow-up (multiple testing-adjusted alpha = 0.00059), most showing an increase. The change in eGFR was inversely related to changes of protein levels for the vast majority of proteins (74%). The change in hemoglobin was significantly related to the change in 40% of the evaluated proteins, with no obvious preference of the direction of these relationships.CONCLUSION: The majority of evaluated proteins increased with aging in adults. Therefore, normal ranges for proteins might be given in age-strata. The increase in protein levels was associated with the degree of reduction in eGFR for the majority of proteins, while no clear pattern was seen for the relationships between the proteins and the change in hemoglobin levels. Studies on changes in urinary proteins are warranted to understand the association between the reduction in eGFR and increase in plasma protein levels.

    View details for DOI 10.1371/journal.pone.0212060

    View details for PubMedID 30802263

  • Clinical and Genetic Determinants of Varicose Veins. Circulation Fukaya, E., Flores, A. M., Lindholm, D., Gustafsson, S., Zanetti, D., Ingelsson, E., Leeper, N. J. 2018; 138 (25): 2869–80

    Abstract

    BACKGROUND: Varicose veins are a common problem with no approved medical therapies. Although it is believed that varicose vein pathogenesis is multifactorial, there is limited understanding of the genetic and environmental factors that contribute to their formation. Large-scale studies of risk factors for varicose veins may highlight important aspects of pathophysiology and identify groups at increased risk for disease.METHODS: We applied machine learning to agnostically search for risk factors of varicose veins in 493519 individuals in the UK Biobank. Predictors were further studied with univariable and multivariable Cox regression analyses (2441 incident events). A genome-wide association study of varicose veins was also performed among 337536 unrelated individuals (9577 cases) of white British descent, followed by expression quantitative loci and pathway analyses. Because height emerged as a new candidate risk factor, we performed mendelian randomization analyses to assess a potential causal role for height in varicose vein development.RESULTS: Machine learning confirmed several known (age, sex, obesity, pregnancy, history of deep vein thrombosis) and identified several new risk factors for varicose vein disease, including height. After adjustment for traditional risk factors in Cox regression, greater height remained independently associated with varicose veins (hazard ratio for upper versus lower quartile, 1.74; 95% CI, 1.51-2.01; P<0.0001). A genome-wide association study identified 30 new genome-wide significant loci, identifying pathways involved in vascular development and skeletal/limb biology. Mendelian randomization analysis provided evidence that increased height is causally related to varicose veins (inverse-variance weighted: odds ratio, 1.26; P=2.07*10-16).CONCLUSIONS: Using data from nearly a half-million individuals, we present a comprehensive genetic and epidemiological study of varicose veins. We identified novel clinical and genetic risk factors that provide pathophysiological insights and could help future improvements of treatment of varicose vein disease.

    View details for DOI 10.1161/CIRCULATIONAHA.118.035584

    View details for PubMedID 30566020

  • Loss of function, missense, and intronic variants in NOTCH1 confer different risks for left ventricular outflow tract obstructive heart defects in two European cohorts. Genetic epidemiology Helle, E., Cordova-Palomera, A., Ojala, T., Saha, P., Potiny, P., Gustafsson, S., Ingelsson, E., Bamshad, M., Nickerson, D., Chong, J. X., University of Washington Center for Mendelian Genomics, Ashley, E., Priest, J. R. 2018

    Abstract

    Loss of function variants in NOTCH1 cause left ventricular outflow tract obstructive defects (LVOTO). However, the risk conferred by rare and noncoding variants in NOTCH1 for LVOTO remains largely uncharacterized. In a cohort of 49 families affected by hypoplastic left heart syndrome, a severe form of LVOTO, we discovered predicted loss of function NOTCH1 variants in 6% of individuals. Rare or low-frequency missense variants were found in 16% of families. To make a quantitative estimate of the genetic risk posed by variants in NOTCH1 for LVOTO, we studied associations of 400 coding and noncoding variants in NOTCH1 in 1,085 cases and 332,788 controls from the UK Biobank. Two rare intronic variants in strong linkage disequilibrium displayed significant association with risk for LVOTO amongst European-ancestry individuals. This result was replicated in an independent analysis of 210 cases and 68,762 controls of non-European and mixed ancestry. In conclusion, carrying rare predicted loss of function variants in NOTCH1 confer significant risk for LVOTO. In addition, the two intronic variants seem to be associated with an increased risk for these defects. Our approach demonstrates the utility of population-based data sets in quantifying the specific risk of individual variants for disease-related phenotypes.

    View details for DOI 10.1002/gepi.22176

    View details for PubMedID 30511478

  • GWAS and colocalization analyses implicate carotid intima-media thickness and carotid plaque loci in cardiovascular outcomes. Nature communications Franceschini, N., Giambartolomei, C., de Vries, P. S., Finan, C., Bis, J. C., Huntley, R. P., Lovering, R. C., Tajuddin, S. M., Winkler, T. W., Graff, M., Kavousi, M., Dale, C., Smith, A. V., Hofer, E., van Leeuwen, E. M., Nolte, I. M., Lu, L., Scholz, M., Sargurupremraj, M., Pitkanen, N., Franzen, O., Joshi, P. K., Noordam, R., Marioni, R. E., Hwang, S., Musani, S. K., Schminke, U., Palmas, W., Isaacs, A., Correa, A., Zonderman, A. B., Hofman, A., Teumer, A., Cox, A. J., Uitterlinden, A. G., Wong, A., Smit, A. J., Newman, A. B., Britton, A., Ruusalepp, A., Sennblad, B., Hedblad, B., Pasaniuc, B., Penninx, B. W., Langefeld, C. D., Wassel, C. L., Tzourio, C., Fava, C., Baldassarre, D., O'Leary, D. H., Teupser, D., Kuh, D., Tremoli, E., Mannarino, E., Grossi, E., Boerwinkle, E., Schadt, E. E., Ingelsson, E., Veglia, F., Rivadeneira, F., Beutner, F., Chauhan, G., Heiss, G., Snieder, H., Campbell, H., Volzke, H., Markus, H. S., Deary, I. J., Jukema, J. W., de Graaf, J., Price, J., Pott, J., Hopewell, J. C., Liang, J., Thiery, J., Engmann, J., Gertow, K., Rice, K., Taylor, K. D., Dhana, K., Kiemeney, L. A., Lind, L., Raffield, L. M., Launer, L. J., Holdt, L. M., Dorr, M., Dichgans, M., Traylor, M., Sitzer, M., Kumari, M., Kivimaki, M., Nalls, M. A., Melander, O., Raitakari, O., Franco, O. H., Rueda-Ochoa, O. L., Roussos, P., Whincup, P. H., Amouyel, P., Giral, P., Anugu, P., Wong, Q., Malik, R., Rauramaa, R., Burkhardt, R., Hardy, R., Schmidt, R., de Mutsert, R., Morris, R. W., Strawbridge, R. J., Wannamethee, S. G., Hagg, S., Shah, S., McLachlan, S., Trompet, S., Seshadri, S., Kurl, S., Heckbert, S. R., Ring, S., Harris, T. B., Lehtimaki, T., Galesloot, T. E., Shah, T., de Faire, U., Plagnol, V., Rosamond, W. D., Post, W., Zhu, X., Zhang, X., Guo, X., Saba, Y., MEGASTROKE Consortium, Dehghan, A., Seldenrijk, A., Morrison, A. C., Hamsten, A., Psaty, B. M., van Duijn, C. M., Lawlor, D. A., Mook-Kanamori, D. O., Bowden, D. W., Schmidt, H., Wilson, J. F., Wilson, J. G., Rotter, J. I., Wardlaw, J. M., Deanfield, J., Halcox, J., Lyytikainen, L., Loeffler, M., Evans, M. K., Debette, S., Humphries, S. E., Volker, U., Gudnason, V., Hingorani, A. D., Bjorkegren, J. L., Casas, J. P., O'Donnell, C. J., Okada, Y., Mishra, A., Rutten-Jacobs, L., Giese, A., van der Laan, S. W., Gretarsdottir, S., Anderson, C. D., Chong, M., Adams, H. H., Ago, T., Almgren, P., Amouyel, P., Ay, H., Bartz, T. M., Benavente, O. R., Bevan, S., Boncoraglio, G. B., Brown, R. D., Butterworth, A. S., Carrera, C., Carty, C. L., Chasman, D. I., Chen, W., Cole, J. W., Cotlarciuc, I., Cruchaga, C., Danesh, J., de Bakker, P. I., DeStefano, A. L., den Hoed, M., Duan, Q., Engelter, S. T., Falcone, G. J., Gottesman, R. F., Grewal, R. P., Gustafsson, S., Haessler, J., Harris, T. B., Hassan, A., Havulinna, A. S., Holliday, E. G., Howard, G., Hsu, F., Hyacinth, H. I., Ikram, M. A., Irvin, M. R., Jian, X., Jimenez-Conde, J., Johnson, J. A., Jukema, J. W., Kanai, M., Keene, K. L., Kissela, B. M., Kleindorfer, D. O., Kooperberg, C., Kubo, M., Lange, L., Langefeld, C. D., Langenberg, C., Lee, J., Lemmens, R., Leys, D., Lewis, C. M., Lin, W., Lindgren, A. G., Lorentzen, E., Magnusson, P. K., Maguire, J., Manichaikul, A., McArdle, P. F., Meschia, J. F., Mosley, T. H., Ninomiya, T., O'Donnell, M. J., Pulit, S. L., Rannikmae, K., Reiner, A. P., Rexrode, K. M., Rich, S. S., Ridker, P. M., Rost, N. S., Rothwell, P. M., Rundek, T., Sacco, R. L., Sakaue, S., Sale, M. M., Salomaa, V., Sapkota, B. R., Schmidt, R., Schmidt, C. O., Schminke, U., Sharma, P., Slowik, A., Sudlow, C. L., Tanislav, C., Tatlisumak, T., Thijs, V. N., Thorleifsson, G., Thorsteinsdottir, U., Tiedt, S., Trompet, S., Walters, M., Wareham, N. J., Wassertheil-Smoller, S., Wiggins, K. L., Yang, Q., Yusuf, S., Pastinen, T., Ruusalepp, A., Schadt, E. E., Koplev, S., Codoni, V., Civelek, M., Smith, N., Tregouet, D. A., Christophersen, I. E., Roselli, C., Lubitz, S. A., Ellinor, P. T., Tai, E. S., Kooner, J. S., Kato, N., He, J., van der Harst, P., Elliott, P., Chambers, J. C., Takeuchi, F., Johnson, A. D., Sanghera, D. K., Melander, O., Jern, C., Strbian, D., Fernandez-Cadenas, I., Longstreth, W. T., Rolfs, A., Hata, J., Woo, D., Rosand, J., Pare, G., Saleheen, D., Stefansson, K., Worrall, B. B., Kittner, S. J., Howson, J. M., Kamatani, Y. 2018; 9 (1): 5141

    Abstract

    Carotid artery intima media thickness (cIMT) and carotid plaque are measures of subclinical atherosclerosis associated with ischemic stroke and coronary heart disease (CHD). Here, we undertake meta-analyses of genome-wide association studies (GWAS) in 71,128 individuals for cIMT, and 48,434 individuals for carotid plaque traits. We identify eight novel susceptibility loci for cIMT, one independent association at the previously-identified PINX1 locus, and one novel locus for carotid plaque. Colocalization analysis with nearby vascular expression quantitative loci (cis-eQTLs) derived from arterial wall and metabolic tissues obtained from patients with CHD identifies candidate genes at two potentially additional loci, ADAMTS9 and LOXL4. LD score regression reveals significant genetic correlations between cIMT and plaque traits, and both cIMT and plaque with CHD, any stroke subtype and ischemic stroke. Our study provides insights into genes and tissue-specific regulatory mechanisms linking atherosclerosis both to its functional genomic origins and its clinical consequences in humans.

    View details for DOI 10.1038/s41467-018-07340-5

    View details for PubMedID 30510157

  • Atrial fibrillation genetic risk differentiates cardioembolic stroke from other stroke subtypes NEUROLOGY-GENETICS Pulit, S. L., Weng, L., McArdle, P. F., Trinquart, L., Choi, S., Mitchell, B. D., Rosand, J., de Bakker, P. W., Benjamin, E. J., Ellinor, P. T., Kittner, S. J., Lubitz, S. A., Anderson, C. D., Christophersen, I. E., Rienstra, M., Roselli, C., Yin, X., Geelhoed, B., Barnard, J., Lin, H., Arking, D. E., Smith, A., Albert, C. M., Chaffin, M., Tucker, N. R., Li, M., Klarin, D., Bihlmeyer, N. A., Low, S., Weeke, P. E., Mueller-Nurasyid, M., Smith, J., Brody, J. A., Niemeijer, M. N., Doerr, M., Trompet, S., Huffman, J., Gustafsson, S., Schurmann, C., Kleber, M. E., Lyytikainen, L., Seppala, I., Malik, R., Horimoto, A. R., Perez, M., Sinisalo, J., Aeschbacher, S., Theriault, S., Yao, J., Radmanesh, F., Weiss, S., Teumer, A., Clauss, S., Deo, R., Rader, D. J., Shah, S., Sun, A., Hopewell, J. C., Debette, S., Chauhan, G., Yang, Q., Worrall, B. B., Pare, G., Kamatani, Y., Hagemeijer, Y. P., Verweij, N., Siland, J. E., Kubo, M., Smith, J. D., Van Wagoner, D. R., Bis, J. C., Perz, S., Psaty, B. M., Ridker, P. M., Magnani, J. W., Harris, T. B., Launer, L. J., Shoemaker, M., Padmanabhan, S., Haessler, J., Bartz, T. M., Waldenberger, M., Lichtner, P., Arendt, M., Krieger, J. E., Kahonen, M., Risch, L., Mansur, A. J., Peters, A., Smith, B. H., Lind, L., Scott, S. A., Lu, Y., Bottinger, E. B., Hernesniemi, J., Lindgren, C. M., Wong, J. A., Huang, J., Eskola, M., Morris, A. P., Ford, I., Reiner, A. P., Delgado, G., Chen, L. Y., Chen, Y., Sandhu, R. K., Li, M., Boerwinkle, E., Eisele, L., Lannfelt, L., Rost, N., Taylor, K. D., Campbell, A., Magnusson, P. K., Porteous, D., Hocking, L. J., Vlachopoulou, E., Pedersen, N. L., Nikus, K., Orho-Melander, M., Hamsten, A., Heeringa, J., Denny, J. C., Kriebel, J., Darbar, D., Newton-Cheh, C., Shaffer, C., Macfarlane, P. W., Heilmann, S., Almgren, P., Huang, P. L., Sotoodehnia, N., Soliman, E. Z., Uitterlinden, A. G., Hofman, A., Franco, O. H., Voelker, U., Joeckel, K., Sinner, M. F., Lin, H. J., Guo, X., Dichgans, M., Ingelsson, E., Kooperberg, C., Melander, O., Loos, R. F., Laurikka, J., Conen, D., van der Harst, P., Lokki, M., Kathiresan, S., Pereira, A., Jukema, J., Hayward, C., Rotter, J., Maerz, W., Lehtimaki, T., Stricker, B. H., Chung, M. K., Felix, S. B., Gudnason, V., Alonso, A., Roden, D. M., Kaeaeb, S., Chasman, D., Heckbert, S. R., Tanaka, T., Lunetta, K. L., Smoller, S., Sorkin, J., Wang, X., Selim, M., Pikula, A., Wolf, P., Seshadri, S., de Bakker, P., Chasman, D., Rexrode, K., Chen, I., Rotter, J., Luke, M., Sale, M., Lee, T., Chang, K., Elkind, M., Goldstein, L., James, M., Breteler, M., O'Donnell, C., Leys, D., Carty, C., Kidwell, C., Olesen, J., Sharma, P., Rich, S., Tatlisumak, T., Happola, O., Bijlenga, P., Soriano, C., Giralt, E., Roquer, J., Jimenez-Conde, J., Cotlarcius, I., Hardy, J., Korostynski, M., Boncoraglio, G., Ballabio, E., Parati, E., Mateusz, A., Urbanik, A., Dziedzic, T., Jagiella, J., Gasowski, J., Wnuk, M., Olszanecki, R., Pera, J., Slowik, A., Juchniewicz, K., Levi, C., Nyquist, P., Cendes, I., Cabral, N., Franca, P., Goncalves, A., Keller, L., Crisby, M., Kostulas, K., Lemmens, R., Ahmadi, K., Opherk, C., Duering, M., Gonik, M., Staals, J., Burri, P., Sadr-Nabavi, A., Romero, J., Biffi, A., Anderson, C., Falcone, G., Brouwers, B., Du, R., Kourkoulis, C., Battey, T., Lubitz, S., Mueller-Myhsok, B., Meschia, J., Brott, T., Pichler, A., Enzinger, C., Schmidt, H., Schmidt, R., Seiler, S., Blanton, S., Yamada, Y., Bersano, A., Rundek, T., Sacco, R., Chan, Y., Gschwendtner, A., Deng, Z., Barr, T., Gwinn, K., Corriveau, R., Singleton, A., Waddy, S., Launer, L., Chen, C., Le, K., Lee, W., Tan, E., Olugbodi, A., Rothwell, P., Schilling, S., Mok, V., Lebedeva, E., Jern, C., Jood, K., Olsson, S., Kim, H., Lee, C., Kilarski, L., Markus, H., Peycke, J., Bevan, S., Sheu, W., Chiou, H., Chern, J., Giraldo, E., Taqi, M., Jain, V., Lam, O., Howard, G., Woo, D., Kittner, S., Mitchell, B., Cole, J., O'Connell, J., Milewicz, D., Illoh, K., Worrall, B., Stine, C., Karaszewski, B., Werring, D., Sofat, R., Smalley, J., Lindgren, A., Hansen, B., Norrving, B., Smith, G., Jose Martin, J., Thijs, V., Klijn, K., van't Hof, F., Algra, A., Macleod, M., Perry, R., Arnett, D., Pezzini, A., Padovani, A., Cramer, S., Fisher, M., Saleheen, D., Broderick, J., Kissela, B., Doney, A., Sudlow, C., Rannikmae, K., Silliman, S., McDonough, C., Walters, M., Pedersen, A., Nakagawa, K., Chang, C., Dobbins, M., McArdle, P., Chang, Y., Brown, R., Brown, D., Holliday, E., Kalaria, R., Maguire, J., Attia, J., Farrall, M., Giese, A., Fornage, M., Majersik, J., Cushman, M., Keene, K., Bennett, S., Tirschwell, D., Psaty, B., Reiner, A., Longstreth, W., Spence, D., Montaner, J., Fernandez-Cadenas, I., Langefeld, C., Bushnell, C., Heitsch, L., Lee, J., Sheth, K., Atrial Fibrillation Genetics Conso, Int Stroke Genetics Consortium 2018; 4 (6): e293

    Abstract

    We sought to assess whether genetic risk factors for atrial fibrillation (AF) can explain cardioembolic stroke risk.We evaluated genetic correlations between a previous genetic study of AF and AF in the presence of cardioembolic stroke using genome-wide genotypes from the Stroke Genetics Network (N = 3,190 AF cases, 3,000 cardioembolic stroke cases, and 28,026 referents). We tested whether a previously validated AF polygenic risk score (PRS) associated with cardioembolic and other stroke subtypes after accounting for AF clinical risk factors.We observed a strong correlation between previously reported genetic risk for AF, AF in the presence of stroke, and cardioembolic stroke (Pearson r = 0.77 and 0.76, respectively, across SNPs with p < 4.4 × 10-4 in the previous AF meta-analysis). An AF PRS, adjusted for clinical AF risk factors, was associated with cardioembolic stroke (odds ratio [OR] per SD = 1.40, p = 1.45 × 10-48), explaining ∼20% of the heritable component of cardioembolic stroke risk. The AF PRS was also associated with stroke of undetermined cause (OR per SD = 1.07, p = 0.004), but no other primary stroke subtypes (all p > 0.1).Genetic risk of AF is associated with cardioembolic stroke, independent of clinical risk factors. Studies are warranted to determine whether AF genetic risk can serve as a biomarker for strokes caused by AF.

    View details for DOI 10.1212/NXG.0000000000000293

    View details for Web of Science ID 000455099800017

    View details for PubMedID 30584597

    View details for PubMedCentralID PMC6283455

  • The association between circulating endostatin levels and incident myocardial infarction. Scandinavian cardiovascular journal : SCJ Ruge, T., Carlsson, A. C., Jansson, J., Soderberg, S., Larsson, A., Arnlov, J. 2018: 1–15

    Abstract

    OBJECTIVE: Increased levels of circulating endostatin have been observed in patients with prevalent ischemic heart disease. However, the association between circulating endostatin, and incident myocardial infarction (MI) is less studied. Our main aim was to study the association between circulating endostatin and incident MI in the community adjusted for established cardiovascular risk factors in men and women.DESIGN: Circulating endostatin was measured in a nested case control study based on three large community-based Swedish cohorts, including 533MI cases, and 1003 age-, sex- and cohort-matched controls. Odds ratios (OR) with 95% confidence intervals (CI) were calculated with adjustments for established cardiovascular risk factors.RESULTS: Higher endostatin was associated with a higher incidence of MI independently of established cardiovascular risk factors (OR 1.19, 95 % CI 1.03-1.37, p=0.02), but this association was abolished after additional adjustment for C-reactive protein. Sex-stratified analyses suggest that the association was substantially stronger in women as compared to men Conclusions: In our community based sample, higher endostatin predicted incident myocardial infarction predominantly in women but not independently of CRP. Thus, our findings do not support a broad utility of endostatin measurements for the prediction of incident myocardial infarction in clinical practice.

    View details for DOI 10.1080/14017431.2018.1547839

    View details for PubMedID 30474426

  • Genome-Wide Association Studies of Estimated Fatty Acid Desaturase Activity in Serum and Adipose Tissue in Elderly Individuals: Associations with Insulin Sensitivity. Nutrients Marklund, M., Morris, A. P., Mahajan, A., Ingelsson, E., Lindgren, C. M., Lind, L., Riserus, U. 2018; 10 (11)

    Abstract

    Fatty acid desaturases (FADS) catalyze the formation of unsaturated fatty acids and have been related to insulin sensitivity (IS). FADS activities differ between tissues and are influenced by genetic factors that may impact the link to IS. Genome-wide association studies of delta-5-desaturase (D5D), delta-6-desaturase (D6D) and stearoyl-CoA desaturase-1 (SCD) activities (estimated by product-to-precursor ratios of fatty acids analyzed by gas chromatography) in serum cholesterol esters (n = 1453) and adipose tissue (n = 783, all men) were performed in two Swedish population-based cohorts. Genome-wide significant associated loci were evaluated for associations with IS measured with a hyperinsulinemic euglycemic clamp (n = 554). Variants at the FADS1 were strongly associated with D5D in both cholesterol esters (p = 1.9 * 10-70) and adipose tissue (p = 1.1 * 10-27). Variants in three further loci were associated with D6D in cholesterol esters (FADS2, p = 3.0 * 10-67; PDXDCI, p = 4.8 * 10-8; and near MC4R, p = 3.7 * 10-8) but no associations with D6D in adipose tissue attained genome-wide significance. One locus was associated with SCD in adipose tissue (PKDL1, p = 2.2 * 10-19). Genetic variants near MC4R were associated with IS (p = 3.8 * 10-3). The FADS cluster was the main genetic determinant of estimated FADS activity. However, fatty acid (FA) ratios in adipose tissue and cholesterol esters represent FADS activities in separate tissues and are thus influenced by different genetic factors with potential varying effects on IS.

    View details for DOI 10.3390/nu10111791

    View details for PubMedID 30453627

  • Publisher Correction: Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits. Nature genetics Evangelou, E., Warren, H. R., Mosen-Ansorena, D., Mifsud, B., Pazoki, R., Gao, H., Ntritsos, G., Dimou, N., Cabrera, C. P., Karaman, I., Ng, F. L., Evangelou, M., Witkowska, K., Tzanis, E., Hellwege, J. N., Giri, A., Velez Edwards, D. R., Sun, Y. V., Cho, K., Gaziano, J. M., Wilson, P. W., Tsao, P. S., Kovesdy, C. P., Esko, T., Magi, R., Milani, L., Almgren, P., Boutin, T., Debette, S., Ding, J., Giulianini, F., Holliday, E. G., Jackson, A. U., Li-Gao, R., Lin, W., Luan, J., Mangino, M., Oldmeadow, C., Prins, B. P., Qian, Y., Sargurupremraj, M., Shah, N., Surendran, P., Theriault, S., Verweij, N., Willems, S. M., Zhao, J., Amouyel, P., Connell, J., de Mutsert, R., Doney, A. S., Farrall, M., Menni, C., Morris, A. D., Noordam, R., Pare, G., Poulter, N. R., Shields, D. C., Stanton, A., Thom, S., Abecasis, G., Amin, N., Arking, D. E., Ayers, K. L., Barbieri, C. M., Batini, C., Bis, J. C., Blake, T., Bochud, M., Boehnke, M., Boerwinkle, E., Boomsma, D. I., Bottinger, E. P., Braund, P. S., Brumat, M., Campbell, A., Campbell, H., Chakravarti, A., Chambers, J. C., Chauhan, G., Ciullo, M., Cocca, M., Collins, F., Cordell, H. J., Davies, G., de Borst, M. H., de Geus, E. J., Deary, I. J., Deelen, J., Del Greco M, F., Demirkale, C. Y., Dorr, M., Ehret, G. B., Elosua, R., Enroth, S., Erzurumluoglu, A. M., Ferreira, T., Franberg, M., Franco, O. H., Gandin, I., Gasparini, P., Giedraitis, V., Gieger, C., Girotto, G., Goel, A., Gow, A. J., Gudnason, V., Guo, X., Gyllensten, U., Hamsten, A., Harris, T. B., Harris, S. E., Hartman, C. A., Havulinna, A. S., Hicks, A. A., Hofer, E., Hofman, A., Hottenga, J., Huffman, J. E., Hwang, S., Ingelsson, E., James, A., Jansen, R., Jarvelin, M., Joehanes, R., Johansson, A., Johnson, A. D., Joshi, P. K., Jousilahti, P., Jukema, J. W., Jula, A., Kahonen, M., Kathiresan, S., Keavney, B. D., Khaw, K., Knekt, P., Knight, J., Kolcic, I., Kooner, J. S., Koskinen, S., Kristiansson, K., Kutalik, Z., Laan, M., Larson, M., Launer, L. J., Lehne, B., Lehtimaki, T., Liewald, D. C., Lin, L., Lind, L., Lindgren, C. M., Liu, Y., Loos, R. J., Lopez, L. M., Lu, Y., Lyytikainen, L., Mahajan, A., Mamasoula, C., Marrugat, J., Marten, J., Milaneschi, Y., Morgan, A., Morris, A. P., Morrison, A. C., Munson, P. J., Nalls, M. A., Nandakumar, P., Nelson, C. P., Niiranen, T., Nolte, I. M., Nutile, T., Oldehinkel, A. J., Oostra, B. A., O'Reilly, P. F., Org, E., Padmanabhan, S., Palmas, W., Palotie, A., Pattie, A., Penninx, B. W., Perola, M., Peters, A., Polasek, O., Pramstaller, P. P., Nguyen, Q. T., Raitakari, O. T., Ren, M., Rettig, R., Rice, K., Ridker, P. M., Ried, J. S., Riese, H., Ripatti, S., Robino, A., Rose, L. M., Rotter, J. I., Rudan, I., Ruggiero, D., Saba, Y., Sala, C. F., Salomaa, V., Samani, N. J., Sarin, A., Schmidt, R., Schmidt, H., Shrine, N., Siscovick, D., Smith, A. V., Snieder, H., Sober, S., Sorice, R., Starr, J. M., Stott, D. J., Strachan, D. P., Strawbridge, R. J., Sundstrom, J., Swertz, M. A., Taylor, K. D., Teumer, A., Tobin, M. D., Tomaszewski, M., Toniolo, D., Traglia, M., Trompet, S., Tuomilehto, J., Tzourio, C., Uitterlinden, A. G., Vaez, A., van der Most, P. J., van Duijn, C. M., Vergnaud, A., Verwoert, G. C., Vitart, V., Volker, U., Vollenweider, P., Vuckovic, D., Watkins, H., Wild, S. H., Willemsen, G., Wilson, J. F., Wright, A. F., Yao, J., Zemunik, T., Zhang, W., Attia, J. R., Butterworth, A. S., Chasman, D. I., Conen, D., Cucca, F., Danesh, J., Hayward, C., Howson, J. M., Laakso, M., Lakatta, E. G., Langenberg, C., Melander, O., Mook-Kanamori, D. O., Palmer, C. N., Risch, L., Scott, R. A., Scott, R. J., Sever, P., Spector, T. D., van der Harst, P., Wareham, N. J., Zeggini, E., Levy, D., Munroe, P. B., Newton-Cheh, C., Brown, M. J., Metspalu, A., Hung, A. M., O'Donnell, C. J., Edwards, T. L., Million Veteran Program, Psaty, B. M., Tzoulaki, I., Barnes, M. R., Wain, L. V., Elliott, P., Caulfield, M. J. 2018

    Abstract

    In the version of this article originally published, the name of author Martin H. de Borst was coded incorrectly in the XML. The error has now been corrected in the HTML version of the paper.

    View details for DOI 10.1038/s41588-018-0297-3

    View details for PubMedID 30429575

  • Genome Analyses of >200,000 Individuals Identify 58 Loci for Chronic Inflammation and Highlight Pathways that Link Inflammation and Complex Disorders. American journal of human genetics Ligthart, S., Vaez, A., Vosa, U., Stathopoulou, M. G., de Vries, P. S., Prins, B. P., Van der Most, P. J., Tanaka, T., Naderi, E., Rose, L. M., Wu, Y., Karlsson, R., Barbalic, M., Lin, H., Pool, R., Zhu, G., Mace, A., Sidore, C., Trompet, S., Mangino, M., Sabater-Lleal, M., Kemp, J. P., Abbasi, A., Kacprowski, T., Verweij, N., Smith, A. V., Huang, T., Marzi, C., Feitosa, M. F., Lohman, K. K., Kleber, M. E., Milaneschi, Y., Mueller, C., Huq, M., Vlachopoulou, E., Lyytikainen, L., Oldmeadow, C., Deelen, J., Perola, M., Zhao, J. H., Feenstra, B., LifeLines Cohort Study, Amini, M., CHARGE Inflammation Working Group, Lahti, J., Schraut, K. E., Fornage, M., Suktitipat, B., Chen, W., Li, X., Nutile, T., Malerba, G., Luan, J., Bak, T., Schork, N., Del Greco M, F., Thiering, E., Mahajan, A., Marioni, R. E., Mihailov, E., Eriksson, J., Ozel, A. B., Zhang, W., Nethander, M., Cheng, Y., Aslibekyan, S., Ang, W., Gandin, I., Yengo, L., Portas, L., Kooperberg, C., Hofer, E., Rajan, K. B., Schurmann, C., den Hollander, W., Ahluwalia, T. S., Zhao, J., Draisma, H. H., Ford, I., Timpson, N., Teumer, A., Huang, H., Wahl, S., Liu, Y., Huang, J., Uh, H., Geller, F., Joshi, P. K., Yanek, L. R., Trabetti, E., Lehne, B., Vozzi, D., Verbanck, M., Biino, G., Saba, Y., Meulenbelt, I., O'Connell, J. R., Laakso, M., Giulianini, F., Magnusson, P. K., Ballantyne, C. M., Hottenga, J. J., Montgomery, G. W., Rivadineira, F., Rueedi, R., Steri, M., Herzig, K., Stott, D. J., Menni, C., Franberg, M., St Pourcain, B., Felix, S. B., Pers, T. H., Bakker, S. J., Kraft, P., Peters, A., Vaidya, D., Delgado, G., Smit, J. H., GroSSmann, V., Sinisalo, J., Seppala, I., Williams, S. R., Holliday, E. G., Moed, M., Langenberg, C., Raikkonen, K., Ding, J., Campbell, H., Sale, M. M., Chen, Y. I., James, A. L., Ruggiero, D., Soranzo, N., Hartman, C. A., Smith, E. N., Berenson, G. S., Fuchsberger, C., Hernandez, D., Tiesler, C. M., Giedraitis, V., Liewald, D., Fischer, K., Mellstrom, D., Larsson, A., Wang, Y., Scott, W. R., Lorentzon, M., Beilby, J., Ryan, K. A., Pennell, C. E., Vuckovic, D., Balkau, B., Concas, M. P., Schmidt, R., Mendes de Leon, C. F., Bottinger, E. P., Kloppenburg, M., Paternoster, L., Boehnke, M., Musk, A. W., Willemsen, G., Evans, D. M., Madden, P. A., Kahonen, M., Kutalik, Z., Zoledziewska, M., Karhunen, V., Kritchevsky, S. B., Sattar, N., Lachance, G., Clarke, R., Harris, T. B., Raitakari, O. T., Attia, J. R., van Heemst, D., Kajantie, E., Sorice, R., Gambaro, G., Scott, R. A., Hicks, A. A., Ferrucci, L., Standl, M., Lindgren, C. M., Starr, J. M., Karlsson, M., Lind, L., Li, J. Z., Chambers, J. C., Mori, T. A., de Geus, E. J., Heath, A. C., Martin, N. G., Auvinen, J., Buckley, B. M., de Craen, A. J., Waldenberger, M., Strauch, K., Meitinger, T., Scott, R. J., McEvoy, M., Beekman, M., Bombieri, C., Ridker, P. M., Mohlke, K. L., Pedersen, N. L., Morrison, A. C., Boomsma, D. I., Whitfield, J. B., Strachan, D. P., Hofman, A., Vollenweider, P., Cucca, F., Jarvelin, M., Jukema, J. W., Spector, T. D., Hamsten, A., Zeller, T., Uitterlinden, A. G., Nauck, M., Gudnason, V., Qi, L., Grallert, H., Borecki, I. B., Rotter, J. I., Marz, W., Wild, P. S., Lokki, M., Boyle, M., Salomaa, V., Melbye, M., Eriksson, J. G., Wilson, J. F., Penninx, B. W., Becker, D. M., Worrall, B. B., Gibson, G., Krauss, R. M., Ciullo, M., Zaza, G., Wareham, N. J., Oldehinkel, A. J., Palmer, L. J., Murray, S. S., Pramstaller, P. P., Bandinelli, S., Heinrich, J., Ingelsson, E., Deary, I. J., Magi, R., Vandenput, L., van der Harst, P., Desch, K. C., Kooner, J. S., Ohlsson, C., Hayward, C., Lehtimaki, T., Shuldiner, A. R., Arnett, D. K., Beilin, L. J., Robino, A., Froguel, P., Pirastu, M., Jess, T., Koenig, W., Loos, R. J., Evans, D. A., Schmidt, H., Smith, G. D., Slagboom, P. E., Eiriksdottir, G., Morris, A. P., Psaty, B. M., Tracy, R. P., Nolte, I. M., Boerwinkle, E., Visvikis-Siest, S., Reiner, A. P., Gross, M., Bis, J. C., Franke, L., Franco, O. H., Benjamin, E. J., Chasman, D. I., Dupuis, J., Snieder, H., Dehghan, A., Alizadeh, B. Z., Alizadeh, B. Z., Boezen, H. M., Franke, L., van der Harst, P., Navis, G., Rots, M., Snieder, H., Swertz, M., Wolffenbuttel, B. H., Wijmenga, C., Benjamin, E., Chasman, D. I., Dehghan, A., Ahluwalia, T. S., Meigs, J., Tracy, R., Alizadeh, B. Z., Ligthart, S., Bis, J., Eiriksdottir, G., Pankratz, N., Gross, M., Rainer, A., Snieder, H., Wilson, J. G., Psaty, B. M., Dupuis, J., Prins, B., Vaso, U., Stathopoulou, M., Franke, L., Lehtimaki, T., Koenig, W., Jamshidi, Y., Siest, S., Abbasi, A., Uitterlinden, A. G., Abdollahi, M., Schnabel, R., Schick, U. M., Nolte, I. M., Kraja, A., Hsu, Y., Tylee, D. S., Zwicker, A., Uher, R., Davey-Smith, G., Morrison, A. C., Hicks, A., van Duijn, C. M., Ward-Caviness, C., Boerwinkle, E., Rotter, J., Rice, K., Lange, L., Perola, M., de Geus, E., Morris, A. P., Makela, K. M., Stacey, D., Eriksson, J., Frayling, T. M., Slagboom, E. P. 2018; 103 (5): 691–706

    Abstract

    C-reactive protein (CRP) is a sensitive biomarker of chronic low-grade inflammation and is associated with multiple complex diseases. The genetic determinants of chronic inflammation remain largely unknown, and the causal role of CRP in several clinical outcomes is debated. We performed two genome-wide association studies (GWASs), on HapMap and 1000 Genomes imputed data, of circulating amounts of CRP by using data from 88 studies comprising 204,402 European individuals. Additionally, we performed in silico functional analyses and Mendelian randomization analyses with several clinical outcomes. The GWAS meta-analyses of CRP revealed 58 distinct genetic loci (p < 5* 10-8). After adjustment for body mass index in the regression analysis, the associations at all except three loci remained. The lead variants at the distinct loci explained up to 7.0% of the variance in circulating amounts of CRP. We identified 66 gene sets that were organized in two substantially correlated clusters, one mainly composed of immune pathways and the other characterized by metabolic pathways in the liver. Mendelian randomization analyses revealed a causal protective effect of CRP on schizophrenia and a risk-increasing effect on bipolar disorder. Our findings provide further insights into the biology of inflammation and could lead to interventions for treating inflammation and its clinical consequences.

    View details for DOI 10.1016/j.ajhg.2018.09.009

    View details for PubMedID 30388399

  • Proteomic profiling of endothelium-dependent vasodilation. Journal of hypertension Lind, L., Sundstrom, J., Arnlov, J., Ingelsson, E. 2018

    Abstract

    OBJECTIVE: As endothelial dysfunction is an early event in atherosclerosis formation, we investigated if proteins previously related to cardiovascular disease also were related to endothelial function using a novel targeted proteomics approach.METHODS: In the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (n = 850-970, all aged 70 years), endothelium-dependent vasodilation (EDV) in the forearm was assessed by intra-arterial infusion of acetylcholine. Flow-mediated vasodilation (FMD) was investigated in the brachial artery by ultrasound. The same investigations were carried out in the Prospective investigation of Obesity, Energy and Metabolism (POEM) study (n = 375-461, all aged 50 years). After strict quality control, 84 cardiovascular-related proteins measured by the proximity extension assay were studied in relation to EDV and FMD in PIVUS (discovery sample) and POEM (validation sample).RESULTS: Of the 15 proteins being significantly related to EDV in PIVUS (false discovery rate <0.025), seven could be replicated in POEM at nominal significance and same effect direction when adjusted for sex and storage time. Of those, only cathepsin D remained significant following further adjustment for traditional cardiovascular risk factors (beta, -0.08; 95% confidence interval, -0.16, -0.01; P = 0.033; change in ln-transformed EDV per 1-SD increase in protein level). No protein was significantly related to FMD.CONCLUSION: Using a discovery/validation approach in two samples, our results indicate an inverse association between plasma cathepsin D levels and endothelial-dependent vasodilation.

    View details for DOI 10.1097/HJH.0000000000001863

    View details for PubMedID 30339551

  • Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nature genetics Mahajan, A., Taliun, D., Thurner, M., Robertson, N. R., Torres, J. M., Rayner, N. W., Payne, A. J., Steinthorsdottir, V., Scott, R. A., Grarup, N., Cook, J. P., Schmidt, E. M., Wuttke, M., Sarnowski, C., Magi, R., Nano, J., Gieger, C., Trompet, S., Lecoeur, C., Preuss, M. H., Prins, B. P., Guo, X., Bielak, L. F., Below, J. E., Bowden, D. W., Chambers, J. C., Kim, Y. J., Ng, M. C., Petty, L. E., Sim, X., Zhang, W., Bennett, A. J., Bork-Jensen, J., Brummett, C. M., Canouil, M., Ec Kardt, K., Fischer, K., Kardia, S. L., Kronenberg, F., Lall, K., Liu, C., Locke, A. E., Luan, J., Ntalla, I., Nylander, V., Schonherr, S., Schurmann, C., Yengo, L., Bottinger, E. P., Brandslund, I., Christensen, C., Dedoussis, G., Florez, J. C., Ford, I., Franco, O. H., Frayling, T. M., Giedraitis, V., Hackinger, S., Hattersley, A. T., Herder, C., Ikram, M. A., Ingelsson, M., Jorgensen, M. E., Jorgensen, T., Kriebel, J., Kuusisto, J., Ligthart, S., Lindgren, C. M., Linneberg, A., Lyssenko, V., Mamakou, V., Meitinger, T., Mohlke, K. L., Morris, A. D., Nadkarni, G., Pankow, J. S., Peters, A., Sattar, N., Stancakova, A., Strauch, K., Taylor, K. D., Thorand, B., Thorleifsson, G., Thorsteinsdottir, U., Tuomilehto, J., Witte, D. R., Dupuis, J., Peyser, P. A., Zeggini, E., Loos, R. J., Froguel, P., Ingelsson, E., Lind, L., Groop, L., Laakso, M., Collins, F. S., Jukema, J. W., Palmer, C. N., Grallert, H., Metspalu, A., Dehghan, A., Kottgen, A., Abecasis, G. R., Meigs, J. B., Rotter, J. I., Marchini, J., Pedersen, O., Hansen, T., Langenberg, C., Wareham, N. J., Stefansson, K., Gloyn, A. L., Morris, A. P., Boehnke, M., McCarthy, M. I. 2018

    Abstract

    We expanded GWAS discovery for type 2 diabetes (T2D) by combining data from 898,130 European-descent individuals (9% cases), after imputation to high-density reference panels. With these data, we (i) extend the inventory of T2D-risk variants (243 loci, 135 newly implicated in T2D predisposition, comprising 403 distinct association signals); (ii) enrich discovery of lower-frequency risk alleles (80 index variants with minor allele frequency <5%, 14 with estimated allelic odds ratio >2); (iii) substantially improve fine-mapping of causal variants (at 51 signals, one variant accounted for >80% posterior probability of association (PPA)); (iv) extend fine-mapping through integration of tissue-specific epigenomic information (islet regulatory annotations extend the number of variants with PPA >80% to 73); (v) highlight validated therapeutic targets (18 genes with associations attributable to coding variants); and (vi) demonstrate enhanced potential for clinical translation (genome-wide chip heritability explains 18% of T2D risk; individuals in the extremes of a T2D polygenic risk score differ more than ninefold in prevalence).

    View details for DOI 10.1038/s41588-018-0241-6

    View details for PubMedID 30297969

  • Genome-wide association study of coronary artery disease among individuals with diabetes: the UK Biobank DIABETOLOGIA Fall, T., Gustafsson, S., Orho-Melander, M., Ingelsson, E. 2018; 61 (10): 2174–79

    Abstract

    Coronary artery disease (CAD) is a common complication among individuals with diabetes. A better understanding of the genetic background of CAD in this population has the potential to suggest novel molecular targets for screening, risk assessment and drug development.We performed a genome-wide association study of CAD in 15,666 unrelated individuals (3,968 CAD cases and 11,698 controls) of white British ancestry with diabetes at inclusion in the UK Biobank study. Our results were compared with results from participants without diabetes.We found genome-wide significant evidence for association with CAD at the previously well-established LPA locus (lead variant: rs74617384; OR 1.38 [95% CI 1.26, 1.51], p = 3.2 × 10-12) and at 9p21 (lead variant: rs10811652; OR 1.19 [95% CI 1.13, 1.26], p = 6.0 × 10-11). Moreover, other variants previously associated with CAD showed similar effects in the participants with and without diabetes, indicating that the genetic architecture of CAD is largely the same.Our results indicate large similarities between the genetic architecture of CAD in participants with and without diabetes. Larger studies are needed to establish whether there are important diabetes-specific CAD loci.

    View details for DOI 10.1007/s00125-018-4686-z

    View details for Web of Science ID 000443448100012

    View details for PubMedID 30003307

  • A Phenome- Wide Association Study of Patatin-like Phospholipase Domain Containing 3 (PNPLA3) I148M Variant in 337,536 Participants of the UK Biobank Study Yeo, Y., Rao, A., Liu, B., Nguyen, M. H., Ingelsson, E. WILEY. 2018: 65A–66A
  • Genetic analysis of over 1 million people identifies 535 new loci associated with blood pressure traits NATURE GENETICS Evangelou, E., Warren, H. R., Mosen-Ansorena, D., Mifsu, B., Pazoki, R., Gao, H., Ntritsos, G., Dimou, N., Cabrer, C. P., Karaman, I., Ng, F., Evangelou, M., Witkowska, K., Tzanis, E., Hellwege, J. N., Giri, A., Edwards, D., Sun, Y., Cho, K., Gaziano, J., Wilson, P. F., Tsao, P. S., Kovesdy, C. P., Esko, T., Magi, R., Milani, L., Almgren, P., Boutin, T., Debette, S., Ding, J., Giulianini, F., Holliday, E. G., Jackson, A. U., Li-Gao, R., Lin, W., Luan, J., Mangino, M., Oldmeadow, C., Prins, B., Qian, Y., Sargurupremraj, M., Shah, N., Surendran, P., Theriault, S., Verweij, N., Willems, S. M., Zhao, J., Amouyel, P., Connell, J., de Mutsert, R., Doney, A. F., Farrall, M., Menni, C., Morris, A. D., Noordam, R., Pare, G., Poulter, N. R., Shields, D. C., Stanton, A., Thom, S., Abecasis, G., Amin, N., Arking, D. E., Ayers, K. L., Barbieri, C. M., Batini, C., Bis, J. C., Blake, T., Bochud, M., Boehnke, M., Boerwinkle, E., Boomsma, D., Bottinger, E. P., Braund, P. S., Brumat, M., Campbell, A., Campbell, H., Chakravarti, A., Chambers, J. C., Chauhan, G., Ciullo, M., Cocca, M., Collins, F., Cordell, H. J., Davies, G., de Borst, M. H., de Geus, E. J., Deary, I. J., Deelen, J., Del Greco, F. M., Demirkale, C., Doerr, M., Ehret, G. B., Elosua, R., Enroth, S., Erzurumluoglu, A., Ferreira, T., Franberg, M., Franco, O. H., Gandin, I., Gasparini, P., Giedraitis, V., Gieger, C., Girotto, G., Goel, A., Gow, A. J., Gudnason, V., Guo, X., Gyllensten, U., Hamsten, A., Harris, T. B., Harris, S. E., Hartman, C. A., Havulinna, A. S., Hicks, A. A., Hofer, E., Hofman, A., Hottenga, J., Huffman, J. E., Hwang, S., Ingelsson, E., James, A., Jansen, R., Jarvelin, M., Joehanes, R., Johansson, A., Johnson, A. D., Joshi, P. K., Jousilahti, P., Jukema, J., Jula, A., Kahonen, M., Kathiresan, S., Keavney, B. D., Khaw, K., Knekt, P., Knight, J., Kolcic, I., Kooner, J. S., Koskinen, S., Kristiansson, K., Kutalik, Z., Laan, M., Larson, M., Launer, L. J., Lehne, B., Lehtimaki, T., Liewald, D. M., Lin, L., Lind, L., Lindgren, C. M., Liu, Y., Loos, R. F., Lopez, L. M., Lu, Y., Lyytikainen, L., Mahajan, A., Mamasoula, C., Marrugat, J., Marten, J., Milaneschi, Y., Morgan, A., Morris, A. P., Morrison, A. C., Munson, P. J., Nalls, M. A., Nandakumar, P., Nelson, C. P., Niiranen, T., Nolte, I. M., Nutile, T., Oldehinkel, A. J., Oostra, B. A., O'Reilly, P. F., Org, E., Padmanabhan, S., Palmas, W., Palotie, A., Pattie, A., Penninx, B. H., Perola, M., Peters, A., Polasek, O., Pramstaller, P. P., Nguyen, Q., Raitakari, O. T., Ren, M., Rettig, R., Rice, K., Ridker, P. M., Ried, J. S., Riese, H., Ripatti, S., Robino, A., Rose, L. M., Rotter, J., Rudan, I., Ruggiero, D., Saba, Y., Sala, C. F., Salomaa, V., Samani, N. J., Sarin, A., Schmidt, R., Schmidt, H., Shrine, N., Siscovick, D., Smith, A., Snieder, H., Sober, S., Sorice, R., Starr, J. M., Stott, D. J., Strachan, D. P., Strawbridge, R. J., Sundstrom, J., Swertz, M. A., Taylor, K. D., Teumer, A., Tobin, M. D., Tomaszewski, M., Toniolo, D., Traglia, M., Trompet, S., Tuomilehto, J., Tzourio, C., Uitterlinden, A. G., Vaez, A., van der Most, P. J., van Duijn, C. M., Vergnaud, A., Verwoert, G. C., Vitart, V., Voelker, U., Vollenweider, P., Vuckovic, D., Watkins, H., Wild, S. H., Willemsen, G., Wilson, J. F., Wright, A. F., Yao, J., Zemunik, T., Zhang, W., Attia, J. R., Butterworth, A. S., Chasman, D., Conen, D., Cucca, F., Danesh, J., Hayward, C., Howson, J. M., Laakso, M., Lakatta, E. G., Langenberg, C., Melander, O., Mook-Kanamori, D. O., Palmer, C. A., Risch, L., Scott, R. A., Scott, R. J., Sever, P., Spector, T. D., van der Harst, P., Wareham, N. J., Zeggini, E., Levy, D., Munroe, P. B., Newton-Cheh, C., Brown, M. J., Metspalu, A., Hung, A. M., O'Donnell, C., Edwards, T. L., Psaty, B. M., Tzoulaki, I., Barnes, M. R., Wain, L., Elliott, P., Caulfield, M. J., Million Vet Program 2018; 50 (10): 1412-+

    Abstract

    High blood pressure is a highly heritable and modifiable risk factor for cardiovascular disease. We report the largest genetic association study of blood pressure traits (systolic, diastolic and pulse pressure) to date in over 1 million people of European ancestry. We identify 535 novel blood pressure loci that not only offer new biological insights into blood pressure regulation but also highlight shared genetic architecture between blood pressure and lifestyle exposures. Our findings identify new biological pathways for blood pressure regulation with potential for improved cardiovascular disease prevention in the future.

    View details for DOI 10.1038/s41588-018-0205-x

    View details for Web of Science ID 000446047000013

    View details for PubMedID 30224653

  • Associations of Circulating Protein Levels With Lipid Fractions in the General Population ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Figarska, S. M., Gustafsson, S., Sundstrom, J., Arnlov, J., Malarstig, A., Elmstahl, S., Fall, T., Lind, L., Ingelsson, E. 2018; 38 (10): 2505–18
  • USE OF PROTEOMICS TO INVESTIGATE BLOOD PRESSURE PROGRESS IN THE ELDERLY Lin Yi-Ting, Fall, T., Gustafsson, S., Ingelsson, E., Arnlov, J., Lind, L., Engstrom, G., Sundstrom, J. LIPPINCOTT WILLIAMS & WILKINS. 2018: E115
  • Epigenetic influences on aging: a longitudinal genome-wide methylation study in old Swedish twins. Epigenetics Wang, Y., Karlsson, R., Lampa, E., Zhang, Q., Hedman, A. K., Almgren, M., Almqvist, C., McRae, A. F., Marioni, R. E., Ingelsson, E., Visscher, P. M., Deary, I. J., Lind, L., Morris, T., Beck, S., Pedersen, N. L., Hagg, S. 2018: 1–13

    Abstract

    Age-related changes in DNA methylation were observed in cross-sectional studies, but longitudinal evidence is still limited. Here, we aimed to characterize longitudinal age-related methylation patterns using 1011 blood samples collected from 385 Swedish twins (age at entry: mean 69 and standard deviation 9.7, 73 monozygotic and 96 dizygotic pairs) up to five times (mean 2.6) over 20 years (mean 8.7). We identified 1316 age-associated methylation sites (P<1.3*10-7) using a longitudinal epigenome-wide association study design. We measured how estimated cellular compositions changed with age and how much they confounded the age effect. We validated the results in two independent longitudinal cohorts, where 118 CpGs were replicated in Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS, 390 samples) (P<3.9*10-5), 594 in Lothian Birth Cohort (LBC, 3018 samples) (P<5.1*10-5) and 63 in both. Functional annotation of age-associated CpGs showed enrichment in CCCTC-binding factor (CTCF) and other transcription factor binding sites. We further investigated genetic influences on methylation and found no interaction between age and genetic effects in the 1316 age-associated CpGs. Moreover, in the same CpGs, methylation differences within twin pairs increased with 6.4% over 10 years, where monozygotic twins had smaller intra-pair differences than dizygotic twins. In conclusion, we show that age-related methylation changes persist in a longitudinal perspective, and are fairly stable across cohorts. The changes are under genetic influence, although this effect is independent of age. Moreover, methylation variability increase over time, especially in age-associated CpGs, indicating the increase of environmental contributions on DNA methylation with age.

    View details for DOI 10.1080/15592294.2018.1526028

    View details for PubMedID 30264654

  • Genetic predictors of testosterone and their associations with cardiovascular disease and risk factors: A Mendelian randomization investigation INTERNATIONAL JOURNAL OF CARDIOLOGY Schooling, C., Luo, S., Yeung, S., Thompson, D. J., Karthikeyan, S., Bolton, T. R., Mason, A. M., Ingelsson, E., Burgess, S. 2018; 267: 171–76

    Abstract

    Testosterone supplementation has been linked to increased cardiovascular disease risk in some observational studies. The causal role of testosterone can be investigated using a Mendelian randomization approach.We assessed genetic associations of variants in two gene regions (SHBG and JMJD1C) with several cardiovascular risk factors (lipids, adiponectin, blood pressure, anthropometric traits) plus male pattern baldness, including control outcomes and potential mediators. We assessed genetic associations with coronary artery disease (CAD) risk in the CARDIoGRAMplusC4D consortium (171,191 individuals including 60,801 cases), and associations with CAD and ischaemic stroke risk in the UK Biobank (367,643 individuals including 25,352 CAD cases and 3650 ischaemic stroke cases). Genetic predictors of increased serum testosterone were associated with lipids, blood pressure, and height. There was some evidence of an association with risk of CAD (SHBG gene region: odds ratio (OR) 0.95 per 1 unit increase in log-transformed testosterone [95% confidence interval: 0.81-1.12, p = 0.55]; JMJD1C gene region: OR 1.24 [1.01-1.51, p = 0.04]) and ischaemic stroke both overall (SHBG: OR 1.05 [0.64, 1.73, p = 0.83]; JMJD1C: OR 2.52 [1.33, 4.77, p = 0.005]) and in men. However, associations with some control outcomes were in the opposite direction to that expected.Sex hormone-related mechanisms appear to be relevant to cardiovascular risk factors and for stroke (particularly for men). However, the extent that these findings are specifically informative about endogenous testosterone or testosterone supplementation is unclear. These findings underline a fundamental limitation for the use of Mendelian randomization where biological knowledge about the function of genetic variants is uncertain.

    View details for DOI 10.1016/j.ijcard.2018.05.051

    View details for Web of Science ID 000436570700040

    View details for PubMedID 29804699

    View details for PubMedCentralID PMC6024225

  • Multi-ethnic genome-wide association study for atrial fibrillation NATURE GENETICS Roselli, C., Chaffin, M. D., Weng, L., Aeschbacher, S., Ahlberg, G., Albert, C. M., Almgren, P., Alonso, A., Anderson, C. D., Aragam, K. G., Arking, D. E., Barnard, J., Bartz, T. M., Benjamin, E. J., Bihlmeyer, N. A., Bis, J. C., Bloom, H. L., Boerwinkle, E., Bottinger, E. B., Brody, J. A., Calkins, H., Campbell, A., Cappola, T. P., Carlquist, J., Chasman, D. I., Chen, L. Y., Chen, Y., Choi, E., Choi, S., Christophersen, I. E., Chung, M. K., Cole, J. W., Conen, D., Cook, J., Crijns, H. J., Cutler, M. J., Damrauer, S. M., Daniels, B. R., Darbar, D., Delgado, G., Denny, J. C., Dichgans, M., Doerr, M., Dudink, E. A., Dudley, S. C., Esa, N., Esko, T., Eskola, M., Fatkin, D., Felix, S. B., Ford, I., Franco, O. H., Geelhoed, B., Grewal, R. P., Gudnason, V., Guo, X., Gupta, N., Gustafsson, S., Gutmann, R., Hamsten, A., Harris, T. B., Hayward, C., Heckbert, S. R., Hernesniemi, J., Hocking, L. J., Hofman, A., Horimoto, A. R., Huang, J., Huang, P. L., Huffman, J., Ingelsson, E., Ipek, E., Ito, K., Jimenez-Conde, J., Johnson, R., Jukema, J., Kaeaeb, S., Kahonen, M., Kamatani, Y., Kane, J. P., Kastrati, A., Kathiresan, S., Katschnig-Winter, P., Kavousi, M., Kessler, T., Kietselaer, B. L., Kirchhof, P., Kleber, M. E., Knight, S., Krieger, J. E., Kubo, M., Launer, L. J., Laurikka, J., Lehtimaki, T., Leineweber, K., Lemaitre, R. N., Li, M., Lim, H., Lin, H. J., Lin, H., Lind, L., Lindgren, C. M., Lokki, M., London, B., Loos, R. F., Low, S., Lu, Y., Lyytikainen, L., Macfarlane, P. W., Magnusson, P. K., Mahajan, A., Malik, R., Mansur, A. J., Marcus, G. M., Margolin, L., Margulies, K. B., Maerz, W., McManus, D. D., Melander, O., Mohanty, S., Montgomery, J. A., Morley, M. P., Morris, A. P., Mueller-Nurasyid, M., Natale, A., Nazarian, S., Neumann, B., Newton-Cheh, C., Niemeijer, M. N., Nikus, K., Nilsson, P., Noordam, R., Oellers, H., Olesen, M. S., Orho-Melander, M., Padmanabhan, S., Pak, H., Pare, G., Pedersen, N. L., Pera, J., Pereira, A., Porteous, D., Psaty, B. M., Pulit, S. L., Pullinger, C. R., Rader, D. J., Refsgaard, L., Ribases, M., Ridker, P. M., Rienstra, M., Risch, L., Roden, D. M., Rosand, J., Rosenberg, M. A., Rost, N., Rotter, J. I., Saba, S., Sandhu, R. K., Schnabel, R. B., Schramm, K., Schunkert, H., Schurman, C., Scott, S. A., Seppala, I., Shaffer, C., Shah, S., Shalaby, A. A., Shim, J., Shoemaker, M., Siland, J. E., Sinisalo, J., Sinner, M. F., Slowik, A., Smith, A. V., Smith, B. H., Smith, J., Smith, J. D., Smith, N. L., Soliman, E. Z., Sotoodehnia, N., Stricker, B. H., Sun, A., Sun, H., Svendsen, J. H., Tanaka, T., Tanriverdi, K., Taylor, K. D., Teder-Laving, M., Teumer, A., Theriault, S., Trompet, S., Tucker, N. R., Tveit, A., Uitterlinden, A. G., Van Der Harst, P., Van Gelder, I. C., Van Wagoner, D. R., Verweij, N., Vlachopoulou, E., Voelker, U., Wang, B., Weeke, P. E., Weijs, B., Weiss, R., Weiss, S., Wells, Q. S., Wiggins, K. L., Wong, J. A., Woo, D., Worrall, B. B., Yang, P., Yao, J., Yoneda, Z. T., Zeller, T., Zeng, L., Lubitz, S. A., Lunetta, K. L., Ellinor, P. T. 2018; 50 (9): 1225-+

    Abstract

    Atrial fibrillation (AF) affects more than 33 million individuals worldwide1 and has a complex heritability2. We conducted the largest meta-analysis of genome-wide association studies (GWAS) for AF to date, consisting of more than half a million individuals, including 65,446 with AF. In total, we identified 97 loci significantly associated with AF, including 67 that were novel in a combined-ancestry analysis, and 3 that were novel in a European-specific analysis. We sought to identify AF-associated genes at the GWAS loci by performing RNA-sequencing and expression quantitative trait locus analyses in 101 left atrial samples, the most relevant tissue for AF. We also performed transcriptome-wide analyses that identified 57 AF-associated genes, 42 of which overlap with GWAS loci. The identified loci implicate genes enriched within cardiac developmental, electrophysiological, contractile and structural pathways. These results extend our understanding of the biological pathways underlying AF and may facilitate the development of therapeutics for AF.

    View details for DOI 10.1038/s41588-018-0133-9

    View details for Web of Science ID 000443151300012

    View details for PubMedID 29892015

  • Multiplex proteomics for prediction of major cardiovascular events in type 2 diabetes DIABETOLOGIA Nowak, C., Carlsson, A. C., Ostgren, C., Nystrom, F. H., Alam, M., Feldreich, T., Sundstrom, J., Carrero, J., Leppert, J., Hedberg, P., Henriksen, E., Cordeiro, A. C., Giedraitis, V., Lind, L., Ingelsson, E., Fall, T., Arnlov, J. 2018; 61 (8): 1748–57

    Abstract

    Multiplex proteomics could improve understanding and risk prediction of major adverse cardiovascular events (MACE) in type 2 diabetes. This study assessed 80 cardiovascular and inflammatory proteins for biomarker discovery and prediction of MACE in type 2 diabetes.We combined data from six prospective epidemiological studies of 30-77-year-old individuals with type 2 diabetes in whom 80 circulating proteins were measured by proximity extension assay. Multivariable-adjusted Cox regression was used in a discovery/replication design to identify biomarkers for incident MACE. We used gradient-boosted machine learning and lasso regularised Cox regression in a random 75% training subsample to assess whether adding proteins to risk factors included in the Swedish National Diabetes Register risk model would improve the prediction of MACE in the separate 25% test subsample.Of 1211 adults with type 2 diabetes (32% women), 211 experienced a MACE over a mean (±SD) of 6.4 ± 2.3 years. We replicated associations (<5% false discovery rate) between risk of MACE and eight proteins: matrix metalloproteinase (MMP)-12, IL-27 subunit α (IL-27a), kidney injury molecule (KIM)-1, fibroblast growth factor (FGF)-23, protein S100-A12, TNF receptor (TNFR)-1, TNFR-2 and TNF-related apoptosis-inducing ligand receptor (TRAIL-R)2. Addition of the 80-protein assay to established risk factors improved discrimination in the separate test sample from 0.686 (95% CI 0.682, 0.689) to 0.748 (95% CI 0.746, 0.751). A sparse model of 20 added proteins achieved a C statistic of 0.747 (95% CI 0.653, 0.842) in the test sample.We identified eight protein biomarkers, four of which are novel, for risk of MACE in community residents with type 2 diabetes, and found improved risk prediction by combining multiplex proteomics with an established risk model. Multiprotein arrays could be useful in identifying individuals with type 2 diabetes who are at highest risk of a cardiovascular event.

    View details for DOI 10.1007/s00125-018-4641-z

    View details for Web of Science ID 000437432200006

    View details for PubMedID 29796748

  • Bioimpedance and New-Onset Heart Failure: A Longitudinal Study of >500 000 Individuals From the General Population JOURNAL OF THE AMERICAN HEART ASSOCIATION Lindholm, D., Fukaya, E., Leeper, N. J., Ingelsson, E. 2018; 7 (13)

    Abstract

    Heart failure constitutes a high burden on patients and society, but although lifetime risk is high, it is difficult to predict without costly or invasive testing. We aimed to establish new risk factors of heart failure, which potentially could enable early diagnosis and preemptive treatment.We applied machine learning in the UK Biobank in an agnostic search of risk factors for heart failure in 500 451 individuals, excluding individuals with prior heart failure. Novel factors were then subjected to several in-depth analyses, including multivariable Cox models of incident heart failure, and assessment of discrimination and calibration. Machine learning confirmed many known and putative risk factors for heart failure and identified several novel candidates. Mean reticulocyte volume appeared as one novel factor and leg bioimpedance another, the latter appearing as the most important new marker. Leg bioimpedance was lower in those who developed heart failure during an up to 9.8-year follow-up. When adjusting for known heart failure risk factors, leg bioimpedance was inversely related to heart failure (hazard ratio [95% confidence interval], 0.60 [0.48-0.73] and 0.75 [0.59-0.94], in age- and sex-adjusted and fully adjusted models, respectively, comparing the upper versus lower quartile). A model including leg bioimpedance, age, sex, and self-reported history of myocardial infarction showed good discrimination for future heart failure hospitalization (Concordance index [C-index]=0.82) and good calibration.Leg bioimpedance is inversely associated with heart failure incidence in the general population. A simple model of exclusively noninvasive measures, combining leg bioimpedance with history of myocardial infarction, age, and sex provides accurate predictive capacity.

    View details for DOI 10.1161/JAHA.118.008970

    View details for Web of Science ID 000452700100033

    View details for PubMedID 29959136

    View details for PubMedCentralID PMC6064899

  • Large-Scale Phenome-Wide Association Study of PCSK9 Variants Demonstrates Protection Against Ischemic Stroke CIRCULATION-GENOMIC AND PRECISION MEDICINE Rao, A. S., Lindholm, D., Rivas, M. A., Knowles, J. W., Montgomery, S. B., Ingelsson, E. 2018; 11 (7): e002162

    Abstract

    PCSK9 inhibition is a potent new therapy for hypercholesterolemia and cardiovascular disease. Although short-term clinical trial results have not demonstrated major adverse effects, long-term data will not be available for some time. Genetic studies in large biobanks offer a unique opportunity to predict drug effects and provide context for the evaluation of future clinical trial outcomes.We tested the association of the PCSK9 missense variant rs11591147 with predefined phenotypes and phenome-wide, in 337 536 individuals of British ancestry in the UK Biobank, with independent discovery and replication. Using a Bayesian statistical method, we leveraged phenotype correlations to evaluate the phenome-wide impact of PCSK9 inhibition with higher power at a finer resolution.The T allele of rs11591147 showed a protective effect on hyperlipidemia (odds ratio, 0.63±0.04; P=2.32×10-38), coronary heart disease (odds ratio, 0.73±0.09; P=1.05×10-6), and ischemic stroke (odds ratio, 0.61±0.18; P=2.40×10-3) and was associated with increased type 2 diabetes mellitus risk adjusted for lipid-lowering medication status (odds ratio, 1.24±0.10; P=1.98×10-7). We did not observe associations with cataracts, heart failure, atrial fibrillation, and cognitive dysfunction. Leveraging phenotype correlations, we observed evidence of a protective association with cerebral infarction and vascular occlusion. These results explore the effects of direct PCSK9 inhibition; off-target effects cannot be predicted using this approach.This result represents the first genetic evidence in a large cohort for the protective effect of PCSK9 inhibition on ischemic stroke and corroborates exploratory evidence from clinical trials. PCSK9 inhibition was not associated with variables other than those related to LDL (low-density lipoprotein) cholesterol, atherosclerosis, and type 2 diabetes mellitus, suggesting that other effects are either small or absent.

    View details for DOI 10.1161/CIRCGEN.118.002162

    View details for Web of Science ID 000447462400006

    View details for PubMedID 29997226

    View details for PubMedCentralID PMC6050027

  • Circulating endostatin and the incidence of heart failure. Scandinavian cardiovascular journal : SCJ Ruge, T., Carlsson, A. C., Ingelsson, E., Riserus, U., Sundstrom, J., Larsson, A., Lind, L., Arnlov, J. 2018: 1–6

    Abstract

    OBJECTIVE: Circulating levels of endostatin are elevated in many underlying conditions leading to heart failure such as hypertension, diabetes, chronic kidney disease and ischemic heart disease. Yet, the association between endostatin and the incidence of heart failure has not been reported previously in the community.DESIGN: We investigated the longitudinal association between serum endostatin levels and incident heart failure in two community-based cohorts of elderly: Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS, n=966; mean age 70 years, 51% women, 81 events, mean follow-up 10 years) and Uppsala Longitudinal Study of Adult Men (ULSAM, n=747 men; mean age 78 years, 98 heart failure events, mean follow-up 8 years). We also investigated the cross-sectional association between endostatin and echocardiographic left ventricular systolic function and diastolic function (ejection fraction and E/A-ratio, respectively).RESULTS: Higher serum endostatin was associated with an increased risk for heart failure in both cohorts after adjustment for established heart failure risk factors, glomerular filtration rate and N-terminal pro-brain natriuretic peptide (NT-proBNP) (PIVUS: multivariable hazard ratio (HR) per 1-standard deviation (SD) increase, HR 1.46 (95%CI, 1.17-1.82, p<.001); ULSAM: HR 1.29 (95%CI, 1.00-1.68, p<.05). In cross-sectional analyses at baseline, higher endostatin was significantly associated with both worsened left ventricular systolic and diastolic function in both cohorts. Conclusion Higher serum endostatin was associated with left ventricular dysfunction and an increased heart failure risk in two community-based cohorts of elderly. Our findings encourage further experimental studies that investigate the role of endostatin in the development of heart failure.

    View details for DOI 10.1080/14017431.2018.1483080

    View details for PubMedID 29893146

  • Glucose challenge metabolomics implicates medium-chain acylcarnitines in insulin resistance SCIENTIFIC REPORTS Nowak, C., Hetty, S., Salihovic, S., Castillejo-Lopez, C., Ganna, A., Cook, N. L., Broeckling, C. D., Prenni, J. E., Shen, X., Giedraitis, V., Arnlov, J., Lind, L., Berne, C., Sundstrom, J., Fall, T., Ingelsson, E. 2018; 8: 8691

    Abstract

    Insulin resistance (IR) predisposes to type 2 diabetes and cardiovascular disease but its causes are incompletely understood. Metabolic challenges like the oral glucose tolerance test (OGTT) can reveal pathogenic mechanisms. We aimed to discover associations of IR with metabolite trajectories during OGTT. In 470 non-diabetic men (age 70.6 ± 0.6 years), plasma samples obtained at 0, 30 and 120 minutes during an OGTT were analyzed by untargeted liquid chromatography-mass spectrometry metabolomics. IR was assessed with the hyperinsulinemic-euglycemic clamp method. We applied age-adjusted linear regression to identify metabolites whose concentration change was related to IR. Nine trajectories, including monounsaturated fatty acids, lysophosphatidylethanolamines and a bile acid, were significantly associated with IR, with the strongest associations observed for medium-chain acylcarnitines C10 and C12, and no associations with L-carnitine or C2-, C8-, C14- or C16-carnitine. Concentrations of C10- and C12-carnitine decreased during OGTT with a blunted decline in participants with worse insulin resistance. Associations persisted after adjustment for obesity, fasting insulin and fasting glucose. In mouse 3T3-L1 adipocytes exposed to different acylcarnitines, we observed blunted insulin-stimulated glucose uptake after treatment with C10- or C12-carnitine. In conclusion, our results identify medium-chain acylcarnitines as possible contributors to IR.

    View details for DOI 10.1038/s41598-018-26701-0

    View details for Web of Science ID 000434252600004

    View details for PubMedID 29875472

    View details for PubMedCentralID PMC5989236

  • Human Genetics of Obesity and Type 2 Diabetes Mellitus: Past, Present, and Future. Circulation. Genomic and precision medicine Ingelsson, E., McCarthy, M. I. 2018; 11 (6): e002090

    Abstract

    Type 2 diabetes mellitus (T2D) and obesity already represent 2 of the most prominent risk factors for cardiovascular disease, and are destined to increase in importance given the global changes in lifestyle. Ten years have passed since the first round of genome-wide association studies for T2D and obesity. During this decade, we have witnessed remarkable developments in human genetics. We have graduated from the despair of candidate gene-based studies that generated few consistently replicated genotype-phenotype associations, to the excitement of an exponential harvest of loci robustly associated with medical outcomes through ever larger genome-wide association study meta-analyses. As well as discovering hundreds of loci, genome-wide association studies have provided transformative insights into the genetic architecture of T2D and other complex traits, highlighting the extent of polygenicity and the tiny effect sizes of many common risk alleles. Genome-wide association studies have also provided a critical starting point for discovering new biology relevant to these traits. Expectations are high that these discoveries will foster development of more effective strategies for intervention, through optimization of precision medicine approaches. In this article, we review current knowledge and provide suggestions for the next steps in genetic research for T2D and obesity. We focus on four areas relevant to precision medicine: genetic architecture, pharmacogenetics and other gene-environment interactions, mechanistic inference, and drug development. As we describe, the genetic architecture of complex traits has major implications for the prospects of precision medicine, rendering some anticipated approaches decidedly unrealistic. We highlight obstacles to the translation of human genetic findings into mechanism inference but are optimistic that, as these are overcome, there is untapped potential for novel drugs and more effective strategies for treating and preventing T2D and obesity.

    View details for DOI 10.1161/CIRCGEN.118.002090

    View details for PubMedID 29899044

  • Clinical and genetic determinants of varicose veins: a prospective, community-based prospective study of similar to 500,000 individuals Fukaya, E., Flores, A., Lindholm, D., Gustafsson, S., Ingelsson, E., Leeper, N. SAGE PUBLICATIONS LTD. 2018: 300
  • A genome-wide association study of IgM antibody against phosphorylcholine: shared genetics and phenotypic relationship to chronic lymphocytic leukemia HUMAN MOLECULAR GENETICS Chen, X., Gustafsson, S., Whitington, T., Borne, Y., Lorentzen, E., Sun, J., Almgren, P., Su, J., Karlsson, R., Song, J., Lu, Y., Zhan, Y., Hagg, S., Svensson, P., Smedby, K. E., Slager, S. L., Ingelsson, E., Lindgren, C. M., Morris, A. P., Melander, O., Karlsson, T., de Faire, U., Caidahl, K., Engstrom, G., Lind, L., Karlsson, M. I., Pedersen, N. L., Frostegard, J., Magnusson, P. E. 2018; 27 (10): 1809–18

    Abstract

    Phosphorylcholine (PC) is an epitope on oxidized low-density lipoprotein (oxLDL), apoptotic cells and several pathogens like Streptococcus pneumoniae. Immunoglobulin M against PC (IgM anti-PC) has the ability to inhibit uptake of oxLDL by macrophages and increase clearance of apoptotic cells. From our genome-wide association studies (GWASs) in four European-ancestry cohorts, six single nucleotide polymorphisms (SNPs) in 11q24.1 were discovered (in 3002 individuals) and replicated (in 646 individuals) to be associated with serum level of IgM anti-PC (the leading SNP rs35923643-G, combined β = 0.19, 95% confidence interval 0.13-0.24, P = 4.3 × 10-11). The haplotype tagged by rs35923643-G (or its proxy SNP rs735665-A) is also known as the top risk allele for chronic lymphocytic leukemia (CLL), and a main increasing allele for general IgM. By using summary GWAS results of IgM anti-PC and CLL in the polygenic risk score (PRS) analysis, PRS on the basis of IgM anti-PC risk alleles positively associated with CLL risk (explained 0.6% of CLL variance, P = 1.2 × 10-15). Functional prediction suggested that rs35923643-G might impede the binding of Runt-related transcription factor 3, a tumor suppressor playing a central role in the immune regulation of cancers. Contrary to the expectations from the shared genetics between IgM anti-PC and CLL, an inverse relationship at the phenotypic level was found in a nested case-control study (30 CLL cases with 90 age- and sex-matched controls), potentially reflecting reverse causation. The suggested function of the top variant as well as the phenotypic association between IgM anti-PC and CLL risk needs replication and motivates further studies.

    View details for DOI 10.1093/hmg/ddy094

    View details for Web of Science ID 000431886200012

    View details for PubMedID 29547969

  • Habitual coffee consumption and cognitive function: a Mendelian randomization meta-analysis in up to 415,530 participants SCIENTIFIC REPORTS Zhou, A., Taylor, A. E., Karhunen, V., Zhan, Y., Rovio, S. P., Lahti, J., Sjogren, P., Byberg, L., Lyall, D. M., Auvinen, J., Lehtimaki, T., Kahonen, M., Hutri-Kahonen, N., Perala, M., Michaelsson, K., Mahajan, A., Lind, L., Power, C., Eriksson, J. G., Raitakari, O. T., Hagg, S., Pedersen, N. L., Veijola, J., Jarvelin, M., Munafo, M. R., Ingelsson, E., Llewellyn, D. J., Hypponen, E. 2018; 8: 7526

    Abstract

    Coffee's long-term effect on cognitive function remains unclear with studies suggesting both benefits and adverse effects. We used Mendelian randomization to investigate the causal relationship between habitual coffee consumption and cognitive function in mid- to later life. This included up to 415,530 participants and 300,760 coffee drinkers from 10 meta-analysed European ancestry cohorts. In each cohort, composite cognitive scores that capture global cognition and memory were computed using available tests. A genetic score derived using CYP1A1/2 (rs2472297) and AHR (rs6968865) was chosen as a proxy for habitual coffee consumption. Null associations were observed when examining the associations of the genetic score with global and memory cognition (β = -0.0007, 95% C.I. -0.009 to 0.008, P = 0.87; β = -0.001, 95% C.I. -0.005 to 0.002, P = 0.51, respectively), with high consistency between studies (Pheterogeneity > 0.4 for both). Domain specific analyses using available cognitive measures in the UK Biobank also did not support effects by habitual coffee intake for reaction time, pairs matching, reasoning or prospective memory (P ≥ 0.05 for all). Despite the power to detect very small effects, our meta-analysis provided no evidence for causal long-term effects of habitual coffee consumption on global cognition or memory.

    View details for DOI 10.1038/s41598-018-25919-2

    View details for Web of Science ID 000431958000003

    View details for PubMedID 29760501

    View details for PubMedCentralID PMC5951917

  • Biological Insights Into Muscular Strength: Genetic Findings in the UK Biobank SCIENTIFIC REPORTS Tikkanen, E., Gustafsson, S., Amar, D., Shcherbina, A., Waggott, D., Ashley, E. A., Ingelsson, E. 2018; 8: 6451

    Abstract

    We performed a large genome-wide association study to discover genetic variation associated with muscular strength, and to evaluate shared genetic aetiology with and causal effects of muscular strength on several health indicators. In our discovery analysis of 223,315 individuals, we identified 101 loci associated with grip strength (P <5 × 10-8). Of these, 64 were associated (P < 0.01 and consistent direction) also in the replication dataset (N = 111,610). eQTL analyses highlighted several genes known to play a role in neuro-developmental disorders or brain function, and the results from meta-analysis showed a significant enrichment of gene expression of brain-related transcripts. Further, we observed inverse genetic correlations of grip strength with cardiometabolic traits, and positive correlation with parents' age of death and education. We also showed that grip strength had shared biological pathways with indicators of frailty, including cognitive performance scores. By use of Mendelian randomization, we provide evidence that higher grip strength is protective of both coronary heart disease (OR = 0.69, 95% CI 0.60-0.79, P < 0.0001) and atrial fibrillation (OR = 0.75, 95% CI 0.62-0.90, P = 0.003). In conclusion, our results show shared genetic aetiology between grip strength, and cardiometabolic and cognitive health; and suggest that maintaining muscular strength could prevent future cardiovascular events.

    View details for DOI 10.1038/s41598-018-24735-y

    View details for Web of Science ID 000430661200018

    View details for PubMedID 29691431

    View details for PubMedCentralID PMC5915424

  • Medical relevance of protein-truncating variants across 337,205 individuals in the UK Biobank study NATURE COMMUNICATIONS DeBoever, C., Tanigawa, Y., Lindholm, M. E., McInnes, G., Lavertu, A., Ingelsson, E., Chang, C., Ashley, E. A., Bustamante, C. D., Daly, M. J., Rivas, M. A. 2018; 9: 1612

    Abstract

    Protein-truncating variants can have profound effects on gene function and are critical for clinical genome interpretation and generating therapeutic hypotheses, but their relevance to medical phenotypes has not been systematically assessed. Here, we characterize the effect of 18,228 protein-truncating variants across 135 phenotypes from the UK Biobank and find 27 associations between medical phenotypes and protein-truncating variants in genes outside the major histocompatibility complex. We perform phenome-wide analyses and directly measure the effect in homozygous carriers, commonly referred to as "human knockouts," across medical phenotypes for genes implicated as being protective against disease or associated with at least one phenotype in our study. We find several genes with strong pleiotropic or non-additive effects. Our results illustrate the importance of protein-truncating variants in a variety of diseases.

    View details for DOI 10.1038/s41467-018-03910-9

    View details for Web of Science ID 000430674000001

    View details for PubMedID 29691392

    View details for PubMedCentralID PMC5915386

  • Associations of Fitness, Physical Activity, Strength, and Genetic Risk With Cardiovascular Disease: Longitudinal Analyses in the UK Biobank Study. Circulation Tikkanen, E., Gustafsson, S., Ingelsson, E. 2018

    Abstract

    Background -Observational studies have shown inverse associations among fitness, physical activity, and cardiovascular disease. However, little is known about these associations in individuals with elevated genetic susceptibility for these diseases. Methods -We estimated associations of grip strength, objective and subjective physical activity, and cardiorespiratory fitness with cardiovascular events and all-cause death in a large cohort of 502635 individuals from the UK Biobank (median follow-up, 6.1 years; interquartile range, 5.4-6.8 years). Then we further examined these associations in individuals with different genetic burden by stratifying individuals based on their genetic risk scores for coronary heart disease and atrial fibrillation. We compared disease risk among individuals in different tertiles of fitness, physical activity, and genetic risk using lowest tertiles as reference. Results -Grip strength, physical activity, and cardiorespiratory fitness showed inverse associations with incident cardiovascular events (coronary heart disease: hazard ratio [HR], 0.79; 95% confidence interval [CI], 0.77- 0.81; HR, 0.95; 95% CI, 0.93-0.97; and HR, 0.68; 95% CI, 0.63-0.74, per SD change, respectively; atrial fibrillation: HR, 0.75; 95% CI, 0.73- 0.76; HR, 0.93; 95% CI, 0.91-0.95; and HR, 0.60; 95% CI, 0.56-0.65, per SD change, respectively). Higher grip strength and cardiorespiratory fitness were associated with lower risk of incident coronary heart disease and atrial fibrillation in each genetic risk score group (Ptrend <0.001 in each genetic risk category). In particular, high levels of cardiorespiratory fitness were associated with 49% lower risk for coronary heart disease (HR, 0.51; 95% CI, 0.38-0.69) and 60% lower risk for atrial fibrillation (HR, 0.40; 95%, CI 0.30-0.55) among individuals at high genetic risk for these diseases. Conclusions - Fitness and physical activity demonstrated inverse associations with incident cardiovascular disease in the general population, as well as in individuals with elevated genetic risk for these diseases.

    View details for DOI 10.1161/CIRCULATIONAHA.117.032432

    View details for PubMedID 29632216

  • Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes NATURE GENETICS Malik, R., Chauhan, G., Traylor, M., Sargurupremraj, M., Okada, Y., Mishra, A., Rutten-Jacobs, L., Giese, A., van der Laan, S. W., Gretarsdottir, S., Anderson, C. D., Chong, M., Adams, H. H., Ago, T., Almgren, P., Amouyel, P., Ay, H., Bartz, T. M., Benavente, O. R., Bevan, S., Boncoraglio, G. B., Brown, R. D., Butterworth, A. S., Carrera, C., Carty, C. L., Chasman, D. I., Chen, W., Cole, J. W., Correa, A., Cotlarciuc, I., Cruchaga, C., Danesh, J., de Bakker, P. W., DeStefano, A. L., den Hoed, M., Duan, Q., Engelter, S. T., Falcone, G. J., Gottesman, R. F., Grewal, R. P., Gudnason, V., Gustafsson, S., Haessler, J., Harris, T. B., Hassan, A., Havulinna, A. S., Heckbert, S. R., Holliday, E. G., Howard, G., Hsu, F., Hyacinth, H. I., Ikram, M., Ingelsson, E., Irvin, M. R., Jian, X., Jimenez-Conde, J., Johnson, J. A., Jukema, J., Kanai, M., Keene, K. L., Kissela, B. M., Kleindorfer, D. O., Kooperberg, C., Kubo, M., Lange, L. A., Langefeld, C. D., Langenberg, C., Launer, L. J., Lee, J., Lemmens, R., Leys, D., Lewis, C. M., Lin, W., Lindgren, A. G., Lorentzen, E., Magnusson, P. K., Maguire, J., Manichaikul, A., McArdle, P. F., Meschia, J. F., Mitchell, B. D., Mosley, T. H., Nalls, M. A., Ninomiya, T., O'Donnell, M. J., Psaty, B. M., Pulit, S. L., Rannikmae, K., Reiner, A. P., Rexrode, K. M., Rice, K., Rich, S. S., Ridker, P. M., Rost, N. S., Rothwell, P. M., Rotter, J. I., Rundek, T., Sacco, R. L., Sakaue, S., Sale, M. M., Salomaa, V., Sapkota, B. R., Schmidt, R., Schmidt, C. O., Schminke, U., Sharma, P., Slowik, A., Sudlow, C. M., Tanislav, C., Tatlisumak, T., Taylor, K. D., Thijs, V. S., Thorleifsson, G., Thorsteinsdottir, U., Tiedt, S., Trompet, S., Tzourio, C., van Duijn, C. M., Walters, M., Wareham, N. J., Wassertheil-Smoller, S., Wilson, J. G., Wiggins, K. L., Yang, Q., Yusuf, S., Bis, J. C., Pastinen, T., Ruusalepp, A., Schadt, E. E., Koplev, S., Bjorkegren, J. M., Codoni, V., Civelek, M., Smith, N. L., Tregouet, D. A., Christophersen, I. E., Roselli, C., Lubitz, S. A., Ellinor, P. T., Tai, E., Kooner, J. S., Kato, N., He, J., van der Harst, P., Elliott, P., Chambers, J. C., Takeuchi, F., Johnson, A. D., Sanghera, D. K., Melander, O., Jern, C., Strbian, D., Fernandez-Cadenas, I., Longstreth, W. T., Rolfs, A., Hata, J., Woo, D., Rosand, J., Pare, G., Hopewell, J. C., Saleheen, D., Stefansson, K., Worrall, B. B., Kittner, S. J., Seshadri, S., Fornage, M., Markus, H. S., Howson, J. M., Kamatani, Y., Debette, S., Dichgans, M., AFGen Consortium, Cohorts Heart Aging Res Genomic, Int Genomics Blood Pressure iGEN, INVENT Consortium, STARNET, BioBank Japan Cooperative Hosp Grp, COMPASS Consortium, EPIC-CVD Consortium, EPIC-InterAct Consortium, Int Stroke Genetics Consortium ISG, METASTROKE Consortium, Neurology Working Grp Charge Con, NINDS Stroke Genetics Network Si, UK Young Lacunar DNA Study, MEGASTROKE Consortium 2018; 50 (4): 524-+

    Abstract

    Stroke has multiple etiologies, but the underlying genes and pathways are largely unknown. We conducted a multiancestry genome-wide-association meta-analysis in 521,612 individuals (67,162 cases and 454,450 controls) and discovered 22 new stroke risk loci, bringing the total to 32. We further found shared genetic variation with related vascular traits, including blood pressure, cardiac traits, and venous thromboembolism, at individual loci (n = 18), and using genetic risk scores and linkage-disequilibrium-score regression. Several loci exhibited distinct association and pleiotropy patterns for etiological stroke subtypes. Eleven new susceptibility loci indicate mechanisms not previously implicated in stroke pathophysiology, with prioritization of risk variants and genes accomplished through bioinformatics analyses using extensive functional datasets. Stroke risk loci were significantly enriched in drug targets for antithrombotic therapy.

    View details for DOI 10.1038/s41588-018-0058-3

    View details for Web of Science ID 000429529300013

    View details for PubMedID 29531354

    View details for PubMedCentralID PMC5968830

  • Refining the accuracy of validated target identification through coding variant fine-mapping in type 2 diabetes NATURE GENETICS Mahajan, A., Wessel, J., Willems, S. M., Zhao, W., Robertson, N. R., Chu, A. Y., Gan, W., Kitajima, H., Taliun, D., Rayner, N., Guo, X., Lu, Y., Li, M., Jensen, R. A., Hu, Y., Huo, S., Lohman, K. K., Zhang, W., Cook, J. P., Prins, B., Flannick, J., Grarup, N., Trubetskoy, V., Kravic, J., Kim, Y., Rybin, D. V., Yaghootkar, H., Mueller-Nurasyid, M., Meidtner, K., Li-Gao, R., Varga, T. V., Marten, J., Li, J., Smith, A., An, P., Ligthart, S., Gustafsson, S., Malerba, G., Demirkan, A., Tajes, J., Steinthorsdottir, V., Wuttke, M., Lecoeur, C., Preuss, M., Bielak, L. F., Graff, M., Highland, H. M., Justice, A. E., Liu, D. J., Marouli, E., Peloso, G., Warren, H. R., Afaq, S., Afzal, S., Ahlqvist, E., Almgren, P., Amin, N., Bang, L. B., Bertoni, A. G., Bombieri, C., Bork-Jensen, J., Brandslund, I., Brody, J. A., Burtt, N. P., Canouil, M., Chen, Y., Cho, Y., Christensen, C., Eastwood, S. V., Eckardt, K., Fischer, K., Gambaro, G., Giedraitis, V., Grove, M. L., de Haan, H. G., Hackinger, S., Hai, Y., Han, S., Tybjaerg-Hansen, A., Hivert, M., Isomaa, B., Jager, S., Jorgensen, M. E., Jorgensen, T., Karajamaki, A., Kim, B., Kim, S., Koistinen, H. A., Kovacs, P., Kriebel, J., Kronenberg, F., Lall, K., Lange, L. A., Lee, J., Lehne, B., Li, H., Lin, K., Linneberg, A., Liu, C., Liu, J., Loh, M., Magi, R., Mamakou, V., McKean-Cowdin, R., Nadkarni, G., Neville, M., Nielsen, S. F., Ntalla, I., Peyser, P. A., Rathmann, W., Rice, K., Rich, S. S., Rode, L., Rolandsson, O., Schonherr, S., Selvin, E., Small, K. S., Stancakova, A., Surendran, P., Taylor, K. D., Teslovich, T. M., Thorand, B., Thorleifsson, G., Tin, A., Tonjes, A., Varbo, A., Witte, D. R., Wood, A. R., Yajnik, P., Yao, J., Yengo, L., Young, R., Amouyel, P., Boeing, H., Boerwinkle, E., Bottinger, E. P., Chowdhury, R., Collins, F. S., Dedoussis, G., Dehghan, A., Deloukas, P., Ferrario, M. M., Ferrieres, J., Florez, J. C., Frossard, P., Gudnason, V., Harris, T. B., Heckbert, S. R., Howson, J. M., Ingelsson, M., Kathiresan, S., Kee, F., Kuusisto, J., Langenberg, C., Launer, L. J., Lindgren, C. M., Mannisto, S., Meitinger, T., Melander, O., Mohlke, K. L., Moitry, M., Morris, A. D., Murray, A. D., de Mutsert, R., Orho-Melander, M., Owen, K. R., Perola, M., Peters, A., Province, M. A., Rasheed, A., Ridker, P. M., Rivadineira, F., Rosendaal, F. R., Rosengren, A. H., Salomaa, V., Sheu, W., Sladek, R., Smith, B. H., Strauch, K., Uitterlinden, A. G., Varma, R., Willer, C. J., Bluher, M., Butterworth, A. S., Chambers, J., Chasman, D. I., Danesh, J., van Duijn, C., Dupuis, J., Franco, O. H., Franks, P. W., Froguel, P., Grallert, H., Groop, L., Han, B., Hansen, T., Hattersley, A. T., Hayward, C., Ingelsson, E., Kardia, S. R., Karpe, F., Kooner, J., Kottgen, A., Kuulasmaa, K., Laakso, M., Lin, X., Lind, L., Liu, Y., Loos, R. F., Marchini, J., Metspalu, A., Mook-Kanamori, D., Nordestgaard, B. G., Palmer, C. A., Pankow, J. S., Pedersen, O., Psaty, B. M., Rauramaa, R., Sattar, N., Schulze, M. B., Soranzo, N., Spector, T. D., Stefansson, K., Stumvoll, M., Thorsteinsdottir, U., Tuomi, T., Tuomilehto, J., Wareham, N. J., Wilson, J. G., Zeggini, E., Scott, R. A., Barroso, I., Frayling, T. M., Goodarzi, M. O., Meigs, J. B., Boehnke, M., Saleheen, D., Morris, A. P., Rotter, J. I., McCarthy, M. I., ExomeBP Consortium, MAGIC Consortium, GIANT Consortium 2018; 50 (4): 559-+

    Abstract

    We aggregated coding variant data for 81,412 type 2 diabetes cases and 370,832 controls of diverse ancestry, identifying 40 coding variant association signals (P < 2.2 × 10-7); of these, 16 map outside known risk-associated loci. We make two important observations. First, only five of these signals are driven by low-frequency variants: even for these, effect sizes are modest (odds ratio ≤1.29). Second, when we used large-scale genome-wide association data to fine-map the associated variants in their regional context, accounting for the global enrichment of complex trait associations in coding sequence, compelling evidence for coding variant causality was obtained for only 16 signals. At 13 others, the associated coding variants clearly represent 'false leads' with potential to generate erroneous mechanistic inference. Coding variant associations offer a direct route to biological insight for complex diseases and identification of validated therapeutic targets; however, appropriate mechanistic inference requires careful specification of their causal contribution to disease predisposition.

    View details for DOI 10.1038/s41588-018-0084-1

    View details for Web of Science ID 000429529300016

    View details for PubMedID 29632382

    View details for PubMedCentralID PMC5898373

  • CONGENITAL HEART DISEASE CONFERS SUBSTANTIAL RISK OF ACQUIRED CARDIOVASCULAR DISEASE AMONGST BRITISH ADULTS Saha, P., Potiny, P., Tcheandjieu, C., Fernandes, S. M., Romfh, A., Bernstein, D., Lui, G. K., Ingelsson, E., Priest, J. ELSEVIER SCIENCE INC. 2018: 553
  • Methylation-based estimated biological age and cardiovascular disease EUROPEAN JOURNAL OF CLINICAL INVESTIGATION Lind, L., Ingelsson, E., Sundstrom, J., Siegbahn, A., Lampa, E. 2018; 48 (2)

    View details for DOI 10.1111/eci.12872

    View details for Web of Science ID 000429997100009

  • Genome-wide association study in 79,366 European-ancestry individuals informs the genetic architecture of 25-hydroxyvitamin D levels NATURE COMMUNICATIONS Jiang, X., O'Reilly, P. F., Aschard, H., Hsu, Y., Richards, J., Dupuis, J., Ingelsson, E., Karasik, D., Pilz, S., Berry, D., Kestenbaum, B., Zheng, J., Luan, J., Sofianopoulou, E., Streeten, E. A., Albanes, D., Lutsey, P. L., Yao, L., Tang, W., Econs, M. J., Wallaschofski, H., Voelzke, H., Zhou, A., Power, C., McCarthy, M. I., Michos, E. D., Boerwinkle, E., Weinstein, S. J., Freedman, N. D., Huang, W., Van Schoor, N. M., van der Velde, N., de Groot, L. M., Enneman, A., Cupples, L., Booth, S. L., Vasan, R. S., Liu, C., Zhou, Y., Ripatti, S., Ohlsson, C., Vandenput, L., Lorentzon, M., Eriksson, J. G., Shea, M., Houston, D. K., Kritchevsky, S. B., Liu, Y., Lohman, K. K., Ferrucci, L., Peacock, M., Gieger, C., Beekman, M., Slagboom, E., Deelen, J., van Heemst, D., Kleber, M. E., Maerz, W., de Boer, I. H., Wood, A. C., Rotter, J. I., Rich, S. S., Robinson-Cohen, C., den Heijer, M., Jarvelin, M., Cavadino, A., Joshi, P. K., Wilson, J. F., Hayward, C., Lind, L., Michaelsson, K., Trompet, S., Zillikens, M., Uitterlinden, A. G., Rivadeneira, F., Broer, L., Zgaga, L., Campbell, H., Theodoratou, E., Farrington, S. M., Timofeeva, M., Dunlop, M. G., Valdes, A. M., Tikkanen, E., Lehtimaki, T., Lyytikainen, L., Kahonen, M., Raitakari, O. T., Mikkila, V., Ikram, M., Sattar, N., Jukema, J., Wareham, N. J., Langenberg, C., Forouhi, N. G., Gundersen, T. E., Khaw, K., Butterworth, A. S., Danesh, J., Spector, T., Wang, T. J., Hypponen, E., Kraft, P., Kiel, D. P. 2018; 9: 260

    Abstract

    Vitamin D is a steroid hormone precursor that is associated with a range of human traits and diseases. Previous GWAS of serum 25-hydroxyvitamin D concentrations have identified four genome-wide significant loci (GC, NADSYN1/DHCR7, CYP2R1, CYP24A1). In this study, we expand the previous SUNLIGHT Consortium GWAS discovery sample size from 16,125 to 79,366 (all European descent). This larger GWAS yields two additional loci harboring genome-wide significant variants (P = 4.7×10-9 at rs8018720 in SEC23A, and P = 1.9×10-14 at rs10745742 in AMDHD1). The overall estimate of heritability of 25-hydroxyvitamin D serum concentrations attributable to GWAS common SNPs is 7.5%, with statistically significant loci explaining 38% of this total. Further investigation identifies signal enrichment in immune and hematopoietic tissues, and clustering with autoimmune diseases in cell-type-specific analysis. Larger studies are required to identify additional common SNPs, and to explore the role of rare or structural variants and gene-gene interactions in the heritability of circulating 25-hydroxyvitamin D levels.

    View details for DOI 10.1038/s41467-017-02662-2

    View details for Web of Science ID 000422650500011

    View details for PubMedID 29343764

    View details for PubMedCentralID PMC5772647

  • Genome-wide Study of Atrial Fibrillation Identifies Seven Risk Loci and Highlights Biological Pathways and Regulatory Elements Involved in Cardiac Development AMERICAN JOURNAL OF HUMAN GENETICS Nielsen, J. B., Fritsche, L. G., Zhou, W., Teslovich, T. M., Holmen, O. L., Gustafsson, S., Gabrielsen, M. E., Schmidt, E. M., Beaumont, R., Wolford, B. N., Lin, M., Brummett, C. M., Preuss, M. H., Refsgaard, L., Bottinger, E. P., Graham, S. E., Surakka, I., Chu, Y., Skogholt, A., Dalen, H., Boyle, A. P., Oral, H., Herron, T. J., Kitzman, J., Jalife, J., Svendsen, J. H., Olesen, M. S., Njolstad, I., Lochen, M., Baras, A., Gottesman, O., Marcketta, A., O'Dushlaine, C., Ritchie, M. D., Wilsgaard, T., Loos, R. F., Frayling, T. M., Boehnke, M., Ingelsson, E., Carey, D. J., Dewey, F. E., Kang, H. M., Abecasis, G. R., Hveem, K., Willer, C. J. 2018; 102 (1): 103–15

    Abstract

    Atrial fibrillation (AF) is a common cardiac arrhythmia and a major risk factor for stroke, heart failure, and premature death. The pathogenesis of AF remains poorly understood, which contributes to the current lack of highly effective treatments. To understand the genetic variation and biology underlying AF, we undertook a genome-wide association study (GWAS) of 6,337 AF individuals and 61,607 AF-free individuals from Norway, including replication in an additional 30,679 AF individuals and 278,895 AF-free individuals. Through genotyping and dense imputation mapping from whole-genome sequencing, we tested almost nine million genetic variants across the genome and identified seven risk loci, including two novel loci. One novel locus (lead single-nucleotide variant [SNV] rs12614435; p = 6.76 × 10-18) comprised intronic and several highly correlated missense variants situated in the I-, A-, and M-bands of titin, which is the largest protein in humans and responsible for the passive elasticity of heart and skeletal muscle. The other novel locus (lead SNV rs56202902; p = 1.54 × 10-11) covered a large, gene-dense chromosome 1 region that has previously been linked to cardiac conduction. Pathway and functional enrichment analyses suggested that many AF-associated genetic variants act through a mechanism of impaired muscle cell differentiation and tissue formation during fetal heart development.

    View details for DOI 10.1016/j.ajhg.2017.12.003

    View details for Web of Science ID 000419305500008

    View details for PubMedID 29290336

    View details for PubMedCentralID PMC5777936

  • Circulating proteins as predictors of incident heart failure in the elderly EUROPEAN JOURNAL OF HEART FAILURE Stenemo, M., Nowak, C., Byberg, L., Sundstrom, J., Giedraitis, V., Lind, L., Ingelsson, E., Fall, T., Arnlov, J. 2018; 20 (1): 55–62

    Abstract

    To identify novel risk markers for incident heart failure using proteomic profiling of 80 proteins previously associated with cardiovascular pathology.Proteomic profiling (proximity extension assay) was performed in two community-based prospective cohorts of elderly individuals without heart failure at baseline: the Prospective Investigation of the Vasculature in Uppsala Seniors [PIVUS, n = 901, median age 70.2 (interquartile range 70.0-70.3) years, 80 events]; and the Uppsala Longitudinal Study of Adult Men [ULSAM, n = 685, median age 77.8 (interquartile range 76.9-78.1) years, 90 events]. Twenty-nine proteins were associated with incident heart failure in the discovery cohort PIVUS after adjustment for age and sex, and correction for multiple testing. Eighteen associations replicated in ULSAM. In pooled analysis of both cohorts, higher levels of nine proteins were associated with incident heart failure after adjustment for established risk factors: growth differentiation factor 15 (GDF-15), T-cell immunoglobulin and mucin domain 1 (TIM-1), tumour necrosis factor-related apoptosis-inducing ligand receptor 2 (TRAIL-R2), spondin-1 (SPON1), matrix metalloproteinase-12 (MMP-12), follistatin (FS), urokinase-type plasminogen activator surface receptor (U-PAR), osteoprotegerin (OPG), and suppression of tumorigenicity 2 (ST2). Of these, GDF-15, U-PAR, MMP-12, TRAIL-R2, SPON1 and FS were associated with worsened echocardiographic left ventricular systolic function at baseline, while only TIM-1 was positively associated with worsened diastolic function (P < 0.02 for all).Proteomic profiling identified several novel associations between proteins involved in apoptosis, inflammation, matrix remodelling, and fibrinolysis with incident heart failure in elderly individuals. Our results encourage additional studies investigating the underlying mechanisms and the clinical utility of our findings.

    View details for DOI 10.1002/ejhf.980

    View details for Web of Science ID 000423809700007

    View details for PubMedID 28967680

  • Role of peroxisome proliferator-activated receptor gamma Pro12Ala polymorphism in human adipose tissue: assessment of adipogenesis and adipocyte glucose and lipid turnover ADIPOCYTE Kamble, P. G., Pereira, M. J., Gustafsson, S., Lundkvist, P., Castillejo-Lopez, C., Fall, T., Ingelsson, E., Eriksson, J. W. 2018; 7 (4): 285–96

    Abstract

    The protective mechanisms of peroxisome proliferator-activated receptor gamma (PPARγ) Pro12Ala polymorphism in type 2 diabetes (T2D) are unclear. We obtained subcutaneous adipose tissue (AT) before and 3 h after oral glucose (OGTT) in carriers and non-carriers of the Ala allele (12 Pro/Pro, 15 Pro/Ala, and 13 Ala/Ala). Adipogenesis, adipocyte glucose uptake and lipolysis as well as PPARγ target gene expression were investigated and compared between the genotype groups. During fasting and post-OGTT, neither basal nor insulin-stimulated adipocyte glucose uptake differed between genotypes. Compared to fasting, a decreased hormone-sensitive lipase gene expression in Pro/Pro (p < 0.05) was accompanied with a higher antilipolytic effect of insulin post-OGTT (p < 0.01). The adipocyte size was similar across groups. Preadipocyte differentiation rates between Pro/Pro and Ala/Ala were unchanged. In conclusion, no major differences in AT differentiation, glucose uptake, lipolysis or expression of PPARγ target genes were observed between different PPARγ Pro12Ala genotypes. Albeit small, our study may suggest that other pathways in AT or effects exerted in other tissues might contribute to the Pro12Ala-mediated protection against T2D.

    View details for DOI 10.1080/21623945.2018.1503030

    View details for Web of Science ID 000450440700007

    View details for PubMedID 30064293

  • Birthweight, Type 2 Diabetes Mellitus, and Cardiovascular Disease: Addressing the Barker Hypothesis With Mendelian Randomization. Circulation. Genomic and precision medicine Zanetti, D., Tikkanen, E., Gustafsson, S., Priest, J. R., Burgess, S., Ingelsson, E. 2018; 11 (6): e002054

    Abstract

    Low birthweight has been associated with a higher risk of hypertension, type 2 diabetes mellitus (T2D), and cardiovascular disease. The Barker hypothesis posits that intrauterine growth restriction resulting in lower birthweight is causal for these diseases, but causality is difficult to infer from observational studies.We performed regression analyses to assess associations of birthweight with cardiovascular disease and T2D in 237 631 individuals from the UK Biobank. Further, we assessed the causal relationship of such associations using Mendelian randomization.In the observational analyses, birthweight showed inverse associations with systolic and diastolic blood pressure (β, -0.83 and -0.26; per raw unit in outcomes and SD change in birthweight; 95% confidence interval [CI], -0.90 to -0.75 and -0.31 to -0.22, respectively), T2D (odds ratio, 0.83; 95% CI, 0.79-0.87), lipid-lowering treatment (odds ratio, 0.84; 95% CI, 0.81-0.86), and coronary artery disease (hazard ratio, 0.85; 95% CI, 0.78-0.94), whereas the associations with adult body mass index and body fat (β, 0.04 and 0.02; per SD change in outcomes and birthweight; 95% CI, 0.03-0.04 and 0.01-0.02, respectively) were positive. The Mendelian randomization analyses indicated inverse causal associations of birthweight with low-density lipoprotein cholesterol, 2-hour glucose, coronary artery disease, and T2D and positive causal association with body mass index but no associations with blood pressure.Our study indicates that lower birthweight, used as a proxy for intrauterine growth retardation, is causally related with increased susceptibility to coronary artery disease and T2D. This causal relationship is not mediated by adult obesity or hypertension.

    View details for DOI 10.1161/CIRCGEN.117.002054

    View details for PubMedID 29875125

  • Genetic Regulatory Mechanisms of Smooth Muscle Cells Map to Coronary Artery Disease Risk Loci. American journal of human genetics Liu, B., Pjanic, M., Wang, T., Nguyen, T., Gloudemans, M., Rao, A., Castano, V. G., Nurnberg, S., Rader, D. J., Elwyn, S., Ingelsson, E., Montgomery, S. B., Miller, C. L., Quertermous, T. 2018

    Abstract

    Coronary artery disease (CAD) is the leading cause of death globally. Genome-wide association studies (GWASs) have identified more than 95 independent loci that influence CAD risk, most of which reside in non-coding regions of the genome. To interpret these loci, we generated transcriptome and whole-genome datasets using human coronary artery smooth muscle cells (HCASMCs) from 52 unrelated donors, as well as epigenomic datasets using ATAC-seq on a subset of 8 donors. Through systematic comparison with publicly available datasets from GTEx and ENCODE projects, we identified transcriptomic, epigenetic, and genetic regulatory mechanisms specific to HCASMCs. We assessed the relevance of HCASMCs to CAD risk using transcriptomic and epigenomic level analyses. By jointly modeling eQTL and GWAS datasets, we identified five genes (SIPA1, TCF21, SMAD3, FES, and PDGFRA) that may modulate CAD risk through HCASMCs, all of which have relevant functional roles in vascular remodeling. Comparison with GTEx data suggests that SIPA1 and PDGFRA influence CAD risk predominantly through HCASMCs, while other annotated genes may have multiple cell and tissue targets. Together, these results provide tissue-specific and mechanistic insights into the regulation of a critical vascular cell type associated with CAD in human populations.

    View details for DOI 10.1016/j.ajhg.2018.08.001

    View details for PubMedID 30146127

  • Protein-altering variants associated with body mass index implicate pathways that control energy intake and expenditure in obesity NATURE GENETICS Turcot, V., Lu, Y., Highland, H. M., Schurmann, C., Justice, A. E., Fine, R. S., Bradfield, J. P., Esko, T., Giri, A., Graff, M., Guo, X., Hendricks, A. E., Karaderi, T., Lempradl, A., Locke, A. E., Mahajan, A., Marouli, E., Sivapalaratnam, S., Young, K. L., Alfred, T., Feitosa, M. F., Masca, N. D., Manning, A. K., Medina-Gomez, C., Mudgal, P., Ng, M. Y., Reiner, A. P., Vedantam, S., Willems, S. M., Winkler, T. W., Abecasis, G., Aben, K. K., Alam, D. S., Alharthi, S. E., Allison, M., Amouyel, P., Asselbergs, F. W., Auer, P. L., Balkau, B., Bang, L. E., Barroso, I., Bastarache, L., Benn, M., Bergmann, S., Bielak, L. F., Blueher, M., Boehnke, M., Boeing, H., Boerwinkle, E., Boeger, C. A., Bork-Jensen, J., Bots, M. L., Bottinger, E. P., Bowden, D. W., Brandslund, I., Breen, G., Brilliant, M. H., Broer, L., Brumat, M., Burt, A. A., Butterworth, A. S., Campbell, P. T., Cappellani, S., Carey, D. J., Catamo, E., Caulfield, M. J., Chambers, J. C., Chasman, D. I., Chen, Y. I., Chowdhury, R., Christensen, C., Chu, A. Y., Cocca, M., Collins, F. S., Cook, J. P., Corley, J., Galbany, J., Cox, A. J., Crosslin, D. S., Cuellar-Partida, G., D'Eustacchio, A., Danesh, J., Davies, G., Bakker, P. W., Groot, M. H., Mutsert, R., Deary, I. J., Dedoussis, G., Demerath, E. W., Heijer, M., Hollander, A. I., Ruijter, H. M., Dennis, J. G., Denny, J. C., Angelantonio, E., Drenos, F., Du, M., Dube, M., Dunning, A. M., Easton, D. F., Edwards, T. L., Ellinghaus, D., Ellinor, P. T., Elliott, P., Evangelou, E., Farmaki, A., Farooqi, I., Faul, J. D., Fauser, S., Feng, S., Ferrannini, E., Ferrieres, J., Florez, J. C., Ford, I., Fornage, M., Franco, O. H., Franke, A., Franks, P. W., Friedrich, N., Frikke-Schmidt, R., Galesloot, T. E., Gan, W., Gandin, I., Gasparini, P., Gibson, J., Giedraitis, V., Gjesing, A. P., Gordon-Larsen, P., Gorski, M., Grabe, H., Grant, S. A., Grarup, N., Griffiths, H. L., Grove, M. L., Gudnason, V., Gustafsson, S., Haessler, J., Hakonarson, H., Hammerschlag, A. R., Hansen, T., Harris, K., Harris, T. B., Hattersley, A. T., Have, C. T., Hayward, C., He, L., Heard-Costa, N. L., Heath, A. C., Heid, I. M., Helgeland, O., Hernesniemi, J., Hewitt, A. W., Holmen, O. L., Hovingh, G., Howson, J. M., Hu, Y., Huang, P. L., Huffman, J. E., Ikram, M., Ingelsson, E., Jackson, A. U., Jansson, J., Jarvik, G. P., Jensen, G. B., Jia, Y., Johansson, S., Jorgensen, M. E., Jorgensen, T., Jukema, J., Kahali, B., Kahn, R. S., Kahonen, M., Kamstrup, P. R., Kanoni, S., Kaprio, J., Karaleftheri, M., Kardia, S. R., Karpe, F., Kathiresan, S., Kee, F., Kiemeney, L. A., Kim, E., Kitajima, H., Komulainen, P., Kooner, J. S., Kooperberg, C., Korhonen, T., Kovacs, P., Kuivaniemi, H., Kutalik, Z., Kuulasmaa, K., Kuusisto, J., Laakso, M., Lakka, T. A., Lamparter, D., Lange, E. M., Lange, L. A., Langenberg, C., Larson, E. B., Lee, N. R., Lehtimaki, T., Lewis, C. E., Li, H., Li, J., Li-Gao, R., Lin, H., Lin, K., Lin, L., Lin, X., Lind, L., Lindstrom, J., Linneberg, A., Liu, C., Liu, D. J., Liu, Y., Lo, K. S., Lophatananon, A., Lotery, A. J., Loukola, A., Luan, J., Lubitz, S. A., Lyytikainen, L., Mannisto, S., Marenne, G., Mazul, A. L., McCarthy, M. I., McKean-Cowdin, R., Medland, S. E., Meidtner, K., Milani, L., Mistry, V., Mitchell, P., Mohlke, K. L., Moilanen, L., Moitry, M., Montgomery, G. W., Mook-Kanamori, D. O., Moore, C., Mori, T. A., Morris, A. D., Morris, A. P., Mueller-Nurasyid, M., Munroe, P. B., Nalls, M. A., Narisu, N., Nelson, C. P., Neville, M., Nielsen, S. F., Nikus, K., Njolstad, P. R., Nordestgaard, B. G., Nyholt, D. R., O'Connel, J. R., O'Donoghue, M. L., Loohuis, L., Ophoff, R. A., Owen, K. R., Packard, C. J., Padmanabhan, S., Palmer, C. A., Palmer, N. D., Pasterkamp, G., Patel, A. P., Pattie, A., Pedersen, O., Peissig, P. L., Peloso, G. M., Pennell, C. E., Perola, M., Perry, J. A., Perry, J. B., Pers, T. H., Person, T. N., Peters, A., Petersen, E. B., Peyser, P. A., Pirie, A., Polasek, O., Polderman, T. J., Puolijoki, H., Raitakari, O. T., Rasheed, A., Rauramaa, R., Reilly, D. F., Renstrom, F., Rheinberger, M., Ridker, P. M., Rioux, J. D., Rivas, M. A., Roberts, D. J., Robertson, N. R., Robino, A., Rolandsson, O., Rudan, I., Ruth, K. S., Saleheen, D., Salomaa, V., Samani, N. J., Sapkota, Y., Sattar, N., Schoen, R. E., Schreiner, P. J., Schulze, M. B., Scott, R. A., Segura-Lepe, M. P., Shah, S. H., Sheu, W., Sim, X., Slater, A. J., Small, K. S., Smith, A. V., Southam, L., Spector, T. D., Speliotes, E. K., Starr, J. M., Stefansson, K., Steinthorsdottir, V., Stirrups, K. E., Strauch, K., Stringham, H. M., Stumvoll, M., Sun, L., Surendran, P., Swift, A. J., Tada, H., Tansey, K. E., Tardif, J., Taylor, K. D., Teumer, A., Thompson, D. J., Thorleifsson, G., Thorsteinsdottir, U., Thuesen, B. H., Toenjes, A., Tromp, G., Trompet, S., Tsafantakis, E., Tuomilehto, J., Tybjaerg-Hansen, A., Tyrer, J. P., Uher, R., Uitterlinden, A. G., Uusitupa, M., Laan, S. W., Duijn, C. M., Leeuwen, N., van Setten, J., Vanhala, M., Varbo, A., Varga, T. V., Varma, R., Edwards, D., Vermeulen, S. H., Veronesi, G., Vestergaard, H., Vitart, V., Vogt, T. F., Voelker, U., Vuckovic, D., Wagenknecht, L. E., Walker, M., Wallentin, L., Wang, F., Wang, C. A., Wang, S., Wang, Y., Ware, E. B., Wareham, N. J., Warren, H. R., Waterworth, D. M., Wessel, J., White, H. D., Willer, C. J., Wilson, J. G., Witte, D. R., Wood, A. R., Wu, Y., Yaghootkar, H., Yao, J., Yao, P., Yerges-Armstrong, L. M., Young, R., Zeggini, E., Zhan, X., Zhang, W., Zhao, J., Zhao, W., Zhou, W., Zondervan, K. T., Rotter, J. I., Pospisilik, J. A., Rivadeneira, F., Borecki, I. B., Deloukas, P., Frayling, T. M., Lettre, G., North, K. E., Lindgren, C. M., Hirschhorn, J. N., Loos, R. F., Chd Exome Consortium, Epic-Cvd Consortium, ExomeBP Consortium, Global Lipids Genetic Consortium, GoT2D Genes Consortium, EPIC InterAct Consortium, Interval Study, ReproGen Consortium, T2D-Genes Consortium, MAGIC Investigators, Understanding Soc Sci Grp 2018; 50 (1): 26-+
  • Dog ownership and the risk of cardiovascular disease and death - a nationwide cohort study SCIENTIFIC REPORTS Mubanga, M., Byberg, L., Nowak, C., Egenvall, A., Magnusson, P. K., Ingelsson, E., Fall, T. 2017; 7: 15821

    Abstract

    Dogs may be beneficial in reducing cardiovascular risk in their owners by providing social support and motivation for physical activity. We aimed to investigate the association of dog ownership with incident cardiovascular disease (CVD) and death in a register-based prospective nation-wide cohort (n = 3,432,153) with up to 12 years of follow-up. Self-reported health and lifestyle habits were available for 34,202 participants in the Swedish Twin Register. Time-to-event analyses with time-updated covariates were used to calculate hazard ratios (HR) with 95% confidence intervals (CI). In single- and multiple-person households, dog ownership (13.1%) was associated with lower risk of death, HR 0.67 (95% CI, 0.65-0.69) and 0.89 (0.87-0.91), respectively; and CVD death, HR 0.64 (0.59-0.70), and 0.85 (0.81-0.90), respectively. In single-person households, dog ownership was inversely associated with cardiovascular outcomes (HR composite CVD 0.92, 95% CI, 0.89-0.94). Ownership of hunting breed dogs was associated with lowest risk of CVD. Further analysis in the Twin Register could not replicate the reduced risk of CVD or death but also gave no indication of confounding by disability, comorbidities or lifestyle factors. In conclusion, dog ownership appears to be associated with lower risk of CVD in single-person households and lower mortality in the general population.

    View details for DOI 10.1038/s41598-017-16118-6

    View details for Web of Science ID 000415658600066

    View details for PubMedID 29150678

    View details for PubMedCentralID PMC5693989

  • Large meta-analysis of genome-wide association studies identifies five loci for lean body mass (vol 8, 80, 2017) NATURE COMMUNICATIONS Zillikens, M., Demissie, S., Hsu, Y., Yerges-Armstrong, L. M., Chou, W., Stolk, L., Livshits, G., Broer, L., Johnson, T., Koller, D. L., Kutalik, Z., Luan, J., Malkin, I., Ried, J. S., Smith, A. V., Thorleifsson, G., Vandenput, L., Zhao, J., Zhang, W., Aghdassi, A., Akesson, K., Amin, N., Baier, L. J., Barroso, I., Bennett, D. A., Bertram, L., Biffar, R., Bochud, M., Boehnke, M., Borecki, I. B., Buchman, A. S., Byberg, L., Campbell, H., Obanda, N., Cauley, J. A., Cawthon, P. M., Cederberg, H., Chen, Z., Cho, N. H., Choi, H., Claussnitzer, M., Collins, F., Cummings, S. R., De Jager, P. L., Demuth, I., Dhonukshe-Rutten, R. M., Diatchenko, L., Eiriksdottir, G., Enneman, A. W., Erdos, M., Eriksson, J. G., Eriksson, J., Estrada, K., Evans, D. S., Feitosa, M. F., Fu, M., Garcia, M., Gieger, C., Girke, T., Glazer, N. L., Grallert, H., Grewal, J., Han, B., Hanson, R. L., Hayward, C., Hofman, A., Hoffman, E. P., Homuth, G., Hsueh, W., Hubal, M. J., Hubbard, A., Huffman, K. M., Husted, L. B., Illig, T., Ingelsson, E., Ittermann, T., Jansson, J., Jordan, J. M., Jula, A., Karlsson, M., Khaw, K., Kilpelanen, T. O., Klopp, N., Kloth, J. L., Koistinen, H. A., Kraus, W. E., Kritchevsky, S., Kuulasmaa, T., Kuusisto, J., Laakso, M., Lahti, J., Lang, T., Langdahl, B. L., Launer, L. J., Lee, J., Lerch, M. M., Lewis, J. R., Lind, L., Lindgren, C., Liu, Y., Liu, T., Liu, Y., Ljunggren, O., Lorentzon, M., Luben, R. N., Maixner, W., McGuigan, F. E., Medina-Gomez, C., Meitinger, T., Melhus, H., Mellstrom, D., Melov, S., Michaelsson, K., Mitchell, B. D., Morris, A. P., Mosekilde, L., Newman, A., Nielson, C. M., O'Connell, J. R., Oostra, B. A., Orwoll, E. S., Palotie, A., Parker, S. J., Peacock, M., Perola, M., Peters, A., Polasek, O., Prince, R. L., Raikkonen, K., Ralston, S. H., Ripatti, S., Robbins, J. A., Rotter, J. I., Rudan, I., Salomaa, V., Satterfield, S., Schadt, E. E., Schipf, S., Scott, L., Sehmi, J., Shen, J., Shin, C., Sigurdsson, G., Smith, S., Soranzo, N., Stancakova, A., Steinhagen-Thiessen, E., Streeten, E. A., Styrkarsdottir, U., Swart, K. A., Tan, S., Tarnopolsky, M. A., Thompson, P., Thomson, C. A., Thorsteinsdottir, U., Tikkanen, E., Tranah, G. J., Tuomilehto, J., van Schoor, N. M., Verma, A., Vollenweider, P., Volzke, H., Wactawski-Wende, J., Walker, M., Weedon, M. N., Welch, R., Wichmann, H., Widen, E., Williams, F. K., Wilson, J. F., Wright, N. C., Xie, W., Yu, L., Zhou, Y., Chambers, J. C., Doring, A., van Duijn, C. M., Econs, M. J., Gudnason, V., Kooner, J. S., Psaty, B. M., Spector, T. D., Stefansson, K., Rivadeneira, F., Uitterlinden, A. G., Wareham, N. J., Ossowski, V., Waterworth, D., Loos, R. F., Karasik, D., Harris, T. B., Ohlsson, C., Kiel, D. P. 2017; 8: 1414

    Abstract

    A correction to this article has been published and is linked from the HTML version of this article.

    View details for DOI 10.1038/s41467-017-01008-2

    View details for Web of Science ID 000414535100001

    View details for PubMedID 29116125

    View details for PubMedCentralID PMC5676783

  • Identification of a novel proinsulin-associated SNP and demonstration that proinsulin is unlikely to be a causal factor in subclinical vascular remodelling using Mendelian randomisation ATHEROSCLEROSIS Strawbridge, R. J., Silveira, A., den Hoed, M., Gustafsson, S., Luan, J., Rybin, D., Dupuis, J., Li-Gao, R., Kavousi, M., Dehghan, A., Haljas, K., Lahti, J., Gadin, J. R., Backlund, A., de Faire, U., Gertow, K., Giral, P., Goel, A., Humphries, S. E., Kurl, S., Langenberg, C., Lannfelt, L. L., Lind, L., Lindgren, C. M., Mannarino, E., Mook-Kanamori, D. O., Morris, A. P., de Mutsert, R., Rauramaa, R., Saliba-Gustafsson, P., Sennblad, B., Smit, A. J., Syvanen, A., Tremoli, E., Veglia, F., Zethelius, B., Bjorck, H. M., Eriksson, J. G., Hofman, A., Franco, O. H., Watkins, H., Jukema, J., Florez, J. C., Wareham, N. J., Meigs, J. B., Ingelsson, E., Baldassarre, D., Hamsten, A., IMPROVE Study Grp 2017; 266: 196–204

    Abstract

    Increased proinsulin relative to insulin levels have been associated with subclinical atherosclerosis (measured by carotid intima-media thickness (cIMT)) and are predictive of future cardiovascular disease (CVD), independently of established risk factors. The mechanisms linking proinsulin to atherosclerosis and CVD are unclear. A genome-wide meta-analysis has identified nine loci associated with circulating proinsulin levels. Using proinsulin-associated SNPs, we set out to use a Mendelian randomisation approach to test the hypothesis that proinsulin plays a causal role in subclinical vascular remodelling.We studied the high CVD-risk IMPROVE cohort (n = 3345), which has detailed biochemical phenotyping and repeated, state-of-the-art, high-resolution carotid ultrasound examinations. Genotyping was performed using Illumina Cardio-Metabo and Immuno arrays, which include reported proinsulin-associated loci. Participants with type 2 diabetes (n = 904) were omitted from the analysis. Linear regression was used to identify proinsulin-associated genetic variants.We identified a proinsulin locus on chromosome 15 (rs8029765) and replicated it in data from 20,003 additional individuals. An 11-SNP score, including the previously identified and the chromosome 15 proinsulin-associated loci, was significantly and negatively associated with baseline IMTmean and IMTmax (the primary cIMT phenotypes) but not with progression measures. However, MR-Eggers refuted any significant effect of the proinsulin-associated 11-SNP score, and a non-pleiotropic SNP score of three variants (including rs8029765) demonstrated no effect on baseline or progression cIMT measures.We identified a novel proinsulin-associated locus and demonstrated that whilst proinsulin levels are associated with cIMT measures, proinsulin per se is unlikely to have a causative effect on cIMT.

    View details for DOI 10.1016/j.atherosclerosis.2017.09.031

    View details for Web of Science ID 000414069700027

    View details for PubMedID 29040868

    View details for PubMedCentralID PMC5679136

  • Vitamin D and cognitive function: A Mendelian randomisation study SCIENTIFIC REPORTS Maddock, J., Zhou, A., Cavadino, A., Kuzma, E., Bao, Y., Smart, M. C., Saum, K., Schoettker, B., Engmann, J., Kjaergaard, M., Karhunen, V., Zhan, Y., Lehtimaki, T., Rovio, S. P., Byberg, L., Lahti, J., Marques-Vidal, P., Sen, A., Perna, L., Schirmer, H., Singh-Manoux, A., Auvinen, J., Hutri-Kahonen, N., Kahonen, M., Kilander, L., Raikkonen, K., Melhus, H., Ingelsson, E., Guessous, I., Petrovic, K. E., Schmidt, H., Schmidt, R., Vollenweider, P., Lind, L., Eriksson, J. G., Michaelsson, K., Raitakari, O. T., Hagg, S., Pedersen, N. L., Herzig, K., Jarvelin, M., Veijola, J., Kivimaki, M., Jorde, R., Brenner, H., Kumari, M., Power, C., Llewellyn, D. J., Hypponen, E. 2017; 7: 13230

    Abstract

    The causal nature of the association between hypovitaminosis D and poor cognitive function in mid- to later-life is uncertain. Using a Mendelian randomisation(MR) approach, we examined the causal relationship between 25(OH)D and cognitive function. Data came from 172,349 participants from 17 cohorts. DHCR7(rs12785878), CYP2R1 rs12794714) and their combined synthesis score were chosen to proxy 25(OH)D. Cognitive tests were standardised into global and memory scores. Analyses were stratified by 25(OH)D tertiles, sex and age. Random effects meta-analyses assessed associations between 25(OH)D and cognitive function. Associations of serum 25(OH)D with global and memory-related cognitive function were non-linear (lower cognitive scores for both low and high 25(OH)D, p curvature ≤ 0.006), with much of the curvature attributed to a single study. DHCR7, CYP2R1, and the synthesis score were associated with small reductions in 25(OH)D per vitamin D-decreasing allele. However, coefficients for associations with global or memory-related cognitive function were non-significant and in opposing directions for DHCR7 and CYP2R1, with no overall association observed for the synthesis score. Coefficients for the synthesis score and global and memory cognition were similar when stratified by 25(OH)D tertiles, sex and age. We found no evidence for serum 25(OH)D concentration as a causal factor for cognitive performance in mid- to later life.

    View details for DOI 10.1038/s41598-017-13189-3

    View details for Web of Science ID 000413048000041

    View details for PubMedID 29038561

    View details for PubMedCentralID PMC5643555

  • New Blood Pressure-Associated Loci Identified in Meta-Analyses of 475000 Individuals CIRCULATION-CARDIOVASCULAR GENETICS Kraja, A. T., Cook, J. P., Warren, H. R., Surendran, P., Liu, C., Evangelou, E., Manning, A. K., Grarup, N., Drenos, F., Sim, X., Smith, A., Amin, N., Blakemore, A. F., Bork-Jensen, J., Brandslund, I., Farmaki, A., Fava, C., Ferreira, T., Herzig, K., Giri, A., Giulianini, F., Grove, M. L., Guo, X., Harris, S. E., Have, C. T., Havulinna, A. S., Zhang, H., Jorgensen, M. E., Karajamaki, A., Kooperberg, C., Linneberg, A., Little, L., Liu, Y., Bonnycastle, L. L., Lu, Y., Magi, R., Mahajan, A., Malerba, G., Marioni, R. E., Mei, H., Menni, C., Morrison, A. C., Padmanabhan, S., Palmas, W., Poveda, A., Rauramaa, R., Rayner, N., Riaz, M., Rice, K., Richard, M. A., Smith, J. A., Southam, L., Stancakova, A., Stirrups, K. E., Tragante, V., Tuomi, T., Tzoulald, I., Varga, T. V., Weiss, S., Yiorkas, A. M., Young, R., Zhang, W., Barnes, M. R., Cabrera, C. P., Gao, H., Boehnke, M., Boerwinkle, E., Chambers, J. C., Connell, J. M., Christensen, C. K., de Boer, R. A., Deary, I. J., Dedoussis, G., Deloukas, P., Dominiczak, A. F., Dorr, M., Joehanes, R., Edwards, T. L., Esko, T., Fornage, M., Franceschini, N., Franks, P. W., Gambaro, G., Groop, L., Hallmans, G., Hansen, T., Hayward, C., Heikki, O., Ingelsson, E., Tuomilehto, J., Jarvelin, M., Kardia, S. R., Karpe, F., Kooner, J. S., Lakka, T. A., Langenberg, C., Lind, L., Loos, R. F., Laakso, M., McCarthy, M. I., Melander, O., Mohlke, K. L., Moris, A. P., Palmer, C. A., Pedersen, O., Polasek, O., Poulter, N. R., Province, M. A., Psaty, B. M., Ridker, P. M., Rotter, J. I., Rudan, I., Salomaa, V., Samani, N. J., Sever, P. J., Skaaby, T., Stafford, J. M., Starr, J. M., van der Harst, P., van der Meer, P., van Duijn, C. M., Vergnaud, A., Gudnason, V., Wareham, N. J., Wilson, J. G., Willer, C. J., Witte, D. R., Zeggini, E., Saleheen, D., Butterworth, A. S., Danesh, J., Asselbergs, F. W., Wain, L. V., Ehret, G. B., Chasman, D. I., Caulfield, M. J., Elliott, P., Lindgren, C. M., Levy, D., Newton-Cheh, C., Munroe, P. B., Howson, J. M., Understanding Soc Sci Grp, CHARGE EXOME BP CHD Exome Exome B, UK Biobank Cardio-Metab Traits Co 2017; 10 (5)

    Abstract

    Genome-wide association studies have recently identified >400 loci that harbor DNA sequence variants that influence blood pressure (BP). Our earlier studies identified and validated 56 single nucleotide variants (SNVs) associated with BP from meta-analyses of exome chip genotype data. An additional 100 variants yielded suggestive evidence of association.Here, we augment the sample with 140 886 European individuals from the UK Biobank, in whom 77 of the 100 suggestive SNVs were available for association analysis with systolic BP or diastolic BP or pulse pressure. We performed 2 meta-analyses, one in individuals of European, South Asian, African, and Hispanic descent (pan-ancestry, ≈475 000), and the other in the subset of individuals of European descent (≈423 000). Twenty-one SNVs were genome-wide significant (P<5×10-8) for BP, of which 4 are new BP loci: rs9678851 (missense, SLC4A1AP), rs7437940 (AFAP1), rs13303 (missense, STAB1), and rs1055144 (7p15.2). In addition, we identified a potentially independent novel BP-associated SNV, rs3416322 (missense, SYNPO2L) at a known locus, uncorrelated with the previously reported SNVs. Two SNVs are associated with expression levels of nearby genes, and SNVs at 3 loci are associated with other traits. One SNV with a minor allele frequency <0.01, (rs3025380 at DBH) was genome-wide significant.We report 4 novel loci associated with BP regulation, and 1 independent variant at an established BP locus. This analysis highlights several candidate genes with variation that alter protein function or gene expression for potential follow-up.

    View details for DOI 10.1161/CIRCGENETICS.117.001778

    View details for Web of Science ID 000424292400015

    View details for PubMedID 29030403

    View details for PubMedCentralID PMC5776077

  • Impact of common genetic determinants of Hemoglobin A1c on type 2 diabetes risk and diagnosis in ancestrally diverse populations: A transethnic genome-wide meta-analysis PLOS MEDICINE Wheeler, E., Leong, A., Liu, C., Hivert, M., Strawbridge, R. J., Podmore, C., Li, M., Yao, J., Sim, X., Hong, J., Chu, A. Y., Zhang, W., Wang, X., Chen, P., Maruthur, N. M., Porneala, B. C., Sharp, S. J., Jia, Y., Kabagambe, E. K., Chang, L., Chen, W., Elks, C. E., Evans, D. S., Fan, Q., Giulianini, F., Go, M., Hottenga, J., Hu, Y., Jackson, A. U., Kanoni, S., Kim, Y., Kleber, M. E., Ladenvall, C., Lecoeur, C., Lim, S., Lu, Y., Mahajan, A., Marzi, C., Nalls, M. A., Navarro, P., Nolte, I. M., Rose, L. M., Rybin, D. V., Sanna, S., Shi, Y., Stram, D. O., Takeuchi, F., Tan, S., van der Most, P. J., Van Vliet-Ostaptchouk, J. V., Wong, A., Yengo, L., Zhao, W., Goel, A., Martinez Larrad, M., Radke, D., Salo, P., Tanaka, T., van Iperen, E. A., Abecasis, G., Afaq, S., Alizadeh, B. Z., Bertoni, A. G., Bonnefond, A., Boettcher, Y., Bottinger, E. P., Campbell, H., Carlson, O. D., Chen, C., Cho, Y., Garvey, W., Gieger, C., Goodarzi, M. O., Grallert, H., Hamsten, A., Hartman, C. A., Herder, C., Hsiung, C., Huang, J., Igase, M., Isono, M., Katsuya, T., Khor, C., Kiess, W., Kohara, K., Kovacs, P., Lee, J., Lee, W., Lehne, B., Li, H., Liu, J., Lobbens, S., Luang, J., Lyssenko, V., Meitinger, T., Miki, T., Miljkovic, I., Moon, S., Mulas, A., Mueller, G., Mueller-Nurasyid, M., Nagaraja, R., Nauck, M., Pankow, J. S., Polasek, O., Prokopenko, I., Ramos, P. S., Rasmussen-Torvik, L., Rathmann, W., Rich, S. S., Robertson, N. R., Roden, M., Roussel, R., Rudan, I., Scott, R. A., Scott, W. R., Sennblad, B., Siscovick, D. S., Strauch, K., Sun, L., Swertz, M., Tajuddin, S. M., Taylor, K. D., Teo, Y., Tham, Y., Toenjes, A., Wareham, N. J., Willemsen, G., Wilsgaard, T., Hingorani, A. D., Egan, J., Ferrucci, L., Hovingh, G., Jula, A., Kivimaki, M., Kumari, M., Njolstad, I., Palmer, C. A., Serrano Rios, M., Stumvoll, M., Watkins, H., Aung, T., Blueher, M., Boehnke, M., Boomsma, D. I., Bornstein, S. R., Chambers, J. C., Chasman, D. I., Chen, Y., Chen, Y., Cheng, C., Cucca, F., de Geus, E. C., Deloukas, P., Evans, M. K., Fornage, M., Friedlander, Y., Froguel, P., Groop, L., Gross, M. D., Harris, T. B., Hayward, C., Heng, C., Ingelsson, E., Kato, N., Kim, B., Koh, W., Kooner, J. S., Koerner, A., Kuh, D., Kuusisto, J., Laakso, M., Lin, X., Liu, Y., Loos, R. F., Magnusson, P. E., Maerz, W., McCarthy, M. I., Oldehinkel, A. J., Ong, K. K., Pedersen, N. L., Pereira, M. A., Peters, A., Ridker, P. M., Sabanayagam, C., Sale, M., Saleheen, D., Saltevo, J., Schwarz, P. H., Sheu, W. H., Snieder, H., Spector, T. D., Tabara, Y., Tuomilehto, J., van Dam, R. M., Wilson, J. G., Wilson, J. F., Wolffenbuttel, B. R., Wong, T., Wu, J., Yuan, J., Zonderman, A. B., Soranzo, N., Guo, X., Roberts, D. J., Florez, J. C., Sladek, R., Dupuis, J., Morris, A. P., Tai, E., Selvin, E., Rotter, J. I., Langenberg, C., Barroso, I., Meigs, J. B., EPIC-CVD Consortium, EPIC-InterAct Consortium, Lifelines Cohort Study 2017; 14 (9): e1002383

    Abstract

    Glycated hemoglobin (HbA1c) is used to diagnose type 2 diabetes (T2D) and assess glycemic control in patients with diabetes. Previous genome-wide association studies (GWAS) have identified 18 HbA1c-associated genetic variants. These variants proved to be classifiable by their likely biological action as erythrocytic (also associated with erythrocyte traits) or glycemic (associated with other glucose-related traits). In this study, we tested the hypotheses that, in a very large scale GWAS, we would identify more genetic variants associated with HbA1c and that HbA1c variants implicated in erythrocytic biology would affect the diagnostic accuracy of HbA1c. We therefore expanded the number of HbA1c-associated loci and tested the effect of genetic risk-scores comprised of erythrocytic or glycemic variants on incident diabetes prediction and on prevalent diabetes screening performance. Throughout this multiancestry study, we kept a focus on interancestry differences in HbA1c genetics performance that might influence race-ancestry differences in health outcomes.Using genome-wide association meta-analyses in up to 159,940 individuals from 82 cohorts of European, African, East Asian, and South Asian ancestry, we identified 60 common genetic variants associated with HbA1c. We classified variants as implicated in glycemic, erythrocytic, or unclassified biology and tested whether additive genetic scores of erythrocytic variants (GS-E) or glycemic variants (GS-G) were associated with higher T2D incidence in multiethnic longitudinal cohorts (N = 33,241). Nineteen glycemic and 22 erythrocytic variants were associated with HbA1c at genome-wide significance. GS-G was associated with higher T2D risk (incidence OR = 1.05, 95% CI 1.04-1.06, per HbA1c-raising allele, p = 3 × 10-29); whereas GS-E was not (OR = 1.00, 95% CI 0.99-1.01, p = 0.60). In Europeans and Asians, erythrocytic variants in aggregate had only modest effects on the diagnostic accuracy of HbA1c. Yet, in African Americans, the X-linked G6PD G202A variant (T-allele frequency 11%) was associated with an absolute decrease in HbA1c of 0.81%-units (95% CI 0.66-0.96) per allele in hemizygous men, and 0.68%-units (95% CI 0.38-0.97) in homozygous women. The G6PD variant may cause approximately 2% (N = 0.65 million, 95% CI 0.55-0.74) of African American adults with T2D to remain undiagnosed when screened with HbA1c. Limitations include the smaller sample sizes for non-European ancestries and the inability to classify approximately one-third of the variants. Further studies in large multiethnic cohorts with HbA1c, glycemic, and erythrocytic traits are required to better determine the biological action of the unclassified variants.As G6PD deficiency can be clinically silent until illness strikes, we recommend investigation of the possible benefits of screening for the G6PD genotype along with using HbA1c to diagnose T2D in populations of African ancestry or groups where G6PD deficiency is common. Screening with direct glucose measurements, or genetically-informed HbA1c diagnostic thresholds in people with G6PD deficiency, may be required to avoid missed or delayed diagnoses.

    View details for DOI 10.1371/journal.pmed.1002383

    View details for Web of Science ID 000411970300003

    View details for PubMedID 28898252

    View details for PubMedCentralID PMC5595282

  • Novel Blood Pressure Locus and Gene Discovery Using Genome-Wide Association Study and Expression Data Sets From Blood and the Kidney HYPERTENSION Wain, L. V., Vaez, A., Jansen, R., Joehanes, R., van der Most, P. J., Erzurumluoglu, A., O'Reilly, P. F., Cabrera, C. P., Warren, H. R., Rose, L. M., Verwoert, G. C., Hottenga, J., Strawbridge, R. J., Esko, T., Arking, D. E., Hwang, S., Guo, X., Kutalik, Z., Trompet, S., Shrine, N., Teumer, A., Ried, J. S., Bis, J. C., Smith, A. V., Amin, N., Nolte, I. M., Lyytikainen, L., Mahajan, A., Wareham, N. J., Hofer, E., Joshi, P. K., Kristiansson, K., Traglia, M., Havulinna, A. S., Goel, A., Nalls, M. A., Sober, S., Vuckovic, D., Luan, J., Del Greco M, F., Ayers, K. L., Marrugat, J., Ruggiero, D., Lopez, L. M., Niiranen, T., Enroth, S., Jackson, A. U., Nelson, C. P., Huffman, J. E., Zhang, W., Marten, J., Gandin, I., Harris, S. E., Zemunik, T., Lu, Y., Evangelou, E., Shah, N., de Borst, M. H., Mangino, M., Prins, B. P., Campbell, A., Li-Gao, R., Chauhan, G., Oldmeadow, C., Abecasis, G., Abedi, M., Barbieri, C. M., Barnes, M. R., Batini, C., Beilby, J., Blake, T., Boehnke, M., Bottinger, E. P., Braund, P. S., Brown, M., Brumat, M., Campbell, H., Chambers, J. C., Cocca, M., Collins, F., Connell, J., Cordell, H. J., Damman, J. J., Davies, G., de Geus, E. J., de Mutsert, R., Deelen, J., Demirkale, Y., Doney, A. F., Dorr, M., Farrall, M., Ferreira, T., Franberg, M., Gao, H., Giedraitis, V., Gieger, C., Giulianini, F., Gow, A. J., Hamsten, A., Harris, T. B., Hofman, A., Holliday, E. G., Hui, J., Jarvelin, M., Johansson, A., Johnson, A. D., Jousilahti, P., Jula, A., Kahonen, M., Kathiresan, S., Khaw, K., Kolcic, I., Koskinen, S., Langenberg, C., Larson, M., Launer, L. J., Lehne, B., Liewald, D. M., Lin, L., Lind, L., Mach, F., Mamasoula, C., Menni, C., Mifsud, B., Milaneschi, Y., Morgan, A., Morris, A. D., Morrison, A. C., Munson, P. J., Nandakumar, P., Quang Tri Nguyen, Nutile, T., Oldehinkel, A. J., Oostra, B. A., Org, E., Padmanabhan, S., Palotie, A., Pare, G., Pattie, A., Penninx, B. H., Poulter, N., Pramstaller, P. P., Raitakari, O. T., Ren, M., Rice, K., Ridker, P. M., Riese, H., Ripatti, S., Robino, A., Rotter, J. I., Rudan, I., Saba, Y., Saint Pierre, A., Sala, C. F., Sarin, A., Schmidt, R., Scott, R., Seelen, M. A., Shields, D. C., Siscovick, D., Sorice, R., Stanton, A., Stott, D. J., Sundstrom, J., Swertz, M., Taylor, K. D., Thom, S., Tzoulaki, I., Tzourio, C., Uitterlinden, A. G., Volker, U., Vollenweider, P., Wild, S., Willemsen, G., Wright, A. F., Yao, J., Theriault, S., Conen, D., Attia, J., Sever, P., Debette, S., Mook-Kanamori, D. O., Zeggini, E., Spector, T. D., van der Harst, P., Palmer, C. A., Vergnaud, A., Loos, R. F., Polasek, O., Starr, J. M., Girotto, G., Hayward, C., Kooner, J. S., Lindgren, C. M., Vitart, V., Samani, N. J., Tuomilehto, J., Gyllensten, U., Knekt, P., Deary, I. J., Ciullo, M., Elosua, R., Keavney, B. D., Hicks, A. A., Scott, R. A., Gasparini, P., Laan, M., Liu, Y., Watkins, H., Hartman, C. A., Salomaa, V., Toniolo, D., Perola, M., Wilson, J. F., Schmidt, H., Zhao, J., Lehtimaki, T., van Duijn, C. M., Gudnason, V., Psaty, B. M., Peters, A., Rettig, R., James, A., Jukema, J., Strachan, D. P., Palmas, W., Metspalu, A., Ingelsson, E., Boomsma, D. I., Franco, O. H., Bochud, M., Newton-Cheh, C., Munroe, P. B., Elliott, P., Chasman, D. I., Chakravarti, A., Knight, J., Morris, A. P., Levy, D., Tobin, M. D., Snieder, H., Caulfield, M. J., Ehret, G. B., Alistair, T. Y., Betholdson, J., Xiao, L., BIOS Consortium, Lifelines Cohort Study, Understanding Soc Sci Grp 2017; 70 (3): E4-+

    Abstract

    Elevated blood pressure is a major risk factor for cardiovascular disease and has a substantial genetic contribution. Genetic variation influencing blood pressure has the potential to identify new pharmacological targets for the treatment of hypertension. To discover additional novel blood pressure loci, we used 1000 Genomes Project-based imputation in 150 134 European ancestry individuals and sought significant evidence for independent replication in a further 228 245 individuals. We report 6 new signals of association in or near HSPB7, TNXB, LRP12, LOC283335, SEPT9, and AKT2, and provide new replication evidence for a further 2 signals in EBF2 and NFKBIA Combining large whole-blood gene expression resources totaling 12 607 individuals, we investigated all novel and previously reported signals and identified 48 genes with evidence for involvement in blood pressure regulation that are significant in multiple resources. Three novel kidney-specific signals were also detected. These robustly implicated genes may provide new leads for therapeutic innovation.

    View details for DOI 10.1161/HYPERTENSIONAHA.117.09438

    View details for Web of Science ID 000407241500001

    View details for PubMedID 28739976

    View details for PubMedCentralID PMC5783787

  • Large meta-analysis of genome-wide association studies identifies five loci for lean body mass NATURE COMMUNICATIONS Zillikens, M., Demissie, S., Hsu, Y., Yerges-Armstrong, L. M., Chou, W., Stolk, L., Livshits, G., Broer, L., Johnson, T., Koller, D. L., Kutalik, Z., Luan, J., Malkin, I., Ried, J. S., Smith, A. V., Thorleifsson, G., Vandenput, L., Zhao, J., Zhang, W., Aghdassi, A., Akesson, K., Amin, N., Baier, L. J., Barroso, I., Bennett, D. A., Bertram, L., Biffar, R., Bochud, M., Boehnke, M., Borecki, I. B., Buchman, A. S., Byberg, L., Campbell, H., Obanda, N., Cauley, J. A., Cawthon, P. M., Cederberg, H., Chen, Z., Cho, N. H., Choi, H., Claussnitzer, M., Collins, F., Cummings, S. R., De Jager, P. L., Demuth, I., Dhonukshe-Rutten, R. M., Diatchenko, L., Eiriksdottir, G., Enneman, A. W., Erdos, M., Eriksson, J. G., Eriksson, J., Estrada, K., Evans, D. S., Feitosa, M. F., Fu, M., Garcia, M., Gieger, C., Girke, T., Glazer, N. L., Grallert, H., Grewal, J., Han, B., Hanson, R. L., Hayward, C., Hofman, A., Hoffman, E. P., Homuth, G., Hsueh, W., Hubal, M. J., Hubbard, A., Huffman, K. M., Husted, L. B., Illig, T., Ingelsson, E., Ittermann, T., Jansson, J., Jordan, J. M., Jula, A., Karlsson, M., Khaw, K., Kilpainen, T. O., Klopp, N., Kloth, J. L., Koistinen, H. A., Kraus, W. E., Kritchevsky, S., Kuulasmaa, T., Kuusisto, J., Laakso, M., Lahti, J., Lang, T., Langdahl, B. L., Launer, L. J., Lee, J., Lerch, M. M., Lewis, J. R., Lind, L., Lindgren, C., Liu, Y., Liu, T., Liu, Y., Ljunggren, O., Lorentzon, M., Luben, R. N., Maixner, W., McGuigan, F. E., Medina-Gomez, C., Meitinger, T., Melhus, H., Mellstrom, D., Melov, S., Michaelsson, K., Mitchell, B. D., Morris, A. P., Mosekilde, L., Newman, A., Nielson, C. M., O'Connell, J. R., Oostra, B. A., Orwoll, E. S., Palotie, A., Parker, S., Peacock, M., Perola, M., Peters, A., Polasek, O., Prince, R. L., Raikkonen, K., Ralston, S. H., Ripatti, S., Robbins, J. A., Rotter, J. I., Rudan, I., Salomaa, V., Satterfield, S., Schadt, E. E., Schipf, S., Scott, L., Sehmi, J., Shen, J., Shin, C., Sigurdsson, G., Smith, S., Soranzo, N., Stancakova, A., Steinhagen-Thiessen, E., Streeten, E. A., Styrkarsdottir, U., Swart, K. A., Tan, S., Tarnopolsky, M. A., Thompson, P., Thomson, C. A., Thorsteinsdottir, U., Tikkanen, E., Tranah, G. J., Tuomilehto, J., van Schoor, N. M., Verma, A., Vollenweider, P., Voelzke, H., Wactawski-Wende, J., Walker, M., Weedon, M. N., Welch, R., Wichman, H., Widen, E., Williams, F. K., Wilson, J. F., Wright, N. C., Xie, W., Yu, L., Zhou, Y., Chambers, J. C., Doring, A., van Duijn, C. M., Econs, M. J., Gudnason, V., Kooner, J. S., Psaty, B. M., Spector, T. D., Stefansson, K., Rivadeneira, F., Uitterlinden, A. G., Wareham, N. J., Ossowski, V., Waterworth, D., Loos, R. F., Karasik, D., Harris, T. B., Ohlsson, C., Kiel, D. P. 2017; 8: 80

    Abstract

    Lean body mass, consisting mostly of skeletal muscle, is important for healthy aging. We performed a genome-wide association study for whole body (20 cohorts of European ancestry with n = 38,292) and appendicular (arms and legs) lean body mass (n = 28,330) measured using dual energy X-ray absorptiometry or bioelectrical impedance analysis, adjusted for sex, age, height, and fat mass. Twenty-one single-nucleotide polymorphisms were significantly associated with lean body mass either genome wide (p < 5 × 10-8) or suggestively genome wide (p < 2.3 × 10-6). Replication in 63,475 (47,227 of European ancestry) individuals from 33 cohorts for whole body lean body mass and in 45,090 (42,360 of European ancestry) subjects from 25 cohorts for appendicular lean body mass was successful for five single-nucleotide polymorphisms in/near HSD17B11, VCAN, ADAMTSL3, IRS1, and FTO for total lean body mass and for three single-nucleotide polymorphisms in/near VCAN, ADAMTSL3, and IRS1 for appendicular lean body mass. Our findings provide new insight into the genetics of lean body mass.Lean body mass is a highly heritable trait and is associated with various health conditions. Here, Kiel and colleagues perform a meta-analysis of genome-wide association studies for whole body lean body mass and find five novel genetic loci to be significantly associated.

    View details for DOI 10.1038/s41467-017-00031-7

    View details for Web of Science ID 000405818900003

    View details for PubMedID 28724990

    View details for PubMedCentralID PMC5517526

  • Large-scale analyses of common and rare variants identify 12 new loci associated with atrial fibrillation. Nature genetics Christophersen, I. E., Rienstra, M., Roselli, C., Yin, X., Geelhoed, B., Barnard, J., Lin, H., Arking, D. E., Smith, A. V., Albert, C. M., Chaffin, M., Tucker, N. R., Li, M., Klarin, D., Bihlmeyer, N. A., Low, S., Weeke, P. E., Müller-Nurasyid, M., Smith, J. G., Brody, J. A., Niemeijer, M. N., Dörr, M., Trompet, S., Huffman, J., Gustafsson, S., Schurmann, C., Kleber, M. E., Lyytikäinen, L., Seppälä, I., Malik, R., Horimoto, A. R., Perez, M., Sinisalo, J., Aeschbacher, S., Thériault, S., Yao, J., Radmanesh, F., Weiss, S., Teumer, A., Choi, S. H., Weng, L., Clauss, S., Deo, R., Rader, D. J., Shah, S. H., Sun, A., Hopewell, J. C., Debette, S., Chauhan, G., Yang, Q., Worrall, B. B., Paré, G., Kamatani, Y., Hagemeijer, Y. P., Verweij, N., Siland, J. E., Kubo, M., Smith, J. D., Van Wagoner, D. R., Bis, J. C., Perz, S., Psaty, B. M., Ridker, P. M., Magnani, J. W., Harris, T. B., Launer, L. J., Shoemaker, M. B., Padmanabhan, S., Haessler, J., Bartz, T. M., Waldenberger, M., Lichtner, P., Arendt, M., Krieger, J. E., Kähönen, M., Risch, L., Mansur, A. J., Peters, A., Smith, B. H., Lind, L., Scott, S. A., Lu, Y., Bottinger, E. B., Hernesniemi, J., Lindgren, C. M., Wong, J. A., Huang, J., Eskola, M., Morris, A. P., Ford, I., Reiner, A. P., Delgado, G., Chen, L. Y., Chen, Y. I., Sandhu, R. K., Li, M., Boerwinkle, E., Eisele, L., Lannfelt, L., Rost, N., Anderson, C. D., Taylor, K. D., Campbell, A., Magnusson, P. K., Porteous, D., Hocking, L. J., Vlachopoulou, E., Pedersen, N. L., Nikus, K., Orho-Melander, M., Hamsten, A., Heeringa, J., Denny, J. C., Kriebel, J., Darbar, D., Newton-Cheh, C., Shaffer, C., Macfarlane, P. W., Heilmann-Heimbach, S., Almgren, P., Huang, P. L., Sotoodehnia, N., Soliman, E. Z., Uitterlinden, A. G., Hofman, A., Franco, O. H., Völker, U., Jöckel, K., Sinner, M. F., Lin, H. J., Guo, X., Dichgans, M., Ingelsson, E., Kooperberg, C., Melander, O., Loos, R. J., Laurikka, J., Conen, D., Rosand, J., van der Harst, P., Lokki, M., Kathiresan, S., Pereira, A., Jukema, J. W., Hayward, C., Rotter, J. I., März, W., Lehtimäki, T., Stricker, B. H., Chung, M. K., Felix, S. B., Gudnason, V., Alonso, A., Roden, D. M., Kääb, S., Chasman, D. I., Heckbert, S. R., Benjamin, E. J., Tanaka, T., Lunetta, K. L., Lubitz, S. A., Ellinor, P. T. 2017; 49 (6): 946-952

    Abstract

    Atrial fibrillation affects more than 33 million people worldwide and increases the risk of stroke, heart failure, and death. Fourteen genetic loci have been associated with atrial fibrillation in European and Asian ancestry groups. To further define the genetic basis of atrial fibrillation, we performed large-scale, trans-ancestry meta-analyses of common and rare variant association studies. The genome-wide association studies (GWAS) included 17,931 individuals with atrial fibrillation and 115,142 referents; the exome-wide association studies (ExWAS) and rare variant association studies (RVAS) involved 22,346 cases and 132,086 referents. We identified 12 new genetic loci that exceeded genome-wide significance, implicating genes involved in cardiac electrical and structural remodeling. Our results nearly double the number of known genetic loci for atrial fibrillation, provide insights into the molecular basis of atrial fibrillation, and may facilitate the identification of new potential targets for drug discovery.

    View details for DOI 10.1038/ng.3843

    View details for PubMedID 28416818

  • Ranking and characterization of established BMI and lipid associated loci as candidates for gene-environment interactions PLOS GENETICS Shungin, D., Deng, W. Q., Varga, T. V., Luan, J., Mihailov, E., Metspalu, A., Morris, A. P., Forouhi, N. G., Lindgren, C., Magnusson, P. E., Pedersen, N. L., Hallmans, G., Chu, A. Y., Justice, A. E., Graff, M., Winkler, T. W., Rose, L. M., Langenberg, C., Cupples, L., Ridker, P. M., Wareham, N. J., Ong, K. K., Loos, R. F., Chasman, D. I., Ingelsson, E., Kilpelainen, T. O., Scott, R. A., Magi, R., Pare, G., Franks, P. W., GIANT Consortium 2017; 13 (6): e1006812

    Abstract

    Phenotypic variance heterogeneity across genotypes at a single nucleotide polymorphism (SNP) may reflect underlying gene-environment (G×E) or gene-gene interactions. We modeled variance heterogeneity for blood lipids and BMI in up to 44,211 participants and investigated relationships between variance effects (Pv), G×E interaction effects (with smoking and physical activity), and marginal genetic effects (Pm). Correlations between Pv and Pm were stronger for SNPs with established marginal effects (Spearman's ρ = 0.401 for triglycerides, and ρ = 0.236 for BMI) compared to all SNPs. When Pv and Pm were compared for all pruned SNPs, only BMI was statistically significant (Spearman's ρ = 0.010). Overall, SNPs with established marginal effects were overrepresented in the nominally significant part of the Pv distribution (Pbinomial <0.05). SNPs from the top 1% of the Pm distribution for BMI had more significant Pv values (PMann-Whitney = 1.46×10-5), and the odds ratio of SNPs with nominally significant (<0.05) Pm and Pv was 1.33 (95% CI: 1.12, 1.57) for BMI. Moreover, BMI SNPs with nominally significant G×E interaction P-values (Pint<0.05) were enriched with nominally significant Pv values (Pbinomial = 8.63×10-9 and 8.52×10-7 for SNP × smoking and SNP × physical activity, respectively). We conclude that some loci with strong marginal effects may be good candidates for G×E, and variance-based prioritization can be used to identify them.

    View details for DOI 10.1371/journal.pgen.1006812

    View details for Web of Science ID 000404512600010

    View details for PubMedID 28614350

    View details for PubMedCentralID PMC5489225

  • An Expanded Genome-Wide Association Study of Type 2 Diabetes in Europeans. Diabetes Scott, R. A., Scott, L. J., Mägi, R., Marullo, L., Gaulton, K. J., Kaakinen, M., Pervjakova, N., Pers, T. H., Johnson, A. D., Eicher, J. D., Jackson, A. U., Ferreira, T., Lee, Y., Ma, C., Steinthorsdottir, V., Thorleifsson, G., Qi, L., Van Zuydam, N. R., Mahajan, A., Chen, H., Almgren, P., Voight, B. F., Grallert, H., Müller-Nurasyid, M., Ried, J. S., Rayner, W. N., Robertson, N., Karssen, L. C., van Leeuwen, E. M., Willems, S. M., Fuchsberger, C., Kwan, P., Teslovich, T. M., Chanda, P., Li, M., Lu, Y., Dina, C., Thuillier, D., Yengo, L., Jiang, L., Sparso, T., Kestler, H. A., Chheda, H., Eisele, L., Gustafsson, S., Frånberg, M., Strawbridge, R. J., Benediktsson, R., Hreidarsson, A. B., Kong, A., Sigurðsson, G., Kerrison, N. D., Luan, J., Liang, L., Meitinger, T., Roden, M., Thorand, B., Esko, T., Mihailov, E., Fox, C., Liu, C., Rybin, D., Isomaa, B., Lyssenko, V., Tuomi, T., Couper, D. J., Pankow, J. S., Grarup, N., Have, C. T., Jørgensen, M. E., Jørgensen, T., Linneberg, A., Cornelis, M. C., van Dam, R. M., Hunter, D. J., Kraft, P., Sun, Q., Edkins, S., Owen, K. R., Perry, J. R., Wood, A. R., Zeggini, E., Tajes-Fernandes, J., Abecasis, G. R., Bonnycastle, L. L., Chines, P. S., Stringham, H. M., Koistinen, H. A., Kinnunen, L., Sennblad, B., Mühleisen, T. W., Nöthen, M. M., Pechlivanis, S., Baldassarre, D., Gertow, K., Humphries, S. E., Tremoli, E., Klopp, N., Meyer, J., Steinbach, G., Wennauer, R., Eriksson, J. G., M?nnistö, S., Peltonen, L., Tikkanen, E., Charpentier, G., Eury, E., Lobbens, S., Gigante, B., Leander, K., McLeod, O., Bottinger, E. P., Gottesman, O., Ruderfer, D., Blüher, M., Kovacs, P., Tonjes, A., Maruthur, N. M., Scapoli, C., Erbel, R., Jöckel, K., Moebus, S., de Faire, U., Hamsten, A., Stumvoll, M., Deloukas, P., Donnelly, P. J., Frayling, T. M., Hattersley, A. T., Ripatti, S., Salomaa, V., Pedersen, N. L., Boehm, B. O., Bergman, R. N., Collins, F. S., Mohlke, K. L., Tuomilehto, J., Hansen, T., Pedersen, O., Barroso, I., Lannfelt, L., Ingelsson, E., Lind, L., Lindgren, C. M., Cauchi, S., Froguel, P., Loos, R. J., Balkau, B., Boeing, H., Franks, P. W., Gurrea, A. B., Palli, D., van der Schouw, Y. T., Altshuler, D., Groop, L. C., Langenberg, C., Wareham, N. J., Sijbrands, E., van Duijn, C. M., Florez, J. C., Meigs, J. B., Boerwinkle, E., Gieger, C., Strauch, K., Metspalu, A., Morris, A. D., Palmer, C. N., Hu, F. B., Thorsteinsdottir, U., Stefansson, K., Dupuis, J., Morris, A. P., Boehnke, M., McCarthy, M. I., Prokopenko, I. 2017

    Abstract

    To characterise type 2 diabetes (T2D) associated variation across the allele frequency spectrum, we conducted a meta-analysis of genome-wide association data from 26,676 T2D cases and 132,532 controls of European ancestry after imputation using the 1000 Genomes multi-ethnic reference panel. Promising association signals were followed-up in additional data sets (of 14,545 or 7,397 T2D cases and 38,994 or 71,604 controls). We identified 13 novel T2D-associated loci (p<5×10(-8)), including variants near the GLP2R, GIP, and HLA-DQA1 genes. Our analysis brought the total number of independent T2D associations to 128 distinct signals at 113 loci. Despite substantially increased sample size and more complete coverage of low-frequency variation, all novel associations were driven by common SNVs. Credible sets of potentially causal variants were generally larger than those based on imputation with earlier reference panels, consistent with resolution of causal signals to common risk haplotypes. Stratification of T2D-associated loci based on T2D-related quantitative trait associations revealed tissue-specific enrichment of regulatory annotations in pancreatic islet enhancers for loci influencing insulin secretion, and in adipocytes, monocytes and hepatocytes for insulin action-associated loci. These findings highlight the predominant role played by common variants of modest effect and the diversity of biological mechanisms influencing T2D pathophysiology.

    View details for DOI 10.2337/db16-1253

    View details for PubMedID 28566273

  • Leveraging Human Genetics to Understand the Relation of LDL Cholesterol with Type 2 Diabetes. Clinical chemistry Ingelsson, E., Knowles, J. W. 2017

    View details for DOI 10.1373/clinchem.2016.268565

    View details for PubMedID 28515096

  • Large-scale genome-wide analysis identifies genetic variants associated with cardiac structure and function JOURNAL OF CLINICAL INVESTIGATION Wild, P. S., Felix, J. F., Schillert, A., Teumer, A., Chen, M., Leening, M. J., Voelker, U., Grossmann, V., Brody, J. A., Irvin, M. R., Shah, S. J., Pramana, S., Lieb, W., Schmidt, R., Stanton, A. V., Malzahn, D., Smith, A. V., Sundstrom, J., Minelli, C., Ruggiero, D., Lyytikainen, L., Tiller, D., Smith, J. G., Monnereau, C., Di Tullio, M. R., Musani, S. K., Morrison, A. C., Pers, T. H., Morley, M., Kleber, M. E., Aragam, J., Benjamin, E. J., Bis, J. C., Bisping, E., Broeckel, U., Cheng, S., Deckers, J. W., Del Greco, F., Edelmann, F., Fornage, M., Franke, L., Friedrich, N., Harris, T. B., Hofer, E., Hofman, A., Huang, J., Hughes, A. D., Kahonen, M., Kruppa, J., Lackner, K. J., Lannfelt, L., Laskowski, R., Launer, L. J., Leosdottir, M., Lin, H., Lindgren, C. M., Loley, C., MacRae, C. A., Mascalzoni, D., Mayet, J., Medenwald, D., Morris, A. P., Mueller, C., Mueller-Nurasyid, M., Nappo, S., Nilsson, P. M., Nuding, S., Nutile, T., Peters, A., Pfeufer, A., Pietzner, D., Pramstaller, P. P., Raitakari, O. T., Rice, K. M., Rivadeneira, F., Rotter, J. I., Ruohonen, S. T., Sacco, R. L., Samdarshi, T. E., Schmidt, H., Sharp, A. S., Shields, D. C., Sorice, R., Sotoodehnia, N., Stricker, B. H., Surendran, P., Thom, S., Toeglhofer, A. M., Uitterlinden, A. G., Wachter, R., Voelzke, H., Ziegler, A., Muenzel, T., Maerz, W., Cappola, T. P., Hirschhorn, J. N., Mitchell, G. F., Smith, N. L., Fox, E. R., Dueker, N. D., Jaddoe, V. W., Melander, O., Russ, M., Lehtimaki, T., Ciullo, M., Hicks, A. A., Lind, L., Gudnason, V., Pieske, B., Barron, A. J., Zweiker, R., Schunkert, H., Ingelsson, E., Liu, K., Arnett, D. K., Psaty, B. M., Blankenberg, S., Larson, M. G., Felix, S. B., Franco, O. H., Zeller, T., Vasan, R. S., Doerr, M. 2017; 127 (5): 1798-1812

    Abstract

    Understanding the genetic architecture of cardiac structure and function may help to prevent and treat heart disease. This investigation sought to identify common genetic variations associated with inter-individual variability in cardiac structure and function.A GWAS meta-analysis of echocardiographic traits was performed, including 46,533 individuals from 30 studies (EchoGen consortium). The analysis included 16 traits of left ventricular (LV) structure, and systolic and diastolic function.The discovery analysis included 21 cohorts for structural and systolic function traits (n = 32,212) and 17 cohorts for diastolic function traits (n = 21,852). Replication was performed in 5 cohorts (n = 14,321) and 6 cohorts (n = 16,308), respectively. Besides 5 previously reported loci, the combined meta-analysis identified 10 additional genome-wide significant SNPs: rs12541595 near MTSS1 and rs10774625 in ATXN2 for LV end-diastolic internal dimension; rs806322 near KCNRG, rs4765663 in CACNA1C, rs6702619 near PALMD, rs7127129 in TMEM16A, rs11207426 near FGGY, rs17608766 in GOSR2, and rs17696696 in CFDP1 for aortic root diameter; and rs12440869 in IQCH for Doppler transmitral A-wave peak velocity. Findings were in part validated in other cohorts and in GWAS of related disease traits. The genetic loci showed associations with putative signaling pathways, and with gene expression in whole blood, monocytes, and myocardial tissue.The additional genetic loci identified in this large meta-analysis of cardiac structure and function provide insights into the underlying genetic architecture of cardiac structure and warrant follow-up in future functional studies.For detailed information per study, see Acknowledgments.

    View details for DOI 10.1172/JCI84840

    View details for Web of Science ID 000400381000022

    View details for PubMedID 28394258

  • Locus Due to Gene-Smoking Interactions. Circulation Saleheen, D., Zhao, W., Young, R., Nelson, C. P., Ho, W. K., Ferguson, J. F., Rasheed, A., Ou, K., Nurnberg, S. T., Bauer, R. C., Goel, A., Do, R., Stewart, A. F., Hartiala, J., Zhang, W., Thorleifsson, G., Strawbridge, R. J., Sinisalo, J., Kanoni, S., Sedaghat, S., Marouli, E., Kristiansson, K., Zhao, J. H., Scott, R., Gauguier, D., Shah, S. H., Smith, A. V., Van Zuydam, N., Cox, A. J., Willenborg, C., Kessler, T., Zeng, L., Province, M. A., Ganna, A., Lind, L., Pedersen, N. L., White, C. C., Joensuu, A., Kleber, M. E., Hall, A. S., März, W., Salomaa, V., O'Donnell, C., Ingelsson, E., Feitosa, M. F., Erdmann, J., Bowden, D. W., Palmer, C. N., Gudnason, V., de Faire, U., Zalloua, P., Wareham, N., Thompson, J. R., Kuulasmaa, K., Dedoussis, G., Perola, M., Dehghan, A., Chambers, J. C., Kooner, J., Allayee, H., Deloukas, P., McPherson, R., Stefansson, K., Schunkert, H., Kathiresan, S., Farrall, M., Frossard, P. M., Rader, D. J., Samani, N., Reilly, M. P. 2017

    Abstract

    Common diseases such as coronary heart disease (CHD) are complex in etiology. The interaction of genetic susceptibility with lifestyle factors may play a prominent role. However, gene-lifestyle interactions for CHD have been difficult to identify. Here, we investigate interaction of smoking behavior, a potent lifestyle factor, with genotypes that have been shown to associate with CHD risk.We analyzed data on 60 919 CHD cases and 80 243 controls from 29 studies for gene-smoking interactions for genetic variants at 45 loci previously reported to be associated with CHD risk. We also studied 5 loci associated with smoking behavior. Study-specific gene-smoking interaction effects were calculated and pooled using fixed-effects meta-analyses. Interaction analyses were declared to be significant at a P value of <1.0×10(-3) (Bonferroni correction for 50 tests).We identified novel gene-smoking interaction for a variant upstream of the ADAMTS7 gene. Every T allele of rs7178051 was associated with lower CHD risk by 12% in never-smokers (P=1.3×10(-16)) in comparison with 5% in ever-smokers (P=2.5×10(-4)), translating to a 60% loss of CHD protection conferred by this allelic variation in people who smoked tobacco (interaction P value=8.7×10(-5)). The protective T allele at rs7178051 was also associated with reduced ADAMTS7 expression in human aortic endothelial cells and lymphoblastoid cell lines. Exposure of human coronary artery smooth muscle cells to cigarette smoke extract led to induction of ADAMTS7. CONCLUSIONS: Allelic variation at rs7178051 that associates with reduced ADAMTS7 expression confers stronger CHD protection in never-smokers than in ever-smokers. Increased vascular ADAMTS7 expression may contribute to the loss of CHD protection in smokers.

    View details for DOI 10.1161/CIRCULATIONAHA.116.022069

    View details for PubMedID 28461624

  • Genome-wide meta-analysis of 241,258 adults accounting for smoking behaviour identifies novel loci for obesity traits NATURE COMMUNICATIONS Justice, A. E., Winkler, T. W., Feitosa, M. F., Graff, M., Fisher, V. A., Young, K., Barata, L., Deng, X., Czajkowski, J., Hadley, D., Ngwa, J. S., Ahluwalia, T. S., Chu, A. Y., Heard-Costa, N. L., Lim, E., Perez, J., Eicher, J. D., Kutalik, Z., Xue, L., Mahajan, A., Renstrom, F., Wu, J., Qi, Q., Ahmad, S., Alfred, T., Amin, N., Bielak, L. F., Bonnefond, A., Bragg, J., Cadby, G., Chittani, M., Coggeshall, S., Corre, T., Direk, N., Eriksson, J., Fischer, K., Gorski, M., Harder, M. N., Horikoshi, M., Huang, T., Huffman, J. E., Jackson, A. U., Justesen, J. M., Kanoni, S., Kinnunen, L., Kleber, M. E., Komulainen, P., Kumari, M., Lim, U., Luan, J., Lyytikainen, L., Mangino, M., Manichaikul, A., Marten, J., Middelberg, R. P., Mueller-Nurasyid, M., Navarro, P., Perusse, L., Pervjakova, N., Sarti, C., Smith, A. V., Smith, J. A., Stancakova, A., Strawbridge, R. J., Stringham, H. M., Sung, Y. J., Tanaka, T., Teumer, A., Trompet, S., van der Laan, S. W., van der Most, P. J., van Vliet-Ostaptchouk, J. V., Vedantam, S. L., Verweij, N., Vink, J. M., Vitart, V., Wu, Y., Yengo, L., Zhang, W., Zhao, J. H., Zimmermann, M. E., Zubair, N., Abecasis, G. R., Adair, L. S., Afaq, S., Afzal, U., Bakker, S. J., Bartz, T. M., Beilby, J., Bergman, R. N., Bergmann, S., Biffar, R., Blangero, J., Boerwinkle, E., Bonnycastle, L. L., Bottinger, E., Braga, D., Buckley, B. M., Buyske, S., Campbell, H., Chambers, J. C., Collins, F. S., Curran, J. E., de Borst, G. J., de Craen, A. J., de Geus, E. J., Dedoussis, G., Delgado, G. E., Den Ruijter, H. M., Eiriksdottir, G., Eriksson, A. L., Esko, T., Faul, J. D., Ford, I., Forrester, T., Gertow, K., Gigante, B., Glorioso, N., Gong, J., Grallert, H., Grammer, T. B., Grarup, N., Haitjema, S., Hallmans, G., Hamsten, A., Hansen, T., Harris, T. B., Hartman, C. A., Hassinen, M., Hastie, N. D., Heath, A. C., Hernandez, D., Hindorff, L., Hocking, L. J., Hollensted, M., Holmen, O. L., Homuth, G., Hottenga, J. J., Huang, J., Hung, J., Hutri-Kahonen, N., Ingelsson, E., James, A. L., Jansson, J., Jarvelin, M., Jhun, M. A., Jorgensen, M. E., Juonala, M., Kahonen, M., Karlsson, M., Koistinen, H. A., Kolcic, I., Kolovou, G., Kooperberg, C., Kramer, B. K., Kuusisto, J., Kvaloy, K., Lakka, T. A., Langenberg, C., Launer, L. J., Leander, K., Lee, N. R., Lind, L., Lindgren, C. M., Linneberg, A., Lobbens, S., Loh, M., Lorentzon, M., Luben, R., Lubke, G., Ludolph-Donislawski, A., Lupoli, S., Madden, P. A., Mannikko, R., Marques-Vidal, P., Martin, N. G., McKenzie, C. A., McKnight, B., Mellstrom, D., Menni, C., Montgomery, G. W., Musk, A. W., Narisu, N., Nauck, M., Nolte, I. M., Oldehinkel, A. J., Olden, M., Ong, K. K., Padmanabhan, S., Peyser, P. A., Pisinger, C., Porteous, D. J., Raitakari, O. T., Rankinen, T., Rao, D. C., Rasmussen-Torvik, L. J., Rawal, R., Rice, T., Ridker, P. M., Rose, L. M., Bien, S. A., Rudan, I., Sanna, S., Sarzynski, M. A., Sattar, N., Savonen, K., Schlessinger, D., Scholtens, S., Schurmann, C., Scott, R. A., Sennblad, B., Siemelink, M. A., Silbernagel, G., Slagboom, P. E., Snieder, H., Staessen, J. A., Stott, D. J., Swertz, M. A., Swift, A. J., Taylor, K. D., Tayo, B. O., Thorand, B., Thuillier, D., Tuomilehto, J., Uitterlinden, A. G., Vandenput, L., Vohl, M., Volzke, H., Vonk, J. M., Waeber, G., Waldenberger, M., Westendorp, R. G., Wild, S., Willemsen, G., Wolffenbuttel, B. H., Wong, A., Wright, A. F., Zhao, W., Zillikens, M. C., Baldassarre, D., Balkau, B., Bandinelli, S., Boger, C. A., Boomsma, D. I., Bouchard, C., Bruinenberg, M., Chasman, D. I., Chen, Y. I., Chines, P. S., Cooper, R. S., Cucca, F., Cusi, D., de Faire, U., Ferrucci, L., Franks, P. W., Froguel, P., Gordon-Larsen, P., Grabe, H., Gudnason, V., Haiman, C. A., Hayward, C., Hveem, K., Johnson, A. D., Jukema, W., Kardia, S. L., Kivimaki, M., Kooner, J. S., Kuh, D., Laakso, M., Lehtimaki, T., Le Marchand, L., Marz, W., McCarthy, M. I., Metspalu, A., Morris, A. P., Ohlsson, C., Palmer, L. J., Pasterkamp, G., Pedersen, O., Peters, A., Peters, U., Polasek, O., Psaty, B. M., Qi, L., Rauramaa, R., Smith, B. H., Sorensen, T. I., Strauch, K., Tiemeier, H., Tremoli, E., van der Harst, P., Vestergaard, H., Vollenweider, P., Wareham, N. J., Weir, D. R., Whitfield, J. B., Wilson, J. F., Tyrrell, J., Frayling, T. M., Barroso, I., Boehnke, M., Deloukas, P., Fox, C. S., Hirschhorn, J. N., Hunter, D. J., Spector, T. D., Strachan, D. P., van Duijn, C. M., Heid, I. M., Mohlke, K. L., Marchini, J., Loos, R. J., Kilpelainen, T. O., Liu, C., Borecki, I. B., North, K. E., Cupples, L. A. 2017; 8

    Abstract

    Few genome-wide association studies (GWAS) account for environmental exposures, like smoking, potentially impacting the overall trait variance when investigating the genetic contribution to obesity-related traits. Here, we use GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI. We identify 23 novel genetic loci, and 9 loci with convincing evidence of gene-smoking interaction (GxSMK) on obesity-related traits. We show consistent direction of effect for all identified loci and significance for 18 novel and for 5 interaction loci in an independent study sample. These loci highlight novel biological functions, including response to oxidative stress, addictive behaviour, and regulatory functions emphasizing the importance of accounting for environment in genetic analyses. Our results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution.

    View details for DOI 10.1038/ncomms14977

    View details for Web of Science ID 000400064600001

    View details for PubMedID 28443625

  • Mapping of 79 loci for 83 plasma protein biomarkers in cardiovascular disease. PLoS genetics Folkersen, L., Fauman, E., Sabater-Lleal, M., Strawbridge, R. J., Frånberg, M., Sennblad, B., Baldassarre, D., Veglia, F., Humphries, S. E., Rauramaa, R., de Faire, U., Smit, A. J., Giral, P., Kurl, S., Mannarino, E., Enroth, S., Johansson, Å., Enroth, S. B., Gustafsson, S., Lind, L., Lindgren, C., Morris, A. P., Giedraitis, V., Silveira, A., Franco-Cereceda, A., Tremoli, E., Gyllensten, U., Ingelsson, E., Brunak, S., Eriksson, P., Ziemek, D., Hamsten, A., Mälarstig, A. 2017; 13 (4)

    Abstract

    Recent advances in highly multiplexed immunoassays have allowed systematic large-scale measurement of hundreds of plasma proteins in large cohort studies. In combination with genotyping, such studies offer the prospect to 1) identify mechanisms involved with regulation of protein expression in plasma, and 2) determine whether the plasma proteins are likely to be causally implicated in disease. We report here the results of genome-wide association (GWA) studies of 83 proteins considered relevant to cardiovascular disease (CVD), measured in 3,394 individuals with multiple CVD risk factors. We identified 79 genome-wide significant (p<5e-8) association signals, 55 of which replicated at P<0.0007 in separate validation studies (n = 2,639 individuals). Using automated text mining, manual curation, and network-based methods incorporating information on expression quantitative trait loci (eQTL), we propose plausible causal mechanisms for 25 trans-acting loci, including a potential post-translational regulation of stem cell factor by matrix metalloproteinase 9 and receptor-ligand pairs such as RANK-RANK ligand. Using public GWA study data, we further evaluate all 79 loci for their causal effect on coronary artery disease, and highlight several potentially causal associations. Overall, a majority of the plasma proteins studied showed evidence of regulation at the genetic level. Our results enable future studies of the causal architecture of human disease, which in turn should aid discovery of new drug targets.

    View details for DOI 10.1371/journal.pgen.1006706

    View details for PubMedID 28369058

  • Alterations in Multiple Lifestyle Factors in Subjects with the Metabolic Syndrome Independently of Obesity METABOLIC SYNDROME AND RELATED DISORDERS Roos, V., Elmstahl, S., Ingelsson, E., Sundstrom, J., Arnlov, J., Lind, L. 2017; 15 (3): 118-123

    Abstract

    Many lifestyle factors have been associated with the metabolic syndrome (MetS). However, most of these studies have not considered the potential impact of obesity and have often only investigated one lifestyle factor at the time. We aimed to investigate the interplay between body mass index (BMI) and MetS with respect to multiple lifestyle factors.BMI and MetS [National Cholesterol Education Program (NCEP)/Adult Treatment Panel III criteria] were assessed in a sample of 18,880 subjects aged 45-75 years from the population-based EpiHealth study. Participants were categorized into six groups according to BMI category (normal weight/BMI <25 kg/m(2), overweight/BMI 25-30 kg/m(2), and obesity/BMI >30 kg/m(2)) and MetS status (+/-, NCEP criteria). A wide range of lifestyle factors related to physical activity, smoking, alcohol, sleep quality, working conditions, quality of life and stress, and eating patterns were assessed using a questionnaire.Prevalent MetS (23% in the sample) was associated with less physical activity (P < 0.0001), more TV watching (P < 0.0001), more years of smoking (P < 0.0001), lower education level (P = 0.007), and experiencing a poor general quality of life (P < 0.0001). These lifestyle factors were all associated with MetS, independently of each other and independently of BMI. Similar results were generated when number of MetS components and presence/absence of individual MetS components were used as outcomes.This cross-sectional study identified alterations in a number of lifestyle factors associated with MetS independently of each other and independently of BMI. Future longitudinal studies are needed to assess causal and temporal relationships between lifestyle factors and MetS development.

    View details for DOI 10.1089/met.2016.0120

    View details for Web of Science ID 000397585500003

    View details for PubMedID 28339343

  • Variant Enriched in the Finnish Population is Associated With Fasting Insulin Levels and Type 2 Diabetes Risk. Diabetes Manning, A., Highland, H. M., Gasser, J., Sim, X., Tukiainen, T., Fontanillas, P., Grarup, N., Rivas, M. A., Mahajan, A., Locke, A. E., Cingolani, P., Pers, T. H., Viñuela, A., Brown, A. A., Wu, Y., Flannick, J., Fuchsberger, C., Gamazon, E. R., Gaulton, K. J., Im, H. K., Teslovich, T. M., Blackwell, T. W., Bork-Jensen, J., Burtt, N. P., Chen, Y., Green, T., Hartl, C., Kang, H. M., Kumar, A., Ladenvall, C., Ma, C., Moutsianas, L., Pearson, R. D., Perry, J. R., Rayner, N. W., Robertson, N. R., Scott, L. J., van de Bunt, M., Eriksson, J. G., Jula, A., Koskinen, S., Lehtimäki, T., Palotie, A., Raitakari, O. T., Jacobs, S. B., Wessel, J., Chu, A. Y., Scott, R. A., Goodarzi, M. O., Blancher, C., Buck, G., Buck, D., Chines, P. S., Gabriel, S., Gjesing, A. P., Groves, C. J., Hollensted, M., Huyghe, J. R., Jackson, A. U., Jun, G., Justesen, J. M., Mangino, M., Murphy, J., Neville, M., Onofrio, R., Small, K. S., Stringham, H. M., Trakalo, J., Banks, E., Carey, J., Carneiro, M. O., DePristo, M., Farjoun, Y., Fennell, T., Goldstein, J. I., Grant, G., Hrabé de Angelis, M., Maguire, J., Neale, B. M., Poplin, R., Purcell, S., Schwarzmayr, T., Shakir, K., Smith, J. D., Strom, T. M., Wieland, T., Lindstrom, J., Brandslund, I., Christensen, C., Surdulescu, G. L., Lakka, T. A., Doney, A. S., Nilsson, P., Wareham, N. J., Langenberg, C., Varga, T. V., Franks, P. W., Rolandsson, O., Rosengren, A. H., Farook, V. S., Thameem, F., Puppala, S., Kumar, S., Lehman, D. M., Jenkinson, C. P., Curran, J. E., Hale, D. E., Fowler, S. P., Arya, R., DeFronzo, R. A., Abboud, H. E., Syvänen, A., Hicks, P. J., Palmer, N. D., Ng, M. C., Bowden, D. W., Freedman, B. I., Esko, T., Mägi, R., Milani, L., Mihailov, E., Metspalu, A., Narisu, N., Kinnunen, L., Bonnycastle, L. L., Swift, A., Pasko, D., Wood, A. R., Fadista, J., Pollin, T. I., Barzilai, N., Atzmon, G., Glaser, B., Thorand, B., Strauch, K., Peters, A., Roden, M., Müller-Nurasyid, M., Liang, L., Kriebel, J., Illig, T., Grallert, H., Gieger, C., Meisinger, C., Lannfelt, L., Musani, S. K., Griswold, M., Taylor, H. A., Wilson, G., Correa, A., Oksa, H., Scott, W. R., Afzal, U., Tan, S., Loh, M., Chambers, J. C., Sehmi, J., Kooner, J. S., Lehne, B., Cho, Y. S., Lee, J., Han, B., Käräjämäki, A., Qi, Q., Qi, L., Huang, J., Hu, F. B., Melander, O., Orho-Melander, M., Below, J. E., Aguilar, D., Wong, T. Y., Liu, J., Khor, C., Chia, K. S., Lim, W. Y., Cheng, C., Chan, E., Tai, E. S., Aung, T., Linneberg, A., Isomaa, B., Meitinger, T., Tuomi, T., Hakaste, L., Kravic, J., Jørgensen, M. E., Lauritzen, T., Deloukas, P., Stirrups, K. E., Owen, K. R., Farmer, A. J., Frayling, T. M., O'Rahilly, S. P., Walker, M., Levy, J. C., Hodgkiss, D., Hattersley, A. T., Kuulasmaa, T., Stancáková, A., Barroso, I., Bharadwaj, D., Chan, J., Chandak, G. R., Daly, M. J., Donnelly, P. J., Ebrahim, S. B., Elliott, P., Fingerlin, T., Froguel, P., Hu, C., Jia, W., Ma, R. C., McVean, G., Park, T., Prabhakaran, D., Sandhu, M., Scott, J., Sladek, R., Tandon, N., Teo, Y. Y., Zeggini, E., Watanabe, R. M., Koistinen, H. A., Kesaniemi, Y. A., Uusitupa, M., Spector, T. D., Salomaa, V., Rauramaa, R., Palmer, C. N., Prokopenko, I., Morris, A. D., Bergman, R. N., Collins, F. S., Lind, L., Ingelsson, E., Tuomilehto, J., Karpe, F., Groop, L., Jørgensen, T., Hansen, T., Pedersen, O., Kuusisto, J., Abecasis, G., Bell, G. I., Blangero, J., Cox, N. J., Duggirala, R., Seielstad, M., Wilson, J. G., Dupuis, J., Ripatti, S., Hanis, C. L., Florez, J. C., Mohlke, K. L., Meigs, J. B., Laakso, M., Morris, A. P., Boehnke, M., Altshuler, D., McCarthy, M. I., Gloyn, A. L., Lindgren, C. M. 2017

    Abstract

    To identify novel coding association signals and facilitate characterization of mechanisms influencing glycemic traits and type 2 diabetes risk, we analyzed 109,215 variants derived from exome array genotyping together with an additional 390,225 variants from exome sequence in up to 39,339 normoglycemic individuals from five ancestry groups. We identified a novel association between the coding variant (p.Pro50Thr) in AKT2 and fasting plasma insulin (FI), a gene in which rare fully penetrant mutations are causal for monogenic glycemic disorders. The low-frequency allele is associated with a 12% increase in FI levels. This variant is present at 1.1% frequency in Finns but virtually absent in individuals from other ancestries. Carriers of the FI-increasing allele had increased 2-h insulin values, decreased insulin sensitivity, and increased risk of type 2 diabetes (odds ratio 1.05). In cellular studies, the AKT2-Thr50 protein exhibited a partial loss of function. We extend the allelic spectrum for coding variants in AKT2 associated with disorders of glucose homeostasis and demonstrate bidirectional effects of variants within the pleckstrin homology domain of AKT2.

    View details for DOI 10.2337/db16-1329

    View details for PubMedID 28341696

  • Association of Pregnancy Complications and Characteristics With Future Risk of Elevated Blood Pressure: The Västerbotten Intervention Program. Hypertension Parikh, N. I., Norberg, M., Ingelsson, E., Cnattingius, S., Vasan, R. S., Domellöf, M., Jansson, J. H., Edstedt Bonamy, A. 2017; 69 (3): 475-483

    Abstract

    Pregnancy characteristics are associated with risk of cardiovascular diseases, but their independent associations with hypertension or blood pressure (BP) levels remain uncertain. We linked the Swedish Medical Birth Register with Västerbotten Intervention Program data (Northern Sweden). Using linear and logistic regression, we related pregnancy factors in any prior pregnancy with BP and hypertension at 40 years of age in 15 896 parous women free of prepregnancy hypertension. Pregnancy factors included parity, age at first delivery, preeclampsia, gestational diabetes mellitus, placental abruption, shortest gestational age small for gestational age baby (

    View details for DOI 10.1161/HYPERTENSIONAHA.116.08121

    View details for PubMedID 28137991

  • Association of Pregnancy Complications and Characteristics With Future Risk of Elevated Blood Pressure The Vasterbotten Intervention Program HYPERTENSION Parikh, N. I., Norberg, M., Ingelsson, E., Cnattingius, S., Vasan, R. S., Domellof, M., Jansson, J. H., Bonamy, A. E. 2017; 69 (3): 475-483
  • Genome-wide association analysis identifies novel blood pressure loci and offers biological insights into cardiovascular risk NATURE GENETICS Warren, H. R., Evangelou, E., Cabrera, C. P., Gao, H., Ren, M., Mifsud, B., Ntalla, I., Surendran, P., Liu, C., Cook, J. P., Kraja, A. T., Drenos, F., Loh, M., Verweij, N., Marten, J., Karaman, I., Lepe, M. P., O'Reilly, P. F., Knight, J., Snieder, H., Kato, N., He, J., Tai, E. S., Said, M. A., Porteous, D., Alver, M., Poulter, N., Farrall, M., Gansevoort, R. T., Padmanabhan, S., Magi, R., Stanton, A., Connell, J., Bakker, S. J., Metspalu, A., Shields, D. C., Thom, S., Brown, M., Sever, P., Esko, T., Hayward, C., van der Harst, P., Saleheen, D., Chowdhury, R., Chambers, J. C., Chasman, D. I., Chakravarti, A., Newton-Cheh, C., Lindgren, C. M., Levy, D., Kooner, J. S., Keavney, B., Tomaszewski, M., Samani, N. J., Howson, J. M., Tobin, M. D., Munroe, P. B., Ehret, G. B., Wain, L. V., Barnes, M. R., Tzoulaki, J., Caulfield, M. J., Elliott, P., Wain, L. V., Vaez, A., Jansen, R., Joehanes, R., van der Most, P. J., Erzurumluoglu, A. M., O'Reilly, P., Cabrera, C. P., Warren, H. R., Rose, L. M., Verwoert, G. C., Hottenga, J., Strawbridge, R. J., Esko, T., Arking, D. E., Hwang, S., Guo, X., Kutalik, Z., Trompet, S., Shrine, N., Teumer, A., Ried, J. S., Bis, J. C., Smith, A. V., Amin, N., Nolte, I. M., Lyytikainen, L., Mahajan, A., Wareham, N. J., Hofer, E., Joshi, P. K., Kristiansson, K., Traglia, M., Havulinna, A. S., Goel, A., Nalls, M. A., Sober, S., Vuckovic, D., Luan, J., Del Greco M, F., Ayers, K. L., Marrugat, J., Ruggiero, D., Lopez, L. M., Niiranen, T., Enroth, S., Jackson, A. U., Nelson, C. P., Huffman, J. E., Zhang, W., Marten, J., Gandin, I., Harris, S. E., Zemonik, T., Lu, Y., Evangelou, E., Shah, N., de Borst, M. H., Mangino, M., Prins, B. P., Campbell, A., Li-Gao, R., Chauhan, G., Oldmeadow, C., Abecasis, G., Abedi, M., Barbieri, C. M., Barnes, M. R., Batini, C., Blake, T., Boehnke, M., Bottinger, E. P., Braund, P. S., Brown, M., Brumat, M., Campbell, H., Chambers, J. C., Cocca, M., Collins, F., Connell, J., Cordell, H. J., Damman, J. J., Davies, G., de Geus, E. J., de Mutsert, R., Deelen, J., Demirkale, Y., Doney, A. S., Dorr, M., Farrall, M., Ferreira, T., Franberg, M., Gao, H., Giedraitis, V., Gieger, C., Giulianini, F., Gow, A. J., Hamsten, A., Harris, T. B., Hofman, A., Holliday, E. G., Jarvelin, M., Johansson, A., Johnson, A. D., Jousilahti, P., Jula, A., Kahonen, M., Kathiresan, S., Khaw, K., Kolcic, I., Koskinen, S., Langenberg, C., Larson, M., Launer, L. J., Lehne, B., Liewald, D. C., Lin, L., Lind, L., Mach, F., Mamasoula, C., Menni, C., Mifsud, B., Milaneschi, Y., Morgan, A., Morris, A. D., Morrison, A. C., Munson, P. J., Nandakumar, P., Quang Tri Nguyen, Q. T., Nutile, T., Oldehinkel, A. J., Oostra, B. A., Org, E., Padmanabhan, S., Palotie, A., Pare, G., Pattie, A., Penninx, B. W., Poulter, N., Pramstaller, P. P., Raitakari, O. T., Ren, M., Rice, K., Ridker, P. M., Riese, H., Ripatti, S., Robino, A., Rotter, J. I., Rudan, I., Saba, Y., Saint Pierre, A., Sala, C. F., Sarin, A., Schmidt, R., Scott, R., Seelen, M. A., Shields, D. C., Siscovick, D., Sorice, R., Stanton, A., Stott, D. J., Sundstrom, J., Swertz, M., Taylor, K. D., Thom, S., Tzoulaki, I., Tzourio, C., Uitterlinden, A. G., Volker, U., Vollenweider, P., Wild, S., Willemsen, G., Wright, A. F., Yao, J., Theriault, S., Conen, D., John, A., Sever, P., Debette, S., Mook-Kanamori, D. O., Zeggini, E., Spector, T. D., van der Harst, P., Palmer, C. N., Vergnaud, A., Loos, R. J., Polasek, O., Starr, J. M., Girotto, G., Hayward, C., Kooner, J. S., Lindgren, C. M., Vitart, V., Samani, N. J., Tuomilehto, J., Gyllensten, U., Knekt, P., Deary, I. J., Ciullo, M., Elosua, R., Keavney, B. D., Hicks, A. A., Scott, R. A., Gasparini, P., Laan, M., Liu, Y., Watkins, H., Hartman, C. A., Salomaa, V., Toniolo, D., Perola, M., Wilson, J. F., Schmidt, H., Zhao, J. H., Lehtimaki, T., van Duijn, C. M., Gudnason, V., Psaty, B. M., Peters, A., Rettig, R., James, A., Jukema, J. W., Strachan, D. P., Palmas, W., Metspalu, A., Ingelsson, E., Boomsma, D. I., Franco, O. H., Bochud, M., Newton-Cheh, C., Munroe, P. B., Elliott, P., Chasman, D. I., Chakravarti, A., Knight, J., Morris, A. P., Levy, D., Tobin, M. D., Snieder, H., Caulfield, M. J., Ehret, G. B. 2017; 49 (3): 403-415

    Abstract

    Elevated blood pressure is the leading heritable risk factor for cardiovascular disease worldwide. We report genetic association of blood pressure (systolic, diastolic, pulse pressure) among UK Biobank participants of European ancestry with independent replication in other cohorts, and robust validation of 107 independent loci. We also identify new independent variants at 11 previously reported blood pressure loci. In combination with results from a range of in silico functional analyses and wet bench experiments, our findings highlight new biological pathways for blood pressure regulation enriched for genes expressed in vascular tissues and identify potential therapeutic targets for hypertension. Results from genetic risk score models raise the possibility of a precision medicine approach through early lifestyle intervention to offset the impact of blood pressure-raising genetic variants on future cardiovascular disease risk.

    View details for DOI 10.1038/ng.3768

    View details for Web of Science ID 000394917800014

    View details for PubMedCentralID PMC5972004

  • Rare and low-frequency coding variants alter human adult height. Nature Marouli, E., Graff, M., Medina-Gomez, C., Lo, K. S., Wood, A. R., Kjaer, T. R., Fine, R. S., Lu, Y., Schurmann, C., Highland, H. M., Rüeger, S., Thorleifsson, G., Justice, A. E., Lamparter, D., Stirrups, K. E., Turcot, V., Young, K. L., Winkler, T. W., Esko, T., Karaderi, T., Locke, A. E., Masca, N. G., Ng, M. C., Mudgal, P., Rivas, M. A., Vedantam, S., Mahajan, A., Guo, X., Abecasis, G., Aben, K. K., Adair, L. S., Alam, D. S., Albrecht, E., Allin, K. H., Allison, M., Amouyel, P., Appel, E. V., Arveiler, D., Asselbergs, F. W., Auer, P. L., Balkau, B., Banas, B., Bang, L. E., Benn, M., Bergmann, S., Bielak, L. F., Blüher, M., Boeing, H., Boerwinkle, E., Böger, C. A., Bonnycastle, L. L., Bork-Jensen, J., Bots, M. L., Bottinger, E. P., Bowden, D. W., Brandslund, I., Breen, G., Brilliant, M. H., Broer, L., Burt, A. A., Butterworth, A. S., Carey, D. J., Caulfield, M. J., Chambers, J. C., Chasman, D. I., Chen, Y. I., Chowdhury, R., Christensen, C., Chu, A. Y., Cocca, M., Collins, F. S., Cook, J. P., Corley, J., Galbany, J. C., Cox, A. J., Cuellar-Partida, G., Danesh, J., Davies, G., de Bakker, P. I., de Borst, G. J., De Denus, S., de Groot, M. C., de Mutsert, R., Deary, I. J., Dedoussis, G., Demerath, E. W., den Hollander, A. I., Dennis, J. G., Di Angelantonio, E., Drenos, F., Du, M., Dunning, A. M., Easton, D. F., Ebeling, T., Edwards, T. L., Ellinor, P. T., Elliott, P., Evangelou, E., Farmaki, A., Faul, J. D., Feitosa, M. F., Feng, S., Ferrannini, E., Ferrario, M. M., Ferrieres, J., Florez, J. C., Ford, I., Fornage, M., Franks, P. W., Frikke-Schmidt, R., Galesloot, T. E., Gan, W., Gandin, I., Gasparini, P., Giedraitis, V., Giri, A., Girotto, G., Gordon, S. D., Gordon-Larsen, P., Gorski, M., Grarup, N., Grove, M. L., Gudnason, V., Gustafsson, S., Hansen, T., Harris, K. M., Harris, T. B., Hattersley, A. T., Hayward, C., He, L., Heid, I. M., Heikkilä, K., Helgeland, Ø., Hernesniemi, J., Hewitt, A. W., Hocking, L. J., Hollensted, M., Holmen, O. L., Hovingh, G. K., Howson, J. M., Hoyng, C. B., Huang, P. L., Hveem, K., Ikram, M. A., Ingelsson, E., Jackson, A. U., Jansson, J., Jarvik, G. P., Jensen, G. B., Jhun, M. A., Jia, Y., Jiang, X., Johansson, S., Jørgensen, M. E., Jørgensen, T., Jousilahti, P., Jukema, J. W., Kahali, B., Kahn, R. S., Kähönen, M., Kamstrup, P. R., Kanoni, S., Kaprio, J., Karaleftheri, M., Kardia, S. L., Karpe, F., Kee, F., Keeman, R., Kiemeney, L. A., Kitajima, H., Kluivers, K. B., Kocher, T., Komulainen, P., Kontto, J., Kooner, J. S., Kooperberg, C., Kovacs, P., Kriebel, J., Kuivaniemi, H., Küry, S., Kuusisto, J., La Bianca, M., Laakso, M., Lakka, T. A., Lange, E. M., Lange, L. A., Langefeld, C. D., Langenberg, C., Larson, E. B., Lee, I., Lehtimäki, T., Lewis, C. E., Li, H., Li, J., Li-Gao, R., Lin, H., Lin, L., Lin, X., Lind, L., Lindström, J., Linneberg, A., Liu, Y., Liu, Y., Lophatananon, A., Luan, J., Lubitz, S. A., Lyytikäinen, L., Mackey, D. A., Madden, P. A., Manning, A. K., Männistö, S., Marenne, G., Marten, J., Martin, N. G., Mazul, A. L., Meidtner, K., Metspalu, A., Mitchell, P., Mohlke, K. L., Mook-Kanamori, D. O., Morgan, A., Morris, A. D., Morris, A. P., Müller-Nurasyid, M., Munroe, P. B., Nalls, M. A., Nauck, M., Nelson, C. P., Neville, M., Nielsen, S. F., Nikus, K., Njølstad, P. R., Nordestgaard, B. G., Ntalla, I., O'Connel, J. R., Oksa, H., Loohuis, L. M., Ophoff, R. A., Owen, K. R., Packard, C. J., Padmanabhan, S., Palmer, C. N., Pasterkamp, G., Patel, A. P., Pattie, A., Pedersen, O., Peissig, P. L., Peloso, G. M., Pennell, C. E., Perola, M., Perry, J. A., Perry, J. R., Person, T. N., Pirie, A., Polasek, O., Posthuma, D., Raitakari, O. T., Rasheed, A., Rauramaa, R., Reilly, D. F., Reiner, A. P., Renström, F., Ridker, P. M., Rioux, J. D., Robertson, N., Robino, A., Rolandsson, O., Rudan, I., Ruth, K. S., Saleheen, D., Salomaa, V., Samani, N. J., Sandow, K., Sapkota, Y., Sattar, N., Schmidt, M. K., Schreiner, P. J., Schulze, M. B., Scott, R. A., Segura-Lepe, M. P., Shah, S., Sim, X., Sivapalaratnam, S., Small, K. S., Smith, A. V., Smith, J. A., Southam, L., Spector, T. D., Speliotes, E. K., Starr, J. M., Steinthorsdottir, V., Stringham, H. M., Stumvoll, M., Surendran, P., 't Hart, L. M., Tansey, K. E., Tardif, J., Taylor, K. D., Teumer, A., Thompson, D. J., Thorsteinsdottir, U., Thuesen, B. H., Tönjes, A., Tromp, G., Trompet, S., Tsafantakis, E., Tuomilehto, J., Tybjaerg-Hansen, A., Tyrer, J. P., Uher, R., Uitterlinden, A. G., Ulivi, S., van der Laan, S. W., van der Leij, A. R., van Duijn, C. M., van Schoor, N. M., van Setten, J., Varbo, A., Varga, T. V., Varma, R., Edwards, D. R., Vermeulen, S. H., Vestergaard, H., Vitart, V., Vogt, T. F., Vozzi, D., Walker, M., Wang, F., Wang, C. A., Wang, S., Wang, Y., Wareham, N. J., Warren, H. R., Wessel, J., Willems, S. M., Wilson, J. G., Witte, D. R., Woods, M. O., Wu, Y., Yaghootkar, H., Yao, J., Yao, P., Yerges-Armstrong, L. M., Young, R., Zeggini, E., Zhan, X., Zhang, W., Zhao, J. H., Zhao, W., Zhao, W., Zheng, H., Zhou, W., Rotter, J. I., Boehnke, M., Kathiresan, S., McCarthy, M. I., Willer, C. J., Stefansson, K., Borecki, I. B., Liu, D. J., North, K. E., Heard-Costa, N. L., Pers, T. H., Lindgren, C. M., Oxvig, C., Kutalik, Z., Rivadeneira, F., Loos, R. J., Frayling, T. M., Hirschhorn, J. N., Deloukas, P., Lettre, G. 2017; 542 (7640): 186-190

    Abstract

    Height is a highly heritable, classic polygenic trait with approximately 700 common associated variants identified through genome-wide association studies so far. Here, we report 83 height-associated coding variants with lower minor-allele frequencies (in the range of 0.1-4.8%) and effects of up to 2 centimetres per allele (such as those in IHH, STC2, AR and CRISPLD2), greater than ten times the average effect of common variants. In functional follow-up studies, rare height-increasing alleles of STC2 (giving an increase of 1-2 centimetres per allele) compromised proteolytic inhibition of PAPP-A and increased cleavage of IGFBP-4 in vitro, resulting in higher bioavailability of insulin-like growth factors. These 83 height-associated variants overlap genes that are mutated in monogenic growth disorders and highlight new biological candidates (such as ADAMTS3, IL11RA and NOX4) and pathways (such as proteoglycan and glycosaminoglycan synthesis) involved in growth. Our results demonstrate that sufficiently large sample sizes can uncover rare and low-frequency variants of moderate-to-large effect associated with polygenic human phenotypes, and that these variants implicate relevant genes and pathways.

    View details for DOI 10.1038/nature21039

    View details for PubMedID 28146470

    View details for PubMedCentralID PMC5302847

  • PCSK9 genetic variants and risk of type 2 diabetes: a mendelian randomisation study. The lancet. Diabetes & endocrinology Schmidt, A. F., Swerdlow, D. I., Holmes, M. V., Patel, R. S., Fairhurst-Hunter, Z., Lyall, D. M., Hartwig, F. P., Horta, B. L., Hyppönen, E., Power, C., Moldovan, M., van Iperen, E., Hovingh, G. K., Demuth, I., Norman, K., Steinhagen-Thiessen, E., Demuth, J., Bertram, L., Liu, T., Coassin, S., Willeit, J., Kiechl, S., Willeit, K., Mason, D., Wright, J., Morris, R., Wanamethee, G., Whincup, P., Ben-Shlomo, Y., McLachlan, S., Price, J. F., Kivimaki, M., Welch, C., Sanchez-Galvez, A., Marques-Vidal, P., Nicolaides, A., Panayiotou, A. G., Onland-Moret, N. C., van der Schouw, Y. T., Matullo, G., Fiorito, G., Guarrera, S., Sacerdote, C., Wareham, N. J., Langenberg, C., Scott, R., Luan, J., Bobak, M., Malyutina, S., Pajak, A., Kubinova, R., Tamosiunas, A., Pikhart, H., Husemoen, L. L., Grarup, N., Pedersen, O., Hansen, T., Linneberg, A., Simonsen, K. S., Cooper, J., Humphries, S. E., Brilliant, M., Kitchner, T., Hakonarson, H., Carrell, D. S., McCarty, C. A., Kirchner, H. L., Larson, E. B., Crosslin, D. R., de Andrade, M., Roden, D. M., Denny, J. C., Carty, C., Hancock, S., Attia, J., Holliday, E., O'Donnell, M., Yusuf, S., Chong, M., Pare, G., van der Harst, P., Said, M. A., Eppinga, R. N., Verweij, N., Snieder, H., Christen, T., Mook-Kanamori, D. O., Gustafsson, S., Lind, L., Ingelsson, E., Pazoki, R., Franco, O., Hofman, A., Uitterlinden, A., Dehghan, A., Teumer, A., Baumeister, S., Dörr, M., Lerch, M. M., Völker, U., Völzke, H., Ward, J., Pell, J. P., Smith, D. J., Meade, T., Maitland-van der Zee, A. H., Baranova, E. V., Young, R., Ford, I., Campbell, A., Padmanabhan, S., Bots, M. L., Grobbee, D. E., Froguel, P., Thuillier, D., Balkau, B., Bonnefond, A., Cariou, B., Smart, M., Bao, Y., Kumari, M., Mahajan, A., Ridker, P. M., Chasman, D. I., Reiner, A. P., Lange, L. A., Ritchie, M. D., Asselbergs, F. W., Casas, J., Keating, B. J., Preiss, D., Hingorani, A. D., Sattar, N. 2017; 5 (2): 97-105

    Abstract

    Statin treatment and variants in the gene encoding HMG-CoA reductase are associated with reductions in both the concentration of LDL cholesterol and the risk of coronary heart disease, but also with modest hyperglycaemia, increased bodyweight, and modestly increased risk of type 2 diabetes, which in no way offsets their substantial benefits. We sought to investigate the associations of LDL cholesterol-lowering PCSK9 variants with type 2 diabetes and related biomarkers to gauge the likely effects of PCSK9 inhibitors on diabetes risk.In this mendelian randomisation study, we used data from cohort studies, randomised controlled trials, case control studies, and genetic consortia to estimate associations of PCSK9 genetic variants with LDL cholesterol, fasting blood glucose, HbA1c, fasting insulin, bodyweight, waist-to-hip ratio, BMI, and risk of type 2 diabetes, using a standardised analysis plan, meta-analyses, and weighted gene-centric scores.Data were available for more than 550 000 individuals and 51 623 cases of type 2 diabetes. Combined analyses of four independent PCSK9 variants (rs11583680, rs11591147, rs2479409, and rs11206510) scaled to 1 mmol/L lower LDL cholesterol showed associations with increased fasting glucose (0·09 mmol/L, 95% CI 0·02 to 0·15), bodyweight (1·03 kg, 0·24 to 1·82), waist-to-hip ratio (0·006, 0·003 to 0·010), and an odds ratio for type diabetes of 1·29 (1·11 to 1·50). Based on the collected data, we did not identify associations with HbA1c (0·03%, -0·01 to 0·08), fasting insulin (0·00%, -0·06 to 0·07), and BMI (0·11 kg/m(2), -0·09 to 0·30).PCSK9 variants associated with lower LDL cholesterol were also associated with circulating higher fasting glucose concentration, bodyweight, and waist-to-hip ratio, and an increased risk of type 2 diabetes. In trials of PCSK9 inhibitor drugs, investigators should carefully assess these safety outcomes and quantify the risks and benefits of PCSK9 inhibitor treatment, as was previously done for statins.British Heart Foundation, and University College London Hospitals NHS Foundation Trust (UCLH) National Institute for Health Research (NIHR) Biomedical Research Centre.

    View details for DOI 10.1016/S2213-8587(16)30396-5

    View details for PubMedID 27908689

    View details for PubMedCentralID PMC5266795

  • Metabolic Syndrome Development During Aging with Special Reference to Obesity Without the Metabolic Syndrome METABOLIC SYNDROME AND RELATED DISORDERS Roos, V., Elmstahl, S., Ingelsson, E., Sundstrom, J., Arnlov, J., Lind, L. 2017; 15 (1): 36-43

    Abstract

    Obesity and its associated metabolic complications continue to increase worldwide. We investigated the development of metabolic syndrome (MetS) during aging in relation to body mass index (BMI) and exercise habits. We assigned special emphasis to the metabolic stability in individuals with obesity, but without MetS, a condition often referred to as metabolically healthy obesity.Cross-sectional analysis was carried out in a sample of 19,129 men and women aged 45-75 years from the EpiHealth study. In addition, longitudinal analyses were carried out in the ULSAM study (2322 men at baseline followed from age 50 to age 77) and in the PIVUS study (1016 men and women at baseline followed from age 70 to age 80). Participants were categorized into six groups according to BMI category (normal weight/BMI <25 kg/m(2), overweight/BMI 25-30 kg/m(2), and obesity/BMI >30 kg/m(2)) and MetS status (+/-, National Cholesterol Education Program criteria).MetS prevalence and number of MetS components increased with age in all three samples. The PIVUS study showed that high baseline BMI, low baseline physical activity, and increasing BMI during follow-up were related to increasing MetS prevalence and increasing numbers of MetS components during follow-up. One-third to half of individuals initially belonging to the obesity without MetS category acquired MetS during aging.MetS prevalence increased during aging, especially in individuals with high BMI, low level of physical activity, and weight gain. Obesity without MetS was not a stable condition over time as many of those individuals gained metabolic disturbances during aging.

    View details for DOI 10.1089/met.2016.0082

    View details for Web of Science ID 000394368100007

  • Transcriptional Dynamics During Human Adipogenesis and Its Link to Adipose Morphology and Distribution DIABETES Ehrlund, A., Mejhert, N., Bjork, C., Andersson, R., Kulyte, A., Astrom, G., Itoh, M., Kawaji, H., Lassmann, T., Daub, C. O., Carninci, P., Forrest, A. R., Hayashizaki, Y., Sandelin, A., Ingelsson, E., Ryden, M., Laurencikiene, J., Arner, P., Arner, E. 2017; 66 (1): 218-230

    View details for DOI 10.2337/db16-0631

    View details for Web of Science ID 000390834300025

  • Association analyses based on false discovery rate implicate new loci for coronary artery disease. Nature genetics Nelson, C. P., Goel, A., Butterworth, A. S., Kanoni, S., Webb, T. R., Marouli, E., Zeng, L., Ntalla, I., Lai, F. Y., Hopewell, J. C., Giannakopoulou, O., Jiang, T., Hamby, S. E., Di Angelantonio, E., Assimes, T. L., Bottinger, E. P., Chambers, J. C., Clarke, R., Palmer, C. N., Cubbon, R. M., Ellinor, P., Ermel, R., Evangelou, E., Franks, P. W., Grace, C., Gu, D., Hingorani, A. D., Howson, J. M., Ingelsson, E., Kastrati, A., Kessler, T., Kyriakou, T., Lehtimäki, T., Lu, X., Lu, Y., März, W., McPherson, R., Metspalu, A., Pujades-Rodriguez, M., Ruusalepp, A., Schadt, E. E., Schmidt, A. F., Sweeting, M. J., Zalloua, P. A., AlGhalayini, K., Keavney, B. D., Kooner, J. S., Loos, R. J., Patel, R. S., Rutter, M. K., Tomaszewski, M., Tzoulaki, I., Zeggini, E., Erdmann, J., Dedoussis, G., Björkegren, J. L., Schunkert, H., Farrall, M., Danesh, J., Samani, N. J., Watkins, H., Deloukas, P. 2017; 49 (9): 1385–91

    Abstract

    Genome-wide association studies (GWAS) in coronary artery disease (CAD) had identified 66 loci at 'genome-wide significance' (P < 5 × 10(-8)) at the time of this analysis, but a much larger number of putative loci at a false discovery rate (FDR) of 5% (refs. 1,2,3,4). Here we leverage an interim release of UK Biobank (UKBB) data to evaluate the validity of the FDR approach. We tested a CAD phenotype inclusive of angina (SOFT; ncases = 10,801) as well as a stricter definition without angina (HARD; ncases = 6,482) and selected cases with the former phenotype to conduct a meta-analysis using the two most recent CAD GWAS. This approach identified 13 new loci at genome-wide significance, 12 of which were on our previous list of loci meeting the 5% FDR threshold, thus providing strong support that the remaining loci identified by FDR represent genuine signals. The 304 independent variants associated at 5% FDR in this study explain 21.2% of CAD heritability and identify 243 loci that implicate pathways in blood vessel morphogenesis as well as lipid metabolism, nitric oxide signaling and inflammation.

    View details for DOI 10.1038/ng.3913

    View details for PubMedID 28714975

  • Genotype-based recall to study metabolic effects of genetic variation: a pilot study of PPARG Pro12Ala carriers UPSALA JOURNAL OF MEDICAL SCIENCES Kamble, P. G., Gustafsson, S., Pereira, M. J., Lundkvist, P., Cook, N., Lind, L., Franks, P. W., Fall, T., Eriksson, J. W., Ingelsson, E. 2017; 122 (4): 234–42

    Abstract

    To assess practical implications of genotype-based recall (GBR) studies, an increasingly popular approach for in-depth characterization of genotype-phenotype relationships.We genotyped 2500 participants from the Swedish EpiHealth cohort and considered loss-of-function and missense variants in genes with relation to cardiometabolic traits as the basis for our GBR study. Therefore, we focused on carriers and non-carriers of the PPARG Pro12Ala (rs1801282) variant, as it is a relatively common variant with a minor allele frequency (MAF) of 0.14. It has also been shown to affect ligand binding and transcription, and carriage of the minor allele (Ala12) is associated with a reduced risk of type 2 diabetes. We re-invited 39 Pro12Pro, 34 Pro12Ala, and 30 Ala12Ala carriers and performed detailed anthropometric and serological assessments.The participation rates in the GBR study were 31%, 44%, and 40%, and accordingly we included 12, 15, and 13 individuals with Pro12Pro, Pro12Ala, and Ala12Ala variants, respectively. There were no differences in anthropometric or metabolic variables among the different genotype groups.Our report highlights that from a practical perspective, GBR can be used to study genotype-phenotype relationships. This approach can prove to be a valuable tool for follow-up findings from large-scale genetic discovery studies by undertaking detailed phenotyping procedures that might not be feasible in large studies. However, our study also illustrates the need for a larger pool of genotyped or sequenced individuals to allow for selection of rare variants with larger effects that can be examined in a GBR study of the present size.

    View details for DOI 10.1080/03009734.2017.1405127

    View details for Web of Science ID 000423294800005

    View details for PubMedID 29303622

    View details for PubMedCentralID PMC5810227

  • Genetic and methylation variation in the CYP2B6 gene is related to circulating p,p '-dde levels in a population-based sample ENVIRONMENT INTERNATIONAL Lind, L., Ng, E., Ingelsson, E., Lindgren, C., Salihovic, S., van Bavel, B., Mahajan, A., Lampa, E., Morris, A. P., Lind, P. M. 2017; 98: 212-218

    Abstract

    Since the metabolism of the organochlorine pesticide dichlorodiphenyltrichloroethane (DDT) is not fully known in humans, we evaluated if circulating levels of a major breakdown product of DDT, p,p'-DDE, were related to genome-wide genetic and methylation variation in a population-based sample.In the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (1016 subjects all aged 70), circulating levels of p,p'-DDE were analyzed by high-resolution chromatography coupled to high-resolution mass spectrometry (HRGC/HRMS). Genetic variants were genotyped and imputed (1000 Genomes reference, March 2012 release). Methylation sites were assayed using the Illumina HumanMethylation450 array in whole blood. A genome-wide association study (GWAS) approach was applied.Evidence for genome-wide significant association with p,p'-DDE levels was observed only for a locus at chromosome 19 corresponding to the CYP2B6 gene (lead SNP rs7260538). Subjects being homozygote for the G allele showed a median level of 472ng/g lipid, while the corresponding level for those being homozygote for the T allele was 192ng/g lipid (p=1.5×10(-31)). An analysis conditioned on the lead SNP disclosed a distinct signal in the same gene (rs7255374, position chr19:41520351; p=2.2×10(-8)). A whole-genome methylation analysis showed one significant relationship vs. p,p'-DDE levels (p=6.2×10(-9)) located 7kb downstream the CYP2B6 gene (cg27089200, position chr19:41531976). This CpG-site was also related to the lead SNP (p=3.8×10(-35)), but mediated only 4% of the effect of the lead SNP on p,p'-DDE levels.Circulating levels of p,p'-DDE were related to genetic variation in the CYP2B6 gene in the general elderly population. DNA methylation in this gene is not closely linked to the p,p'-DDE levels.

    View details for DOI 10.1016/j.envint.2016.11.010

    View details for Web of Science ID 000389913500025

    View details for PubMedID 27839851

    View details for PubMedCentralID PMC5152752

  • Multiethnic genome-wide meta-analysis of ectopic fat depots identifies loci associated with adipocyte development and differentiation NATURE GENETICS Chu, A. Y., Deng, X., Fisher, V. A., Drong, A., Zhang, Y., Feitosa, M. F., Liu, C., Weeks, O., Choh, A. C., Duan, Q., Dyer, T. D., Eicher, J. D., Guo, X., Heard-Costa, N. L., Kacprowski, T., Kent, J. W., Lange, L. A., Liu, X., Lohman, K., Lu, L., Mahajan, A., O'Connell, J. R., Parihar, A., Peralta, J. M., Smith, A. V., Zhang, Y., Homuth, G., Kissebah, A. H., Kullberg, J., Laqua, R., Launer, L. J., Nauck, M., Olivier, M., Peyser, P. A., Terry, J. G., Wojczynski, M. K., Yao, J., Bielak, L. F., Blangero, J., Borecki, I. B., Bowden, D. W., Carr, J. J., Czerwinski, S. A., Ding, J., Friedrich, N., Gudnason, V., Harris, T. B., Ingelsson, E., Johnson, A. D., Kardia, S. L., Langefeld, C. D., Lind, L., Liu, Y., Mitchell, B. D., Morris, A. P., Mosley, T. H., Rotter, J. I., Shuldiner, A. R., Towne, B., Voelzke, H., Wallaschofski, H., Wilson, J. G., Allison, M., Lindgren, C. M., Goessling, W., Cupples, L. A., Steinhauser, M. L., Fox, C. S. 2017; 49 (1): 125-130

    Abstract

    Variation in body fat distribution contributes to the metabolic sequelae of obesity. The genetic determinants of body fat distribution are poorly understood. The goal of this study was to gain new insights into the underlying genetics of body fat distribution by conducting sample-size-weighted fixed-effects genome-wide association meta-analyses in up to 9,594 women and 8,738 men of European, African, Hispanic and Chinese ancestry, with and without sex stratification, for six traits associated with ectopic fat (hereinafter referred to as ectopic-fat traits). In total, we identified seven new loci associated with ectopic-fat traits (ATXN1, UBE2E2, EBF1, RREB1, GSDMB, GRAMD3 and ENSA; P < 5 × 10(-8); false discovery rate < 1%). Functional analysis of these genes showed that loss of function of either Atxn1 or Ube2e2 in primary mouse adipose progenitor cells impaired adipocyte differentiation, suggesting physiological roles for ATXN1 and UBE2E2 in adipogenesis. Future studies are necessary to further explore the mechanisms by which these genes affect adipocyte biology and how their perturbations contribute to systemic metabolic disease.

    View details for DOI 10.1038/ng.3738

    View details for Web of Science ID 000390976600018

    View details for PubMedID 27918534

  • Association of Body Mass Index with DNA Methylation and Gene Expression in Blood Cells and Relations to Cardiometabolic Disease: A Mendelian Randomization Approach. PLoS medicine Mendelson, M. M., Marioni, R. E., Joehanes, R., Liu, C., Hedman, Å. K., Aslibekyan, S., Demerath, E. W., Guan, W., Zhi, D., Yao, C., Huan, T., Willinger, C., Chen, B., Courchesne, P., Multhaup, M., Irvin, M. R., Cohain, A., Schadt, E. E., Grove, M. L., Bressler, J., North, K., Sundström, J., Gustafsson, S., Shah, S., McRae, A. F., Harris, S. E., Gibson, J., Redmond, P., Corley, J., Murphy, L., Starr, J. M., Kleinbrink, E., Lipovich, L., Visscher, P. M., Wray, N. R., Krauss, R. M., Fallin, D., Feinberg, A., Absher, D. M., Fornage, M., Pankow, J. S., Lind, L., Fox, C., Ingelsson, E., Arnett, D. K., Boerwinkle, E., Liang, L., Levy, D., Deary, I. J. 2017; 14 (1)

    Abstract

    The link between DNA methylation, obesity, and adiposity-related diseases in the general population remains uncertain.We conducted an association study of body mass index (BMI) and differential methylation for over 400,000 CpGs assayed by microarray in whole-blood-derived DNA from 3,743 participants in the Framingham Heart Study and the Lothian Birth Cohorts, with independent replication in three external cohorts of 4,055 participants. We examined variations in whole blood gene expression and conducted Mendelian randomization analyses to investigate the functional and clinical relevance of the findings. We identified novel and previously reported BMI-related differential methylation at 83 CpGs that replicated across cohorts; BMI-related differential methylation was associated with concurrent changes in the expression of genes in lipid metabolism pathways. Genetic instrumental variable analysis of alterations in methylation at one of the 83 replicated CpGs, cg11024682 (intronic to sterol regulatory element binding transcription factor 1 [SREBF1]), demonstrated links to BMI, adiposity-related traits, and coronary artery disease. Independent genetic instruments for expression of SREBF1 supported the findings linking methylation to adiposity and cardiometabolic disease. Methylation at a substantial proportion (16 of 83) of the identified loci was found to be secondary to differences in BMI. However, the cross-sectional nature of the data limits definitive causal determination.We present robust associations of BMI with differential DNA methylation at numerous loci in blood cells. BMI-related DNA methylation and gene expression provide mechanistic insights into the relationship between DNA methylation, obesity, and adiposity-related diseases.

    View details for DOI 10.1371/journal.pmed.1002215

    View details for PubMedID 28095459

    View details for PubMedCentralID PMC5240936

  • Sensitivity Analyses for Robust Causal Inference from Mendelian Randomization Analyses with Multiple Genetic Variants EPIDEMIOLOGY Burgess, S., Bowden, J., Fall, T., Ingelsson, E., Thompson, S. G. 2017; 28 (1): 30-42
  • Epigenetic Patterns in Blood Associated With Lipid Traits Predict Incident Coronary Heart Disease Events and Are Enriched for Results From Genome-Wide Association Studies. Circulation. Cardiovascular genetics Hedman, Å. K., Mendelson, M. M., Marioni, R. E., Gustafsson, S., Joehanes, R., Irvin, M. R., Zhi, D., Sandling, J. K., Yao, C., Liu, C., Liang, L., Huan, T., McRae, A. F., Demissie, S., Shah, S., Starr, J. M., Cupples, L. A., Deloukas, P., Spector, T. D., Sundström, J., Krauss, R. M., Arnett, D. K., Deary, I. J., Lind, L., Levy, D., Ingelsson, E. 2017; 10 (1)

    Abstract

    Genome-wide association studies have identified loci influencing circulating lipid concentrations in humans; further information on novel contributing genes, pathways, and biology may be gained through studies of epigenetic modifications.To identify epigenetic changes associated with lipid concentrations, we assayed genome-wide DNA methylation at cytosine-guanine dinucleotides (CpGs) in whole blood from 2306 individuals from 2 population-based cohorts, with replication of findings in 2025 additional individuals. We identified 193 CpGs associated with lipid levels in the discovery stage (P<1.08E-07) and replicated 33 (at Bonferroni-corrected P<0.05), including 25 novel CpGs not previously associated with lipids. Genes at lipid-associated CpGs were enriched in lipid and amino acid metabolism processes. A differentially methylated locus associated with triglycerides and high-density lipoprotein cholesterol (HDL-C; cg27243685; P=8.1E-26 and 9.3E-19) was associated with cis-expression of a reverse cholesterol transporter (ABCG1; P=7.2E-28) and incident cardiovascular disease events (hazard ratio per SD increment, 1.38; 95% confidence interval, 1.15-1.66; P=0.0007). We found significant cis-methylation quantitative trait loci at 64% of the 193 CpGs with an enrichment of signals from genome-wide association studies of lipid levels (PTC=0.004, PHDL-C=0.008 and Ptriglycerides=0.00003) and coronary heart disease (P=0.0007). For example, genome-wide significant variants associated with low-density lipoprotein cholesterol and coronary heart disease at APOB were cis-methylation quantitative trait loci for a low-density lipoprotein cholesterol-related differentially methylated locus.We report novel associations of DNA methylation with lipid levels, describe epigenetic mechanisms related to previous genome-wide association studies discoveries, and provide evidence implicating epigenetic regulation of reverse cholesterol transport in blood in relation to occurrence of cardiovascular disease events.

    View details for DOI 10.1161/CIRCGENETICS.116.001487

    View details for PubMedID 28213390

    View details for PubMedCentralID PMC5331877

  • Statistical power considerations in genotype-based recall randomized controlled trials SCIENTIFIC REPORTS Atabaki-Pasdar, N., Ohlsson, M., Shungin, D., Kurbasic, A., Ingelsson, E., Pearson, E. R., Ali, A., Franks, P. W. 2016; 6

    Abstract

    Randomized controlled trials (RCT) are often underpowered for validating gene-treatment interactions. Using published data from the Diabetes Prevention Program (DPP), we examined power in conventional and genotype-based recall (GBR) trials. We calculated sample size and statistical power for gene-metformin interactions (vs. placebo) using incidence rates, gene-drug interaction effect estimates and allele frequencies reported in the DPP for the rs8065082 SLC47A1 variant, a metformin transported encoding locus. We then calculated statistical power for interactions between genetic risk scores (GRS), metformin treatment and intensive lifestyle intervention (ILI) given a range of sampling frames, clinical trial sample sizes, interaction effect estimates, and allele frequencies; outcomes were type 2 diabetes incidence (time-to-event) and change in small LDL particles (continuous outcome). Thereafter, we compared two recruitment frameworks: GBR (participants recruited from the extremes of a GRS distribution) and conventional sampling (participants recruited without explicit emphasis on genetic characteristics). We further examined the influence of outcome measurement error on statistical power. Under most simulated scenarios, GBR trials have substantially higher power to observe gene-drug and gene-lifestyle interactions than same-sized conventional RCTs. GBR trials are becoming popular for validation of gene-treatment interactions; our analyses illustrate the strengths and weaknesses of this design.

    View details for DOI 10.1038/srep37307

    View details for Web of Science ID 000388480300001

    View details for PubMedID 27886175

  • DNA methylation patterns associated with oxidative stress in an ageing population BMC MEDICAL GENOMICS Hedman, A. K., Zilmer, M., Sundstrom, J., Lind, L., Ingelsson, E. 2016; 9

    Abstract

    Oxidative stress has been related to type 2 diabetes (T2D) and cardiovascular disease (CVD), the leading global cause of death. Contributions of environmental factors such as oxidative stress on complex traits and disease may be partly mediated through changes in epigenetic marks (e.g. DNA methylation). Studies relating differential methylation with intermediate phenotypes and disease endpoints may be useful in identifying additional candidate genes and mechanisms involved in disease.To investigate the role of epigenetic variation in oxidative stress marker levels and subsequent development of CVD and T2D, we performed analyses of genome-wide DNA methylation in blood, ten markers of oxidative stress (total glutathione [TGSH], reduced glutathione [GSH], oxidised glutathione [GSSG], GSSG to GSH ratio, homocysteine [HCY], oxidised low-density lipoprotein (oxLDL), antibodies against oxLDL [OLAB], conjugated dienes [CD], baseline conjugated dienes [BCD]-LDL and total antioxidant capacity [TAOC]) and incident disease in up to 966 age-matched individuals.In total, we found 66 cytosine-guanine (CpG) sites associated with one or more oxidative stress markers (false discovery rate [FDR] <0.05). These sites were enriched in regulatory regions of the genome. Genes annotated to CpG sites showed enrichment in annotation clusters relating to phospho-metabolism and proteins with pleckstrin domains. We investigated the contribution of oxidative stress-associated CpGs to development of cardiometabolic disease. Methylation variation at CpGs in the 3'-UTR of HIST1H4D (cg08170869; histone cluster 1, H4d) and in the body of DVL1 (cg03465880; dishevelled-1) were associated with incident T2D events during 10 years of follow-up (all permutation p-values <0.01), indicating a role of epigenetic regulation in oxidative stress processes leading to development or progression of diabetes. Methylation QTL (meQTL) analysis showed significant associations with genetic sequence variants in cis at 28 (42%) of oxidative stress phenotype-associated sites (FDR < 0.05). Integrating cis-meQTLs with genotype-phenotype associations indicated that genetic effects on oxidative stress phenotype at one locus (cg07547695; BCL2L11) may be mediated through DNA methylation.In conclusion, we report novel associations of DNA methylation with oxidative stress, some of which also show evidence of a relation with T2D incidence.

    View details for DOI 10.1186/s12920-016-0235-0

    View details for Web of Science ID 000388539300001

    View details for PubMedID 27884142

  • Novel genetic loci associated with long-term deterioration in blood lipid concentrations and coronary artery disease in European adults. International journal of epidemiology Varga, T. V., Kurbasic, A., Aine, M., Eriksson, P., Ali, A., Hindy, G., Gustafsson, S., Luan, J., Shungin, D., Chen, Y., Schulz, C., Nilsson, P. M., Hallmans, G., Barroso, I., Deloukas, P., Langenberg, C., Scott, R. A., Wareham, N. J., Lind, L., Ingelsson, E., Melander, O., Orho-Melander, M., Renström, F., Franks, P. W. 2016

    Abstract

    Cross-sectional genome-wide association studies have identified hundreds of loci associated with blood lipids and related cardiovascular traits, but few genetic association studies have focused on long-term changes in blood lipids.Participants from the GLACIER Study (Nmax = 3492) were genotyped with the MetaboChip array, from which 29 387 SNPs (single nucleotide polymorphisms; replication, fine-mapping regions and wildcard SNPs for lipid traits) were extracted for association tests with 10-year change in total cholesterol (ΔTC) and triglycerides (ΔTG). Four additional prospective cohort studies (MDC, PIVUS, ULSAM, MRC Ely; Nmax = 8263 participants) were used for replication. We conducted an in silico look-up for association with coronary artery disease (CAD) in the Coronary ARtery DIsease Genome-wide Replication and Meta-analysis (CARDIoGRAMplusC4D) Consortium (N ∼ 190 000) and functional annotation for the top ranking variants.In total, 956 variants were associated (P < 0.01) with either ΔTC or ΔTG in GLACIER. In GLACIER, chr19:50121999 at APOE was associated with ΔTG and multiple SNPs in the APOA1/A4/C3/A5 region at genome-wide significance (P < 5 × 10(-8)), whereas variants in four loci, DOCK7, BRE, SYNE1 and KCNIP1, reached study-wide significance (P < 1.7 × 10(-6)). The rs7412 variant at APOE was associated with ΔTC in GLACIER (P < 1.7 × 10(-6)). In pooled analyses of all cohorts, 139 SNPs at six and five loci were associated with ΔTC and for ΔTG, respectively (P < 10(-3)). Of these, a variant at CAPN3 (P = 1.2 × 10(-4)), multiple variants at HPR (Pmin = 1.5 × 10(-6)) and a variant at SIX5 (P = 1.9 × 10(-4)) showed evidence for association with CAD.We identified seven novel genomic regions associated with long-term changes in blood lipids, of which three also raise CAD risk.

    View details for PubMedID 27864399

  • A DNA methylation biomarker of alcohol consumption. Molecular psychiatry Liu, C., Marioni, R. E., Hedman, Å. K., Pfeiffer, L., Tsai, P., Reynolds, L. M., JUST, A. C., Duan, Q., Boer, C. G., Tanaka, T., Elks, C. E., Aslibekyan, S., Brody, J. A., Kühnel, B., Herder, C., Almli, L. M., Zhi, D., Wang, Y., Huan, T., Yao, C., Mendelson, M. M., Joehanes, R., Liang, L., Love, S., Guan, W., Shah, S., McRae, A. F., Kretschmer, A., Prokisch, H., Strauch, K., Peters, A., Visscher, P. M., Wray, N. R., Guo, X., Wiggins, K. L., Smith, A. K., Binder, E. B., Ressler, K. J., IRVIN, M. R., Absher, D. M., Hernandez, D., Ferrucci, L., Bandinelli, S., Lohman, K., Ding, J., Trevisi, L., Gustafsson, S., Sandling, J. H., Stolk, L., Uitterlinden, A. G., Yet, I., Castillo-Fernandez, J. E., Spector, T. D., Schwartz, J. D., Vokonas, P., Lind, L., Li, Y., Fornage, M., Arnett, D. K., Wareham, N. J., Sotoodehnia, N., Ong, K. K., van Meurs, J. B., Conneely, K. N., Baccarelli, A. A., Deary, I. J., Bell, J. T., North, K. E., Liu, Y., Waldenberger, M., London, S. J., Ingelsson, E., Levy, D. 2016

    Abstract

    The lack of reliable measures of alcohol intake is a major obstacle to the diagnosis and treatment of alcohol-related diseases. Epigenetic modifications such as DNA methylation may provide novel biomarkers of alcohol use. To examine this possibility, we performed an epigenome-wide association study of methylation of cytosine-phosphate-guanine dinucleotide (CpG) sites in relation to alcohol intake in 13 population-based cohorts (ntotal=13 317; 54% women; mean age across cohorts 42-76 years) using whole blood (9643 European and 2423 African ancestries) or monocyte-derived DNA (588 European, 263 African and 400 Hispanic ancestry) samples. We performed meta-analysis and variable selection in whole-blood samples of people of European ancestry (n=6926) and identified 144 CpGs that provided substantial discrimination (area under the curve=0.90-0.99) for current heavy alcohol intake (⩾42 g per day in men and ⩾28 g per day in women) in four replication cohorts. The ancestry-stratified meta-analysis in whole blood identified 328 (9643 European ancestry samples) and 165 (2423 African ancestry samples) alcohol-related CpGs at Bonferroni-adjusted P<1 × 10(-7). Analysis of the monocyte-derived DNA (n=1251) identified 62 alcohol-related CpGs at P<1 × 10(-7). In whole-blood samples of people of European ancestry, we detected differential methylation in two neurotransmitter receptor genes, the γ-Aminobutyric acid-A receptor delta and γ-aminobutyric acid B receptor subunit 1; their differential methylation was associated with expression levels of a number of genes involved in immune function. In conclusion, we have identified a robust alcohol-related DNA methylation signature and shown the potential utility of DNA methylation as a clinically useful diagnostic test to detect current heavy alcohol consumption.Molecular Psychiatry advance online publication, 15 November 2016; doi:10.1038/mp.2016.192.

    View details for DOI 10.1038/mp.2016.192

    View details for PubMedID 27843151

  • Effects of cigarette smoking on cardiovascular-related protein profiles in two community-based cohort studies. Atherosclerosis Huang, B., Svensson, P., Ärnlöv, J., Sundström, J., Lind, L., Ingelsson, E. 2016; 254: 52-58

    Abstract

    Cardiovascular diseases account for the largest fraction of smoking-induced deaths. Studies of smoking in relation to cardiovascular-related protein markers can provide novel insights into the biological effects of smoking. We investigated the associations between cigarette smoking and 80 protein markers known to be related to cardiovascular diseases in two community-based cohorts, the Prospective Study of the Vasculature in Uppsala Seniors (PIVUS, n = 969, 50% women, all aged 70 years) and the Uppsala Longitudinal Study of Adult Men (ULSAM, n = 717, all men aged 77 years).Smoking status was self-reported and defined as current smoker, former smoker or never-smoker. Levels of the 80 proteins were measured using the proximity extension assay, a novel PCR-based proteomics technique.We found 30 proteins to be significantly associated with current cigarette smoking in PIVUS (FDR<5%); and ten were replicated in ULSAM (p < 0.05). Matrix metalloproteinase-12 (MMP-12), growth/differentiation factor 15 (GDF-15), urokinase plasminogen activator surface receptor (uPAR), TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2), lectin-like oxidized LDL receptor 1 (LOX-1), hepatocyte growth factor (HGF), matrix metalloproteinase-10 (MMP-10) and matrix metalloproteinase-1 (MMP-1) were positively associated, while endothelial cell-specific molecule 1 (ESM-1) and interleukin-27 subunit alpha (IL27-A) showed inverse associations. All of them remained significant in a subset of individuals without manifest cardiovascular disease.The findings of the present study suggest that cigarette smoking may interfere with several essential parts of the atherosclerosis process, as evidenced by associations with protein markers representing endothelial dysfunction, inflammation, neointimal formation, foam cell formation and plaque instability.

    View details for DOI 10.1016/j.atherosclerosis.2016.09.014

    View details for PubMedID 27684606

  • Transcriptional Dynamics During Human Adipogenesis and its Link to Adipose Morphology and Distribution. Diabetes Ehrlund, A., Mejhert, N., Björk, C., Andersson, R., Kulyté, A., Åström, G., Itoh, M., Kawaji, H., Lassmann, T., Daub, C. O., Carninci, P., Forrest, A. R., Hayashizaki, Y., Sandelin, A., Ingelsson, E., Consortium, F., Rydén, M., Laurencikiene, J., Arner, P., Arner, E. 2016

    Abstract

    White adipose tissue (WAT) can develop into several phenotypes with different pathophysiological impact on type 2 diabetes. To better understand the adipogenic process, the transcriptional events that occur during in vitro differentiation of human adipocytes were investigated and the findings linked to WAT phenotypes. Single-molecule transcriptional profiling provided a detailed map of the expressional changes of genes, enhancers, and long noncoding RNAs, where different types of transcripts share common dynamics during differentiation. Common signatures include early downregulated, transient, and late induced transcripts, all of which are linked to distinct developmental processes during adipogenesis. Enhancers expressed during adipogenesis overlap significantly with genetic variants associated with WAT distribution. Transiently expressed and late induced genes are associated with hypertrophic WAT (few but large fat cells), a phenotype closely linked to insulin resistance and type 2 diabetes. Transcription factors that are expressed early or transiently affect differentiation and adipocyte function and are controlled by several well-known upstream regulators such as glucocorticosteroids, insulin, cAMP, and thyroid hormones. Taken together, our results suggest a complex but highly coordinated regulation of adipogenesis.

    View details for PubMedID 27803022

  • Metabolic Syndrome Development During Aging with Special Reference to Obesity Without the Metabolic Syndrome. Metabolic syndrome and related disorders Roos, V., Elmståhl, S., Ingelsson, E., Sundström, J., Ärnlöv, J., Lind, L. 2016: -?

    Abstract

    Obesity and its associated metabolic complications continue to increase worldwide. We investigated the development of metabolic syndrome (MetS) during aging in relation to body mass index (BMI) and exercise habits. We assigned special emphasis to the metabolic stability in individuals with obesity, but without MetS, a condition often referred to as metabolically healthy obesity.Cross-sectional analysis was carried out in a sample of 19,129 men and women aged 45-75 years from the EpiHealth study. In addition, longitudinal analyses were carried out in the ULSAM study (2322 men at baseline followed from age 50 to age 77) and in the PIVUS study (1016 men and women at baseline followed from age 70 to age 80). Participants were categorized into six groups according to BMI category (normal weight/BMI <25 kg/m(2), overweight/BMI 25-30 kg/m(2), and obesity/BMI >30 kg/m(2)) and MetS status (+/-, National Cholesterol Education Program criteria).MetS prevalence and number of MetS components increased with age in all three samples. The PIVUS study showed that high baseline BMI, low baseline physical activity, and increasing BMI during follow-up were related to increasing MetS prevalence and increasing numbers of MetS components during follow-up. One-third to half of individuals initially belonging to the obesity without MetS category acquired MetS during aging.MetS prevalence increased during aging, especially in individuals with high BMI, low level of physical activity, and weight gain. Obesity without MetS was not a stable condition over time as many of those individuals gained metabolic disturbances during aging.

    View details for PubMedID 27754771

  • No Association of Coronary Artery Disease with X-Chromosomal Variants in Comprehensive International Meta-Analysis SCIENTIFIC REPORTS Loley, C., Alver, M., Assimes, T. L., Bjonnes, A., Goel, A., Gustafsson, S., Hernesniemi, J., Hopewell, J. C., Kanoni, S., Kleber, M. E., Lau, K. W., Lu, Y., Lyytikainen, L., Nelson, C. P., Nikpay, M., Qu, L., Salfati, E., Scholz, M., Tukiainen, T., Willenborg, C., Won, H., Zeng, L., Zhang, W., Anand, S. S., Beutner, F., Bottinger, E. P., Clarke, R., Dedoussis, G., Do, R., Esko, T., Eskola, M., Farrall, M., Gauguier, D., Giedraitis, V., Granger, C. B., Hall, A. S., Hamsten, A., Hazen, S. L., Huang, J., Kahonen, M., Kyriakou, T., Laaksonen, R., Lind, L., Lindgren, C., Magnusson, P. K., Marouli, E., Mihailov, E., Morris, A. P., Nikus, K., Pedersen, N., Rallidis, L., Salomaa, V., Shah, S. H., Stewart, A. F., Thompson, J. R., Zalloua, P. A., Chambers, J. C., Collins, R., Ingelsson, E., Iribarren, C., Karhunen, P. J., Kooner, J. S., Lehtimaki, T., Loos, R. J., Maerz, W., McPherson, R., Metspalu, A., Reilly, M. P., Ripatti, S., Sanghera, D. K., Thiery, J., Watkins, H., Deloukas, P., Kathiresan, S., Samani, N. J., Schunkert, H., Erdmann, J., Koenig, I. R. 2016; 6

    Abstract

    In recent years, genome-wide association studies have identified 58 independent risk loci for coronary artery disease (CAD) on the autosome. However, due to the sex-specific data structure of the X chromosome, it has been excluded from most of these analyses. While females have 2 copies of chromosome X, males have only one. Also, one of the female X chromosomes may be inactivated. Therefore, special test statistics and quality control procedures are required. Thus, little is known about the role of X-chromosomal variants in CAD. To fill this gap, we conducted a comprehensive X-chromosome-wide meta-analysis including more than 43,000 CAD cases and 58,000 controls from 35 international study cohorts. For quality control, sex-specific filters were used to adequately take the special structure of X-chromosomal data into account. For single study analyses, several logistic regression models were calculated allowing for inactivation of one female X-chromosome, adjusting for sex and investigating interactions between sex and genetic variants. Then, meta-analyses including all 35 studies were conducted using random effects models. None of the investigated models revealed genome-wide significant associations for any variant. Although we analyzed the largest-to-date sample, currently available methods were not able to detect any associations of X-chromosomal variants with CAD.

    View details for DOI 10.1038/srep35278

    View details for Web of Science ID 000385004200001

    View details for PubMedID 27731410

    View details for PubMedCentralID PMC5059659

  • Genome-wide association study of caffeine metabolites provides new insights to caffeine metabolism and dietary caffeine-consumption behavior. Human molecular genetics Cornelis, M. C., Kacprowski, T., Menni, C., Gustafsson, S., Pivin, E., Adamski, J., Artati, A., Eap, C. B., Ehret, G., Friedrich, N., Ganna, A., Guessous, I., Homuth, G., Lind, L., Magnusson, P. K., Mangino, M., Pedersen, N. L., Pietzner, M., Suhre, K., Völzke, H., Bochud, M., Spector, T. D., Grabe, H. J., Ingelsson, E. 2016

    Abstract

    Caffeine is the most widely consumed psychoactive substance in the world and presents with wide interindividual variation in metabolism. This variation may modify potential adverse or beneficial effects of caffeine on health. We conducted a genome-wide association study (GWAS) of plasma caffeine, paraxanthine, theophylline, theobromine and paraxanthine/caffeine ratio among up to 9,876 individuals of European ancestry from six population-based studies. A single SNP at 6p23 (near CD83) and several SNPs at 7p21 (near AHR), 15q24 (near CYP1A2) and 19q13.2 (near CYP2A6) met GW-significance (P < 5 × 10(-8)) and were associated with one or more metabolites. Variants at 7p21 and 15q24 associated with higher plasma caffeine and lower plasma paraxanthine/caffeine (slow caffeine metabolism) were previously associated with lower coffee and caffeine consumption behavior in GWAS. Variants at 19q13.2 associated with higher plasma paraxanthine/caffeine (slow paraxanthine metabolism) were also associated with lower coffee consumption in the UK Biobank (n = 94 343, P < 1.0 × 10(-6)). Variants at 2p24 (in GCKR), 4q22 (in ABCG2) and 7q11.23 (near POR) that were previously associated with coffee consumption in GWAS were nominally associated with plasma caffeine or its metabolites. Taken together, we have identified genetic factors contributing to variation in caffeine metabolism and confirm an important modulating role of systemic caffeine levels in dietary caffeine consumption behavior. Moreover, candidate genes identified encode proteins with important clinical functions that extend beyond caffeine metabolism.

    View details for PubMedID 27702941

  • Non-targeted metabolomics combined with genetic analyses identifies bile acid synthesis and phospholipid metabolism as being associated with incident type 2 diabetes. Diabetologia Fall, T., Salihovic, S., Brandmaier, S., Nowak, C., Ganna, A., Gustafsson, S., Broeckling, C. D., Prenni, J. E., Kastenmüller, G., Peters, A., Magnusson, P. K., Wang-Sattler, R., Giedraitis, V., Berne, C., Gieger, C., Pedersen, N. L., Ingelsson, E., Lind, L. 2016; 59 (10): 2114-2124

    Abstract

    Identification of novel biomarkers for type 2 diabetes and their genetic determinants could lead to improved understanding of causal pathways and improve risk prediction.In this study, we used data from non-targeted metabolomics performed using liquid chromatography coupled with tandem mass spectrometry in three Swedish cohorts (Uppsala Longitudinal Study of Adult Men [ULSAM], n = 1138; Prospective Investigation of the Vasculature in Uppsala Seniors [PIVUS], n = 970; TwinGene, n = 1630). Metabolites associated with impaired fasting glucose (IFG) and/or prevalent type 2 diabetes were assessed for associations with incident type 2 diabetes in the three cohorts followed by replication attempts in the Cooperative Health Research in the Region of Augsburg (KORA) S4 cohort (n = 855). Assessment of the association of metabolite-regulating genetic variants with type 2 diabetes was done using data from a meta-analysis of genome-wide association studies.Out of 5961 investigated metabolic features, 1120 were associated with prevalent type 2 diabetes and IFG and 70 were annotated to metabolites and replicated in the three cohorts. Fifteen metabolites were associated with incident type 2 diabetes in the four cohorts combined (358 events) following adjustment for age, sex, BMI, waist circumference and fasting glucose. Novel findings included associations of higher values of the bile acid deoxycholic acid and monoacylglyceride 18:2 and lower concentrations of cortisol with type 2 diabetes risk. However, adding metabolites to an existing risk score improved model fit only marginally. A genetic variant within the CYP7A1 locus, encoding the rate-limiting enzyme in bile acid synthesis, was found to be associated with lower concentrations of deoxycholic acid, higher concentrations of LDL-cholesterol and lower type 2 diabetes risk. Variants in or near SGPP1, GCKR and FADS1/2 were associated with diabetes-associated phospholipids and type 2 diabetes.We found evidence that the metabolism of bile acids and phospholipids shares some common genetic origin with type 2 diabetes.Metabolomics data have been deposited in the Metabolights database, with accession numbers MTBLS93 (TwinGene), MTBLS124 (ULSAM) and MTBLS90 (PIVUS).

    View details for DOI 10.1007/s00125-016-4041-1

    View details for PubMedID 27406814

  • Trans-ancestry meta-analyses identify rare and common variants associated with blood pressure and hypertension NATURE GENETICS Surendran, P., Drenos, F., Young, R., Warren, H., Cook, J. P., Manning, A. K., Grarup, N., Sim, X., Barnes, D. R., Witkowska, K., Staley, J. R., Tragante, V., Tukiainen, T., Yaghootkar, H., Masca, N., Freitag, D. F., Ferreira, T., Giannakopoulou, O., Tinker, A., Harakalova, M., Mihailov, E., Liu, C., Kraja, A. T., Nielsen, S. F., Rasheed, A., Samue, M., Zhao, W., Bonnycastle, L. L., Jackson, A. U., Narisu, N., Swift, A. J., Southam, L., Marten, J., Huyghe, J. R., Stancakova, A., Fava, C., Ohlsson, T., Matchan, A., Stirrups, K. E., Bork-Jensen, J., Gjesing, A. P., Kontto, J., Perola, M., Shaw-Hawkins, S., Havulinna, A. S., Zhang, H., Donnelly, L. A., Groves, C. J., Rayner, N. W., Neville, M. J., Robertson, N. R., Yiorkas, A. M., Herzig, K., Kajantie, E., Zhang, W., Willems, S. M., Lannfelt, L., Malerba, G., Soranzo, N., Trabetti, E., Verweij, N., Evangelou, E., Moayyeri, A., Vergnaud, A., Nelson, C. P., Poveda, A., Varga, T. V., Caslake, M., de Craen, A. J., Trompet, S., Luan, J., Scott, R. A., Harris, S. E., Liewald, D. C., Marioni, R., Menni, C., Farmaki, A., Hallmans, G., Renstrom, F., Huffman, J. E., Hassinen, M., Burgess, S., Vasan, R. S., Felix, J. F., Uria-Nickelsen, M., Malarstign, A., Reilly, D. F., Hoek, M., Vogt, T. F., Lin, H., Lieb, W., Traylor, M., Markus, H. S., Highland, H. M., Justice, A. E., Marouli, E., Lindstrom, J., Uusitupa, M., Komulainen, P., Lakka, T. A., Rauramaa, R., Polasek, O., Rudan, I., Rolandsson, O., Franks, P. W., Dedoussis, G., Spector, T. D., Jousilahti, P., Mannisto, S., Deary, I. J., Starr, J. M., Langenberg, C., Wareham, N. J., Brown, M. J., Dominiczak, A. F., Connell, J. M., Jukema, J. W., Sattar, N., Ford, I., Packard, C. J., Esko, T., Magi, R., Metspalu, A., de Boer, R. A., van der Meer, P., van der Harst, P., Gambaro, G., Ingelsson, E., Lind, L., de Bakker, P. I., Numans, M. E., Brandslund, I., Christensen, C., Petersen, E. R., Korpi-Hyovalti, E., Oksa, H., Chambers, J. C., Kooner, J. S., Blakemore, A. I., Franks, S., Jarvelin, M., Husemoen, L. L., Linneberg, A., Skaaby, T., Thuesen, B., Karpe, F., Tuomilehto, J., Doney, A. S., Morris, A. D., Palmer, C. N., Holmen, O. L., Hveem, K., Willer, C. J., Tuomi, T., Groop, L., Karajamaki, A., Palotie, A., Ripatti, S., Salomaa, V., Alam, D. S., Majmnder, A. A., Di Angelantonio, E., Chowdhury, R., McCarthy, M. I., Poulter, N., Stanton, A. V., Sever, P., Amouyel, P., Arveiler, D., Blankenberg, S., Ferrieres, J., Kee, F., Kuulasmaa, K., Muller-Nurasyid, M., Veronesi, G., Virtamo, J., Deloukas, P., Elliott, P., Zeggini, E., Kathiresan, S., Melander, O., Kuusisto, J., Laakso, M., Padmanabhan, S., Porteous, D. J., Hayward, C., Scotland, G., Collins, F. S., Mohlke, K. L., Hansen, T., Pedersen, O., Boehnke, M., Stringham, H. M., Frossard, P., Newton-Cheh, C., Tobin, M. D., Nordestgaard, B. G., Caulfield, M. J., Mahajan, A., Morris, A. P., Tomaszewski, M., Samani, N. J., Saleheen, D., Asselbergs, F. W., Lindgren, C. M., Danesh, J., Wain, L. V., Butterworth, A. S., Howson, J. M., Munroe, P. B. 2016; 48 (10): 1151-1161

    Abstract

    High blood pressure is a major risk factor for cardiovascular disease and premature death. However, there is limited knowledge on specific causal genes and pathways. To better understand the genetics of blood pressure, we genotyped 242,296 rare, low-frequency and common genetic variants in up to 192,763 individuals and used ∼155,063 samples for independent replication. We identified 30 new blood pressure- or hypertension-associated genetic regions in the general population, including 3 rare missense variants in RBM47, COL21A1 and RRAS with larger effects (>1.5 mm Hg/allele) than common variants. Multiple rare nonsense and missense variant associations were found in A2ML1, and a low-frequency nonsense variant in ENPEP was identified. Our data extend the spectrum of allelic variation underlying blood pressure traits and hypertension, provide new insights into the pathophysiology of hypertension and indicate new targets for clinical intervention.

    View details for DOI 10.1038/ng.3654

    View details for Web of Science ID 000384391600010

    View details for PubMedID 27618447

    View details for PubMedCentralID PMC5056636

  • Genome-Wide Association Study of the Modified Stumvoll Insulin Sensitivity Index Identifies BCL2 and FAM19A2 as Novel Insulin Sensitivity Loci. Diabetes Walford, G. A., Gustafsson, S., Rybin, D., Stancáková, A., Chen, H., Liu, C., Hong, J., Jensen, R. A., Rice, K., Morris, A. P., Mägi, R., Tönjes, A., Prokopenko, I., Kleber, M. E., Delgado, G., Silbernagel, G., Jackson, A. U., Appel, E. V., Grarup, N., Lewis, J. P., Montasser, M. E., Landenvall, C., Staiger, H., Luan, J., Frayling, T. M., Weedon, M. N., Xie, W., Morcillo, S., Martínez-Larrad, M. T., Biggs, M. L., Chen, Y. I., Corbaton-Anchuelo, A., Færch, K., Gómez-Zumaquero, J. M., Goodarzi, M. O., Kizer, J. R., Koistinen, H. A., Leong, A., Lind, L., Lindgren, C., Machicao, F., Manning, A. K., Martín-Núñez, G. M., Rojo-Martínez, G., Rotter, J. I., Siscovick, D. S., Zmuda, J. M., Zhang, Z., Serrano-Rios, M., Smith, U., Soriguer, F., Hansen, T., Jørgensen, T. J., Linnenberg, A., Pedersen, O., Walker, M., Langenberg, C., Scott, R. A., Wareham, N. J., Fritsche, A., Häring, H., Stefan, N., Groop, L., O'Connell, J. R., Boehnke, M., Bergman, R. N., Collins, F. S., Mohlke, K. L., Tuomilehto, J., März, W., Kovacs, P., Stumvoll, M., Psaty, B. M., Kuusisto, J., Laakso, M., Meigs, J. B., Dupuis, J., Ingelsson, E., Florez, J. C. 2016; 65 (10): 3200-3211

    Abstract

    Genome-wide association studies (GWAS) have found few common variants that influence fasting measures of insulin sensitivity. We hypothesized that a GWAS of an integrated assessment of fasting and dynamic measures of insulin sensitivity would detect novel common variants. We performed a GWAS of the modified Stumvoll Insulin Sensitivity Index (ISI) within the Meta-Analyses of Glucose and Insulin-Related Traits Consortium. Discovery for genetic association was performed in 16,753 individuals, and replication was attempted for the 23 most significant novel loci in 13,354 independent individuals. Association with ISI was tested in models adjusted for age, sex, and BMI and in a model analyzing the combined influence of the genotype effect adjusted for BMI and the interaction effect between the genotype and BMI on ISI (model 3). In model 3, three variants reached genome-wide significance: rs13422522 (NYAP2; P = 8.87 × 10(-11)), rs12454712 (BCL2; P = 2.7 × 10(-8)), and rs10506418 (FAM19A2; P = 1.9 × 10(-8)). The association at NYAP2 was eliminated by conditioning on the known IRS1 insulin sensitivity locus; the BCL2 and FAM19A2 associations were independent of known cardiometabolic loci. In conclusion, we identified two novel loci and replicated known variants associated with insulin sensitivity. Further studies are needed to clarify the causal variant and function at the BCL2 and FAM19A2 loci.

    View details for DOI 10.2337/db16-0199

    View details for PubMedID 27416945

    View details for PubMedCentralID PMC5033262

  • Effect of Insulin Resistance on Monounsaturated Fatty Acid Levels: A Multi-cohort Non-targeted Metabolomics and Mendelian Randomization Study. PLoS genetics Nowak, C., Salihovic, S., Ganna, A., Brandmaier, S., Tukiainen, T., Broeckling, C. D., Magnusson, P. K., Prenni, J. E., Wang-Sattler, R., Peters, A., Strauch, K., Meitinger, T., Giedraitis, V., Ärnlöv, J., Berne, C., Gieger, C., Ripatti, S., Lind, L., Pedersen, N. L., Sundström, J., Ingelsson, E., Fall, T. 2016; 12 (10)

    Abstract

    Insulin resistance (IR) and impaired insulin secretion contribute to type 2 diabetes and cardiovascular disease. Both are associated with changes in the circulating metabolome, but causal directions have been difficult to disentangle. We combined untargeted plasma metabolomics by liquid chromatography/mass spectrometry in three non-diabetic cohorts with Mendelian Randomization (MR) analysis to obtain new insights into early metabolic alterations in IR and impaired insulin secretion. In up to 910 elderly men we found associations of 52 metabolites with hyperinsulinemic-euglycemic clamp-measured IR and/or β-cell responsiveness (disposition index) during an oral glucose tolerance test. These implicated bile acid, glycerophospholipid and caffeine metabolism for IR and fatty acid biosynthesis for impaired insulin secretion. In MR analysis in two separate cohorts (n = 2,613) followed by replication in three independent studies profiled on different metabolomics platforms (n = 7,824 / 8,961 / 8,330), we discovered and replicated causal effects of IR on lower levels of palmitoleic acid and oleic acid. A trend for a causal effect of IR on higher levels of tyrosine reached significance only in meta-analysis. In one of the largest studies combining "gold standard" measures for insulin responsiveness with non-targeted metabolomics, we found distinct metabolic profiles related to IR or impaired insulin secretion. We speculate that the causal effects on monounsaturated fatty acid levels could explain parts of the raised cardiovascular disease risk in IR that is independent of diabetes development.

    View details for DOI 10.1371/journal.pgen.1006379

    View details for PubMedID 27768686

  • The genetics of blood pressure regulation and its target organs from association studies in 342,415 individuals NATURE GENETICS Ehret, G. B., Ferreira, T., Chasman, D. I., Jackson, A. U., Schmidt, E. M., Johnson, T., Thorleifsson, G., Luan, J., Donnelly, L. A., Kanoni, S., Petersen, A. -., Pihurl, V., Strawbridge, R. J., Shungin, D., Hughes, M. F., Meirelles, O., Kaakinen, M., Bouatia-Naji, N., Kristiansson, K., Shah, S., Kleber, M. E., Guo, X., Lyytikainen, L., Fava, C., Eriksson, N., Nolte, I. M., Magnusson, P. K., Salfati, E. L., Rallidis, L. S., Theusch, E., Smith, A. J., Folkersen, L., Witkowska, K., Pers, T. H., Joehanes, R., Kim, S. K., Lataniotis, L., Jansen, R., Johnson, A. D., Warren, H., Kim, Y. J., Zhao, W., Wu, Y., Tayo, B. O., Bochud, M., Absher, D., Adair, L. S., Amin, N., Arkingl, D. E., Axelsson, T., Baldassarre, D., Balkau, B., Bandinelli, S., Barnes, M. R., Barroso, I., Bevan, S., Bis, J. C., Bjornsdottir, G., Boehnke, M., Boerwinkle, E., Bonnycastle, L. L., Boomsma, D. I., Bornstein, S. R., Brown, M. J., Burnier, M., Cabrera, C. P., Chambers, J. C., Chang, I., Cheng, C., Chines, P. S., Chung, R., Collins, F. S., Connell, J. M., Doring, A., Dallongeville, J., Danesh, J., de Faire, U., Delgado, G., Dominiczak, A. F., Doney, A. S., Drenos, F., Edkins, S., Eicher, J. D., Elosua, R., Enroth, S., Erdmann, J., Eriksson, P., Esko, T., Evangelou, E., Evans, A., Fai, T., Farra, M., Felixl, J. F., Ferrieres, J., Ferrucci, L., Fornage, M., Forrester, T., Franceschinil, N., Franco, O. H., Franco-Cereceda, A., Fraser, R. M., Ganesh, S. K., Gao, H., Gertow, K., Gianfagna, F., Gigante, B., Giulianini, F., Goe, A., Goodall, A. H., Goodarzi, M., Gorski, M., Grassler, J., Groves, C. J., Gudnason, V., Gyllensten, U., Hallmans, G., Hartikainen, A., Hassinen, M., Havulinna, A. S., Hayward, C., Hercberg, S., Herzig, K., Hicks, A. A., Hingorani, A. D., Hirschhorn, J. N., Hofmanl, A., Holmen, J., Holmen, O. L., Hottenga, J., Howard, P., Hsiung, C. A., Hunt, S. C., Ikram, M. A., Illig, T., Iribarren, C., Jensen, R. A., Kahonen, M., Kang, H. M., Kathiresan, S., Keating, B. J., Khaw, K., Kim, Y. K., Kim, E., Kivimaki, M., Klopp, N., Kolovou, G., Komulainen, P., Kooner, J. S., Kosova, G., Krauss, R. M., Kuh, D., Kutalik, Z., Kuusisto, J., Kvaloy, K., Lakka, T. A., Lee, N. R., Lee, I., Lee, W., Levy, D., Li, X., Liang, K., Lin, H., Lin, L., Lindstrom, J., Lobbens, S., Mannisto, S., Muller, G., Muller-Nurasyid, M., Mach, F., Markus, H. S., Marouli, E., McCarthy, M. I., McKenzie, C. A., Meneton, P., Menni, C., Metspalu, A., Mijatovic, V., Moilanen, L., Montasser, M. E., Morris, A. D., Morrison, A. C., Mulas, A., Nagaraja, R., Narisu, N., Nikus, K., O'Donnell, C. J., O'Reilly, P. F., Ong, K. K., Paccaud, F., Palmer, C. D., Parsa, A., Pedersen, N. L., Penninx, B. W., Perola, M., Peters, A., Poulter, N., Pramstaller, P. P., Psaty, B. M., Quertermous, T., Rao, D. C., Rasheed, A., Rayner, N. W., Renstrom, F., Rettig, R., Rice, K. M., Roberts, R., Rose, L. M., Rossouw, J., Samani, N. J., Sanna, S., Saramies, J., Schunkert, H., Sebert, S., Sheu, W. H., Shin, Y., Sim, X., Smit, J. H., Smith, A. V., Sosa, M. X., Spector, T. D., Stancakova, A., Stanton, A. V., Stirrups, K. E., Stringham, H. M., Sundstrom, J., Swift, A. J., Syvanen, A., Tai, E., Tanaka, T., Tarasov, K. V., Teumer, A., Thorsteinsdottir, U., Tobin, M. D., Tremoli, E., Uitterlinden, A. G., Uusitupa, M., Vaez, A., Vaidya, D., van Duijn, C. M., van Iperen, E. P., Vasan, R. S., Verwoert, G. C., Virtamo, J., Vitart, V., Voight, B. F., Vollenweider, P., Wagner, A., Wain, L. V., Wareham, N. J., Watldns, H., Weder, A. B., Westra, H. J., Wilks, R., Wilsgaard, T., Wilson, J. F., Wong, T. Y., Yang, T., Yao, J., Yengo, L., Zhang, W., Zhao, J. H., Zhu, X., Bovet, P., Cooper, R. S., Mohlke, K. L., Saleheen, D., Lee, J., Elliott, P., Gierman, H. J., Willer, C. J., Franke, L., Hovingh, G. K., Taylor, K. D., Dedoussis, G., Sever, P., Wong, A., Lind, L., Assimes, T. L., Njolstad, I., Schwarz, P. E., Langenberg, C., Snieder, H., Caulfield, M. J., Melander, E., Laakso, M., Saltevo, J., Rauramaa, R., Tuomilehto, J., Ingelsson, E., Lehtimaki, T., Hveem, K., Palmas, W., Marz, W., Kumar, M., Salomaa, V., Chen, Y. I., Rotter, J. I., Froguel, P., Jarvelin, M., Lakatta, E. G., Kuulasmaa, K., Franks, P. W., Hamsten, A., Wichmann, H., Palmer, C. N., Stefansson, K., Ridker, P. M., Loos, R. J., Chalcravarti, A., Deloukas, P., Morris, A. P., Newton-Cheh, C., Munroe, P. B. 2016; 48 (10): 1171-1184

    Abstract

    To dissect the genetic architecture of blood pressure and assess effects on target organ damage, we analyzed 128,272 SNPs from targeted and genome-wide arrays in 201,529 individuals of European ancestry, and genotypes from an additional 140,886 individuals were used for validation. We identified 66 blood pressure-associated loci, of which 17 were new; 15 harbored multiple distinct association signals. The 66 index SNPs were enriched for cis-regulatory elements, particularly in vascular endothelial cells, consistent with a primary role in blood pressure control through modulation of vascular tone across multiple tissues. The 66 index SNPs combined in a risk score showed comparable effects in 64,421 individuals of non-European descent. The 66-SNP blood pressure risk score was significantly associated with target organ damage in multiple tissues but with minor effects in the kidney. Our findings expand current knowledge of blood pressure-related pathways and highlight tissues beyond the classical renal system in blood pressure regulation.

    View details for DOI 10.1038/ng.3667

    View details for Web of Science ID 000384391600012

    View details for PubMedCentralID PMC5042863

  • The genetics of blood pressure regulation and its target organs from association studies in 342,415 individuals. Nature genetics Ehret, G. B., Ferreira, T., Chasman, D. I., Jackson, A. U., Schmidt, E. M., Johnson, T., Thorleifsson, G., Luan, J., Donnelly, L. A., Kanoni, S., Petersen, A., Pihur, V., Strawbridge, R. J., Shungin, D., Hughes, M. F., Meirelles, O., Kaakinen, M., Bouatia-Naji, N., Kristiansson, K., Shah, S., Kleber, M. E., Guo, X., Lyytikäinen, L., Fava, C., Eriksson, N., Nolte, I. M., Magnusson, P. K., Salfati, E. L., Rallidis, L. S., Theusch, E., Smith, A. J., Folkersen, L., Witkowska, K., Pers, T. H., Joehanes, R., Kim, S. K., Lataniotis, L., Jansen, R., Johnson, A. D., Warren, H., Kim, Y. J., Zhao, W., Wu, Y., Tayo, B. O., Bochud, M., Absher, D., Adair, L. S., Amin, N., Arking, D. E., Axelsson, T., Baldassarre, D., Balkau, B., Bandinelli, S., Barnes, M. R., Barroso, I., Bevan, S., Bis, J. C., Bjornsdottir, G., Boehnke, M., Boerwinkle, E., Bonnycastle, L. L., Boomsma, D. I., Bornstein, S. R., Brown, M. J., Burnier, M., Cabrera, C. P., Chambers, J. C., Chang, I., Cheng, C., Chines, P. S., Chung, R., Collins, F. S., Connell, J. M., Döring, A., Dallongeville, J., Danesh, J., de Faire, U., Delgado, G., Dominiczak, A. F., Doney, A. S., Drenos, F., Edkins, S., Eicher, J. D., Elosua, R., Enroth, S., Erdmann, J., Eriksson, P., Esko, T., Evangelou, E., Evans, A., Fall, T., Farrall, M., Felix, J. F., Ferrières, J., Ferrucci, L., Fornage, M., Forrester, T., Franceschini, N., Franco, O. H., Franco-Cereceda, A., Fraser, R. M., Ganesh, S. K., Gao, H., Gertow, K., Gianfagna, F., Gigante, B., Giulianini, F., Goel, A., Goodall, A. H., Goodarzi, M. O., Gorski, M., Gräßler, J., Groves, C. J., Gudnason, V., Gyllensten, U., Hallmans, G., Hartikainen, A., Hassinen, M., Havulinna, A. S., Hayward, C., Hercberg, S., Herzig, K., Hicks, A. A., Hingorani, A. D., Hirschhorn, J. N., Hofman, A., Holmen, J., Holmen, O. L., Hottenga, J., Howard, P., Hsiung, C. A., Hunt, S. C., Ikram, M. A., Illig, T., Iribarren, C., Jensen, R. A., Kähönen, M., Kang, H. M., Kathiresan, S., Keating, B. J., Khaw, K., Kim, Y. K., Kim, E., Kivimaki, M., Klopp, N., Kolovou, G., Komulainen, P., Kooner, J. S., Kosova, G., Krauss, R. M., Kuh, D., Kutalik, Z., Kuusisto, J., Kvaløy, K., Lakka, T. A., Lee, N. R., Lee, I., Lee, W., Levy, D., Li, X., Liang, K., Lin, H., Lin, L., Lindström, J., Lobbens, S., Männistö, S., Müller, G., Müller-Nurasyid, M., Mach, F., Markus, H. S., Marouli, E., McCarthy, M. I., McKenzie, C. A., Meneton, P., Menni, C., Metspalu, A., Mijatovic, V., Moilanen, L., Montasser, M. E., Morris, A. D., Morrison, A. C., Mulas, A., Nagaraja, R., Narisu, N., Nikus, K., O'Donnell, C. J., O'Reilly, P. F., Ong, K. K., Paccaud, F., Palmer, C. D., Parsa, A., Pedersen, N. L., Penninx, B. W., Perola, M., Peters, A., Poulter, N., Pramstaller, P. P., Psaty, B. M., Quertermous, T., Rao, D. C., Rasheed, A., Rayner, N. W., Renström, F., Rettig, R., Rice, K. M., Roberts, R., Rose, L. M., Rossouw, J., Samani, N. J., Sanna, S., Saramies, J., Schunkert, H., Sebert, S., Sheu, W. H., Shin, Y., Sim, X., Smit, J. H., Smith, A. V., Sosa, M. X., Spector, T. D., Stancáková, A., Stanton, A. V., Stirrups, K. E., Stringham, H. M., Sundstrom, J., Swift, A. J., Syvänen, A., Tai, E., Tanaka, T., Tarasov, K. V., Teumer, A., Thorsteinsdottir, U., Tobin, M. D., Tremoli, E., Uitterlinden, A. G., Uusitupa, M., Vaez, A., Vaidya, D., van Duijn, C. M., van Iperen, E. P., Vasan, R. S., Verwoert, G. C., Virtamo, J., Vitart, V., Voight, B. F., Vollenweider, P., Wagner, A., Wain, L. V., Wareham, N. J., Watkins, H., Weder, A. B., Westra, H., Wilks, R., Wilsgaard, T., Wilson, J. F., Wong, T. Y., Yang, T., Yao, J., Yengo, L., Zhang, W., Zhao, J. H., Zhu, X., Bovet, P., Cooper, R. S., Mohlke, K. L., Saleheen, D., Lee, J., Elliott, P., Gierman, H. J., Willer, C. J., Franke, L., Hovingh, G. K., Taylor, K. D., Dedoussis, G., Sever, P., Wong, A., Lind, L., Assimes, T. L., Njølstad, I., Schwarz, P. E., Langenberg, C., Snieder, H., Caulfield, M. J., Melander, O., Laakso, M., Saltevo, J., Rauramaa, R., Tuomilehto, J., Ingelsson, E., Lehtimäki, T., Hveem, K., Palmas, W., März, W., Kumari, M., Salomaa, V., Chen, Y. I., Rotter, J. I., Froguel, P., Jarvelin, M., Lakatta, E. G., Kuulasmaa, K., Franks, P. W., Hamsten, A., Wichmann, H., Palmer, C. N., Stefansson, K., Ridker, P. M., Loos, R. J., Chakravarti, A., Deloukas, P., Morris, A. P., Newton-Cheh, C., Munroe, P. B. 2016; 48 (10): 1171-1184

    Abstract

    To dissect the genetic architecture of blood pressure and assess effects on target organ damage, we analyzed 128,272 SNPs from targeted and genome-wide arrays in 201,529 individuals of European ancestry, and genotypes from an additional 140,886 individuals were used for validation. We identified 66 blood pressure-associated loci, of which 17 were new; 15 harbored multiple distinct association signals. The 66 index SNPs were enriched for cis-regulatory elements, particularly in vascular endothelial cells, consistent with a primary role in blood pressure control through modulation of vascular tone across multiple tissues. The 66 index SNPs combined in a risk score showed comparable effects in 64,421 individuals of non-European descent. The 66-SNP blood pressure risk score was significantly associated with target organ damage in multiple tissues but with minor effects in the kidney. Our findings expand current knowledge of blood pressure-related pathways and highlight tissues beyond the classical renal system in blood pressure regulation.

    View details for DOI 10.1038/ng.3667

    View details for PubMedID 27618452

    View details for PubMedCentralID PMC5042863

  • Enabling Efficient and Confident Annotation of LC-MS Metabolomics Data through MS1 Spectrum and Time Prediction ANALYTICAL CHEMISTRY Broeckling, C. D., Ganna, A., Layer, M., Brown, K., Sutton, B., Ingelsson, E., Peers, G., Prenni, J. E. 2016; 88 (18): 9226-9234

    Abstract

    Liquid chromatography coupled to electrospray ionization-mass spectrometry (LC-ESI-MS) is a versatile and robust platform for metabolomic analysis. However, while ESI is a soft ionization technique, in-source phenomena including multimerization, nonproton cation adduction, and in-source fragmentation complicate interpretation of MS data. Here, we report chromatographic and mass spectrometric behavior of 904 authentic standards collected under conditions identical to a typical nontargeted profiling experiment. The data illustrate that the often high level of complexity in MS spectra is likely to result in misinterpretation during the annotation phase of the experiment and a large overestimation of the number of compounds detected. However, our analysis of this MS spectral library data indicates that in-source phenomena are not random but depend at least in part on chemical structure. These nonrandom patterns enabled predictions to be made as to which in-source signals are likely to be observed for a given compound. Using the authentic standard spectra as a training set, we modeled the in-source phenomena for all compounds in the Human Metabolome Database to generate a theoretical in-source spectrum and retention time library. A novel spectral similarity matching platform was developed to facilitate efficient spectral searching for nontargeted profiling applications. Taken together, this collection of experimental spectral data, predictive modeling, and informatic tools enables more efficient, reliable, and transparent metabolite annotation.

    View details for DOI 10.1021/acs.analchem.6b02479

    View details for Web of Science ID 000384038400041

    View details for PubMedID 27560453

  • Trans-ethnic Fine Mapping Highlights Kidney-Function Genes Linked to Salt Sensitivity. American journal of human genetics Mahajan, A., Rodan, A. R., Le, T. H., Gaulton, K. J., Haessler, J., Stilp, A. M., Kamatani, Y., Zhu, G., Sofer, T., Puri, S., Schellinger, J. N., Chu, P., Cechova, S., Van Zuydam, N., Arnlov, J., Flessner, M. F., Giedraitis, V., Heath, A. C., Kubo, M., Larsson, A., Lindgren, C. M., Madden, P. A., Montgomery, G. W., Papanicolaou, G. J., Reiner, A. P., Sundström, J., Thornton, T. A., Lind, L., Ingelsson, E., Cai, J., Martin, N. G., Kooperberg, C., Matsuda, K., Whitfield, J. B., Okada, Y., Laurie, C. C., Morris, A. P., Franceschini, N. 2016; 99 (3): 636-646

    Abstract

    We analyzed genome-wide association studies (GWASs), including data from 71,638 individuals from four ancestries, for estimated glomerular filtration rate (eGFR), a measure of kidney function used to define chronic kidney disease (CKD). We identified 20 loci attaining genome-wide-significant evidence of association (p < 5 × 10(-8)) with kidney function and highlighted that allelic effects on eGFR at lead SNPs are homogeneous across ancestries. We leveraged differences in the pattern of linkage disequilibrium between diverse populations to fine-map the 20 loci through construction of "credible sets" of variants driving eGFR association signals. Credible variants at the 20 eGFR loci were enriched for DNase I hypersensitivity sites (DHSs) in human kidney cells. DHS credible variants were expression quantitative trait loci for NFATC1 and RGS14 (at the SLC34A1 locus) in multiple tissues. Loss-of-function mutations in ancestral orthologs of both genes in Drosophila melanogaster were associated with altered sensitivity to salt stress. Renal mRNA expression of Nfatc1 and Rgs14 in a salt-sensitive mouse model was also reduced after exposure to a high-salt diet or induced CKD. Our study (1) demonstrates the utility of trans-ethnic fine mapping through integration of GWASs involving diverse populations with genomic annotation from relevant tissues to define molecular mechanisms by which association signals exert their effect and (2) suggests that salt sensitivity might be an important marker for biological processes that affect kidney function and CKD in humans.

    View details for DOI 10.1016/j.ajhg.2016.07.012

    View details for PubMedID 27588450

    View details for PubMedCentralID PMC5011075

  • Cystatin C and Cardiovascular Disease: A Mendelian Randomization Study. Journal of the American College of Cardiology van der Laan, S. W., Fall, T., Soumaré, A., Teumer, A., Sedaghat, S., Baumert, J., Zabaneh, D., van Setten, J., Isgum, I., Galesloot, T. E., Arpegård, J., Amouyel, P., Trompet, S., Waldenberger, M., Dörr, M., Magnusson, P. K., Giedraitis, V., Larsson, A., Morris, A. P., Felix, J. F., Morrison, A. C., Franceschini, N., Bis, J. C., Kavousi, M., O'Donnell, C., Drenos, F., Tragante, V., Munroe, P. B., Malik, R., Dichgans, M., Worrall, B. B., Erdmann, J., Nelson, C. P., Samani, N. J., Schunkert, H., Marchini, J., Patel, R. S., Hingorani, A. D., Lind, L., Pedersen, N. L., de Graaf, J., Kiemeney, L. A., Baumeister, S. E., Franco, O. H., Hofman, A., Uitterlinden, A. G., Koenig, W., Meisinger, C., Peters, A., Thorand, B., Jukema, J. W., Eriksen, B. O., Toft, I., Wilsgaard, T., Onland-Moret, N. C., van der Schouw, Y. T., Debette, S., Kumari, M., Svensson, P., van der Harst, P., Kivimaki, M., Keating, B. J., Sattar, N., Dehghan, A., Reiner, A. P., Ingelsson, E., Den Ruijter, H. M., de Bakker, P. I., Pasterkamp, G., Ärnlöv, J., Holmes, M. V., Asselbergs, F. W. 2016; 68 (9): 934-945

    Abstract

    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.The aim of this study was to use Mendelian randomization to investigate whether cystatin C is causally related to CVD in the general population.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.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.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.

    View details for DOI 10.1016/j.jacc.2016.05.092

    View details for PubMedID 27561768

  • The genetic architecture of type 2 diabetes NATURE Fuchsberger, C., Flannick, J., Teslovich, T. M., Mahajan, A., Agarwala, V., Gaulton, K. J., Ma, C., Fontanillas, P., Moutsianas, L., McCarthy, D. J., Rivas, M. A., Perry, J. R., Sim, X., Blackwell, T. W., Robertson, N. R., Rayner, N. W., Cingolani, P., Locke, A. E., Tajes, J. F., Highland, H. M., Dupuis, J., Chines, P. S., Lindgren, C. M., Hartl, C., Jackson, A. U., Chen, H., Huyghe, J. R., van de Bunt, M., Pearson, R. D., Kumar, A., Mueller-Nurasyid, M., Grarup, N., Stringham, H. M., Gamazon, E. R., Lee, J., Chen, Y., Scott, R. A., Below, J. E., Chen, P., Huang, J., Go, M. J., Stitzel, M. L., Pasko, D., Parker, S. C., Varga, T. V., Green, T., Beer, N. L., Day-Williams, A. G., Ferreira, T., Fingerlin, T., Horikoshi, M., Hu, C., Huh, I., Ikram, M. K., Kim, B., Kim, Y., Kim, Y. J., Kwon, M., Lee, J., Lee, S., Lin, K., Maxwell, T. J., Nagai, Y., Wang, X., Welch, R. P., Yoon, J., Zhang, W., Barzilai, N., Voight, B. F., Han, B., Jenkinson, C. P., Kuulasmaa, T., Kuusisto, J., Manning, A., Ng, M. C., Palmer, N. D., Balkau, B., Stancakova, A., Abboud, H. E., Boeing, H., Giedraitis, V., Prabhakaran, D., Gottesman, O., Scott, J., Carey, J., Kwan, P., Grant, G., Smith, J. D., Neale, B. M., Purcell, S., Butterworth, A. S., Howson, J. M., Lee, H. M., Lu, Y., Kwak, S., Zhao, W., Danesh, J., Lam, V. K., Park, K. S., Saleheen, D., So, W. Y., Tam, C. H., Afzal, U., Aguilar, D., Arya, R., Aung, T., Chan, E., Navarro, C., Cheng, C., Palli, D., Correa, A., Curran, J. E., Rybin, D., Farook, V. S., Fowler, S. P., Freedman, B. I., Griswold, M., Hale, D. E., Hicks, P. J., Khor, C., Kumar, S., Lehne, B., Thuillier, D., Lim, W. Y., Liu, J., van der Schouw, Y. T., Loh, M., Musani, S. K., Puppala, S., Scott, W. R., Yengo, L., Tan, S., Taylor, H. A., Thameem, F., Wilson, G., Wong, T. Y., Njolstad, P. R., Levy, J. C., Mangino, M., Bonnycastle, L. L., Schwarzmayr, T., Fadista, J., Surdulescu, G. L., Herder, C., Groves, C. J., Wieland, T., Bork-Jensen, J., Brandslund, I., Christensen, C., Koistinen, H. A., Doney, A. S., Kinnunen, L., Esko, T., Farmer, A. J., Hakaste, L., Hodgkiss, D., Kravic, J., Lyssenko, V., Hollensted, M., Jorgensen, M. E., Jorgensen, T., Ladenvall, C., Justesen, J. M., Karajamaki, A., Kriebel, J., Rathmann, W., Lannfelt, L., Lauritzen, T., Narisu, N., Linneberg, A., Melander, O., Milani, L., Neville, M., Orho-Melander, M., Qi, L., Qi, Q., Roden, M., Rolandsson, O., Swift, A., Rosengren, A. H., Stirrups, K., Wood, A. R., Mihailov, E., Blancher, C., Carneiro, M. O., Maguire, J., Poplin, R., Shakir, K., Fennell, T., DePristo, M., de Angelis, M. H., Deloukas, P., Gjesing, A. P., Jun, G., Nilsson, P., Murphy, J., Onofrio, R., Thorand, B., Hansen, T., Meisinger, C., Hu, F. B., Isomaa, B., Karpe, F., Liang, L., Peters, A., Huth, C., O'Rahilly, S. P., Palmer, C. N., Pedersen, O., Rauramaa, R., Tuomilehto, J., Salomaa, V., Watanabe, R. M., Syvanen, A., Bergman, R. N., Bharadwaj, D., Bottinger, E. P., Cho, Y. S., Chandak, G. R., Chan, J. C., Chia, K. S., Daly, M. J., Ebrahim, S. B., Langenberg, C., Elliott, P., Jablonski, K. A., Lehman, D. M., Jia, W., Ma, R. C., Pollin, T. I., Sandhu, M., Tandon, N., Froguel, P., Barroso, I., Teo, Y. Y., Zeggini, E., Loos, R. J., Small, K. S., Ried, J. S., DeFronzo, R. A., Grallert, H., Glaser, B., Metspalu, A., Wareham, N. J., Walker, M., Banks, E., Gieger, C., Ingelsson, E., Im, H. K., Illig, T., Franks, P. W., Buck, G., Trakalo, J., Buck, D., Prokopenko, I., Magi, R., Lind, L., Farjoun, Y., Owen, K. R., Gloyn, A. L., Strauch, K., Tuomi, T., Kooner, J. S., Lee, J., Park, T., Donnelly, P., Morris, A. D., Hattersley, A. T., Bowden, D. W., Collins, F. S., Atzmon, G., Chambers, J. C., Spector, T. D., Laakso, M., Strom, T. M., Bell, G. I., Blangero, J., Duggirala, R., Tai, E. S., McVean, G., Hanis, C. L., Wilson, J. G., Seielstad, M., Frayling, T. M., Meigs, J. B., Cox, N. J., Sladek, R., Lander, E. S., Gabriel, S., Burtt, N. P., Mohlke, K. L., Meitinger, T., Groop, L., Abecasis, G., Florez, J. C., Scott, L. J., Morris, A. P., Kang, H. M., Boehnke, M., Altshuler, D., McCarthy, M. I. 2016; 536 (7614): 41-?

    Abstract

    The genetic architecture of common traits, including the number, frequency, and effect sizes of inherited variants that contribute to individual risk, has been long debated. Genome-wide association studies have identified scores of common variants associated with type 2 diabetes, but in aggregate, these explain only a fraction of the heritability of this disease. Here, to test the hypothesis that lower-frequency variants explain much of the remainder, the GoT2D and T2D-GENES consortia performed whole-genome sequencing in 2,657 European individuals with and without diabetes, and exome sequencing in 12,940 individuals from five ancestry groups. To increase statistical power, we expanded the sample size via genotyping and imputation in a further 111,548 subjects. Variants associated with type 2 diabetes after sequencing were overwhelmingly common and most fell within regions previously identified by genome-wide association studies. Comprehensive enumeration of sequence variation is necessary to identify functional alleles that provide important clues to disease pathophysiology, but large-scale sequencing does not support the idea that lower-frequency variants have a major role in predisposition to type 2 diabetes.

    View details for DOI 10.1038/nature18642

    View details for Web of Science ID 000380999200026

    View details for PubMedID 27398621

  • ?-3 Polyunsaturated Fatty Acid Biomarkers and Coronary Heart Disease: Pooling Project of 19 Cohort Studies. JAMA internal medicine Del Gobbo, L. C., Imamura, F., Aslibekyan, S., Marklund, M., Virtanen, J. K., Wennberg, M., Yakoob, M. Y., Chiuve, S. E., Dela Cruz, L., Frazier-Wood, A. C., Fretts, A. M., Guallar, E., Matsumoto, C., Prem, K., Tanaka, T., Wu, J. H., Zhou, X., Helmer, C., Ingelsson, E., Yuan, J., Barberger-Gateau, P., Campos, H., Chaves, P. H., Djoussé, L., Giles, G. G., Gómez-Aracena, J., Hodge, A. M., Hu, F. B., Jansson, J., Johansson, I., Khaw, K., Koh, W., Lemaitre, R. N., Lind, L., Luben, R. N., Rimm, E. B., Risérus, U., Samieri, C., Franks, P. W., Siscovick, D. S., Stampfer, M., Steffen, L. M., Steffen, B. T., Tsai, M. Y., van Dam, R. M., Voutilainen, S., Willett, W. C., Woodward, M., Mozaffarian, D. 2016; 176 (8): 1155-1166

    Abstract

    The role of ω-3 polyunsaturated fatty acids for primary prevention of coronary heart disease (CHD) remains controversial. Most prior longitudinal studies evaluated self-reported consumption rather than biomarkers.To evaluate biomarkers of seafood-derived eicosapentaenoic acid (EPA; 20:5ω-3), docosapentaenoic acid (DPA; 22:5ω-3), and docosahexaenoic acid (DHA; 22:6ω-3) and plant-derived α-linolenic acid (ALA; 18:3ω-3) for incident CHD.A global consortium of 19 studies identified by November 2014.Available prospective (cohort, nested case-control) or retrospective studies with circulating or tissue ω-3 biomarkers and ascertained CHD.Each study conducted standardized, individual-level analysis using harmonized models, exposures, outcomes, and covariates. Findings were centrally pooled using random-effects meta-analysis. Heterogeneity was examined by age, sex, race, diabetes, statins, aspirin, ω-6 levels, and FADS desaturase genes.Incident total CHD, fatal CHD, and nonfatal myocardial infarction (MI).The 19 studies comprised 16 countries, 45 637 unique individuals, and 7973 total CHD, 2781 fatal CHD, and 7157 nonfatal MI events, with ω-3 measures in total plasma, phospholipids, cholesterol esters, and adipose tissue. Median age at baseline was 59 years (range, 18-97 years), and 28 660 (62.8%) were male. In continuous (per 1-SD increase) multivariable-adjusted analyses, the ω-3 biomarkers ALA, DPA, and DHA were associated with a lower risk of fatal CHD, with relative risks (RRs) of 0.91 (95% CI, 0.84-0.98) for ALA, 0.90 (95% CI, 0.85-0.96) for DPA, and 0.90 (95% CI, 0.84-0.96) for DHA. Although DPA was associated with a lower risk of total CHD (RR, 0.94; 95% CI, 0.90-0.99), ALA (RR, 1.00; 95% CI, 0.95-1.05), EPA (RR, 0.94; 95% CI, 0.87-1.02), and DHA (RR, 0.95; 95% CI, 0.91-1.00) were not. Significant associations with nonfatal MI were not evident. Associations appeared generally stronger in phospholipids and total plasma. Restricted cubic splines did not identify evidence of nonlinearity in dose responses.On the basis of available studies of free-living populations globally, biomarker concentrations of seafood and plant-derived ω-3 fatty acids are associated with a modestly lower incidence of fatal CHD.

    View details for DOI 10.1001/jamainternmed.2016.2925

    View details for PubMedID 27357102

  • Identification of additional risk loci for stroke and small vessel disease: a meta-analysis of genome-wide association studies LANCET NEUROLOGY Chauhan, G., Arnold, C. R., Chu, A. Y., Fornage, M., Reyahi, A., Bis, J. C., Havulinna, A. S., Sargurupremraj, M., Smith, A. V., Adams, H. H., Choi, S. H., Pulit, S. L., Trompet, S., Garcia, M. E., Manichaikul, A., Teumer, A., Gustafsson, S., Bartz, T. M., Bellenguez, C., Vidal, J. S., Jian, X., Kjartansson, O., Wiggins, K. L., Satizabal, C. L., Xue, F., Ripatti, S., Liu, Y., Deelen, J., Den Hoed, M., Bevan, S., Hopewell, J. C., Malik, R., Heckbert, S. R., Rice, K., Smith, N. L., Levi, C., Sharma, P., Sudlow, C. L., Nik, A. M., Cole, J. W., Schmidt, R., Meschia, J., Thijs, V., Lindgren, A., Melander, O., Grewal, R. P., Sacco, R. L., Rundek, T., Rothwell, P. M., Arnett, D. K., Jern, C., Johnson, J. A., Benavente, O. R., Wassertheil-Smoller, S., Lee, J., Wong, Q., Aparicio, H. J., Engelter, S. T., Kloss, M., Leys, D., Pezzini, A., Buring, J. E., Ridker, P. M., Berr, C., Dartigues, J., Hamsten, A., Magnusson, P. K., Traylor, M., Pedersen, N. L., Lannfelt, L., Lind, L., Lindgren, C. M., Morris, A. P., Jimenez-Conde, J., Montaner, J., Radmanesh, F., Slowik, A., Woo, D., Hofman, A., Koudstaal, P. J., Portegies, M. L., Uitterlinden, A. G., de Craen, A. J., Ford, I., Jukema, J. W., Stott, D. J., Allen, N. B., Sale, M. M., Johnson, A. D., Bennett, D. A., De Jager, P. L., White, C. C., Grabe, H. J., Markus, M. R., Schminke, U., Boncoraglio, G. B., Clarke, R., Kamatani, Y., Dallongeville, J., Lopez, O. L., Rotter, J. I., Nails, M. A., Gottesman, R. F., Griswold, M. E., Knopman, D. S., Windham, B. G., Beiser, A., Markus, H. S., Vartiainen, E., French, C. R., Dichgans, M., Pastinen, T., Lathrop, M., Gudnason, V., Kurth, T., Psaty, B. M., Harris, T. B., Rich, S. S., DeStefano, A. L., Schmidt, C. O., Worrall, B. B., Rosand, J., Salomaa, V., Mosley, T. H., Ingelsson, E., van Duijn, C. M., Tzourio, C., Rexrode, K. M., Lehmann, O. J., Launer, L. J., Ikram, M. A., Carlsson, P., Chasman, D. I., Childs, S. J., Longstreth Junior, W. T., Seshadri, S., Debette, S. 2016; 15 (7): 695-707
  • The metabolic fingerprint of p,p '-DDE and HCB exposure in humans ENVIRONMENT INTERNATIONAL Salihovic, S., Ganna, A., Fall, T., Broeckling, C. D., Prenni, J. E., van Bavel, B., Lind, P. M., Ingelsson, E., Lind, L. 2016; 88: 60-66

    Abstract

    Dichlorodiphenyldichloroethylene (p,p'-DDE) and hexachlorobenzene (HCB) are organochlorine pesticides with well-known endocrine disrupting properties. Exposure to p,p'-DDE and HCB concerns human populations worldwide and has been linked to metabolic disorders such as obesity and type 2 diabetes, but details about these associations in humans from the general population are largely unknown.We investigated the associations between p,p'-DDE and HCB exposure and global metabolomic profiles in serum samples from 1016 participants from the Swedish population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study.HCB and p,p'-DDE levels were determined using gas chromatography coupled to high-resolution mass spectrometry (GC-HRMS). Metabolite levels were determined by using a non-targeted metabolomics approach with ultra-performance liquid chromatography coupled to time-of- flight mass spectrometry (UPLC-TOFMS). Association analyses were performed using multivariate linear regression.We found circulating levels of p,p-DDE and HCB to be significantly associated with circulating levels of 16 metabolites following adjustment for age, sex, education level, exercise habits, smoking, energy intake, and alcohol intake. The majority of the 16 metabolites belong to lipid metabolism pathways and include fatty acids, glycerophospholipids, sphingolipids, and glycerolipids. Overall, p,p'-DDE and HCB levels were found to be correlated to different metabolites, which suggests that different metabolic fingerprints may be related to circulating levels of these two pesticides.Our findings establish a link between human exposure to organochlorine pesticides and metabolites of key metabolic processes mainly related to human lipid metabolism.

    View details for DOI 10.1016/j.envint.2015.12.015

    View details for Web of Science ID 000371359300009

    View details for PubMedID 26720637

  • Six Novel Loci Associated with Circulating VEGF Levels Identified by a Meta-analysis of Genome-Wide Association Studies PLOS GENETICS Choi, S. H., Ruggiero, D., Sorice, R., Song, C., Nutile, T., Smith, A. V., Concas, M. P., Traglia, M., Barbieri, C., Ndiaye, N. C., Stathopoulou, M. G., Lagou, V., Maestrale, G. B., Sala, C., Debette, S., Kovacs, P., Lind, L., Lamont, J., Fitzgerald, P., Toenjes, A., Gudnason, V., Toniolo, D., Pirastu, M., Bellenguez, C., Vasan, R. S., Ingelsson, E., Leutenegger, A., Johnson, A. D., DeStefano, A. L., Visvikis-Siest, S., Seshadri, S., Ciullo, M. 2016; 12 (2)

    Abstract

    Vascular endothelial growth factor (VEGF) is an angiogenic and neurotrophic factor, secreted by endothelial cells, known to impact various physiological and disease processes from cancer to cardiovascular disease and to be pharmacologically modifiable. We sought to identify novel loci associated with circulating VEGF levels through a genome-wide association meta-analysis combining data from European-ancestry individuals and using a dense variant map from 1000 genomes imputation panel. Six discovery cohorts including 13,312 samples were analyzed, followed by in-silico and de-novo replication studies including an additional 2,800 individuals. A total of 10 genome-wide significant variants were identified at 7 loci. Four were novel loci (5q14.3, 10q21.3, 16q24.2 and 18q22.3) and the leading variants at these loci were rs114694170 (MEF2C, P = 6.79 x 10(-13)), rs74506613 (JMJD1C, P = 1.17 x 10(-19)), rs4782371 (ZFPM1, P = 1.59 x 10(-9)) and rs2639990 (ZADH2, P = 1.72 x 10(-8)), respectively. We also identified two new independent variants (rs34528081, VEGFA, P = 1.52 x 10(-18); rs7043199, VLDLR-AS1, P = 5.12 x 10(-14)) at the 3 previously identified loci and strengthened the evidence for the four previously identified SNPs (rs6921438, LOC100132354, P = 7.39 x 10(-1467); rs1740073, C6orf223, P = 2.34 x 10(-17); rs6993770, ZFPM2, P = 2.44 x 10(-60); rs2375981, KCNV2, P = 1.48 x 10(-100)). These variants collectively explained up to 52% of the VEGF phenotypic variance. We explored biological links between genes in the associated loci using Ingenuity Pathway Analysis that emphasized their roles in embryonic development and function. Gene set enrichment analysis identified the ERK5 pathway as enriched in genes containing VEGF associated variants. eQTL analysis showed, in three of the identified regions, variants acting as both cis and trans eQTLs for multiple genes. Most of these genes, as well as some of those in the associated loci, were involved in platelet biogenesis and functionality, suggesting the importance of this process in regulation of VEGF levels. This work also provided new insights into the involvement of genes implicated in various angiogenesis related pathologies in determining circulating VEGF levels. The understanding of the molecular mechanisms by which the identified genes affect circulating VEGF levels could be important in the development of novel VEGF-related therapies for such diseases.

    View details for DOI 10.1371/journal.pgen.1005874

    View details for Web of Science ID 000372554100047

    View details for PubMedID 26910538

  • Prediction impact curve is a new measure integrating intervention effects in the evaluation of risk models JOURNAL OF CLINICAL EPIDEMIOLOGY Campbell, W., Ganna, A., Ingelsson, E., Janssens, A. C. 2016; 69: 89-95

    Abstract

    We propose a new measure of assessing the performance of risk models, the area under the prediction impact curve (auPIC), which quantifies the performance of risk models in terms of their average health impact in the population.Using simulated data, we explain how the prediction impact curve (PIC) estimates the percentage of events prevented when a risk model is used to assign high-risk individuals to an intervention. We apply the PIC to the Atherosclerosis Risk in Communities (ARIC) Study to illustrate its application toward prevention of coronary heart disease.We estimated that if the ARIC cohort received statins at baseline, 5% of events would be prevented when the risk model was evaluated at a cutoff threshold of 20% predicted risk compared to 1% when individuals were assigned to the intervention without the use of a model. By calculating the auPIC, we estimated that an average of 15% of events would be prevented when considering performance across the entire interval.We conclude that the PIC is a clinically meaningful measure for quantifying the expected health impact of risk models that supplements existing measures of model performance.

    View details for DOI 10.1016/j.jclinepi.2015.06.011

    View details for Web of Science ID 000367127600012

    View details for PubMedID 26119889

  • Genome-wide meta-analysis uncovers novel loci influencing circulating leptin levels. Nature communications Kilpeläinen, T. O., Carli, J. F., Skowronski, A. A., Sun, Q., Kriebel, J., Feitosa, M. F., Hedman, Å. K., Drong, A. W., Hayes, J. E., Zhao, J., Pers, T. H., Schick, U., Grarup, N., Kutalik, Z., Trompet, S., Mangino, M., Kristiansson, K., Beekman, M., Lyytikäinen, L., Eriksson, J., Henneman, P., Lahti, J., Tanaka, T., Luan, J., Greco M, F. D., Pasko, D., Renström, F., Willems, S. M., Mahajan, A., Rose, L. M., Guo, X., Liu, Y., Kleber, M. E., Pérusse, L., Gaunt, T., Ahluwalia, T. S., Ju Sung, Y., Ramos, Y. F., Amin, N., Amuzu, A., Barroso, I., Bellis, C., Blangero, J., Buckley, B. M., Böhringer, S., I Chen, Y., de Craen, A. J., Crosslin, D. R., Dale, C. E., Dastani, Z., Day, F. R., Deelen, J., Delgado, G. E., Demirkan, A., Finucane, F. M., Ford, I., Garcia, M. E., Gieger, C., Gustafsson, S., Hallmans, G., Hankinson, S. E., Havulinna, A. S., Herder, C., Hernandez, D., Hicks, A. A., Hunter, D. J., Illig, T., Ingelsson, E., Ioan-Facsinay, A., Jansson, J., Jenny, N. S., Jørgensen, M. E., Jørgensen, T., Karlsson, M., Koenig, W., Kraft, P., Kwekkeboom, J., Laatikainen, T., Ladwig, K., Leduc, C. A., Lowe, G., Lu, Y., Marques-Vidal, P., Meisinger, C., Menni, C., Morris, A. P., Myers, R. H., Männistö, S., Nalls, M. A., Paternoster, L., Peters, A., Pradhan, A. D., Rankinen, T., Rasmussen-Torvik, L. J., Rathmann, W., Rice, T. K., Brent Richards, J., Ridker, P. M., Sattar, N., Savage, D. B., Söderberg, S., Timpson, N. J., Vandenput, L., van Heemst, D., Uh, H., Vohl, M., Walker, M., Wichmann, H., Widén, E., Wood, A. R., Yao, J., Zeller, T., Zhang, Y., Meulenbelt, I., Kloppenburg, M., Astrup, A., Sørensen, T. I., Sarzynski, M. A., Rao, D. C., Jousilahti, P., Vartiainen, E., Hofman, A., Rivadeneira, F., Uitterlinden, A. G., Kajantie, E., Osmond, C., Palotie, A., Eriksson, J. G., Heliövaara, M., Knekt, P. B., Koskinen, S., Jula, A., Perola, M., Huupponen, R. K., Viikari, J. S., Kähönen, M., Lehtimäki, T., Raitakari, O. T., Mellström, D., Lorentzon, M., Casas, J. P., Bandinelli, S., März, W., Isaacs, A., van Dijk, K. W., van Duijn, C. M., Harris, T. B., Bouchard, C., Allison, M. A., Chasman, D. I., Ohlsson, C., Lind, L., Scott, R. A., Langenberg, C., Wareham, N. J., Ferrucci, L., Frayling, T. M., Pramstaller, P. P., Borecki, I. B., Waterworth, D. M., Bergmann, S., Waeber, G., Vollenweider, P., Vestergaard, H., Hansen, T., Pedersen, O., Hu, F. B., Eline Slagboom, P., Grallert, H., Spector, T. D., Jukema, J. W., Klein, R. J., Schadt, E. E., Franks, P. W., Lindgren, C. M., Leibel, R. L., Loos, R. J. 2016; 7: 10494-?

    Abstract

    Leptin is an adipocyte-secreted hormone, the circulating levels of which correlate closely with overall adiposity. Although rare mutations in the leptin (LEP) gene are well known to cause leptin deficiency and severe obesity, no common loci regulating circulating leptin levels have been uncovered. Therefore, we performed a genome-wide association study (GWAS) of circulating leptin levels from 32,161 individuals and followed up loci reaching P<10(-6) in 19,979 additional individuals. We identify five loci robustly associated (P<5 × 10(-8)) with leptin levels in/near LEP, SLC32A1, GCKR, CCNL1 and FTO. Although the association of the FTO obesity locus with leptin levels is abolished by adjustment for BMI, associations of the four other loci are independent of adiposity. The GCKR locus was found associated with multiple metabolic traits in previous GWAS and the CCNL1 locus with birth weight. Knockdown experiments in mouse adipose tissue explants show convincing evidence for adipogenin, a regulator of adipocyte differentiation, as the novel causal gene in the SLC32A1 locus influencing leptin levels. Our findings provide novel insights into the regulation of leptin production by adipose tissue and open new avenues for examining the influence of variation in leptin levels on adiposity and metabolic health.

    View details for DOI 10.1038/ncomms10494

    View details for PubMedID 26833098

    View details for PubMedCentralID PMC4740377

  • New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk. Nature communications Lu, Y., Day, F. R., Gustafsson, S., Buchkovich, M. L., Na, J., Bataille, V., Cousminer, D. L., Dastani, Z., Drong, A. W., Esko, T., Evans, D. M., Falchi, M., Feitosa, M. F., Ferreira, T., Hedman, Å. K., Haring, R., Hysi, P. G., Iles, M. M., Justice, A. E., Kanoni, S., Lagou, V., Li, R., Li, X., Locke, A., Lu, C., Mägi, R., Perry, J. R., Pers, T. H., Qi, Q., Sanna, M., Schmidt, E. M., Scott, W. R., Shungin, D., Teumer, A., Vinkhuyzen, A. A., Walker, R. W., Westra, H., Zhang, M., Zhang, W., Zhao, J. H., Zhu, Z., Afzal, U., Ahluwalia, T. S., Bakker, S. J., Bellis, C., Bonnefond, A., Borodulin, K., Buchman, A. S., Cederholm, T., Choh, A. C., Choi, H. J., Curran, J. E., de Groot, L. C., De Jager, P. L., Dhonukshe-Rutten, R. A., Enneman, A. W., Eury, E., Evans, D. S., Forsen, T., Friedrich, N., Fumeron, F., Garcia, M. E., Gärtner, S., Han, B., Havulinna, A. S., Hayward, C., Hernandez, D., Hillege, H., Ittermann, T., Kent, J. W., Kolcic, I., Laatikainen, T., Lahti, J., Mateo Leach, I., Lee, C. G., Lee, J., Liu, T., Liu, Y., Lobbens, S., Loh, M., Lyytikäinen, L., Medina-Gomez, C., Michaëlsson, K., Nalls, M. A., Nielson, C. M., Oozageer, L., Pascoe, L., Paternoster, L., Polašek, O., Ripatti, S., Sarzynski, M. A., Shin, C. s., Narancic, N. S., Spira, D., Srikanth, P., Steinhagen-Thiessen, E., Sung, Y. J., Swart, K. M., Taittonen, L., Tanaka, T., Tikkanen, E., van der Velde, N., van Schoor, N. M., Verweij, N., Wright, A. F., Yu, L., Zmuda, J. M., Eklund, N., Forrester, T., Grarup, N., Jackson, A. U., Kristiansson, K., Kuulasmaa, T., Kuusisto, J., Lichtner, P., Luan, J., Mahajan, A., Männistö, S., Palmer, C. D., Ried, J. S., Scott, R. A., Stancáková, A., Wagner, P. J., Demirkan, A., Döring, A., Gudnason, V., Kiel, D. P., Kühnel, B., Mangino, M., McKnight, B., Menni, C., O'Connell, J. R., Oostra, B. A., Shuldiner, A. R., Song, K., Vandenput, L., van Duijn, C. M., Vollenweider, P., White, C. C., Boehnke, M., Boettcher, Y., Cooper, R. S., Forouhi, N. G., Gieger, C., Grallert, H., Hingorani, A., Jørgensen, T., Jousilahti, P., Kivimaki, M., Kumari, M., Laakso, M., Langenberg, C., Linneberg, A., Luke, A., McKenzie, C. A., Palotie, A., Pedersen, O., Peters, A., Strauch, K., Tayo, B. O., Wareham, N. J., Bennett, D. A., Bertram, L., Blangero, J., Blüher, M., Bouchard, C., Campbell, H., Cho, N. H., Cummings, S. R., Czerwinski, S. A., Demuth, I., Eckardt, R., Eriksson, J. G., Ferrucci, L., Franco, O. H., Froguel, P., Gansevoort, R. T., Hansen, T., Harris, T. B., Hastie, N., Heliövaara, M., Hofman, A., Jordan, J. M., Jula, A., Kähönen, M., Kajantie, E., Knekt, P. B., Koskinen, S., Kovacs, P., Lehtimäki, T., Lind, L., Liu, Y., Orwoll, E. S., Osmond, C., Perola, M., Pérusse, L., Raitakari, O. T., Rankinen, T., Rao, D. C., Rice, T. K., Rivadeneira, F., Rudan, I., Salomaa, V., Sørensen, T. I., Stumvoll, M., Tönjes, A., Towne, B., Tranah, G. J., Tremblay, A., Uitterlinden, A. G., van der Harst, P., Vartiainen, E., Viikari, J. S., Vitart, V., Vohl, M., Völzke, H., Walker, M., Wallaschofski, H., Wild, S., Wilson, J. F., Yengo, L., Bishop, D. T., Borecki, I. B., Chambers, J. C., Cupples, L. A., Dehghan, A., Deloukas, P., Fatemifar, G., Fox, C., Furey, T. S., Franke, L., Han, J., Hunter, D. J., Karjalainen, J., Karpe, F., Kaplan, R. C., Kooner, J. S., McCarthy, M. I., Murabito, J. M., Morris, A. P., Bishop, J. A., North, K. E., Ohlsson, C., Ong, K. K., Prokopenko, I., Richards, J. B., Schadt, E. E., Spector, T. D., Widén, E., Willer, C. J., Yang, J., Ingelsson, E., Mohlke, K. L., Hirschhorn, J. N., Pospisilik, J. A., Zillikens, M. C., Lindgren, C., Kilpeläinen, T. O., Loos, R. J. 2016; 7: 10495-?

    Abstract

    To increase our understanding of the genetic basis of adiposity and its links to cardiometabolic disease risk, we conducted a genome-wide association meta-analysis of body fat percentage (BF%) in up to 100,716 individuals. Twelve loci reached genome-wide significance (P<5 × 10(-8)), of which eight were previously associated with increased overall adiposity (BMI, BF%) and four (in or near COBLL1/GRB14, IGF2BP1, PLA2G6, CRTC1) were novel associations with BF%. Seven loci showed a larger effect on BF% than on BMI, suggestive of a primary association with adiposity, while five loci showed larger effects on BMI than on BF%, suggesting association with both fat and lean mass. In particular, the loci more strongly associated with BF% showed distinct cross-phenotype association signatures with a range of cardiometabolic traits revealing new insights in the link between adiposity and disease risk.

    View details for DOI 10.1038/ncomms10495

    View details for PubMedID 26833246

    View details for PubMedCentralID PMC4740398

  • Large-scale non-targeted metabolomic profiling in three human population-based studies METABOLOMICS Ganna, A., Fall, T., Salihovic, S., Lee, W., Broeckling, C. D., Kumar, J., Hagg, S., Stenemo, M., Magnusson, P. K., Prenni, J. E., Lind, L., Pawitan, Y., Ingelsson, E. 2016; 12 (1)
  • Protein Biomarkers for Insulin Resistance and Type 2 Diabetes Risk in Two Large Community Cohorts DIABETES Nowak, C., Sundstrom, J., Gustafsson, S., Giedraitis, V., Lind, L., Ingelsson, E., Fall, T. 2016; 65 (1): 276-284

    Abstract

    Insulin resistance (IR) is a precursor of type 2 diabetes (T2D), and improved risk prediction and understanding of the pathogenesis are needed. We used a novel high-throughput 92-protein assay to identify circulating biomarkers for HOMA of IR in two cohorts of community residents without diabetes (n = 1,367) (mean age 73 ± 3.6 years). Adjusted linear regression identified cathepsin D and confirmed six proteins (leptin, renin, interleukin-1 receptor antagonist [IL-1ra], hepatocyte growth factor, fatty acid-binding protein 4, and tissue plasminogen activator [t-PA]) as IR biomarkers. Mendelian randomization analysis indicated a positive causal effect of IR on t-PA concentrations. Two biomarkers, IL-1ra (hazard ratio [HR] 1.28, 95% CI 1.03-1.59) and t-PA (HR 1.30, 1.02-1.65) were associated with incident T2D, and t-PA predicted 5-year transition to hyperglycemia (odds ratio 1.30, 95% CI 1.02-1.65). Additional adjustment for fasting glucose rendered both coefficients insignificant and revealed an association between renin and T2D (HR 0.79, 0.62-0.99). LASSO regression suggested a risk model including IL-1ra, t-PA, and the Framingham Offspring Study T2D score, but prediction improvement was nonsignificant (difference in C-index 0.02, 95% CI -0.08 to 0.12) over the T2D score only. In conclusion, proteomic blood profiling indicated cathepsin D as a new IR biomarker and suggested a causal effect of IR on t-PA.

    View details for DOI 10.2337/db15-0881

    View details for Web of Science ID 000367424900029

    View details for PubMedID 26420861

  • Genome-wide genetic homogeneity between sexes and populations for human height and body mass index HUMAN MOLECULAR GENETICS Yang, J., Bakshi, A., Zhu, Z., Hemani, G., Vinkhuyzen, A. A., Nolte, I. M., van Vliet-Ostaptchouk, J. V., Snieder, H., Study, L. C., Esko, T., Milani, L., Maegi, R., Metspalu, A., Hamsten, A., Magnusson, P. K., Pedersen, N. L., Ingelsson, E., Visscher, P. M. 2015; 24 (25): 7445-7449

    View details for DOI 10.1093/hmg/ddv443

    View details for Web of Science ID 000368373600023

  • Genome-wide genetic homogeneity between sexes and populations for human height and body mass index. Human molecular genetics Yang, J., Bakshi, A., Zhu, Z., Hemani, G., Vinkhuyzen, A. A., Nolte, I. M., van Vliet-Ostaptchouk, J. V., Snieder, H., Esko, T., Milani, L., Mägi, R., Metspalu, A., Hamsten, A., Magnusson, P. K., Pedersen, N. L., Ingelsson, E., Visscher, P. M. 2015; 24 (25): 7445-7449

    Abstract

    Sex-specific genetic effects have been proposed to be an important source of variation for human complex traits. Here we use two distinct genome-wide methods to estimate the autosomal genetic correlation (rg) between men and women for human height and body mass index (BMI), using individual-level (n = ∼44 000) and summary-level (n = ∼133 000) data from genome-wide association studies. Results are consistent and show that the between-sex genetic correlation is not significantly different from unity for both traits. In contrast, we find evidence of genetic heterogeneity between sexes for waist-hip ratio (rg = ∼0.7) and between populations for BMI (rg = ∼0.9 between Europe and the USA) but not for height. The lack of evidence for substantial genetic heterogeneity for body size is consistent with empirical findings across traits and species.

    View details for DOI 10.1093/hmg/ddv443

    View details for PubMedID 26494901

  • Genetic fine mapping and genomic annotation defines causal mechanisms at type 2 diabetes susceptibility loci NATURE GENETICS Gaulton, K. J., Ferreira, T., Lee, Y., Raimondo, A., Maegi, R., Reschen, M. E., Mahajan, A., Locke, A., Rayner, N. W., Robertson, N., Scott, R. A., Prokopenko, I., Scott, L. J., Green, T., Sparso, T., Thuillier, D., Yengo, L., Grallert, H., Wahl, S., Franberg, M., Strawbridge, R. J., Kestler, H., Chheda, H., Eisele, L., Gustafsson, S., Steinthorsdottir, V., Thorleifsson, G., Qi, L., Karssen, L. C., van Leeuwen, E. M., Willems, S. M., Li, M., Chen, H., Fuchsberger, C., Kwan, P., Ma, C., Linderman, M., Lu, Y., Thomsen, S. K., Rundle, J. K., Beer, N. L., van de Bunt, M., Chalisey, A., Kang, H. M., Voight, B. F., Abecasis, G. R., Almgren, P., Baldassarre, D., Balkau, B., Benediktsson, R., Blueher, M., Boeing, H., Bonnycastle, L. L., Bottinger, E. P., Burtt, N. P., Carey, J., Charpentier, G., Chines, P. S., Cornelis, M. C., Couper, D. J., Crenshaw, A. T., van Dam, R. M., Doney, A. S., Dorkhan, M., Edkins, S., Eriksson, J. G., Esko, T., Eury, E., Fadista, J., Flannick, J., Fontanillas, P., Fox, C., Franks, P. W., Gertow, K., Gieger, C., Gigante, B., Gottesman, O., Grant, G. B., Grarup, N., Groves, C. J., Hassinen, M., Have, C. T., Herder, C., Holmen, O. L., Hreidarsson, A. B., Humphries, S. E., Hunter, D. J., Jackson, A. U., Jonsson, A., Jorgensen, M. E., Jorgensen, T., Kao, W. L., Kerrison, N. D., Kinnunen, L., Klopp, N., Kong, A., Kovacs, P., Kraft, P., Kravic, J., Langford, C., Leander, K., Liang, L., Lichtner, P., Lindgren, C. M., Lindholm, E., Linneberg, A., Liu, C., Lobbens, S., Luan, J., Lyssenko, V., Mannisto, S., McLeod, O., Meyer, J., Mihailov, E., Mirza, G., Muehleisen, T. W., Mueller-Nurasyid, M., Navarro, C., Noethen, M. M., Oskolkov, N. N., Owen, K. R., Palli, D., Pechlivanis, S., Peltonen, L., Perry, J. R., Platou, C. G., Roden, M., Ruderfer, D., Rybin, D., van der Schouw, Y. T., Sennblad, B., Sigurdsson, G., Stancakova, A., Steinbach, G., Storm, P., Strauch, K., Stringham, H. M., Sun, Q., Thorand, B., Tikkanen, E., Tonjes, A., Trakalo, J., Tremoli, E., Tuomi, T., Wennauer, R., Wiltshire, S., Wood, A. R., Zeggini, E., Dunham, I., Birney, E., Pasquali, L., Ferrer, J., Loos, R. J., Dupuis, J., Florez, J. C., Boerwinkle, E., Pankow, J. S., Van Duijn, C., Sijbrands, E., Meigs, J. B., Hu, F. B., Thorsteinsdottir, U., Stefansson, K., Lakka, T. A., Rauramaa, R., Stumvoll, M., Pedersen, N. L., Lind, L., Keinanen-Kiukaanniemi, S. M., Korpi-Hyovalti, E., Saaristo, T. E., Saltevo, J., Kuusisto, J., Laakso, M., Metspalu, A., Erbel, R., Joecke, K., Moebus, S., Ripatti, S., Salomaa, V., Ingelsson, E., Boehm, B. O., Bergman, R. N., Collins, F. S., Mohlke, K. L., Koistinen, H., Tuomilehto, J., Hveem, K., Njolstad, I., Deloukas, P., Donnelly, P. J., Frayling, T. M., Hattersley, A. T., de Faire, U., Hamsten, A., Illig, T., Peters, A., Cauchi, S., Sladek, R., Froguel, P., Hansen, T., Pedersen, O., Morris, A. D., Palmer, C. N., Kathiresan, S., Melander, O., Nilsson, P. M., Groop, L. C., Barroso, I., Langenberg, C., Wareham, N. J., O'Callaghan, C. A., Gloyn, A. L., Altshuler, D., Boehnke, M., Teslovich, T. M., McCarthy, M. I., Morris, A. P. 2015; 47 (12): 1415-?

    Abstract

    We performed fine mapping of 39 established type 2 diabetes (T2D) loci in 27,206 cases and 57,574 controls of European ancestry. We identified 49 distinct association signals at these loci, including five mapping in or near KCNQ1. 'Credible sets' of the variants most likely to drive each distinct signal mapped predominantly to noncoding sequence, implying that association with T2D is mediated through gene regulation. Credible set variants were enriched for overlap with FOXA2 chromatin immunoprecipitation binding sites in human islet and liver cells, including at MTNR1B, where fine mapping implicated rs10830963 as driving T2D association. We confirmed that the T2D risk allele for this SNP increases FOXA2-bound enhancer activity in islet- and liver-derived cells. We observed allele-specific differences in NEUROD1 binding in islet-derived cells, consistent with evidence that the T2D risk allele increases islet MTNR1B expression. Our study demonstrates how integration of genetic and genomic information can define molecular mechanisms through which variants underlying association signals exert their effects on disease.

    View details for DOI 10.1038/ng.3437

    View details for Web of Science ID 000365813200013

    View details for PubMedID 26551672

    View details for PubMedCentralID PMC4666734

  • Sixteen new lung function signals identified through 1000 Genomes Project reference panel imputation NATURE COMMUNICATIONS Artigas, M. S., Wain, L. V., Miller, S., Kheirallah, A. K., Huffman, J. E., Ntalla, I., Shrine, N., Obeidat, M., Trochet, H., McArdle, W. L., Alves, A. C., Hui, J., Zhao, J. H., Joshi, P. K., Teumer, A., Albrecht, E., Imboden, M., Rawal, R., Lopez, L. M., Marten, J., Enroth, S., Surakka, I., Polasek, O., Lyytikainen, L., Granell, R., Hysi, P. G., Flexeder, C., Mahajan, A., Beilby, J., Bosse, Y., Brandsma, C., Campbell, H., Gieger, C., Glaeser, S., Gonzalez, J. R., Grallert, H., Hammond, C. J., Harris, S. E., Hartikainen, A., Heliovaara, M., Henderson, J., Hocking, L., Horikoshi, M., Hutri-Kahonen, N., Ingelsson, E., Johansson, A., Kemp, J. P., Kolcic, I., Kumar, A., Lind, L., Melen, E., Musk, A. W., Navarro, P., Nickle, D. C., Padmanabhan, S., Raitakari, O. T., Ried, J. S., Ripatti, S., Schulz, H., Scott, R. A., Sin, D. D., Starr, J. M., Vinuela, A., Voelzke, H., Wild, S. H., Wright, A. F., Zemunik, T., Jarvis, D. L., Spector, T. D., Evans, D. M., Lehtimaki, T., Vitart, V., Kahonen, M., Gyllensten, U., Rudan, I., Deary, I. J., Karrasch, S., Probst-Hensch, N. M., Heinrich, J., Stubbe, B., Wilson, J. F., Wareham, N. J., James, A. L., Morris, A. P., Jarvelin, M., Hayward, C., Sayers, I., Strachan, D. P., Hall, I. P., Tobin, M. D., Deloukas, P., Hansell, A. L., Hubbard, R., Jackson, V. E., Marchini, J., Pavord, I., Thomson, N. C., Zeggini, E. 2015; 6

    Abstract

    Lung function measures are used in the diagnosis of chronic obstructive pulmonary disease. In 38,199 European ancestry individuals, we studied genome-wide association of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC with 1000 Genomes Project (phase 1)-imputed genotypes and followed up top associations in 54,550 Europeans. We identify 14 novel loci (P<5 × 10(-8)) in or near ENSA, RNU5F-1, KCNS3, AK097794, ASTN2, LHX3, CCDC91, TBX3, TRIP11, RIN3, TEKT5, LTBP4, MN1 and AP1S2, and two novel signals at known loci NPNT and GPR126, providing a basis for new understanding of the genetic determinants of these traits and pulmonary diseases in which they are altered.

    View details for DOI 10.1038/ncomms9658

    View details for Web of Science ID 000367589100001

    View details for PubMedCentralID PMC4686825

  • Gene-based meta-analysis of genome-wide association studies implicates new loci involved in obesity HUMAN MOLECULAR GENETICS Hagg, S., Ganna, A., van der Laan, S. W., Esko, T., Pers, T. H., Locke, A. E., Berndt, S. I., Justice, A. E., Kahali, B., Siemelink, M. A., Pasterkamp, G., Strachan, D. P., Speliotes, E. K., North, K. E., Loos, R. J., Hirschhorn, J. N., Pawitan, Y., Ingelsson, E. 2015; 24 (23): 6849-6860

    Abstract

    To date, genome-wide association studies (GWASs) have identified >100 loci with single variants associated with body mass index (BMI). This approach may miss loci with high allelic heterogeneity; therefore, the aim of the present study was to use gene-based meta-analysis to identify regions with high allelic heterogeneity to discover additional obesity susceptibility loci. We included GWAS data from 123 865 individuals of European descent from 46 cohorts in Stage 1 and Metabochip data from additional 103 046 individuals from 43 cohorts in Stage 2, all within the Genetic Investigation of ANthropometric Traits (GIANT) consortium. Each cohort was tested for association between ∼2.4 million (Stage 1) or ∼200 000 (Stage 2) imputed or genotyped single variants and BMI, and summary statistics were subsequently meta-analyzed in 17 941 genes. We used the 'VErsatile Gene-based Association Study' (VEGAS) approach to assign variants to genes and to calculate gene-based P-values based on simulations. The VEGAS method was applied to each cohort separately before a gene-based meta-analysis was performed. In Stage 1, two known (FTO and TMEM18) and six novel (PEX2, MTFR2, SSFA2, IARS2, CEP295 and TXNDC12) loci were associated with BMI (P < 2.8 × 10(-6) for 17 941 gene tests). We confirmed all loci, and six of them were gene-wide significant in Stage 2 alone. We provide biological support for the loci by pathway, expression and methylation analyses. Our results indicate that gene-based meta-analysis of GWAS provides a useful strategy to find loci of interest that were not identified in standard single-marker analyses due to high allelic heterogeneity.

    View details for DOI 10.1093/hmg/ddv379

    View details for Web of Science ID 000368371600023

    View details for PubMedID 26376864

  • Population genetic differentiation of height and body mass index across Europe NATURE GENETICS Robinson, M. R., Hemani, G., Medina-Gomez, C., Mezzavilla, M., Esko-, T., Shakhbazov, K., Powell, J. E., Vinkhuyzen, A., Berndt, S. I., Gustafsson, S., Justice, A. E., Kahali, B., Locke, A. E., Pers, T. H., Vedantam, S., Wood, A. R., van Rheenen, W., Andreassen, O. A., Gasparini, P., Metspalu, A., Van den Berg, L. H., Veldink, J. H., Rivadeneira, F., Werge, T. M., Abecasis, G. R., Boomsma, D. I., Chasman, D. I., de Geus, E. J., Frayling, T. M., Hirschhorn, J. N., Hottenga, J. J., Ingelsson, E., Loos, R. J., Magnusson, P. K., Martin, N. G., Montgomery, G. W., North, K. E., Pedersen, N. L., Spector, T. D., Speliotes, E. K., Goddard, M. E., Yang, J., Visscher, P. M. 2015; 47 (11): 1357-?

    Abstract

    Across-nation differences in the mean values for complex traits are common, but the reasons for these differences are unknown. Here we find that many independent loci contribute to population genetic differences in height and body mass index (BMI) in 9,416 individuals across 14 European countries. Using discovery data on over 250,000 individuals and unbiased effect size estimates from 17,500 sibling pairs, we estimate that 24% (95% credible interval (CI) = 9%, 41%) and 8% (95% CI = 4%, 16%) of the captured additive genetic variance for height and BMI, respectively, reflect population genetic differences. Population genetic divergence differed significantly from that in a null model (height, P < 3.94 × 10(-8); BMI, P < 5.95 × 10(-4)), and we find an among-population genetic correlation for tall and slender individuals (r = -0.80, 95% CI = -0.95, -0.60), consistent with correlated selection for both phenotypes. Observed differences in height among populations reflected the predicted genetic means (r = 0.51; P < 0.001), but environmental differences across Europe masked genetic differentiation for BMI (P < 0.58).

    View details for DOI 10.1038/ng.3401

    View details for Web of Science ID 000363988200021

    View details for PubMedID 26366552

  • Early Exposure to Dogs and Farm Animals and the Risk of Childhood Asthma JAMA PEDIATRICS Fall, T., Lundholm, C., Ortqvist, A. K., Fall, K., Fang, F., Hedhammar, A., Kampe, O., Ingelsson, E., Almqvist, C. 2015; 169 (11)

    Abstract

    The association between early exposure to animals and childhood asthma is not clear, and previous studies have yielded contradictory results.To determine whether exposure to dogs and farm animals confers a risk of asthma.In a nationwide cohort study, the association between early exposure to dogs and farm animals and the risk of asthma was evaluated and included all children born in Sweden from January 1, 2001, to December 31, 2010 (N = 1,011,051), using registry data on dog and farm registration, asthma medication, diagnosis, and confounders for parents and their children. The association was assessed as the odds ratio (OR) for a current diagnosis of asthma at age 6 years for school-aged children and as the hazard ratio (HR) for incident asthma at ages 1 to 5 years for preschool-aged children. Data were analyzed from January 1, 2007, to September 30, 2012.Living with a dog or farm animal.Childhood asthma diagnosis and medication used.Of the 1,011,051 children born during the study period, 376,638 preschool-aged (53,460 [14.2%] exposed to dogs and 1729 [0.5%] exposed to farm animals) and 276,298 school-aged children (22,629 [8.2%] exposed to dogs and 958 [0.3%] exposed to farm animals) were included in the analyses. Of these, 18,799 children (5.0%) in the preschool-aged children's cohort experienced an asthmatic event before baseline, and 28,511 cases of asthma and 906,071 years at risk were recorded during follow-up (incidence rate, 3.1 cases per 1000 years at risk). In the school-aged children's cohort, 11,585 children (4.2%) experienced an asthmatic event during the seventh year of life. Dog exposure during the first year of life was associated with a decreased risk of asthma in school-aged children (OR, 0.87; 95% CI, 0.81-0.93) and in preschool-aged children 3 years or older (HR, 0.90; 95% CI, 0.83-0.99) but not in children younger than 3 years (HR, 1.03; 95% CI, 1.00-1.07). Results were comparable when analyzing only first-born children. Farm animal exposure was associated with a reduced risk of asthma in both school-aged children and preschool-aged children (OR, 0.48; 95% CI, 0.31-0.76, and HR, 0.69; 95% CI, 0.56-0.84), respectively.In this study, the data support the hypothesis that exposure to dogs and farm animals during the first year of life reduces the risk of asthma in children at age 6 years. This information might be helpful in decision making for families and physicians on the appropriateness and timing of early animal exposure.

    View details for DOI 10.1001/jamapediatrics.2015.3219

    View details for Web of Science ID 000364430000002

    View details for PubMedID 26523822

  • A comprehensive 1000 Genomes-based genome-wide association meta-analysis of coronary artery disease NATURE GENETICS Nikpay, M., Goel, A., Won, H., Hall, L. M., Willenborg, C., Kanoni, S., Saleheen, D., Kyriakou, T., Nelson, C. P., Hopewell, J. C., Webb, T. R., Zeng, L., Dehghan, A., Alver, M., Armasu, S. M., Auro, K., Bjonnes, A., Chasman, D. I., Chen, S., Ford, I., Franceschini, N., Gieger, C., Grace, C., Gustafsson, S., Huang, J., Hwang, S., Kim, Y. K., Kleber, M. E., Lau, K. W., Lu, X., Lu, Y., Lyytikainen, L., Mihailov, E., Morrison, A. C., Pervjakova, N., Qu, L., Rose, L. M., Salfati, E., Saxena, R., Scholz, M., Smith, A. V., Tikkanen, E., Uitterlinden, A., Yang, X., Zhang, W., Zhao, W., de Andrade, M., de Vries, P. S., Van Zuydam, N. R., Anand, S. S., Bertram, L., Beutner, F., Dedoussis, G., Frossard, P., Gauguier, D., Goodall, A. H., Gottesman, O., Haber, M., Han, B., Huang, J., Jalilzadeh, S., Kessler, T., Koenig, I. R., Lannfelt, L., Lieb, W., Lind, L., Lindgren, C. M., Lokki, M., Magnusson, P. K., Mallick, N. H., Mehra, N., Meitinger, T., Memon, F., Morris, A. P., Nieminen, M. S., Pedersen, N. L., Peters, A., Rallidis, L. S., Rasheed, A., Samuel, M., Shah, S. H., Sinisalo, J., Stirrups, K. E., Trompet, S., Wang, L., Zaman, K. S., Ardissino, D., Boerwinkle, E., Borecki, I. B., Bottinger, E. P., Buring, J. E., Chambers, J. C., Collins, R., Cupples, L. A., Danesh, J., Demuth, I., Elosua, R., Epstein, S. E., Esko, T., Feitosa, M. F., Franco, O. H., Franzosi, M. G., Granger, C. B., Gu, D., Gudnason, V., Hall, A. S., Hamsten, A., Harris, T. B., Hazen, S. L., Hengstenberg, C., Hofman, A., Ingelsson, E., Iribarren, C., Jukema, J. W., Karhunen, P. J., Kim, B., Kooner, J. S., Kullo, I. J., Lehtimaki, T., Loos, R. J., Melander, O., Metspalu, A., Maerz, W., Palmer, C. N., Perola, M., Quertermous, T., Rader, D. J., Ridker, P. M., Ripatti, S., Roberts, R., Salomaa, V., Sanghera, D. K., Schwartz, S. M., Seedorf, U., Stewart, A. F., Stott, D. J., Thiery, J., Zalloua, P. A., O'Donnell, C. J., Reilly, M. P., Assimes, T. L., Thompson, J. R., Erdmann, J., Clarke, R., Watkins, H., Kathiresan, S., McPherson, R., Deloukas, P., Schunkert, H., Samani, N. J., Farrall, M. 2015; 47 (10): 1121-?

    Abstract

    Existing knowledge of genetic variants affecting risk of coronary artery disease (CAD) is largely based on genome-wide association study (GWAS) analysis of common SNPs. Leveraging phased haplotypes from the 1000 Genomes Project, we report a GWAS meta-analysis of ∼185,000 CAD cases and controls, interrogating 6.7 million common (minor allele frequency (MAF) > 0.05) and 2.7 million low-frequency (0.005 < MAF < 0.05) variants. In addition to confirming most known CAD-associated loci, we identified ten new loci (eight additive and two recessive) that contain candidate causal genes newly implicating biological processes in vessel walls. We observed intralocus allelic heterogeneity but little evidence of low-frequency variants with larger effects and no evidence of synthetic association. Our analysis provides a comprehensive survey of the fine genetic architecture of CAD, showing that genetic susceptibility to this common disease is largely determined by common SNPs of small effect size.

    View details for DOI 10.1038/ng.3396

    View details for Web of Science ID 000361969900007

    View details for PubMedID 26343387

    View details for PubMedCentralID PMC4589895

  • The Influence of Age and Sex on Genetic Associations with Adult Body Size and Shape: A Large-Scale Genome-Wide Interaction Study PLOS GENETICS Winkler, T. W., Justice, A. E., Graff, M., Barata, L., Feitosa, M. F., Chu, S., Czajkowski, J., Esko, T., Fall, T., Kilpelainen, T. O., Lu, Y., Magi, R., Mihailov, E., Pers, T. H., Rueeger, S., Teumer, A., Ehret, G. B., Ferreira, T., Heard-Costa, N. L., Karjalainen, J., Lagou, V., Mahajan, A., Neinast, M. D., Prokopenko, I., Simino, J., Teslovich, T. M., Jansen, R., Westra, H., White, C. C., Absher, D., Ahluwalia, T. S., Ahmad, S., Albrecht, E., Alves, A. C., Bragg-Gresham, J. L., de Craen, A. J., Bis, J. C., Bonnefond, A., Boucher, G., Cadby, G., Cheng, Y., Chiang, C. W., Delgado, G., Demirkan, A., Dueker, N., Eklund, N., Eiriksdottir, G., Eriksson, J., Feenstra, B., Fischer, K., Frau, F., Galesloot, T. E., Geller, F., Goel, A., Gorski, M., Grammer, T. B., Gustafsson, S., Haitjema, S., Hottenga, J., Huffman, J. E., Jackson, A. U., Jacobs, K. B., Johansson, A., Kaakinen, M., Kleber, M. E., Lahti, J., Leach, I. M., Lehne, B., Liu, Y., Lo, K. S., Lorentzon, M., Luan, J., Madden, P. A., Mangino, M., McKnight, B., Medina-Gomez, C., Monda, K. L., Montasser, M. E., Mueller, G., Mueller-Nurasyid, M., Nolte, I. M., Panoutsopoulou, K., Pascoe, L., Paternoster, L., Rayner, N. W., Renstrom, F., Rizzi, F., Rose, L. M., Ryan, K. A., Salo, P., Sanna, S., Scharnagl, H., Shi, J., Smith, A. V., Southam, L., Stancakova, A., Steinthorsdottir, V., Strawbridge, R. J., Sung, Y. J., Tachmazidou, I., Tanaka, T., Thorleifsson, G., Trompet, S., Pervjakova, N., Tyrer, J. P., Vandenput, L., van der Laan, S. W., van der Velde, N., van Setten, J., van Vliet-Ostaptchouk, J. V., Verweij, N., Vlachopoulou, E., Waite, L. L., Wang, S. R., Wang, Z., Wild, S. H., Willenborg, C., Wilson, J. F., Wong, A., Yang, J., Yengo, L., Yerges-Armstrong, L. M., Yu, L., Zhang, W., Zhao, J. H., Andersson, E. A., Bakker, S. J., Baldassarre, D., Banasik, K., Barcella, M., Barlassina, C., Bellis, C., Benaglio, P., Blangero, J., Blueher, M., Bonnet, F., Bonnycastle, L. L., Boyd, H. A., Bruinenberg, M., Buchman, A. S., Campbell, H., Chen, Y. I., Chines, P. S., Claudi-Boehm, S., Cole, J., Collins, F. S., de Geus, E. J., de Groot, L. C., Dimitriou, M., Duan, J., Enroth, S., Eury, E., Farmaki, A., Forouhi, N. G., Friedrich, N., Gejman, P. V., Gigante, B., Glorioso, N., Go, A. S., Gottesman, O., Graessler, J., Grallert, H., Grarup, N., Gu, Y., Broer, L., Ham, A. C., Hansen, T., Harris, T. B., Hartman, C. A., Hassinen, M., Hastie, N., Hattersley, A. T., Heath, A. C., Henders, A. K., Hernandez, D., Hillege, H., Holmen, O., Hovingh, K. G., Hui, J., Husemoen, L. L., Hutri-Kahonen, N., Hysi, P. G., Illig, T., De Jager, P. L., Jalilzadeh, S., Jorgensen, T., Jukema, J. W., Juonala, M., Kanoni, S., Karaleftheri, M., Khaw, K. T., Kinnunen, L., Kittner, S. J., Koenig, W., Kolcic, I., Kovacs, P., Krarup, N. T., Kratzer, W., Krueger, J., Kuh, D., Kumari, M., Kyriakou, T., Langenberg, C., Lannfelt, L., Lanzani, C., Lotay, V., Launer, L. J., Leander, K., Lindstrom, J., Linneberg, A., Liu, Y., Lobbens, S., Luben, R., Lyssenko, V., Mannisto, S., Magnusson, P. K., McArdle, W. L., Menni, C., Merger, S., Milani, L., Montgomery, G. W., Morris, A. P., Narisu, N., Nelis, M., Ong, K. K., Palotie, A., Perusse, L., Pichler, I., Pilia, M. G., Pouta, A., Rheinberger, M., Ribel-Madsen, R., Richards, M., Rice, K. M., Rice, T. K., Rivolta, C., Salomaa, V., Sanders, A. R., Sarzynski, M. A., Scholtens, S., Scott, R. A., Scott, W. R., Sebert, S., Sengupta, S., Sennblad, B., Seufferlein, T., Silveira, A., Slagboom, P. E., Smit, J. H., Sparso, T. H., Stirrups, K., Stolk, R. P., Stringham, H. M., Swertz, M. A., Swift, A. J., Syvanen, A., Tan, S., Thorand, B., Toenjes, A., Tremblay, A., Tsafantakis, E., van der Most, P. J., Voelker, U., Vohl, M., Vonk, J. M., Waldenberger, M., Walker, R. W., Wennauer, R., Widen, E., Willemsen, G., Wilsgaard, T., Wright, A. F., Zillikens, M. C., van Dijk, S. C., van Schoor, N. M., Asselbergs, F. W., de Bakker, P. I., Beckmann, J. S., Beilby, J., Bennett, D. A., Bergman, R. N., Bergmann, S., Boeger, C. A., Boehm, B. O., Boerwinkle, E., Boomsma, D. I., Bornstein, S. R., Bottinger, E. P., Bouchard, C., Chambers, J. C., Chanock, S. J., Chasman, D. I., Cucca, F., Cusi, D., Dedoussis, G., Erdmann, J., Eriksson, J. G., Evans, D. A., de Faire, U., Farrall, M., Ferrucci, L., Ford, I., Franke, L., Franks, P. W., Froguel, P., Gansevoort, R. T., Gieger, C., Gronberg, H., Gudnason, V., Gyllensten, U., Hall, P., Hamsten, A., van der Harst, P., Hayward, C., Heliovaara, M., Hengstenberg, C., Hicks, A. A., Hingorani, A., Hofman, A., Hu, F., Huikuri, H. V., Hveem, K., James, A. L., Jordan, J. M., Jula, A., Kaehoenen, M., Kajantie, E., Kathiresan, S., Kiemeney, L. A., Kivimaki, M., Knekt, P. B., Koistinen, H. A., Kooner, J. S., Koskinen, S., Kuusisto, J., Maerz, W., Martin, N. G., Laakso, M., Lakka, T. A., Lehtimaki, T., Lettre, G., Levinson, D. F., Lind, L., Lokki, M., Mantyselka, P., Melbye, M., Metspalu, A., Mitchell, B. D., Moll, F. L., Murray, J. C., Musk, A. W., Nieminen, M. S., Njolstad, I., Ohlsson, C., Oldehinkel, A. J., Oostra, B. A., Palmer, L. J., Pankow, J. S., Pasterkamp, G., Pedersen, N. L., Pedersen, O., Penninx, B. W., Perola, M., Peters, A., Polasek, O., Pramstaller, P. P., Psaty, B. M., Qi, L., Quertermous, T., Raitakari, O. T., Rankinen, T., Rauramaa, R., Ridker, P. M., Rioux, J. D., Rivadeneira, F., Rotter, J. I., Rudan, I., Den Ruijter, H. M., Saltevo, J., Sattar, N., Schunkert, H., Schwarz, P. E., Shuldiner, A. R., Sinisalo, J., Snieder, H., Sorensen, T. I., Spector, T. D., Staessen, J. A., Stefania, B., Thorsteinsdottir, U., Stumvoll, M., Tardif, J., Tremoli, E., Tuomilehto, J., Uitterlinden, A. G., Uusitupa, M., Verbeek, A. L., Vermeulen, S. H., Viikari, J. S., Vitart, V., Voelzke, H., Vollenweider, P., Waeber, G., Walker, M., Wallaschofski, H., Wareham, N. J., Watkins, H., Zeggini, E., Chakravarti, A., Clegg, D. J., Cupples, L. A., Gordon-Larsen, P., Jaquish, C. E., Rao, D. C., Abecasis, G. R., Assimes, T. L., Barroso, I., Berndt, S. I., Boehnke, M., Deloukas, P., Fox, C. S., Groop, L. C., Hunter, D. J., Ingelsson, E., Kaplan, R. C., McCarthy, M. I., Mohlke, K. L., O'Connell, J. R., Schlessinger, D., Strachan, D. P., Stefansson, K., van Duijn, C. M., Hirschhorn, J. N., Lindgren, C. M., Heid, I. M., North, K. E., Borecki, I. B., Kutalik, Z., Loos, R. J. 2015; 11 (10)

    View details for DOI 10.1371/journal.pgen.1005378

    View details for Web of Science ID 000364401600002

    View details for PubMedID 26426971

  • Genetic variance estimation with imputed variants finds negligible missing heritability for human height and body mass index. Nature genetics Yang, J., Bakshi, A., Zhu, Z., Hemani, G., Vinkhuyzen, A. A., Lee, S. H., Robinson, M. R., Perry, J. R., Nolte, I. M., van Vliet-Ostaptchouk, J. V., Snieder, H., Esko, T., Milani, L., Mägi, R., Metspalu, A., Hamsten, A., Magnusson, P. K., Pedersen, N. L., Ingelsson, E., Soranzo, N., Keller, M. C., Wray, N. R., Goddard, M. E., Visscher, P. M. 2015; 47 (10): 1114-1120

    Abstract

    We propose a method (GREML-LDMS) to estimate heritability for human complex traits in unrelated individuals using whole-genome sequencing data. We demonstrate using simulations based on whole-genome sequencing data that ∼97% and ∼68% of variation at common and rare variants, respectively, can be captured by imputation. Using the GREML-LDMS method, we estimate from 44,126 unrelated individuals that all ∼17 million imputed variants explain 56% (standard error (s.e.) = 2.3%) of variance for height and 27% (s.e. = 2.5%) of variance for body mass index (BMI), and we find evidence that height- and BMI-associated variants have been under natural selection. Considering the imperfect tagging of imputation and potential overestimation of heritability from previous family-based studies, heritability is likely to be 60-70% for height and 30-40% for BMI. Therefore, the missing heritability is small for both traits. For further discovery of genes associated with complex traits, a study design with SNP arrays followed by imputation is more cost-effective than whole-genome sequencing at current prices.

    View details for DOI 10.1038/ng.3390

    View details for PubMedID 26323059

    View details for PubMedCentralID PMC4589513

  • Use of a proximity extension assay proteomics chip to discover new biomarkers for human atherosclerosis ATHEROSCLEROSIS Lind, L., Arnlov, J., Lindahl, B., Siegbahn, A., Sundstrom, J., Ingelsson, E. 2015; 242 (1): 205-210

    Abstract

    We used a proteomics array to simultaneously measure multiple proteins that have been suggested to be associated with atherosclerosis and related them to plaque prevalence in carotid arteries in a human population-based study.In the Prospective Study of the Vasculature in Uppsala Seniors (PIVUS; n = 931, 50% women, all aged 70 years), the number of carotid arteries with plaques was recorded by ultrasound. Levels of 82 proteins were assessed in plasma by a proximity extension assay (Proseek Multiplex CVD, Olink Bioscience, Uppsala, Sweden) and related to carotid measures in a regression framework.Following adjustment for multiple testing with Bonferroni correction, seven of the proteins were significantly related to the number of carotid arteries affected by plaques in sex-adjusted models (osteoprotegrin, T-cell immunoglobulin and mucin domain (TIM)-1, growth/differentiation factor 15 (GDF-15), matrix metalloprotease-12 (MMP-12), renin, tumor necrosis factor ligand superfamily member 14 (TNFSF14) and growth hormone). Of these, renin (odds ratio [OR], 1.30; 95% confidence interval [CI], 1.13-1.49 per standard deviation increase), growth hormone (OR, 1.24; 95% CI, 1.08-1.43), osteoprotegerin (OR, 1.22; 95% CI, 1.05-1.43) and TNFSF14 (OR, 1.17; 95% CI, 1.01-1.35) were related to plaque prevalence independently of each other and traditional cardiovascular risk factors.A novel targeted proteomics approach using the proximity extension technique discovered several new associations of candidate proteins with carotid artery plaque prevalence in a large human sample.

    View details for DOI 10.1016/j.atherosclerosis.2015.07.023

    View details for Web of Science ID 000360100900032

    View details for PubMedID 26204497

  • Genome-wide association study of toxic metals and trace elements reveals novel associations HUMAN MOLECULAR GENETICS Ng, E., Lind, P. M., Lindgren, C., Ingelsson, E., Mahajan, A., Morris, A., Lind, L. 2015; 24 (16): 4739-4745

    Abstract

    The accumulation of toxic metals in the human body is influenced by exposure and mechanisms involved in metabolism, some of which may be under genetic control. This is the first genome-wide association study to investigate variants associated with whole blood levels of a range of toxic metals. Eleven toxic metals and trace elements (aluminium, cadmium, cobalt, copper, chromium, mercury, manganese, molybdenum, nickel, lead and zinc) were assayed in a cohort of 949 individuals using mass spectrometry. DNA samples were genotyped on the Infinium Omni Express bead microarray and imputed up to reference panels from the 1000 Genomes Project. Analyses revealed two regions associated with manganese level at genome-wide significance, mapping to 4q24 and 1q41. The lead single nucleotide polymorphism (SNP) in the 4q24 locus was rs13107325 (P-value = 5.1 × 10(-11), β = -0.77), located in an exon of SLC39A8, which encodes a protein involved in manganese and zinc transport. The lead SNP in the 1q41 locus is rs1776029 (P-value = 2.2 × 10(-14), β = -0.46). The SNP lies within the intronic region of SLC30A10, another transporter protein. Among other metals, the loci 6q14.1 and 3q26.32 were associated with cadmium and mercury levels (P = 1.4 × 10(-10), β = -1.2 and P = 1.8 × 10(-9), β = -1.8, respectively). Whole blood measurements of toxic metals are associated with genetic variants in metal transporter genes and others. This is relevant in inferring metabolic pathways of metals and identifying subsets of individuals who may be more susceptible to metal toxicity.

    View details for DOI 10.1093/hmg/ddv190

    View details for Web of Science ID 000361315400021

    View details for PubMedID 26025379

  • Gene x dietary pattern interactions in obesity: analysis of up to 68 317 adults of European ancestry HUMAN MOLECULAR GENETICS Nettleton, J. A., Follis, J. L., Ngwa, J. S., Smith, C. E., Ahmad, S., Tanaka, T., Wojczynski, M. K., Voortman, T., Lemaitre, R. N., Kristiansson, K., Nuotio, M., Houston, D. K., Perala, M., Qi, Q., Sonestedt, E., Manichaikul, A., Kanoni, S., Ganna, A., Mikkila, V., North, K. E., Siscovick, D. S., Harald, K., McKeown, N. M., Johansson, I., Rissanen, H., Liu, Y., Lahti, J., Hu, F. B., Bandinelli, S., Rukh, G., Rich, S., Booij, L., Dmitriou, M., Ax, E., Raitakari, O., Mukamal, K., Mannisto, S., Hallmans, G., Jula, A., Ericson, U., Jacobs, D. R., van Rooij, F. J., Deloukas, P., Sjogren, P., Kahonen, M., Djousse, L., Perola, M., Barroso, I., Hofman, A., Stirrups, K., Viikari, J., Uitterlinden, A. G., Kalafati, I. P., Franco, O. H., Mozaffarian, D., Salomaa, V., Borecki, I. B., Knekt, P., Kritchevsky, S. B., Eriksson, J. G., Dedoussis, G. V., Qi, L., Ferrucci, L., Orho-Melander, M., Zillikens, M. C., Ingelsson, E., Lehtimaki, T., Renstrom, F., Cupples, L. A., Loos, R. J., Franks, P. W. 2015; 24 (16): 4728-4738

    Abstract

    Obesity is highly heritable. Genetic variants showing robust associations with obesity traits have been identified through genome-wide association studies. We investigated whether a composite score representing healthy diet modifies associations of these variants with obesity traits. Totally, 32 body mass index (BMI)- and 14 waist-hip ratio (WHR)-associated single nucleotide polymorphisms were genotyped, and genetic risk scores (GRS) were calculated in 18 cohorts of European ancestry (n = 68 317). Diet score was calculated based on self-reported intakes of whole grains, fish, fruits, vegetables, nuts/seeds (favorable) and red/processed meats, sweets, sugar-sweetened beverages and fried potatoes (unfavorable). Multivariable adjusted, linear regression within each cohort followed by inverse variance-weighted, fixed-effects meta-analysis was used to characterize: (a) associations of each GRS with BMI and BMI-adjusted WHR and (b) diet score modification of genetic associations with BMI and BMI-adjusted WHR. Nominally significant interactions (P = 0.006-0.04) were observed between the diet score and WHR-GRS (but not BMI-GRS), two WHR loci (GRB14 rs10195252; LYPLAL1 rs4846567) and two BMI loci (LRRN6C rs10968576; MTIF3 rs4771122), for the respective BMI-adjusted WHR or BMI outcomes. Although the magnitudes of these select interactions were small, our data indicated that associations between genetic predisposition and obesity traits were stronger with a healthier diet. Our findings generate interesting hypotheses; however, experimental and functional studies are needed to determine their clinical relevance.

    View details for DOI 10.1093/hmg/ddv186

    View details for Web of Science ID 000361315400020

    View details for PubMedID 25994509

  • Sparse estimation of gene-gene interactions in prediction models. Statistical methods in medical research Lee, S., Pawitan, Y., Ingelsson, E., Lee, Y. 2015

    Abstract

    Current assessment of gene-gene interactions is typically based on separate parallel analysis, where each interaction term is tested separately, while less attention has been paid on simultaneous estimation of interaction terms in a prediction model. As the number of interaction terms grows fast, sparse estimation is desirable from statistical and interpretability reasons. There is a large literature on sparse estimation, but there is a natural hierarchy between the interaction and its corresponding main effects that requires special considerations. We describe random-effect models that impose sparse estimation of interactions under both strong and weak-hierarchy constraints. We develop an estimation procedure based on the hierarchical-likelihood argument and show that the modelling approach is equivalent to a penalty-based method, with the advantage of the models being more transparent and flexible. We compare the procedure with some standard methods in a simulation study and illustrate its application in an analysis of gene-gene interaction model to predict body-mass index.

    View details for PubMedID 26265764

  • 5 year mortality predictors in 498 103 UK Biobank participants: a prospective population-based study LANCET Ganna, A., Ingelsson, E. 2015; 386 (9993): 533-540

    Abstract

    To our knowledge, a systematic comparison of predictors of mortality in middle-aged to elderly individuals has not yet been done. We investigated predictors of mortality in UK Biobank participants during a 5 year period. We aimed to investigate the associations between most of the available measurements and 5 year all-cause and cause-specific mortality, and to develop and validate a prediction score for 5 year mortality using only self-reported information.Participants were enrolled in the UK Biobank from April, 2007, to July, 2010, from 21 assessment centres across England, Wales, and Scotland with standardised procedures. In this prospective population-based study, we assessed sex-specific associations of 655 measurements of demographics, health, and lifestyle with all-cause mortality and six cause-specific mortality categories in UK Biobank participants using the Cox proportional hazard model. We excluded variables that were missing in more than 80% of the participants and all cardiorespiratory fitness test measurements because summary data were not available. Validation of the prediction score was done in participants enrolled at the Scottish centres. UK life tables and census information were used to calibrate the score to the overall UK population.About 500,000 participants were included in the UK Biobank. We excluded participants with more than 80% variables missing (n=746). Of 498,103 UK Biobank participants included (54% of whom were women) aged 37-73 years, 8532 (39% of whom were women) died during a median follow-up of 4·9 years (IQR 4·33-5·22). Self-reported health (C-index including age 0·74 [95% CI 0·73-0·75]) was the strongest predictor of all-cause mortality in men and a previous cancer diagnosis (0·73 [0·72-0·74]) was the strongest predictor of all-cause mortality in women. When excluding individuals with major diseases or disorders (Charlson comorbidity index >0; n=355 043), measures of smoking habits were the strongest predictors of all-cause mortality. The prognostic score including 13 self-reported predictors for men and 11 for women achieved good discrimination (0·80 [0·77-0·83] for men and 0·79 [0·76-0·83] for women) and significantly outperformed the Charlson comorbidity index (p<0·0001 in men and p=0·0007 in women). A dedicated website allows the interactive exploration of all results along with calculation of individual risk through an online questionnaire.Measures that can simply be obtained by questionnaires and without physical examination were the strongest predictors of all-cause mortality in the UK Biobank population. The prediction score we have developed accurately predicts 5 year all-cause mortality and can be used by individuals to improve health awareness, and by health professionals and organisations to identify high-risk individuals and guide public policy.Knut and Alice Wallenberg Foundation and the Swedish Research Council.

    View details for DOI 10.1016/S0140-6736(15)60175-1

    View details for Web of Science ID 000359431900027

    View details for PubMedID 26049253

  • Sex-Specific Effects of Adiponectin on Carotid Intima-Media Thickness and Incident Cardiovascular Disease JOURNAL OF THE AMERICAN HEART ASSOCIATION Persson, J., Strawbridge, R. J., McLeod, O., Gertow, K., Silveira, A., Baldassarre, D., Van Zuydam, N., Shah, S., Fava, C., Gustafsson, S., Veglia, F., Sennblad, B., Larsson, M., Sabater-Lleal, M., Leander, K., Gigante, B., Tabak, A., Kivimaki, M., Kauhanen, J., Rauramaa, R., Smit, A. J., Mannarino, E., Giral, P., Humphries, S. E., Tremoli, E., de Faire, U., Lind, L., Ingelsson, E., Hedblad, B., Melander, O., Kumari, M., Hingorani, A., Morris, A. D., Palmer, C. N., Lundman, P., Ohrvik, J., Soderberg, S. 2015; 4 (8)

    Abstract

    Plasma adiponectin levels have previously been inversely associated with carotid intima-media thickness (IMT), a marker of subclinical atherosclerosis. In this study, we used a sex-stratified Mendelian randomization approach to investigate whether adiponectin has a causal protective influence on IMT.Baseline plasma adiponectin concentration was tested for association with baseline IMT, IMT progression over 30 months, and occurrence of cardiovascular events within 3 years in 3430 participants (women, n=1777; men, n=1653) with high cardiovascular risk but no prevalent disease. Plasma adiponectin levels were inversely associated with baseline mean bifurcation IMT after adjustment for established risk factors (β=-0.018, P<0.001) in men but not in women (β=-0.006, P=0.185; P for interaction=0.061). Adiponectin levels were inversely associated with progression of mean common carotid IMT in men (β=-0.0022, P=0.047), whereas no association was seen in women (0.0007, P=0.475; P for interaction=0.018). Moreover, we observed that adiponectin levels were inversely associated with coronary events in women (hazard ratio 0.57, 95% CI 0.37 to 0.87) but not in men (hazard ratio 0.82, 95% CI 0.54 to 1.25). A gene score of adiponectin-raising alleles in 6 loci, reported recently in a large multi-ethnic meta-analysis, was inversely associated with baseline mean bifurcation IMT in men (β=-0.0008, P=0.004) but not in women (β=-0.0003, P=0.522; P for interaction=0.007).This report provides some evidence for adiponectin protecting against atherosclerosis, with effects being confined to men; however, compared with established cardiovascular risk factors, the effect of plasma adiponectin was modest. Further investigation involving mechanistic studies is warranted.

    View details for DOI 10.1161/JAHA.115.001853

    View details for Web of Science ID 000364150900006

    View details for PubMedID 26276317

  • Directional dominance on stature and cognition in diverse human populations NATURE Joshi, P. K., Esko, T., Mattsson, H., Eklund, N., Gandin, I., Nutile, T., Jackson, A. U., Schurmann, C., Smith, A. V., Zhang, W., Okada, Y., Stancakova, A., Faul, J. D., Zhao, W., Bartz, T. M., Concas, M. P., Franceschini, N., Enroth, S., Vitart, V., Trompet, S., Guo, X., Chasman, D. I., O'Connel, J. R., Corre, T., Nongmaithem, S. S., Chen, Y., Mangino, M., Ruggiero, D., Traglia, M., Farmaki, A., Kacprowski, T., Bjonnes, A., van der Spek, A., Wu, Y., Giri, A. K., Yanek, L. R., Wang, L., Hofer, E., Rietveld, C. A., McLeod, O., Cornelis, M. C., Pattaro, C., Verweij, N., Baumbach, C., Abdellaoui, A., Warren, H. R., Vuckovic, D., Mei, H., Bouchard, C., Perry, J. R., Cappellani, S., Mirza, S. S., Benton, M. C., Broeckel, U., Medland, S. E., Lind, P., Malerba, G., Drong, A., Yengo, L., Bielak, L. F., Zhi, D., van der Most, P. J., Shriner, D., Maegi, R., Hemani, G., Karaderi, T., Wang, Z., Liu, T., Demuth, I., Zhao, J. H., Meng, W., Lataniotis, L., van der Laan, S. W., Bradfield, J. P., Wood, A. R., Bonnefond, A., Ahluwalia, T. S., Hall, L., Salvi, E., Yazar, S., Carstensen, L., de Haan, H. G., Abney, M., Afzal, U., Allison, M. A., Amin, N., Asselbergs, F. W., Bakker, S. J., Barr, R. G., Baumeister, S. E., Benjamin, D. J., Bergmann, S., Boerwinkle, E., Bottinger, E. P., Campbell, A., Chakravarti, A., Chan, Y., Chanock, S. J., Chen, C., Chen, Y. I., Collins, F. S., Connell, J., Correa, A., Cupples, L. A., Smith, G. D., Davies, G., Doerr, M., Ehret, G., Ellis, S. B., Feenstra, B., Feitosa, M. F., Ford, I., Fox, C. S., Frayling, T. M., Friedrich, N., Geller, F., Scotland, G., Gillham-Nasenya, I., Gottesman, O., Graff, M., Grodstein, F., Gu, C., Haley, C., Hammond, C. J., Harris, S. E., Harris, T. B., Hastie, N. D., Heard-Costa, N. L., Heikkila, K., Hocking, L. J., Homuth, G., Hottenga, J., Huang, J., Huffman, J. E., Hysi, P. G., Ikram, M. A., Ingelsson, E., Joensuu, A., Johansson, A., Jousilahti, P., Jukema, J. W., Kahonen, M., Kamatani, Y., Kanoni, S., Kerr, S. M., Khan, N. M., Koellinger, P., Koistinen, H. A., Kooner, M. K., Kubo, M., Kuusisto, J., Lahti, J., Launer, L. J., Lea, R. A., Lehne, B., Lehtimaki, T., Liewald, D. C., Lind, L., Loh, M., Lokki, M., London, S. J., Loomis, S. J., Loukola, A., Lu, Y., Lumley, T., Lundqvist, A., Mannisto, S., Marques-Vidal, P., Masciullo, C., Matchan, A., Mathias, R. A., Matsuda, K., Meigs, J. B., Meisinger, C., Meitinger, T., Menni, C., Mentch, F. D., Mihailov, E., Milani, L., Montasser, M. E., Montgomery, G., Morrison, A., Myers, R. H., Nadukuru, R., Navarro, P., Nelis, M., Nieminen, M. S., Nolte, I. M., O'Connor, G. T., Ogunniyi, A., Padmanabhan, S., Palmas, W. R., Pankow, J. S., Patarcic, I., Pavani, F., Peyser, P. A., Pietilainen, K., Poulter, N., Prokopenko, I., Ralhan, S., Redmond, P., Rich, S. S., Rissanen, H., Robino, A., Rose, L. M., Rose, R., Sala, C., Salako, B., Salomaa, V., Sarin, A., Saxena, R., Schmidt, H., Scott, L. J., Scott, W. R., Sennblad, B., Seshadri, S., Sever, P., Shrestha, S., Smith, B. H., Smith, J. A., Soranzo, N., Sotoodehnia, N., Southam, L., Stanton, A. V., Stathopoulou, M. G., Strauch, K., Strawbridge, R. J., Suderman, M. J., Tandon, N., Tang, S., Taylor, K. D., Tayo, B. O., Toeglhofer, A. M., Tomaszewski, M., Tsernikova, N., Tuomilehto, J., Uitterlinden, A. G., Vaidya, D., Vlieg, A. v., van Setten, J., Vasankari, T., Vedantam, S., Vlachopoulou, E., Vozzi, D., Vuoksimaa, E., Waldenberger, M., Ware, E. B., Wentworth-Shields, W., Whitfield, J. B., Wild, S., Willemsen, G., Yajnik, C. S., Yao, J., Zaza, G., Zhu, X., Salem, R. M., Melbye, M., Bisgaard, H., Samani, N. J., Cusi, D., Mackey, D. A., Cooper, R. S., Froguel, P., Pasterkamp, G., Grant, S. F., Hakonarson, H., Ferrucci, L., Scott, R. A., Morris, A. D., Palmer, C. N., Dedoussis, G., Deloukas, P., Bertram, L., Lindenberger, U., Berndt, S. I., Lindgren, C. M., Timpson, N. J., Toenjes, A., Munroe, P. B., Sorensen, T. I., Rotimi, C. N., Arnett, D. K., Oldehinkel, A. J., Kardia, S. L., Balkau, B., Gambaro, G., Morris, A. P., Eriksson, J. G., Wright, M. J., Martin, N. G., Hunt, S. C., Starr, J. M., Deary, I. J., Griffiths, L. R., Tiemeier, H., Pirastu, N., Kaprio, J., Wareham, N. J., Perusse, L., Wilson, J. G., Girotto, G., Caulfield, M. J., Raitakari, O., Boomsma, D. I., Gieger, C., van der Harst, P., Hicks, A. A., Kraft, P., Sinisalo, J., Knekt, P., Johannesson, M., Magnusson, P. K., Hamsten, A., Schmidt, R., Borecki, I. B., Vartiainen, E., Becker, D. M., Bharadwaj, D., Mohlke, K. L., Boehnke, M., van Duijn, C. M., Sanghera, D. K., Teumer, A., Zeggini, E., Metspalu, A., Gasparini, P., Ulivi, S., Ober, C., Toniolo, D., Rudan, I., Porteous, D. J., Ciullo, M., Spector, T. D., Hayward, C., Dupuis, J., Loos, R. J., Wright, A. F., Chandak, G. R., Vollenweider, P., Shuldiner, A. R., Ridker, P. M., Rotter, J. I., Sattar, N., Gyllensten, U., North, K. E., Pirastu, M., Psaty, B. M., Weir, D. R., Laakso, M., Gudnason, V., Takahashi, A., Chambers, J. C., Kooner, J. S., Strachan, D. P., Campbell, H., Hirschhorn, J. N., Perola, M., Polasek, O., Wilson, J. F. 2015; 523 (7561): 459-U176

    Abstract

    Homozygosity has long been associated with rare, often devastating, Mendelian disorders, and Darwin was one of the first to recognize that inbreeding reduces evolutionary fitness. However, the effect of the more distant parental relatedness that is common in modern human populations is less well understood. Genomic data now allow us to investigate the effects of homozygosity on traits of public health importance by observing contiguous homozygous segments (runs of homozygosity), which are inferred to be homozygous along their complete length. Given the low levels of genome-wide homozygosity prevalent in most human populations, information is required on very large numbers of people to provide sufficient power. Here we use runs of homozygosity to study 16 health-related quantitative traits in 354,224 individuals from 102 cohorts, and find statistically significant associations between summed runs of homozygosity and four complex traits: height, forced expiratory lung volume in one second, general cognitive ability and educational attainment (P < 1 × 10(-300), 2.1 × 10(-6), 2.5 × 10(-10) and 1.8 × 10(-10), respectively). In each case, increased homozygosity was associated with decreased trait value, equivalent to the offspring of first cousins being 1.2 cm shorter and having 10 months' less education. Similar effect sizes were found across four continental groups and populations with different degrees of genome-wide homozygosity, providing evidence that homozygosity, rather than confounding, directly contributes to phenotypic variance. Contrary to earlier reports in substantially smaller samples, no evidence was seen of an influence of genome-wide homozygosity on blood pressure and low density lipoprotein cholesterol, or ten other cardio-metabolic traits. Since directional dominance is predicted for traits under directional evolutionary selection, this study provides evidence that increased stature and cognitive function have been positively selected in human evolution, whereas many important risk factors for late-onset complex diseases may not have been.

    View details for DOI 10.1038/nature14618

    View details for Web of Science ID 000358378900036

    View details for PubMedCentralID PMC4516141

  • Discovery and Fine-Mapping of Glycaemic and Obesity-Related Trait Loci Using High-Density Imputation PLOS GENETICS Horikoshi, M., Maegi, R., van de Bunt, M., Surakka, I., Sarin, A., Mahajan, A., Marullo, L., Thorleifsson, G., Haegg, S., Hottenga, J., Ladenvall, C., Ried, J. S., Winkler, T. W., Willems, S. M., Pervjakova, N., Esko, T., Beekman, M., Nelson, C. P., Willenborg, C., Wiltshire, S., Ferreira, T., Fernandez, J., Gaulton, K. J., Steinthorsdottir, V., Hamsten, A., Magnusson, P. K., Willemsen, G., Milaneschi, Y., Robertson, N. R., Groves, C. J., Bennett, A. J., Lehtimaeki, T., Viikari, J. S., Rung, J., Lyssenko, V., Perola, M., Heid, I. M., Herder, C., Grallert, H., Mueller-Nurasyid, M., Roden, M., Hypponen, E., Isaacs, A., van Leeuwen, E. M., Karssen, L. C., Mihailov, E., Houwing-Duistermaat, J. J., de Craen, A. J., Deelen, J., Havulinna, A. S., Blades, M., Hengstenberg, C., Erdmann, J., Schunkert, H., Kaprio, J., Tobin, M. D., Samani, N. J., Lind, L., Salomaa, V., Lindgren, C. M., Slagboom, P. E., Metspalu, A., van Duijn, C. M., Eriksson, J. G., Peters, A., Gieger, C., Jula, A., Groop, L., Raitakari, O. T., Power, C., Penninx, B. W., de Geus, E., Smit, J. H., Boomsma, D. I., Pedersen, N. L., Ingelsson, E., Thorsteinsdottir, U., Stefansson, K., Ripatti, S., Prokopenko, I., McCarthy, M. I., Morris, A. P. 2015; 11 (7)

    Abstract

    Reference panels from the 1000 Genomes (1000G) Project Consortium provide near complete coverage of common and low-frequency genetic variation with minor allele frequency ≥0.5% across European ancestry populations. Within the European Network for Genetic and Genomic Epidemiology (ENGAGE) Consortium, we have undertaken the first large-scale meta-analysis of genome-wide association studies (GWAS), supplemented by 1000G imputation, for four quantitative glycaemic and obesity-related traits, in up to 87,048 individuals of European ancestry. We identified two loci for body mass index (BMI) at genome-wide significance, and two for fasting glucose (FG), none of which has been previously reported in larger meta-analysis efforts to combine GWAS of European ancestry. Through conditional analysis, we also detected multiple distinct signals of association mapping to established loci for waist-hip ratio adjusted for BMI (RSPO3) and FG (GCK and G6PC2). The index variant for one association signal at the G6PC2 locus is a low-frequency coding allele, H177Y, which has recently been demonstrated to have a functional role in glucose regulation. Fine-mapping analyses revealed that the non-coding variants most likely to drive association signals at established and novel loci were enriched for overlap with enhancer elements, which for FG mapped to promoter and transcription factor binding sites in pancreatic islets, in particular. Our study demonstrates that 1000G imputation and genetic fine-mapping of common and low-frequency variant association signals at GWAS loci, integrated with genomic annotation in relevant tissues, can provide insight into the functional and regulatory mechanisms through which their effects on glycaemic and obesity-related traits are mediated.

    View details for DOI 10.1371/journal.pgen.1005230

    View details for Web of Science ID 000360622200002

    View details for PubMedID 26132169

  • Rediscovery rate estimation for assessing the validation of significant findings in high-throughput studies BRIEFINGS IN BIOINFORMATICS Ganna, A., Lee, D., Ingelsson, E., Pawitan, Y. 2015; 16 (4): 563-575

    Abstract

    It is common and advised practice in biomedical research to validate experimental or observational findings in a population different from the one where the findings were initially assessed. This practice increases the generalizability of the results and decreases the likelihood of reporting false-positive findings. Validation becomes critical when dealing with high-throughput experiments, where the large number of tests increases the chance to observe false-positive results. In this article, we review common approaches to determine statistical thresholds for validation and describe the factors influencing the proportion of significant findings from a 'training' sample that are replicated in a 'validation' sample. We refer to this proportion as rediscovery rate (RDR). In high-throughput studies, the RDR is a function of false-positive rate and power in both the training and validation samples. We illustrate the application of the RDR using simulated data and real data examples from metabolomics experiments. We further describe an online tool to calculate the RDR using t-statistics. We foresee two main applications. First, if the validation study has not yet been collected, the RDR can be used to decide the optimal combination between the proportion of findings taken to validation and the size of the validation study. Secondly, if a validation study has already been done, the RDR estimated using the training data can be compared with the observed RDR from the validation data; hence, the success of the validation study can be assessed.

    View details for DOI 10.1093/bib/bbu033

    View details for Web of Science ID 000359083200002

    View details for PubMedID 25256289

  • Using Genetic Variants to Assess the Relationship Between Circulating Lipids and Type 2 Diabetes DIABETES Fall, T., Xie, W., Poon, W., Yaghootkar, H., Maegi, R., Knowles, J. W., Lyssenko, V., Weedon, M., Frayling, T. M., Ingelsson, E. 2015; 64 (7): 2676-2684

    Abstract

    The effects of dyslipidemia on the risk of type 2 diabetes (T2D) and related traits are not clear. We used regression models and 140 lipid-associated genetic variants to estimate associations between circulating HDL cholesterol (HDL-C), LDL cholesterol (LDL-C), and triglycerides and T2D and related traits. Each genetic test was corrected for effects of variants on the other two lipid types and surrogates of adiposity. We used the largest data sets available: 34,840 T2D case and 114,981 control subjects from the DIAGRAM (DIAbetes Genetics Replication And Meta-analysis) consortium and up to 133,010 individuals without diabetes for insulin secretion and sensitivity from the MAGIC (Meta-Analyses of Glucose and Insulin-related traits Consortium) and GENESIS (GENEticS of Insulin Sensitivity) studies. Eight of 21 associations between groups of variants and diabetes traits were significant at the nominal level, including those between genetically determined lower HDL-C (β = -0.12, P = 0.03) and T2D and genetically determined lower LDL-C (β = -0.21, P = 5 × 10(-6)) and T2D. Although some of these may represent causal associations, we discuss why caution must be used when using Mendelian randomization in the context of circulating lipid levels and diabetes traits. In conclusion, we found evidence of links between genetic variants associated with lipids and T2D, but deeper knowledge of the underlying genetic mechanisms of specific lipid variants is needed before drawing definite conclusions about causality based on Mendelian randomization methodology.

    View details for DOI 10.2337/db14-1710

    View details for Web of Science ID 000356934000049

    View details for PubMedID 25948681

  • Genome-wide association study of plasma levels of polychlorinated biphenyls disclose an association with the CYP2B6 gene in a population-based sample ENVIRONMENTAL RESEARCH Ng, E., Salihovic, S., Lind, P. M., Mahajan, A., Syvanen, A., Axelsson, T., Ingelsson, E., Lindgren, C. M., van Bavel, B., Morris, A. P., Lind, L. 2015; 140: 95-101

    Abstract

    Polychlorinated biphenyls (PCBs) are a group of man-made environmental pollutants which accumulate in humans with adverse health effects. To date, very little effort has been devoted to the study of the metabolism of PCBs on a genome-wide level.Here, we conducted a genome-wide association study (GWAS) to identify genomic regions involved in the metabolism of PCBs.Plasma levels of 16 PCBs ascertained in a cohort of elderly individuals from Sweden (n=1016) were measured using gas chromatography-high resolution mass spectrophotometry (GC-HRMS). DNA samples were genotyped on the Infinium Omni Express bead microarray, and imputed up to reference panels from the 1000 Genomes Project. Association testing was performed in a linear regression framework under an additive model.Plasma levels of PCB-99 demonstrated genome-wide significant association with single nucleotide polymorphisms (SNPs) mapping to chromosome 19q13.2. The SNP with the strongest association was rs8109848 (p=3.7×10(-13)), mapping to an intronic region of CYP2B6. Moreover, when all PCBs were conditioned on PCB-99, further signals were revealed for PCBs -74, -105 and -118, mapping to the same genomic region. The lead SNPs were rs8109848 (p=3.8×10(-12)) for PCB-118, rs4802104 (p=1.4×10(-9)) for PCB-74 and rs4803413 (p=2.5×10(-9)) for PCB-105, all of which map to CYP2B6.In our study, we found plasma levels of four lower-chlorinated PCBs to be significantly associated with the genetic region mapping to the CYP2B6 locus. These findings show that CYP2B6 is of importance for the metabolism of PCBs in humans, and may help to identify individuals who may be susceptible to PCB toxicity.

    View details for DOI 10.1016/j.envres.2015.03.022

    View details for Web of Science ID 000357904100011

    View details for PubMedID 25839716

  • Contribution of common non-synonymous variants in PCSK1 to body mass index variation and risk of obesity: a systematic review and meta-analysis with evidence from up to 331 175 individuals HUMAN MOLECULAR GENETICS Nead, K. T., Li, A., Wehner, M. R., Neupane, B., Gustafsson, S., Butterworth, A., Engert, J. C., Davis, A. D., Hegele, R. A., Miller, R., Den Hoed, M., Khaw, K., Kilpelaeinen, T. O., Wareham, N., Edwards, T. L., Hallmans, G., Varga, T. V., Kardia, S. L., Smith, J. A., Zhao, W., Faul, J. D., Weir, D., Mi, J., Xi, B., Quinteros, S. C., Cooper, C., Sayer, A. A., Jameson, K., Grontved, A., Fornage, M., Sidney, S., Hanis, C. L., Highland, H. M., Haering, H., Heni, M., Lasky-Su, J., Weiss, S. T., Gerhard, G. S., Still, C., Melka, M. M., Pausova, Z., Paus, T., Grant, S. F., Hakonarson, H., Price, R. A., Wang, K., Scherag, A., Hebebrand, J., Hinney, A., Franks, P. W., Frayling, T. M., McCarthy, M. I., Hirschhorn, J. N., Loos, R. J., Ingelsson, E., Gerstein, H. C., Yusuf, S., Beyene, J., Anand, S. S., Meyre, D. 2015; 24 (12): 3582-3594

    Abstract

    Polymorphisms rs6232 and rs6234/rs6235 in PCSK1 have been associated with extreme obesity [e.g. body mass index (BMI) ≥ 40 kg/m(2)], but their contribution to common obesity (BMI ≥ 30 kg/m(2)) and BMI variation in a multi-ethnic context is unclear. To fill this gap, we collected phenotypic and genetic data in up to 331 175 individuals from diverse ethnic groups. This process involved a systematic review of the literature in PubMed, Web of Science, Embase and the NIH GWAS catalog complemented by data extraction from pre-existing GWAS or custom-arrays in consortia and single studies. We employed recently developed global meta-analytic random-effects methods to calculate summary odds ratios (OR) and 95% confidence intervals (CIs) or beta estimates and standard errors (SE) for the obesity status and BMI analyses, respectively. Significant associations were found with binary obesity status for rs6232 (OR = 1.15, 95% CI 1.06-1.24, P = 6.08 × 10(-6)) and rs6234/rs6235 (OR = 1.07, 95% CI 1.04-1.10, P = 3.00 × 10(-7)). Similarly, significant associations were found with continuous BMI for rs6232 (β = 0.03, 95% CI 0.00-0.07; P = 0.047) and rs6234/rs6235 (β = 0.02, 95% CI 0.00-0.03; P = 5.57 × 10(-4)). Ethnicity, age and study ascertainment significantly modulated the association of PCSK1 polymorphisms with obesity. In summary, we demonstrate evidence that common gene variation in PCSK1 contributes to BMI variation and susceptibility to common obesity in the largest known meta-analysis published to date in genetic epidemiology.

    View details for DOI 10.1093/hmg/ddv097

    View details for Web of Science ID 000355674400024

    View details for PubMedID 25784503

    View details for PubMedCentralID PMC4498155

  • The impact of low-frequency and rare variants on lipid levels NATURE GENETICS Surakka, I., Horikoshi, M., Magi, R., Sarin, A., Mahajan, A., Lagou, V., Marullo, L., Ferreira, T., Miraglio, B., Timonen, S., Kettunen, J., Pirinen, M., Karjalainen, J., Thorleifsson, G., Hagg, S., Hottenga, J., Isaacs, A., Ladenvall, C., Beekman, M., Esko, T., Ried, J. S., Nelson, C. P., Willenborg, C., Gustafsson, S., Westra, H., Blades, M., de Craen, A. J., de Geus, E. J., Deelen, J., Grallert, H., Hamsten, A., Havulinna, A. S., Hengstenberg, C., Houwing-Duistermaat, J. J., Hypponen, E., Karssen, L. C., Lehtimaki, T., Lyssenko, V., Magnusson, P. K., Mihailov, E., Muller-Nurasyid, M., Mpindi, J., Pedersen, N. L., Penninx, B. W., Perola, M., Pers, T. H., Peters, A., Rung, J., Smit, J. H., Steinthorsdottir, V., Tobin, M. D., Tsernikova, N., van Leeuwen, E. M., Viikari, J. S., Willems, S. M., Willemsen, G., Schunkert, H., Erdmann, J., Samani, N. J., Kaprio, J., Lind, L., Gieger, C., Metspalu, A., Slagboom, P. E., Groop, L., van Duijn, C. M., Eriksson, J. G., Jula, A., Salomaa, V., Boomsma, D. I., Power, C., Raitakari, O. T., Ingelsson, E., Jarvelin, M., Thorsteinsdottir, U., Franke, L., Ikonen, E., Kallioniemi, O., Pietiainen, V., Lindgren, C. M., Stefansson, K., Palotie, A., McCarthy, M. I., Morris, A. P., Prokopenko, I., Ripatti, S. 2015; 47 (6): 589-597

    Abstract

    Using a genome-wide screen of 9.6 million genetic variants achieved through 1000 Genomes Project imputation in 62,166 samples, we identify association to lipid traits in 93 loci, including 79 previously identified loci with new lead SNPs and 10 new loci, 15 loci with a low-frequency lead SNP and 10 loci with a missense lead SNP, and 2 loci with an accumulation of rare variants. In six loci, SNPs with established function in lipid genetics (CELSR2, GCKR, LIPC and APOE) or candidate missense mutations with predicted damaging function (CD300LG and TM6SF2) explained the locus associations. The low-frequency variants increased the proportion of variance explained, particularly for low-density lipoprotein cholesterol and total cholesterol. Altogether, our results highlight the impact of low-frequency variants in complex traits and show that imputation offers a cost-effective alternative to resequencing.

    View details for DOI 10.1038/ng.3300

    View details for Web of Science ID 000355386500009

    View details for PubMedID 25961943

  • Age- and Sex-Specific Causal Effects of Adiposity on Cardiovascular Risk Factors DIABETES Fall, T., Hagg, S., Ploner, A., Maegi, R., Fischer, K., Draisma, H. H., Sarin, A., Benyamin, B., Ladenvall, C., Akerlund, M., Kals, M., Esko, T., Nelson, C. P., Kaakinen, M., Huikari, V., Mangino, M., Meirhaeghe, A., Kristiansson, K., Nuotio, M., Kobl, M., Grallert, H., Dehghan, A., Kuningas, M., de Vries, P. S., de Bruijn, R. F., Willems, S. M., Heikkila, K., Silventoinen, K., Pietilainen, K. H., Legry, V., Giedraitis, V., Goumidi, L., Syvanen, A., Strauch, K., Koenig, W., Lichtner, P., Herder, C., Palotie, A., Menni, C., Uitterlinden, A. G., Kuulasmaa, K., Havulinna, A. S., Moreno, L. A., Gonzalez-Gross, M., Evans, A., Tregouet, D., Yarnell, J. W., Virtamo, J., Ferrieres, J., Veronesi, G., Perola, M., Arveiler, D., Brambilla, P., Lind, L., Kaprio, J., Hofman, A., Stricker, B. H., van Duijn, C. M., Ikram, M. A., Franco, O. H., Cottel, D., Dallongeville, J., Hall, A. S., Jula, A., Tobin, M. D., Penninx, B. W., Peters, A., Gieger, C., Samani, N. J., Montgomery, G. W., Whiteld, J. B., Martin, N. G., Groop, L., Spector, T. D., Magnusson, P. K., Amouyel, P., Boomsma, D. I., Nilsson, P. M., Jarvelin, M., Lyssenko, V., Metspalu, A., Strachan, D. P., Salomaa, V., Ripatti, S., Pedersen, N. L., Prokopenko, I., McCarthy, M. I., Ingelsson, E. 2015; 64 (5): 1841-1852

    Abstract

    Observational studies have reported different effects of adiposity on cardiovascular risk factors across age and sex. Since cardiovascular risk factors are enriched in obese individuals, it has not been easy to dissect the effects of adiposity from those of other risk factors. We used a Mendelian randomization approach, applying a set of 32 genetic markers to estimate the causal effect of adiposity on blood pressure, glycemic indices, circulating lipid levels, and markers of inflammation and liver disease in up to 67,553 individuals. All analyses were stratified by age (cutoff 55 years of age) and sex. The genetic score was associated with BMI in both nonstratified analysis (P = 2.8 × 10(-107)) and stratified analyses (all P < 3.3 × 10(-30)). We found evidence of a causal effect of adiposity on blood pressure, fasting levels of insulin, C-reactive protein, interleukin-6, HDL cholesterol, and triglycerides in a nonstratified analysis and in the <55-year stratum. Further, we found evidence of a smaller causal effect on total cholesterol (P for difference = 0.015) in the ≥55-year stratum than in the <55-year stratum, a finding that could be explained by biology, survival bias, or differential medication. In conclusion, this study extends previous knowledge of the effects of adiposity by providing sex- and age-specific causal estimates on cardiovascular risk factors.

    View details for DOI 10.2337/db14-0988

    View details for Web of Science ID 000353431200039

    View details for PubMedID 25712996

  • Variants in ELL2 influencing immunoglobulin levels associate with multiple myeloma NATURE COMMUNICATIONS Swaminathan, B., Thorleifsson, G., Joud, M., Ali, M., Johnsson, E., Ajore, R., Sulem, P., Halvarsson, B., Eyjolfsson, G., Haraldsdottir, V., Hultman, C., Ingelsson, E., Kristinsson, S. Y., Kahler, A. K., Lenhoff, S., Masson, G., Mellqvist, U., Mansson, R., Nelander, S., Olafsson, I., Siguroardottir, O., Steingrimsdottir, H., Vangsted, A., Vogel, U., Waage, A., Nahi, H., Gudbjartsson, D. F., Rafnar, T., Turesson, I., Gullberg, U., Stefansson, K., Hansson, M., Thorsteinsdottir, U., Nilsson, B. 2015; 6

    Abstract

    Multiple myeloma (MM) is characterized by an uninhibited, clonal growth of plasma cells. While first-degree relatives of patients with MM show an increased risk of MM, the genetic basis of inherited MM susceptibility is incompletely understood. Here we report a genome-wide association study in the Nordic region identifying a novel MM risk locus at ELL2 (rs56219066T; odds ratio (OR)=1.25; P=9.6 × 10(-10)). This gene encodes a stoichiometrically limiting component of the super-elongation complex that drives secretory-specific immunoglobulin mRNA production and transcriptional regulation in plasma cells. We find that the MM risk allele harbours a Thr298Ala missense variant in an ELL2 domain required for transcription elongation. Consistent with a hypomorphic effect, we find that the MM risk allele also associates with reduced levels of immunoglobulin A (IgA) and G (IgG) in healthy subjects (P=8.6 × 10(-9) and P=6.4 × 10(-3), respectively) and, potentially, with an increased risk of bacterial meningitis (OR=1.30; P=0.0024).

    View details for DOI 10.1038/ncomms8213

    View details for Web of Science ID 000355537600004

    View details for PubMedID 26007630

  • Genome-wide meta-analysis identifies six novel loci associated with habitual coffee consumption MOLECULAR PSYCHIATRY Cornelis, M. C., Byrne, E. M., Esko, T., Nalls, M. A., Ganna, A., Paynter, N., Monda, K. L., Amin, N., Fischer, K., Renstrom, F., Ngwa, J. S., Huikari, V., Cavadino, A., Nolte, I. M., Teumer, A., Yu, K., Marques-Vidal, P., Rawal, R., Manichaikul, A., Wojczynski, M. K., Vink, J. M., Zhao, J. H., Burlutsky, G., Lahti, J., Mikkila, V., Lemaitre, R. N., Eriksson, J., Musani, S. K., Tanaka, T., Geller, F., Luan, J., Hui, J., Maegi, R., Dimitriou, M., Garcia, M. E., Ho, W., Wright, M. J., Rose, L. M., Magnusson, P. K., Pedersen, N. L., Couper, D., Oostra, B. A., Hofman, A., Ikram, M. A., Tiemeier, H. W., Uitterlinden, A. G., Van Rooij, F. J., Barroso, I., Johansson, I., Xue, L., Kaakinen, M., Milani, L., Power, C., Snieder, H., Stolk, R. P., Baumeister, S. E., Biffar, R., Gu, F., Bastardot, F., Kutalik, Z., Jacobs, D. R., Forouhi, N. G., MIHAILOV, E., Lind, L., Lindgren, C., Michaelsson, K., Morris, A., Jensen, M., Khaw, K., Luben, R. N., Wang, J. J., Mannisto, S., Perala, M., Kahonen, M., Lehtimaki, T., Viikari, J., Mozaffarian, D., Mukamal, K., Psaty, B. M., Doering, A., Heath, A. C., Montgomery, G. W., Dahmen, N., Carithers, T., Tucker, K. L., Ferrucci, L., Boyd, H. A., Melbye, M., Treur, J. L., Mellstrom, D., Hottenga, J. J., Prokopenko, I., Toenjes, A., Deloukas, P., Kanoni, S., Lorentzon, M., Houston, D. K., Liu, Y., Danesh, J., Rasheed, A., Mason, M. A., Zonderman, A. B., Franke, L., Kristal, B. S., Karjalainen, J., Reed, D. R., Westra, H., Evans, M. K., Saleheen, D., Harris, T. B., Dedoussis, G., Curhan, G., Stumvoll, M., Beilby, J., Pasquale, L. R., Feenstra, B., Bandinelli, S., Ordovas, J. M., Chan, A. T., Peters, U., Ohlsson, C., Gieger, C., Martin, N. G., Waldenberger, M., Siscovick, D. S., Raitakari, O., Eriksson, J. G., Mitchell, P., Hunter, D. J., Kraft, P., Rimm, E. B., Boomsma, D. I., Borecki, I. B., Loos, R. J., Wareham, N. J., Vollenweider, P., Caporaso, N., Grabe, H. J., Neuhouser, M. L., Wolffenbuttel, B. H., Hu, F. B., Hyppoenen, E., Jarvelin, M., Cupples, L. A., Franks, P. W., Ridker, P. M., van Duijn, C. M., Heiss, G., Metspalu, A., North, K. E., Ingelsson, E., Nettleton, J. A., van Dam, R. M., Chasman, D. I. 2015; 20 (5): 647-656

    Abstract

    Coffee, a major dietary source of caffeine, is among the most widely consumed beverages in the world and has received considerable attention regarding health risks and benefits. We conducted a genome-wide (GW) meta-analysis of predominately regular-type coffee consumption (cups per day) among up to 91,462 coffee consumers of European ancestry with top single-nucleotide polymorphisms (SNPs) followed-up in ~30 062 and 7964 coffee consumers of European and African-American ancestry, respectively. Studies from both stages were combined in a trans-ethnic meta-analysis. Confirmed loci were examined for putative functional and biological relevance. Eight loci, including six novel loci, met GW significance (log10Bayes factor (BF)>5.64) with per-allele effect sizes of 0.03-0.14 cups per day. Six are located in or near genes potentially involved in pharmacokinetics (ABCG2, AHR, POR and CYP1A2) and pharmacodynamics (BDNF and SLC6A4) of caffeine. Two map to GCKR and MLXIPL genes related to metabolic traits but lacking known roles in coffee consumption. Enhancer and promoter histone marks populate the regions of many confirmed loci and several potential regulatory SNPs are highly correlated with the lead SNP of each. SNP alleles near GCKR, MLXIPL, BDNF and CYP1A2 that were associated with higher coffee consumption have previously been associated with smoking initiation, higher adiposity and fasting insulin and glucose but lower blood pressure and favorable lipid, inflammatory and liver enzyme profiles (P<5 × 10(-8)).Our genetic findings among European and African-American adults reinforce the role of caffeine in mediating habitual coffee consumption and may point to molecular mechanisms underlying inter-individual variability in pharmacological and health effects of coffee.

    View details for DOI 10.1038/mp.2014.107

    View details for Web of Science ID 000353706400016

    View details for PubMedID 25288136

    View details for PubMedCentralID PMC4388784

  • Genetically Determined Height and Coronary Artery Disease NEW ENGLAND JOURNAL OF MEDICINE Nelson, C. P., Hamby, S. E., Saleheen, D., Hopewell, J. C., Zeng, L., Assimes, T. L., Kanoni, S., WILLENBORG, C., Burgess, S., Amouyel, P., Anand, S., Blankenberg, S., Boehm, B. O., Clarke, R. J., Collins, R., Dedoussis, G., Farrall, M., Franks, P. W., Groop, L., Hall, A. S., Hamsten, A., Hengstenberg, C., Hovingh, G. K., Ingelsson, E., Kathiresan, S., Kee, F., Koenig, I. R., Kooner, J., Lehtimaeki, T., Maerz, W., Mcpherson, R., Metspalu, A., NIEMINEN, M. S., O'Donnell, C. J., Palmer, C. N., Peters, A., Perola, M., Reilly, M. P., Ripatti, S., Roberts, R., Salomaa, V., Shah, S. H., Schreiber, S., Siegbahn, A., Thorsteinsdottir, U., Veronesi, G., Wareham, N., Willer, C. J., Zalloua, P. A., Erdmann, J., Deloukas, P., Watkins, H., Schunkert, H., Danesh, J., Thompson, J. R., Samani, N. J. 2015; 372 (17): 1608-1618

    Abstract

    The nature and underlying mechanisms of an inverse association between adult height and the risk of coronary artery disease (CAD) are unclear.We used a genetic approach to investigate the association between height and CAD, using 180 height-associated genetic variants. We tested the association between a change in genetically determined height of 1 SD (6.5 cm) with the risk of CAD in 65,066 cases and 128,383 controls. Using individual-level genotype data from 18,249 persons, we also examined the risk of CAD associated with the presence of various numbers of height-associated alleles. To identify putative mechanisms, we analyzed whether genetically determined height was associated with known cardiovascular risk factors and performed a pathway analysis of the height-associated genes.We observed a relative increase of 13.5% (95% confidence interval [CI], 5.4 to 22.1; P<0.001) in the risk of CAD per 1-SD decrease in genetically determined height. There was a graded relationship between the presence of an increased number of height-raising variants and a reduced risk of CAD (odds ratio for height quartile 4 versus quartile 1, 0.74; 95% CI, 0.68 to 0.84; P<0.001). Of the 12 risk factors that we studied, we observed significant associations only with levels of low-density lipoprotein cholesterol and triglycerides (accounting for approximately 30% of the association). We identified several overlapping pathways involving genes associated with both development and atherosclerosis.There is a primary association between a genetically determined shorter height and an increased risk of CAD, a link that is partly explained by the association between shorter height and an adverse lipid profile. Shared biologic processes that determine achieved height and the development of atherosclerosis may explain some of the association. (Funded by the British Heart Foundation and others.).

    View details for DOI 10.1056/NEJMoa1404881

    View details for Web of Science ID 000353294000006

    View details for PubMedID 25853659

  • GWAS-identified loci for coronary heart disease are associated with intima-media thickness and plaque presence at the carotid artery bulb ATHEROSCLEROSIS den Hoed, M., Strawbridge, R. J., Almgren, P., Gustafsson, S., Axelsson, T., Engstrom, G., de Faire, U., Hedblad, B., Humphries, S. E., Lindgren, C. M., Morris, A. P., Ostling, G., Syvanen, A., Tremoli, E., Hamsten, A., Ingelsson, E., Melander, O., Lind, L. 2015; 239 (2): 304-310

    Abstract

    Large-scale genome-wide association studies (GWAS) have so far identified 45 loci that are robustly associated with coronary heart disease (CHD) in data from adult men and women of European descent.To examine whether the CHD-associated loci are associated with measures of atherosclerosis in data from up to 9582 individuals of European ancestry.Forty-five SNPs representing the CHD-associated loci were genotyped in middle-aged to elderly individuals of European descent from four independent population-based studies (IMPROVE, MDC-CC, ULSAM and PIVUS). Intima-media thickness (IMT) was measured by external B-mode ultrasonography at the far wall of the bulb (sinus) and common carotid artery. Plaque presence was defined as a maximal IMT of the bulb >1.5 mm. We meta-analysed single-SNP associations across the four studies, and combined them in a genetic predisposition score. We subsequently examined the association of the genetic predisposition score with prevalent CHD and the three indices of atherosclerosis, adjusting for sex, age and Framingham risk factors.As anticipated, the genetic predisposition score was associated with prevalent CHD, with each additional risk allele increasing the odds of disease by 5.5% (p = 4.1 × 10(-6)). Moreover, each additional CHD-risk allele across the 45 loci was associated with a 0.24% increase in IMT (p = 4.0 × 10(-3)), and with a 2.8% increased odds of plaque presence (p = 7.4 × 10(-6)) at the far wall of the bulb. The genetic predisposition score was not associated with IMT of the common carotid artery (p = 0.47).Our results suggest that the association between the 45 previously identified loci and CHD at least partly acts through atherosclerosis.

    View details for DOI 10.1016/j.atherosclerosis.2015.01.032

    View details for Web of Science ID 000351061600004

    View details for PubMedID 25682028

  • Adiposity as a cause of cardiovascular disease: a Mendelian randomization study INTERNATIONAL JOURNAL OF EPIDEMIOLOGY Haegg, S., Fall, T., Ploner, A., Maegi, R., Fischer, K., Draisma, H. H., Kals, M., de Vries, P. S., Dehghan, A., Willems, S. M., Sarin, A., Kristiansson, K., Nuotio, M., Havulinna, A. S., de Bruijn, R. F., Ikram, M. A., Kuningas, M., Stricker, B. H., Franco, O. H., Benyamin, B., Gieger, C., Hall, A. S., Huikari, V., Jula, A., Jarvelin, M., Kaakinen, M., Kaprio, J., Kobl, M., Mangino, M., Nelson, C. P., Palotie, A., Samani, N. J., Spector, T. D., Strachan, D. P., Tobin, M. D., Whitfield, J. B., Uitterlinden, A. G., Salomaa, V., Syvanen, A., Kuulasmaa, K., Magnusson, P. K., Esko, T., Hofman, A., de Geus, E. J., Lind, L., Giedraitis, V., Perola, M., Evans, A., Ferrieres, J., Virtamo, J., Kee, F., Tregouet, D., Arveiler, D., Amouyel, P., Gianfagna, F., Brambilla, P., Ripatti, S., van Duijn, C. M., Metspalu, A., Prokopenko, I., McCarthy, M. I., Pedersen, N. L., Ingelsson, E. 2015; 44 (2): 578-586

    Abstract

    Adiposity, as indicated by body mass index (BMI), has been associated with risk of cardiovascular diseases in epidemiological studies. We aimed to investigate if these associations are causal, using Mendelian randomization (MR) methods.The associations of BMI with cardiovascular outcomes [coronary heart disease (CHD), heart failure and ischaemic stroke], and associations of a genetic score (32 BMI single nucleotide polymorphisms) with BMI and cardiovascular outcomes were examined in up to 22,193 individuals with 3062 incident cardiovascular events from nine prospective follow-up studies within the ENGAGE consortium. We used random-effects meta-analysis in an MR framework to provide causal estimates of the effect of adiposity on cardiovascular outcomes.There was a strong association between BMI and incident CHD (HR = 1.20 per SD-increase of BMI, 95% CI, 1.12-1.28, P = 1.9.10(-7)), heart failure (HR = 1.47, 95% CI, 1.35-1.60, P = 9.10(-19)) and ischaemic stroke (HR = 1.15, 95% CI, 1.06-1.24, P = 0.0008) in observational analyses. The genetic score was robustly associated with BMI (β = 0.030 SD-increase of BMI per additional allele, 95% CI, 0.028-0.033, P = 3.10(-107)). Analyses indicated a causal effect of adiposity on development of heart failure (HR = 1.93 per SD-increase of BMI, 95% CI, 1.12-3.30, P = 0.017) and ischaemic stroke (HR = 1.83, 95% CI, 1.05-3.20, P = 0.034). Additional cross-sectional analyses using both ENGAGE and CARDIoGRAMplusC4D data showed a causal effect of adiposity on CHD.Using MR methods, we provide support for the hypothesis that adiposity causes CHD, heart failure and, previously not demonstrated, ischaemic stroke.

    View details for DOI 10.1093/ije/dyv094

    View details for Web of Science ID 000357106100020

    View details for PubMedID 26016847

  • Identification and validation of N-acetyltransferase 2 as an insulin sensitivity gene JOURNAL OF CLINICAL INVESTIGATION Knowles, J. W., Xie, W., Zhang, Z., Chennemsetty, I., Assimes, T. L., Paananen, J., Hansson, O., Pankow, J., Goodarzi, M. O., Carcamo-Orive, I., Morris, A. P., Chen, Y. I., Maekinen, V., Ganna, A., Mahajan, A., Guo, X., Abbasi, F., Greenawalt, D. M., Lum, P., Molony, C., Lind, L., Lindgren, C., Raffel, L. J., Tsao, P. S., Schadt, E. E., Rotter, J. I., Sinaiko, A., Reaven, G., Yang, X., Hsiung, C. A., Groop, L., Cordell, H. J., Laakso, M., Hao, K., Ingelsson, E., Frayling, T. M., Weedon, M. N., Walker, M., Quertermous, T. 2015; 125 (4): 1739-1751

    Abstract

    Decreased insulin sensitivity, also referred to as insulin resistance (IR), is a fundamental abnormality in patients with type 2 diabetes and a risk factor for cardiovascular disease. While IR predisposition is heritable, the genetic basis remains largely unknown. The GENEticS of Insulin Sensitivity consortium conducted a genome-wide association study (GWAS) for direct measures of insulin sensitivity, such as euglycemic clamp or insulin suppression test, in 2,764 European individuals, with replication in an additional 2,860 individuals. The presence of a nonsynonymous variant of N-acetyltransferase 2 (NAT2) [rs1208 (803A>G, K268R)] was strongly associated with decreased insulin sensitivity that was independent of BMI. The rs1208 "A" allele was nominally associated with IR-related traits, including increased fasting glucose, hemoglobin A1C, total and LDL cholesterol, triglycerides, and coronary artery disease. NAT2 acetylates arylamine and hydrazine drugs and carcinogens, but predicted acetylator NAT2 phenotypes were not associated with insulin sensitivity. In a murine adipocyte cell line, silencing of NAT2 ortholog Nat1 decreased insulin-mediated glucose uptake, increased basal and isoproterenol-stimulated lipolysis, and decreased adipocyte differentiation, while Nat1 overexpression produced opposite effects. Nat1-deficient mice had elevations in fasting blood glucose, insulin, and triglycerides and decreased insulin sensitivity, as measured by glucose and insulin tolerance tests, with intermediate effects in Nat1 heterozygote mice. Our results support a role for NAT2 in insulin sensitivity.

    View details for DOI 10.1172/JCI74592

    View details for Web of Science ID 000352248600037

    View details for PubMedID 25798622

  • Genetic studies of body mass index yield new insights for obesity biology. Nature Locke, A. E., Kahali, B., Berndt, S. I., Justice, A. E., Pers, T. H., Day, F. R., Powell, C., Vedantam, S., Buchkovich, M. L., Yang, J., Croteau-Chonka, D. C., Esko, T., Fall, T., Ferreira, T., Gustafsson, S., Kutalik, Z., Luan, J., Mägi, R., Randall, J. C., Winkler, T. W., Wood, A. R., Workalemahu, T., Faul, J. D., Smith, J. A., Hua Zhao, J., Zhao, W., Chen, J., Fehrmann, R., Hedman, Å. K., Karjalainen, J., Schmidt, E. M., Absher, D., Amin, N., Anderson, D., Beekman, M., Bolton, J. L., Bragg-Gresham, J. L., Buyske, S., Demirkan, A., Deng, G., Ehret, G. B., Feenstra, B., Feitosa, M. F., Fischer, K., Goel, A., Gong, J., Jackson, A. U., Kanoni, S., Kleber, M. E., Kristiansson, K., Lim, U., Lotay, V., Mangino, M., Mateo Leach, I., Medina-Gomez, C., Medland, S. E., Nalls, M. A., Palmer, C. D., Pasko, D., Pechlivanis, S., Peters, M. J., Prokopenko, I., Shungin, D., Stancáková, A., Strawbridge, R. J., Ju Sung, Y., Tanaka, T., Teumer, A., Trompet, S., van der Laan, S. W., van Setten, J., van Vliet-Ostaptchouk, J. V., Wang, Z., Yengo, L., Zhang, W., Isaacs, A., Albrecht, E., Ärnlöv, J., Arscott, G. M., Attwood, A. P., Bandinelli, S., Barrett, A., Bas, I. N., Bellis, C., Bennett, A. J., Berne, C., Blagieva, R., Blüher, M., Böhringer, S., Bonnycastle, L. L., Böttcher, Y., Boyd, H. A., Bruinenberg, M., Caspersen, I. H., Ida Chen, Y., Clarke, R., Warwick Daw, E., de Craen, A. J., Delgado, G., Dimitriou, M., Doney, A. S., Eklund, N., Estrada, K., Eury, E., Folkersen, L., Fraser, R. M., Garcia, M. E., Geller, F., Giedraitis, V., Gigante, B., Go, A. S., Golay, A., Goodall, A. H., Gordon, S. D., Gorski, M., Grabe, H., Grallert, H., Grammer, T. B., Gräßler, J., Grönberg, H., Groves, C. J., Gusto, G., Haessler, J., Hall, P., Haller, T., Hallmans, G., Hartman, C. A., Hassinen, M., Hayward, C., Heard-Costa, N. L., Helmer, Q., Hengstenberg, C., Holmen, O., Hottenga, J., James, A. L., Jeff, J. M., Johansson, Å., Jolley, J., Juliusdottir, T., Kinnunen, L., Koenig, W., Koskenvuo, M., Kratzer, W., Laitinen, J., Lamina, C., Leander, K., Lee, N. R., Lichtner, P., Lind, L., Lindström, J., Sin Lo, K., Lobbens, S., Lorbeer, R., Lu, Y., Mach, F., Magnusson, P. K., Mahajan, A., McArdle, W. L., McLachlan, S., Menni, C., Merger, S., Mihailov, E., Milani, L., Moayyeri, A., Monda, K. L., Morken, M. A., Mulas, A., Müller, G., Müller-Nurasyid, M., Musk, A. W., Nagaraja, R., Nöthen, M. M., Nolte, I. M., Pilz, S., Rayner, N. W., Renstrom, F., Rettig, R., Ried, J. S., Ripke, S., Robertson, N. R., Rose, L. M., Sanna, S., Scharnagl, H., Scholtens, S., Schumacher, F. R., Scott, W. R., Seufferlein, T., Shi, J., Vernon Smith, A., Smolonska, J., Stanton, A. V., Steinthorsdottir, V., Stirrups, K., Stringham, H. M., Sundström, J., Swertz, M. A., Swift, A. J., Syvänen, A., Tan, S., Tayo, B. O., Thorand, B., Thorleifsson, G., Tyrer, J. P., Uh, H., Vandenput, L., Verhulst, F. C., Vermeulen, S. H., Verweij, N., Vonk, J. M., Waite, L. L., Warren, H. R., Waterworth, D., Weedon, M. N., Wilkens, L. R., Willenborg, C., Wilsgaard, T., Wojczynski, M. K., Wong, A., Wright, A. F., Zhang, Q., Brennan, E. P., Choi, M., Dastani, Z., Drong, A. W., Eriksson, P., Franco-Cereceda, A., Gådin, J. R., Gharavi, A. G., Goddard, M. E., Handsaker, R. E., Huang, J., Karpe, F., Kathiresan, S., Keildson, S., Kiryluk, K., Kubo, M., Lee, J., Liang, L., Lifton, R. P., Ma, B., McCarroll, S. A., McKnight, A. J., Min, J. L., Moffatt, M. F., Montgomery, G. W., Murabito, J. M., Nicholson, G., Nyholt, D. R., Okada, Y., Perry, J. R., Dorajoo, R., Reinmaa, E., Salem, R. M., Sandholm, N., Scott, R. A., Stolk, L., Takahashi, A., Tanaka, T., van't Hooft, F. M., Vinkhuyzen, A. A., Westra, H., Zheng, W., Zondervan, K. T., Heath, A. C., Arveiler, D., Bakker, S. J., Beilby, J., Bergman, R. N., Blangero, J., Bovet, P., Campbell, H., Caulfield, M. J., Cesana, G., Chakravarti, A., Chasman, D. I., Chines, P. S., Collins, F. S., Crawford, D. C., Adrienne Cupples, L., Cusi, D., Danesh, J., de Faire, U., Den Ruijter, H. M., Dominiczak, A. F., Erbel, R., Erdmann, J., Eriksson, J. G., Farrall, M., Felix, S. B., Ferrannini, E., Ferrières, J., Ford, I., Forouhi, N. G., Forrester, T., Franco, O. H., Gansevoort, R. T., Gejman, P. V., Gieger, C., Gottesman, O., Gudnason, V., Gyllensten, U., Hall, A. S., Harris, T. B., Hattersley, A. T., Hicks, A. A., Hindorff, L. A., Hingorani, A. D., Hofman, A., Homuth, G., Kees Hovingh, G., Humphries, S. E., Hunt, S. C., Hyppönen, E., Illig, T., Jacobs, K. B., Jarvelin, M., Jöckel, K., Johansen, B., Jousilahti, P., Wouter Jukema, J., Jula, A. M., Kaprio, J., Kastelein, J. J., Keinanen-Kiukaanniemi, S. M., Kiemeney, L. A., Knekt, P., Kooner, J. S., Kooperberg, C., Kovacs, P., Kraja, A. T., Kumari, M., Kuusisto, J., Lakka, T. A., Langenberg, C., Le Marchand, L., Lehtimäki, T., Lyssenko, V., Männistö, S., Marette, A., Matise, T. C., McKenzie, C. A., McKnight, B., Moll, F. L., Morris, A. D., Morris, A. P., Murray, J. C., Nelis, M., Ohlsson, C., Oldehinkel, A. J., Ong, K. K., Madden, P. A., Pasterkamp, G., Peden, J. F., Peters, A., Postma, D. S., Pramstaller, P. P., Price, J. F., Qi, L., Raitakari, O. T., Rankinen, T., Rao, D. C., Rice, T. K., Ridker, P. M., Rioux, J. D., Ritchie, M. D., Rudan, I., Salomaa, V., Samani, N. J., Saramies, J., Sarzynski, M. A., Schunkert, H., Schwarz, P. E., Sever, P., Shuldiner, A. R., Sinisalo, J., Stolk, R. P., Strauch, K., Tönjes, A., Trégouët, D., Tremblay, A., Tremoli, E., Virtamo, J., Vohl, M., Völker, U., Waeber, G., Willemsen, G., Witteman, J. C., Zillikens, M. C., Adair, L. S., Amouyel, P., Asselbergs, F. W., Assimes, T. L., Bochud, M., Boehm, B. O., Boerwinkle, E., Bornstein, S. R., Bottinger, E. P., Bouchard, C., Cauchi, S., Chambers, J. C., Chanock, S. J., Cooper, R. S., de Bakker, P. I., Dedoussis, G., Ferrucci, L., Franks, P. W., Froguel, P., Groop, L. C., Haiman, C. A., Hamsten, A., Hui, J., Hunter, D. J., Hveem, K., Kaplan, R. C., Kivimaki, M., Kuh, D., Laakso, M., Liu, Y., Martin, N. G., März, W., Melbye, M., Metspalu, A., Moebus, S., Munroe, P. B., Njølstad, I., Oostra, B. A., Palmer, C. N., Pedersen, N. L., Perola, M., Pérusse, L., Peters, U., Power, C., Quertermous, T., Rauramaa, R., Rivadeneira, F., Saaristo, T. E., Saleheen, D., Sattar, N., Schadt, E. E., Schlessinger, D., Eline Slagboom, P., Snieder, H., Spector, T. D., Thorsteinsdottir, U., Stumvoll, M., Tuomilehto, J., Uitterlinden, A. G., Uusitupa, M., van der Harst, P., Walker, M., Wallaschofski, H., Wareham, N. J., Watkins, H., Weir, D. R., Wichmann, H., Wilson, J. F., Zanen, P., Borecki, I. B., Deloukas, P., Fox, C. S., Heid, I. M., O'Connell, J. R., Strachan, D. P., Stefansson, K., van Duijn, C. M., Abecasis, G. R., Franke, L., Frayling, T. M., McCarthy, M. I., Visscher, P. M., Scherag, A., Willer, C. J., Boehnke, M., Mohlke, K. L., Lindgren, C. M., Beckmann, J. S., Barroso, I., North, K. E., Ingelsson, E., Hirschhorn, J. N., Loos, R. J., Speliotes, E. K. 2015; 518 (7538): 197-206

    Abstract

    Obesity is heritable and predisposes to many diseases. To understand the genetic basis of obesity better, here we conduct a genome-wide association study and Metabochip meta-analysis of body mass index (BMI), a measure commonly used to define obesity and assess adiposity, in up to 339,224 individuals. This analysis identifies 97 BMI-associated loci (P < 5 × 10(-8)), 56 of which are novel. Five loci demonstrate clear evidence of several independent association signals, and many loci have significant effects on other metabolic phenotypes. The 97 loci account for ∼2.7% of BMI variation, and genome-wide estimates suggest that common variation accounts for >20% of BMI variation. Pathway analyses provide strong support for a role of the central nervous system in obesity susceptibility and implicate new genes and pathways, including those related to synaptic function, glutamate signalling, insulin secretion/action, energy metabolism, lipid biology and adipogenesis.

    View details for DOI 10.1038/nature14177

    View details for PubMedID 25673413

    View details for PubMedCentralID PMC4382211

  • New genetic loci link adipose and insulin biology to body fat distribution. Nature Shungin, D., Winkler, T. W., Croteau-Chonka, D. C., Ferreira, T., Locke, A. E., Mägi, R., Strawbridge, R. J., Pers, T. H., Fischer, K., Justice, A. E., Workalemahu, T., Wu, J. M., Buchkovich, M. L., Heard-Costa, N. L., Roman, T. S., Drong, A. W., Song, C., Gustafsson, S., Day, F. R., Esko, T., Fall, T., Kutalik, Z., Luan, J., Randall, J. C., Scherag, A., Vedantam, S., Wood, A. R., Chen, J., Fehrmann, R., Karjalainen, J., Kahali, B., Liu, C., Schmidt, E. M., Absher, D., Amin, N., Anderson, D., Beekman, M., Bragg-Gresham, J. L., Buyske, S., Demirkan, A., Ehret, G. B., Feitosa, M. F., Goel, A., Jackson, A. U., Johnson, T., Kleber, M. E., Kristiansson, K., Mangino, M., Mateo Leach, I., Medina-Gomez, C., Palmer, C. D., Pasko, D., Pechlivanis, S., Peters, M. J., Prokopenko, I., Stancáková, A., Ju Sung, Y., Tanaka, T., Teumer, A., van Vliet-Ostaptchouk, J. V., Yengo, L., Zhang, W., Albrecht, E., Ärnlöv, J., Arscott, G. M., Bandinelli, S., Barrett, A., Bellis, C., Bennett, A. J., Berne, C., Blüher, M., Böhringer, S., Bonnet, F., Böttcher, Y., Bruinenberg, M., Carba, D. B., Caspersen, I. H., Clarke, R., Daw, E. W., Deelen, J., Deelman, E., Delgado, G., Doney, A. S., Eklund, N., Erdos, M. R., Estrada, K., Eury, E., Friedrich, N., Garcia, M. E., Giedraitis, V., Gigante, B., Go, A. S., Golay, A., Grallert, H., Grammer, T. B., Gräßler, J., Grewal, J., Groves, C. J., Haller, T., Hallmans, G., Hartman, C. A., Hassinen, M., Hayward, C., Heikkilä, K., Herzig, K., Helmer, Q., Hillege, H. L., Holmen, O., Hunt, S. C., Isaacs, A., Ittermann, T., James, A. L., Johansson, I., Juliusdottir, T., Kalafati, I., Kinnunen, L., Koenig, W., Kooner, I. K., Kratzer, W., Lamina, C., Leander, K., Lee, N. R., Lichtner, P., Lind, L., Lindström, J., Lobbens, S., Lorentzon, M., Mach, F., Magnusson, P. K., Mahajan, A., McArdle, W. L., Menni, C., Merger, S., Mihailov, E., Milani, L., Mills, R., Moayyeri, A., Monda, K. L., Mooijaart, S. P., Mühleisen, T. W., Mulas, A., Müller, G., Müller-Nurasyid, M., Nagaraja, R., Nalls, M. A., Narisu, N., Glorioso, N., Nolte, I. M., Olden, M., Rayner, N. W., Renstrom, F., Ried, J. S., Robertson, N. R., Rose, L. M., Sanna, S., Scharnagl, H., Scholtens, S., Sennblad, B., Seufferlein, T., Sitlani, C. M., Vernon Smith, A., Stirrups, K., Stringham, H. M., Sundström, J., Swertz, M. A., Swift, A. J., Syvänen, A., Tayo, B. O., Thorand, B., Thorleifsson, G., Tomaschitz, A., Troffa, C., van Oort, F. V., Verweij, N., Vonk, J. M., Waite, L. L., Wennauer, R., Wilsgaard, T., Wojczynski, M. K., Wong, A., Zhang, Q., Hua Zhao, J., Brennan, E. P., Choi, M., Eriksson, P., Folkersen, L., Franco-Cereceda, A., Gharavi, A. G., Hedman, Å. K., Hivert, M., Huang, J., Kanoni, S., Karpe, F., Keildson, S., Kiryluk, K., Liang, L., Lifton, R. P., Ma, B., McKnight, A. J., McPherson, R., Metspalu, A., Min, J. L., Moffatt, M. F., Montgomery, G. W., Murabito, J. M., Nicholson, G., Nyholt, D. R., Olsson, C., Perry, J. R., Reinmaa, E., Salem, R. M., Sandholm, N., Schadt, E. E., Scott, R. A., Stolk, L., Vallejo, E. E., Westra, H., Zondervan, K. T., Amouyel, P., Arveiler, D., Bakker, S. J., Beilby, J., Bergman, R. N., Blangero, J., Brown, M. J., Burnier, M., Campbell, H., Chakravarti, A., Chines, P. S., Claudi-Boehm, S., Collins, F. S., Crawford, D. C., Danesh, J., de Faire, U., de Geus, E. J., Dörr, M., Erbel, R., Eriksson, J. G., Farrall, M., Ferrannini, E., Ferrières, J., Forouhi, N. G., Forrester, T., Franco, O. H., Gansevoort, R. T., Gieger, C., Gudnason, V., Haiman, C. A., Harris, T. B., Hattersley, A. T., Heliövaara, M., Hicks, A. A., Hingorani, A. D., Hoffmann, W., Hofman, A., Homuth, G., Humphries, S. E., Hyppönen, E., Illig, T., Jarvelin, M., Johansen, B., Jousilahti, P., Jula, A. M., Kaprio, J., Kee, F., Keinanen-Kiukaanniemi, S. M., Kooner, J. S., Kooperberg, C., Kovacs, P., Kraja, A. T., Kumari, M., Kuulasmaa, K., Kuusisto, J., Lakka, T. A., Langenberg, C., Le Marchand, L., Lehtimäki, T., Lyssenko, V., Männistö, S., Marette, A., Matise, T. C., McKenzie, C. A., McKnight, B., Musk, A. W., Möhlenkamp, S., Morris, A. D., Nelis, M., Ohlsson, C., Oldehinkel, A. J., Ong, K. K., Palmer, L. J., Penninx, B. W., Peters, A., Pramstaller, P. P., Raitakari, O. T., Rankinen, T., Rao, D. C., Rice, T. K., Ridker, P. M., Ritchie, M. D., Rudan, I., Salomaa, V., Samani, N. J., Saramies, J., Sarzynski, M. A., Schwarz, P. E., Shuldiner, A. R., Staessen, J. A., Steinthorsdottir, V., Stolk, R. P., Strauch, K., Tönjes, A., Tremblay, A., Tremoli, E., Vohl, M., Völker, U., Vollenweider, P., Wilson, J. F., Witteman, J. C., Adair, L. S., Bochud, M., Boehm, B. O., Bornstein, S. R., Bouchard, C., Cauchi, S., Caulfield, M. J., Chambers, J. C., Chasman, D. I., Cooper, R. S., Dedoussis, G., Ferrucci, L., Froguel, P., Grabe, H., Hamsten, A., Hui, J., Hveem, K., Jöckel, K., Kivimaki, M., Kuh, D., Laakso, M., Liu, Y., März, W., Munroe, P. B., Njølstad, I., Oostra, B. A., Palmer, C. N., Pedersen, N. L., Perola, M., Pérusse, L., Peters, U., Power, C., Quertermous, T., Rauramaa, R., Rivadeneira, F., Saaristo, T. E., Saleheen, D., Sinisalo, J., Slagboom, P. E., Snieder, H., Spector, T. D., Thorsteinsdottir, U., Stumvoll, M., Tuomilehto, J., Uitterlinden, A. G., Uusitupa, M., van der Harst, P., Veronesi, G., Walker, M., Wareham, N. J., Watkins, H., Wichmann, H., Abecasis, G. R., Assimes, T. L., Berndt, S. I., Boehnke, M., Borecki, I. B., Deloukas, P., Franke, L., Frayling, T. M., Groop, L. C., Hunter, D. J., Kaplan, R. C., O'Connell, J. R., Qi, L., Schlessinger, D., Strachan, D. P., Stefansson, K., van Duijn, C. M., Willer, C. J., Visscher, P. M., Yang, J., Hirschhorn, J. N., Zillikens, M. C., McCarthy, M. I., Speliotes, E. K., North, K. E., Fox, C. S., Barroso, I., Franks, P. W., Ingelsson, E., Heid, I. M., Loos, R. J., Cupples, L. A., Morris, A. P., Lindgren, C. M., Mohlke, K. L. 2015; 518 (7538): 187-196

    Abstract

    Body fat distribution is a heritable trait and a well-established predictor of adverse metabolic outcomes, independent of overall adiposity. To increase our understanding of the genetic basis of body fat distribution and its molecular links to cardiometabolic traits, here we conduct genome-wide association meta-analyses of traits related to waist and hip circumferences in up to 224,459 individuals. We identify 49 loci (33 new) associated with waist-to-hip ratio adjusted for body mass index (BMI), and an additional 19 loci newly associated with related waist and hip circumference measures (P < 5 × 10(-8)). In total, 20 of the 49 waist-to-hip ratio adjusted for BMI loci show significant sexual dimorphism, 19 of which display a stronger effect in women. The identified loci were enriched for genes expressed in adipose tissue and for putative regulatory elements in adipocytes. Pathway analyses implicated adipogenesis, angiogenesis, transcriptional regulation and insulin resistance as processes affecting fat distribution, providing insight into potential pathophysiological mechanisms.

    View details for DOI 10.1038/nature14132

    View details for PubMedID 25673412

    View details for PubMedCentralID PMC4338562

  • Skeletal muscle morphology and risk of cardiovascular disease in elderly men EUROPEAN JOURNAL OF PREVENTIVE CARDIOLOGY Andersen, K., Lind, L., Ingelsson, E., Arnlov, J., Byberg, L., Michaelsson, K., Sundstrom, J. 2015; 22 (2): 231-239

    Abstract

    While it is well known that physical inactivity is a major risk factor for cardiovascular disease, there is still a search for the mechanisms by which exercise exerts its positive effect. Skeletal muscle fibre type can be affected to some extent by exercise, and different fibre types possess different anti-inflammatory and glucometabolic properties that may influence cardiovascular disease risk.Population-based cohort study.We investigated relations of skeletal muscle morphology to risk of cardiovascular events in a sample of 466 71-year-old men without cardiovascular disease, of which 295 were physically active (strenuous physical activity at least 3 h/week).During a median of 13.1 years of follow up, 173 major cardiovascular events occurred. Among physically active men, 10% higher proportion of type-I (slow-twitch oxidative) fibres was associated with a hazard ratio (HR) of 0.84 (95% confidence interval 0.74-0.95) for cardiovascular events, and 10% higher proportion of type-IIx (fast-twitch glycolytic) fibres was associated with a HR of 1.24 (1.06-1.45), adjusting for age. Similar results were observed in several sets of multivariable-adjusted models. No association of muscle fibre type with risk of cardiovascular events was observed among physically inactive men.Higher skeletal muscle proportion of type-I fibres was associated with lower risk of cardiovascular events and a higher proportion of type-IIx fibres was associated with higher risk of cardiovascular events. These relations were only observed in physically active men. Skeletal muscle fibre composition may be a mediator of the protective effects of exercise against cardiovascular disease.

    View details for DOI 10.1177/2047487313506828

    View details for Web of Science ID 000348115400014

    View details for PubMedID 24092874

  • Mutagenesis. Smoking is associated with mosaic loss of chromosome Y. Science Dumanski, J. P., Rasi, C., Lönn, M., Davies, H., Ingelsson, M., Giedraitis, V., Lannfelt, L., Magnusson, P. K., Lindgren, C. M., Morris, A. P., Cesarini, D., Johannesson, M., Tiensuu Janson, E., Lind, L., Pedersen, N. L., Ingelsson, E., Forsberg, L. A. 2015; 347 (6217): 81-83

    Abstract

    Tobacco smoking is a risk factor for numerous disorders, including cancers affecting organs outside the respiratory tract. Epidemiological data suggest that smoking is a greater risk factor for these cancers in males compared with females. This observation, together with the fact that males have a higher incidence of and mortality from most non-sex-specific cancers, remains unexplained. Loss of chromosome Y (LOY) in blood cells is associated with increased risk of nonhematological tumors. We demonstrate here that smoking is associated with LOY in blood cells in three independent cohorts [TwinGene: odds ratio (OR) = 4.3, 95% confidence interval (CI) = 2.8 to 6.7; Uppsala Longitudinal Study of Adult Men: OR = 2.4, 95% CI = 1.6 to 3.6; and Prospective Investigation of the Vasculature in Uppsala Seniors: OR = 3.5, 95% CI = 1.4 to 8.4] encompassing a total of 6014 men. The data also suggest that smoking has a transient and dose-dependent mutagenic effect on LOY status. The finding that smoking induces LOY thus links a preventable risk factor with the most common acquired human mutation.

    View details for DOI 10.1126/science.1262092

    View details for PubMedID 25477213

  • Smoking is associated with mosaic loss of chromosome Y SCIENCE Dumanski, J. P., Rasi, C., Lonn, M., Davies, H., Ingelsson, M., Giedraitis, V., Lannfelt, L., Magnusson, P. K., Lindgren, C. M., Morris, A. P., Cesarini, D., Johannesson, M., Janson, E. T., Lind, L., Pedersen, N. L., Ingelsson, E., Forsberg, L. A. 2015; 347 (6217): 81-83
  • Identification and Functional Characterization of G6PC2 Coding Variants Influencing Glycemic Traits Define an Effector Transcript at the G6PC2-ABCB11 Locus PLOS GENETICS Mahajan, A., Sim, X., Ng, H. J., Manning, A., Rivas, M. A., Highland, H. M., Locke, A. E., Grarup, N., Im, H. K., Cingolani, P., Flannick, J., Fontanillas, P., Fuchsberger, C., Gaulton, K. J., Teslovich, T. M., Rayner, N. W., Robertson, N. R., Beer, N. L., Rundle, J. K., Bork-Jensen, J., Ladenvall, C., Blancher, C., Buck, D., Buck, G., Burtt, N. P., Gabriel, S., Gjesing, A. P., Groves, C. J., Hollensted, M., Huyghe, J. R., Jackson, A. U., Jun, G., Justesen, J. M., Mangino, M., Murphy, J., Neville, M., Onofrio, R., Small, K. S., Stringham, H. M., Syvanen, A., Trakalo, J., Abecasis, G., Bell, G. I., Blangero, J., Cox, N. J., Duggirala, R., Hanis, C. L., Seielstad, M., Wilson, J. G., Christensen, C., Brandslund, I., Rauramaa, R., Surdulescu, G. L., Doney, A. S., Lannfelt, L., Linneberg, A., Isomaa, B., Tuomi, T., Jorgensen, M. E., Jorgensen, T., Kuusisto, J., Uusitupa, M., Salomaa, V., Spector, T. D., Morris, A. D., Palmer, C. N., Collins, F. S., Mohlke, K. L., Bergman, R. N., Ingelsson, E., Lind, L., Tuomilehto, J., Hansen, T., Watanabe, R. M., Prokopenko, I., Dupuis, J., Karpe, F., Groop, L., Laakso, M., Pedersen, O., Florez, J. C., Morris, A. P., Altshuler, D., Meigs, J. B., Boehnke, M., McCarthy, M. I., Lindgren, C. M., Gloyn, A. L. 2015; 11 (1)

    Abstract

    Genome wide association studies (GWAS) for fasting glucose (FG) and insulin (FI) have identified common variant signals which explain 4.8% and 1.2% of trait variance, respectively. It is hypothesized that low-frequency and rare variants could contribute substantially to unexplained genetic variance. To test this, we analyzed exome-array data from up to 33,231 non-diabetic individuals of European ancestry. We found exome-wide significant (P<5×10-7) evidence for two loci not previously highlighted by common variant GWAS: GLP1R (p.Ala316Thr, minor allele frequency (MAF)=1.5%) influencing FG levels, and URB2 (p.Glu594Val, MAF = 0.1%) influencing FI levels. Coding variant associations can highlight potential effector genes at (non-coding) GWAS signals. At the G6PC2/ABCB11 locus, we identified multiple coding variants in G6PC2 (p.Val219Leu, p.His177Tyr, and p.Tyr207Ser) influencing FG levels, conditionally independent of each other and the non-coding GWAS signal. In vitro assays demonstrate that these associated coding alleles result in reduced protein abundance via proteasomal degradation, establishing G6PC2 as an effector gene at this locus. Reconciliation of single-variant associations and functional effects was only possible when haplotype phase was considered. In contrast to earlier reports suggesting that, paradoxically, glucose-raising alleles at this locus are protective against type 2 diabetes (T2D), the p.Val219Leu G6PC2 variant displayed a modest but directionally consistent association with T2D risk. Coding variant associations for glycemic traits in GWAS signals highlight PCSK1, RREB1, and ZHX3 as likely effector transcripts. These coding variant association signals do not have a major impact on the trait variance explained, but they do provide valuable biological insights.

    View details for DOI 10.1371/journal.pgen.1004876

    View details for Web of Science ID 000349314600012

    View details for PubMedID 25625282

  • Low-frequency and rare exome chip variants associate with fasting glucose and type 2 diabetes susceptibility NATURE COMMUNICATIONS Wessel, J., Chu, A. Y., Willems, S. M., Wang, S., Yaghootkar, H., Brody, J. A., Dauriz, M., Hivert, M., Raghavan, S., Lipovich, L., Hidalgo, B., Fox, K., Huffman, J. E., An, P., Lu, Y., Rasmussen-Torvik, L. J., Grarup, N., Ehm, M. G., Li, L., Baldridge, A. S., Stancakova, A., Abrol, R., Besse, C., Boland, A., Bork-Jensen, J., Fornage, M., Freitag, D. F., Garcia, M. E., Guo, X., Hara, K., Isaacs, A., Jakobsdottir, J., Lange, L. A., Layton, J. C., Li, M., Zhao, J. H., Meidtner, K., Morrison, A. C., Nalls, M. A., Peters, M. J., Sabater-Lleal, M., Schurmann, C., Silveira, A., Smith, A. V., Southam, L., Stoiber, M. H., Strawbridge, R. J., Taylor, K. D., Varga, T. V., Allin, K. H., Amin, N., Aponte, J. L., Aung, T., Barbieri, C., Bihlmeyer, N. A., Boehnke, M., Bombieri, C., Bowden, D. W., Burns, S. M., Chen, Y., Chen, Y., Cheng, C., Correa, A., Czajkowski, J., Dehghan, A., Ehret, G. B., Eiriksdottir, G., Escher, S. A., Farmaki, A., Franberg, M., Gambaro, G., Giulianini, F., Goddard, W. A., Goel, A., Gottesman, O., Grove, M. L., Gustafsson, S., Hai, Y., Hallmans, G., Heo, J., Hoffmann, P., Ikram, M. K., Jensen, R. A., Jorgensen, M. E., Jorgensen, T., Karaleftheri, M., Khor, C. C., Kirkpatrick, A., Kraja, A. T., Kuusisto, J., Lange, E. M., Lee, I. T., Lee, W., Leong, A., Liao, J., Liu, C., Liu, Y., Lindgren, C. M., Linneberg, A., Malerba, G., Mamakou, V., Marouli, E., Maruthur, N. M., Matchan, A., McKean-Cowdin, R., McLeod, O., Metcalf, G. A., Mohlke, K. L., Muzny, D. M., Ntalla, I., Palmer, N. D., Pasko, D., Peter, A., Rayner, N. W., Renstroem, F., Rice, K., Sala, C. F., Sennblad, B., Serafetinidis, I., Smith, J. A., Soranzo, N., Speliotes, E. K., Stahl, E. A., Stirrups, K., Tentolouris, N., Thanopoulou, A., Torres, M., Traglia, M., Tsafantakis, E., Javad, S., Yanek, L. R., Zengini, E., Becker, D. M., Bis, J. C., Brown, J. B., Cupples, L. A., Hansen, T., Ingelsson, E., Karter, A. J., Lorenzo, C., Mathias, R. A., Norris, J. M., Peloso, G. M., Sheu, W. H., Toniolo, D., Vaidya, D., Varma, R., Wagenknecht, L. E., Boeing, H., Bottinger, E. P., Dedoussis, G., Deloukas, P., Ferrannini, E., Franco, O. H., Franks, P. W., Gibbs, R. A., Gudnason, V., Hamsten, A., Harris, T. B., Hattersley, A. T., Hayward, C., Hofman, A., Jansson, J., Langenberg, C., Launer, L. J., Levy, D., Oostra, B. A., O'Donnell, C. J., O'Rahilly, S., Padmanabhan, S., Pankow, J. S., Polasek, O., Province, M. A., Rich, S. S., Ridker, P. M., Rudan, I., Schulze, M. B., Smith, B. H., Uitterlinden, A. G., Walker, M., Watkins, H., Wong, T. Y., Zeggini, E., Laakso, M., Borecki, I. B., Chasman, D. I., Pedersen, O., Psaty, B. M., Tai, E. S., van Duijn, C. M., Wareham, N. J., Waterworth, D. M., Boerwinkle, E., Kao, W. H., Florez, J. C., Loos, R. J., Wilson, J. G., Frayling, T. M., Siscovick, D. S., Dupuis, J., Rotter, J. I., Meigs, J. B., Scott, R. A., Goodarzi, M. O., Sharp, S. J., Forouhi, N. G., Kerrison, N. D., Lucarelli, D. M., Sims, M., Barroso, I., McCarthy, M. I., Arriola, L., Balkau, B., Barricarte, A., Gonzalez, C., Grioni, S., Kaaks, R., Key, T. J., Navarro, C., Nilsson, P. M., Overvad, K., Palli, D., Panico, S., Quiros, J. R., Rolandsson, O., Sacerdote, C., Sanchez, M., Slimani, N., Tjonneland, A., Tumino, R., van der A, D. L., van der Schouw, Y. T., Riboli, E. 2015; 6

    Abstract

    Fasting glucose and insulin are intermediate traits for type 2 diabetes. Here we explore the role of coding variation on these traits by analysis of variants on the HumanExome BeadChip in 60,564 non-diabetic individuals and in 16,491 T2D cases and 81,877 controls. We identify a novel association of a low-frequency nonsynonymous SNV in GLP1R (A316T; rs10305492; MAF=1.4%) with lower FG (β=-0.09±0.01 mmol l(-1), P=3.4 × 10(-12)), T2D risk (OR[95%CI]=0.86[0.76-0.96], P=0.010), early insulin secretion (β=-0.07±0.035 pmolinsulin mmolglucose(-1), P=0.048), but higher 2-h glucose (β=0.16±0.05 mmol l(-1), P=4.3 × 10(-4)). We identify a gene-based association with FG at G6PC2 (pSKAT=6.8 × 10(-6)) driven by four rare protein-coding SNVs (H177Y, Y207S, R283X and S324P). We identify rs651007 (MAF=20%) in the first intron of ABO at the putative promoter of an antisense lncRNA, associating with higher FG (β=0.02±0.004 mmol l(-1), P=1.3 × 10(-8)). Our approach identifies novel coding variant associations and extends the allelic spectrum of variation underlying diabetes-related quantitative traits and T2D susceptibility.

    View details for DOI 10.1038/ncomms6897

    View details for Web of Science ID 000348741700001

    View details for PubMedID 25631608

  • Large-scale Metabolomic Profiling Identifies Novel Biomarkers for Incident Coronary Heart Disease PLOS GENETICS Ganna, A., Salihovic, S., Sundstrom, J., Broeckling, C. D., Hedman, A. K., Magnusson, P. K., Pedersen, N. L., Larsson, A., Siegbahn, A., Zilmer, M., Prenni, J., Arnlov, J., Lind, L., Fall, T., Ingelsson, E. 2014; 10 (12)

    Abstract

    Analyses of circulating metabolites in large prospective epidemiological studies could lead to improved prediction and better biological understanding of coronary heart disease (CHD). We performed a mass spectrometry-based non-targeted metabolomics study for association with incident CHD events in 1,028 individuals (131 events; 10 y. median follow-up) with validation in 1,670 individuals (282 events; 3.9 y. median follow-up). Four metabolites were replicated and independent of main cardiovascular risk factors [lysophosphatidylcholine 18∶1 (hazard ratio [HR] per standard deviation [SD] increment = 0.77, P-value<0.001), lysophosphatidylcholine 18∶2 (HR = 0.81, P-value<0.001), monoglyceride 18∶2 (MG 18∶2; HR = 1.18, P-value = 0.011) and sphingomyelin 28∶1 (HR = 0.85, P-value = 0.015)]. Together they contributed to moderate improvements in discrimination and re-classification in addition to traditional risk factors (C-statistic: 0.76 vs. 0.75; NRI: 9.2%). MG 18∶2 was associated with CHD independently of triglycerides. Lysophosphatidylcholines were negatively associated with body mass index, C-reactive protein and with less evidence of subclinical cardiovascular disease in additional 970 participants; a reverse pattern was observed for MG 18∶2. MG 18∶2 showed an enrichment (P-value = 0.002) of significant associations with CHD-associated SNPs (P-value = 1.2×10-7 for association with rs964184 in the ZNF259/APOA5 region) and a weak, but positive causal effect (odds ratio = 1.05 per SD increment in MG 18∶2, P-value = 0.05) on CHD, as suggested by Mendelian randomization analysis. In conclusion, we identified four lipid-related metabolites with evidence for clinical utility, as well as a causal role in CHD development.

    View details for DOI 10.1371/journal.pgen.1004801

    View details for Web of Science ID 000346649900014

    View details for PubMedID 25502724

  • Common Genetic Variants Highlight the Role of Insulin Resistance and Body Fat Distribution in Type 2 Diabetes, Independent of Obesity DIABETES Scott, R. A., Fall, T., Pasko, D., Barker, A., Sharp, S. J., Arriola, L., Balkau, B., Barricarte, A., Barroso, I., Boeing, H., Clavel-Chapelon, F., Crowe, F. L., Dekker, J. M., Fagherazzi, G., Ferrannini, E., Forouhi, N. G., Franks, P. W., Gavrila, D., Giedraitis, V., Grioni, S., Groop, L. C., Kaaks, R., Key, T. J., Kuehn, T., Lotta, L. A., Nilsson, P. M., Overvad, K., Palli, D., Panico, S., Quiros, J. R., Rolandsson, O., Roswall, N., Sacerdote, C., Sala, N., Sanchez, M., Schulze, M. B., Siddiq, A., Slimani, N., Sluijs, I., Spijkerman, A. M., Tjonneland, A., Tumino, R., van der A, D. L., Yaghootkar, H., McCarthy, M. I., Semple, R. K., Riboli, E., Walker, M., Ingelsson, E., Frayling, T. M., Savage, D. B., Langenberg, C., Wareham, N. J. 2014; 63 (12): 4378-4387

    Abstract

    We aimed to validate genetic variants as instruments for insulin resistance and secretion, to characterize their association with intermediate phenotypes, and to investigate their role in type 2 diabetes (T2D) risk among normal-weight, overweight, and obese individuals. We investigated the association of genetic scores with euglycemic-hyperinsulinemic clamp- and oral glucose tolerance test-based measures of insulin resistance and secretion and a range of metabolic measures in up to 18,565 individuals. We also studied their association with T2D risk among normal-weight, overweight, and obese individuals in up to 8,124 incident T2D cases. The insulin resistance score was associated with lower insulin sensitivity measured by M/I value (β in SDs per allele [95% CI], -0.03 [-0.04, -0.01]; P = 0.004). This score was associated with lower BMI (-0.01 [-0.01, -0.0]; P = 0.02) and gluteofemoral fat mass (-0.03 [-0.05, -0.02; P = 1.4 × 10(-6)) and with higher alanine transaminase (0.02 [0.01, 0.03]; P = 0.002) and γ-glutamyl transferase (0.02 [0.01, 0.03]; P = 0.001). While the secretion score had a stronger association with T2D in leaner individuals (Pinteraction = 0.001), we saw no difference in the association of the insulin resistance score with T2D among BMI or waist strata (Pinteraction > 0.31). While insulin resistance is often considered secondary to obesity, the association of the insulin resistance score with lower BMI and adiposity and with incident T2D even among individuals of normal weight highlights the role of insulin resistance and ectopic fat distribution in T2D, independently of body size.

    View details for DOI 10.2337/db14-0319

    View details for Web of Science ID 000345335500046

    View details for PubMedID 24947364

  • The association between glomerular filtration rate and left ventricular function in two independent community-based cohorts of elderly NEPHROLOGY DIALYSIS TRANSPLANTATION Nerpin, E., Ingelsson, E., Riserus, U., Sundstrom, J., Andren, B., Jobs, E., Larsson, A., Lars, L., Arnlov, J. 2014; 29 (11): 2069-2074

    Abstract

    The cardiorenal syndrome, the detrimental bi-directional interplay between symptomatic heart failure and chronic kidney disease, is a major clinical challenge. Nonetheless, it is unknown if this interplay begins already at an asymptomatic stage. Therefore we investigated whether the glomerular filtration rate (GFR) is associated with left ventricular function in participants free from clinical heart failure and with a left ventricular ejection fraction (LVEF) >40% and with pre-specified sub-group analyses in individuals with a GFR >60 mL/min/m(2).Two independent community-based cohorts were used; the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS; n = 911; 50% women; mean age: 70 years) and the Uppsala Longitudinal Study of Adult Men (ULSAM; n = 538; mean age: 71 years). We investigated cross-sectional association between cystatin C-based GFR (estimated glomerular function [eGFR]) and systolic (LVEF), diastolic- (isovolumic relaxation time [IVRT]) and global left ventricular function (myocardial performance index [MPI]) determined by echocardiography.In both PIVUS and ULSAM, higher eGFR was significantly associated with higher LVEF (P = 0.004 [PIVUS] and P = 0.005 [ULSAM]). In PIVUS, higher eGFR was significantly associated with lower IVRT (P = 0.001) and MPI (P = 0.006), in age- and sex-adjusted models. After further adjustment for cardiovascular risk factors, the association between higher eGFR and higher LVEF was still statistically significant (P = 0.008 [PIVUS] and P = 0.02 [ULSAM]). In PIVUS, the age- and sex-adjusted association between eGFR and left ventricular function was similar in participants with eGFR >60 mL/min/m(2).Our data suggest that the interplay between kidney and heart function begins prior to the development of symptomatic heart failure and kidney disease.

    View details for DOI 10.1093/ndt/gfu199

    View details for Web of Science ID 000344625400015

    View details for PubMedID 24916339

  • Fat Mass and Obesity-Associated Gene (FTO) Is Linked to Higher Plasma Levels of the Hunger Hormone Ghrelin and Lower Serum Levels of the Satiety Hormone Leptin in Older Adults DIABETES Benedict, C., Axelsson, T., Soederberg, S., Larsson, A., Ingelsson, E., Lind, L., Schioeth, H. B. 2014; 63 (11): 3955-3959

    Abstract

    The mechanisms through which common polymorphisms in the fat mass and obesity-associated gene (FTO) drive the development of obesity in humans are poorly understood. Using cross-sectional data from 985 older people (50% females) who participated at age 70 years in the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS), circulating levels of ghrelin and leptin were measured after an overnight fast. In addition, subjects were genotyped for FTO rs17817449 (AA, n = 345 [35%]; AC/CA, n = 481 [48.8%]; CC, n = 159 [16.1%]). Linear regression analyses controlling for sex, self-reported physical activity level, fasting plasma glucose, and BMI were used. A positive relationship between the number of FTO C risk alleles and plasma ghrelin levels was found (P = 0.005; relative plasma ghrelin difference between CC and AA carriers = ∼ 9%). In contrast, serum levels of the satiety-enhancing hormone leptin were inversely linked to the number of FTO C risk alleles (P = 0.001; relative serum leptin difference between CC and AA carriers = ∼ 11%). These associations were also found when controlling for waist circumference. The present findings suggest that FTO may facilitate weight gain in humans by shifting the endocrine balance from the satiety hormone leptin toward the hunger-promoting hormone ghrelin.

    View details for DOI 10.2337/db14-0470

    View details for Web of Science ID 000343966100043

    View details for PubMedID 24898142

  • Influence of persistent organic pollutants on oxidative stress in population-based samples CHEMOSPHERE Kumar, J., Lind, P. M., Salihovic, S., van Bavel, B., Lind, L., Ingelsson, E. 2014; 114: 303-309
  • Soluble tumor necrosis factor receptor 1 (sTNFR1) is associated with increased total mortality due to cancer and cardiovascular causes - Findings from two community based cohorts of elderly ATHEROSCLEROSIS Carlsson, A. C., Juhlin, C. C., Larsson, T. E., Larsson, A., Ingelsson, E., Sundstrom, J., Lind, L., Arnlov, J. 2014; 237 (1): 236-242

    Abstract

    Experimental evidence support soluble receptors for tumor necrosis factor alpha as important mediators of the underlying pathology leading to cardiovascular disease and cancer. However, prospective data concerning the relation between circulating soluble tumor necrosis factor receptor-1 (sTNFR1) and mortality in humans are lacking. We aimed to explore and validate the association between sTNFR1 and mortality, and to explore the influence of other established risk factors for mortality, including other inflammatory markers.The association between serum sTNFR1and the risk for mortality was investigated in two community-based cohorts of elderly: the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS; women 50%, n = 1005, mean age 70 years, median follow-up 7.9 years) and the Uppsala Longitudinal Study of Adult Men (ULSAM, n = 775, mean age 77 years, median follow-up 8.1 years).In total, 101 participants in PIVUS and 274 in ULSAM died during follow-up. In multivariable Cox regression models adjusted for inflammation, lifestyle and established cardiovascular risk factors, one standard deviation (SD) higher sTNFR1 was associated with a hazard ratio (HR) for mortality of 1.37, 95% confidence interval (CI) 1.17-1.60, in PIVUS and HR 1.22, 95% CI 1.10-1.37 in ULSAM. Moreover, circulatings TNFR1 was associated with cardiovascular mortality (HR per SD of sTNFR1, 1.24, 95% CI 1.07-1.44) and cancer mortality (HR per SD of sTNFR1, 1.32, 95% CI 1.11-1.57) in the ULSAM cohort. High levels of sTNFR1 identified individuals with increased risk of mortality among those with high as well as low levels of systemic inflammation.An association between circulating sTNFR1 and an increased risk for mortality was found and validated in two independent community-based cohorts. The future clinical role of sTNFR1 to identify high risk patients for adverse outcomes and mortality has yet to be determined.

    View details for DOI 10.1016/j.atherosclerosis.2014.09.005

    View details for Web of Science ID 000345011300037

    View details for PubMedID 25255422

  • Influence of persistent organic pollutants on oxidative stress in population-based samples. Chemosphere Kumar, J., Monica Lind, P., Salihovic, S., van Bavel, B., Lind, L., Ingelsson, E. 2014; 114: 303-309

    Abstract

    Persistent organic pollutants (POPs) are a large group of chemicals widely used and produced in various industrial applications. Many cell culture/animal studies have shown that POPs can induce oxidative stress. Since such data is lacking in humans, we conducted a large population-based study to analyze associations between POPs and oxidative stress markers. We measured following POPs; 16 polychlorinated biphenyls (PCBs), 5 organochlorine (OC) pesticides, octachlorinated dibenzo-p-dioxin, and polybrominated diphenyl ether 47, and oxidative stress markers; homocysteine, reduced [GSH] and oxidized glutathione [GSSG], glutathione ratio [GSSG/GSH], total glutathione, oxidized low-density lipoprotein [ox-LDL], ox-LDL antibodies, conjugated dienes, baseline conjugated dienes of LDL, and total anti-oxidative capacity in plasma samples collected from 992 70-year old individuals (50% women) from the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) cohort. Linear regression analyses were performed to study the associations between oxidative stress markers and summary measures of POPs including the total toxic equivalence (TEQ), sums of PCBs and OC pesticides (main exposures) while adjusting for potential confounders. In multivariable-adjusted analyses, sum of PCBs showed strong associations with ox-LDL (β=0.94; P=2.9*10(-6)). Further, sum of PCBs showed association with glutathione-related markers (GSSG: β=-0.01; P=6.0*10(-7); GSSG/GSH: β=-0.002; P=9.7*10(-10)), although in reverse direction. Other summary measures did not show any significant association with these markers. In our study of elderly individuals from the general population, we show that plasma levels of POPs are associated with markers of increased oxidative stress thereby suggesting that even low dose background exposure to POPs may be involved in oxidative stress.

    View details for DOI 10.1016/j.chemosphere.2014.05.013

    View details for PubMedID 25113216

  • Defining the role of common variation in the genomic and biological architecture of adult human height NATURE GENETICS Wood, A. R., Esko, T., Yang, J., Vedantam, S., Pers, T. H., Gustafsson, S., Chun, A. Y., Estrada, K., Luan, J., Kutalik, Z., Amin, N., Buchkovich, M. L., Croteau-Chonka, D. C., Day, F. R., Duan, Y., Fall, T., Fehrmann, R., Ferreira, T., Jackson, A. U., Karjalainen, J., Lo, K. S., Locke, A. E., Maegi, R., Mihailov, E., Porcu, E., Randall, J. C., Scherag, A., Vinkhuyzen, A. A., Westra, H., Winkler, T. W., Workalemahu, T., Zhao, J. H., Absher, D., Albrecht, E., Anderson, D., Baron, J., Beekman, M., Demirkan, A., Ehret, G. B., Feenstra, B., Feitosa, M. F., Fischer, K., Fraser, R. M., Goel, A., Gong, J., Justice, A. E., Kanoni, S., Kleber, M. E., Kristiansson, K., Lim, U., Lotay, V., Lui, J. C., Mangino, M., Leach, I. M., Medina-Gomez, C., Nalls, M. A., Nyholt, D. R., Palmer, C. D., Pasko, D., Pechlivanis, S., Prokopenko, I., Ried, J. S., Ripke, S., Shungin, D., Stancakova, A., Strawbridge, R. J., Sung, Y. J., Tanaka, T., Teumer, A., Trompet, S., van der Laan, S. W., van Setten, J., van Vliet-Ostaptchouk, J. V., Wang, Z., Yengo, L., Zhang, W., Afzal, U., Arnloev, J., Arscott, G. M., Bandinelli, S., Barrett, A., Bellis, C., Bennett, A. J., Berne, C., Blueher, M., Bolton, J. L., Boettcher, Y., Boyd, H. A., Bruinenberg, M., Buckley, B. M., Buyske, S., Caspersen, I. H., Chines, P. S., Clarke, R., Claudi-Boehm, S., Cooper, M., Daw, E. W., de Jong, P. A., Deelen, J., Delgado, G., Denny, J. C., Dhonukshe-Rutten, R., Dimitriou, M., Doney, A. S., Doerr, M., Eklund, N., Eury, E., Folkersen, L., Garcia, M. E., Geller, F., Giedraitis, V., Go, A. S., Grallert, H., Grammer, T. B., Graessler, J., Groenberg, H., de Groot, L. C., Groves, C. J., Haessler, J., Hall, P., Haller, T., Hallmans, G., Hannemann, A., Hartman, C. A., Hassinen, M., Hayward, C., Heard-Costa, N. L., Helmer, Q., Hemani, G., Henders, A. K., Hillege, H. L., Hlatky, M. A., Hoffmann, W., Hoffmann, P., Holmen, O., Houwing-Duistermaat, J. J., Illig, T., Isaacs, A., James, A. L., Jeff, J., Johansen, B., Johansson, A., Jolley, J., Juliusdottir, T., Junttila, J., Kho, A. N., Kinnunen, L., Klopp, N., Kocher, T., Kratzer, W., Lichtner, P., Lind, L., Lindstroem, J., Lobbens, S., Lorentzon, M., Lu, Y., Lyssenko, V., Magnusson, P. K., Mahajan, A., Maillard, M., McArdle, W. L., McKenzie, C. A., McLachlan, S., McLaren, P. J., Menni, C., Merger, S., Milani, L., Moayyeri, A., Monda, K. L., Morken, M. A., Mueller, G., Mueller-Nurasyid, M., Musk, A. W., Narisu, N., Nauck, M., Nolte, I. M., Noethen, M. M., Oozageer, L., Pilz, S., Rayner, N. W., Renstrom, F., Robertson, N. R., Rose, L. M., Roussel, R., Sanna, S., Scharnagl, H., Scholtens, S., Schumacher, F. R., Schunkert, H., Scott, R. A., Sehmi, J., Seufferlein, T., Shin, J., Silventoinen, K., Smit, J. H., Smith, A. V., Smolonska, J., Stanton, A. V., Stirrups, K., Stott, D. J., Stringham, H. M., Sundstrom, J., Swertz, M. A., Syvanen, A., Tayo, B. O., Thorleifsson, G., Tyrer, J. P., Van Dijk, S., van Schoor, N. M., van der Velde, N., van Heemst, D., van Oort, F. V., Vermeulen, S. H., Verweij, N., Vonk, J. M., Waite, L. L., Waldenberger, M., Wennauer, R., Wilkens, L. R., Willenborg, C., Wilsgaard, T., Wojczynski, M. K., Wong, A., Wright, A. F., Zhang, Q., Arveiler, D., Bakker, S. J., Beilby, J., Bergman, R. N., Bergmann, S., Biffar, R., Blangero, J., Boomsma, D. I., Bornstein, S. R., Bovet, P., Brambilla, P., Brown, M. J., Campbell, H., Caulfield, M. J., Chakravarti, A., Collins, R., Collins, F. S., Crawford, D. C., Cupples, L. A., Danesh, J., de Faire, U., Den Ruijter, H. M., Erbel, R., Erdmann, J., Eriksson, J. G., Farrall, M., Ferrannini, E., Ferrieres, J., Ford, I., Forouhi, N. G., Forrester, T., Gansevoort, R. T., Gejman, P. V., Gieger, C., Golay, A., Gottesman, O., Gudnason, V., Gyllensten, U., Haas, D. W., Hall, A. S., Harris, T. B., Hattersley, A. T., Heath, A. C., Hengstenberg, C., Hicks, A. A., Hindorff, L. A., Hingorani, A. D., Hofman, A., Hovingh, G. K., Humphries, S. E., Hunt, S. C., Hypponen, E., Jacobs, K. B., Jarvelin, M., Jousilahti, P., Jula, A. M., Kaprio, J., Kastelein, J. J., Kayser, M., Kee, F., Keinanen-Kiukaanniemi, S. M., Kiemeney, L. A., Kooner, J. S., Kooperberg, C., Koskinen, S., Kovacs, P., Kraja, A. T., Kumari, M., Kuusisto, J., Lakka, T. A., Langenberg, C., Le Marchand, L., Lehtimaki, T., Lupoli, S., Madden, P. A., Mannisto, S., Manunta, P., Marette, A., Matise, T. C., McKnight, B., Meitinger, T., Moll, F. L., Montgomery, G. W., Morris, A. D., Morris, A. P., Murray, J. C., Nelis, M., Ohlsson, C., Oldehinkel, A. J., Ong, K. K., Ouwehand, W. H., Pasterkamp, G., Peters, A., Pramstaller, P. P., Price, J. F., Qi, L., Raitakari, O. T., Rankinen, T., Rao, D. C., Rice, T. K., Ritchie, M., Rudan, I., Salomaa, V., Samani, N. J., Saramies, J., Sarzynski, M. A., Schwarz, P. E., Sebert, S., Sever, P., Shuldiner, A. R., Sinisalo, J., Steinthorsdottir, V., Stolk, R. P., Tardif, J., Toenjes, A., Tremblay, A., Tremoli, E., Virtamo, J., Vohl, M., Amouyel, P., Asselbergs, F. W., Assimes, T. L., Bochud, M., Boehm, B. O., Boerwinkle, E., Bottinger, E. P., Bouchard, C., Cauchi, S., Chambers, J. C., Chanock, S. J., Cooper, R. S., de Bakker, P. I., Dedoussis, G., Ferrucci, L., Franks, P. W., Froguel, P., Groop, L. C., Haiman, C. A., Hamsten, A., Hayes, M. G., Hui, J., Hunter, D. J., Hveem, K., Jukema, J. W., Kaplan, R. C., Kivimaki, M., Kuh, D., Laakso, M., Liu, Y., Martin, N. G., Maerz, W., Melbye, M., Moebus, S., Munroe, P. B., Njolstad, I., Oostra, B. A., Palmer, C. N., Pedersen, N. L., Perola, M., Perusse, L., Peters, U., Powell, J. E., Power, C., Quertermous, T., Rauramaa, R., Reinmaa, E., Ridker, P. M., Rivadeneira, F., Rotter, J. I., Saaristo, T. E., Saleheen, D., Schlessinger, D., Slagboom, P. E., Snieder, H., Spector, T. D., Strauch, K., Stumvoll, M., Tuomilehto, J., Uusitupa, M., van der Harst, P., Voelzke, H., Walker, M., Wareham, N. J., Watkins, H., Wichmann, H., Wilson, J. F., Zanen, P., Deloukas, P., Heid, I. M., Lindgren, C. M., Mohlke, K. L., Speliotes, E. K., Thorsteinsdottir, U., Barroso, I., Fox, C. S., North, K. E., Strachan, D. P., Beckmann, J. S., Berndt, S. I., Boehnke, M., Borecki, I. B., McCarthy, M. I., Metspalu, A., Stefansson, K., Uitterlinden, A. G., van Duijn, C. M., Franke, L., Willer, C. J., Price, A. L., Lettre, G., Loos, R. J., Weedon, M. N., Ingelsson, E., O'Connell, J. R., Abecasis, G. R., Chasman, D. I., Goddard, M. E., Visscher, P. M., Hirschhorn, J. N., Frayling, T. M. 2014; 46 (11): 1173-1186

    Abstract

    Using genome-wide data from 253,288 individuals, we identified 697 variants at genome-wide significance that together explained one-fifth of the heritability for adult height. By testing different numbers of variants in independent studies, we show that the most strongly associated ∼2,000, ∼3,700 and ∼9,500 SNPs explained ∼21%, ∼24% and ∼29% of phenotypic variance. Furthermore, all common variants together captured 60% of heritability. The 697 variants clustered in 423 loci were enriched for genes, pathways and tissue types known to be involved in growth and together implicated genes and pathways not highlighted in earlier efforts, such as signaling by fibroblast growth factors, WNT/β-catenin and chondroitin sulfate-related genes. We identified several genes and pathways not previously connected with human skeletal growth, including mTOR, osteoglycin and binding of hyaluronic acid. Our results indicate a genetic architecture for human height that is characterized by a very large but finite number (thousands) of causal variants.

    View details for DOI 10.1038/ng.3097

    View details for Web of Science ID 000344131900008

  • Parent-of-origin-specific allelic associations among 106 genomic loci for age at menarche NATURE Perry, J. R., Day, F., Elks, C. E., Sulem, P., Thompson, D. J., Ferreira, T., He, C., Chasman, D. I., Esko, T., Thorleifsson, G., Albrecht, E., Ang, W. Q., Corre, T., Cousminer, D. L., Feenstra, B., Franceschini, N., Ganna, A., Johnson, A. D., Kjellqvist, S., Lunetta, K. L., McMahon, G., Nolte, I. M., Paternoster, L., Porcu, E., Smith, A. V., Stolk, L., Teumer, A., Tsernikova, N., Tikkanen, E., Ulivi, S., Wagner, E. K., Amin, N., Bierut, L. J., Byrne, E. M., Hottenga, J., Koller, D. L., Mangino, M., Pers, T. H., Yerges-Armstrong, L. M., Zhao, J. H., Andrulis, I. L., Anton-Culver, H., Atsma, F., Bandinelli, S., Beckmann, M. W., Benitez, J., Blomqvist, C., Bojesen, S. E., Bolla, M. K., Bonanni, B., Brauch, H., Brenner, H., Buring, J. E., Chang-Claude, J., Chanock, S., Chen, J., Chenevix-Trench, G., Collee, J. M., Couch, F. J., Couper, D., Coviello, A. D., Cox, A., Czene, K., D'Adamo, A. P., Smith, G. D., De Vivo, I., Demerath, E. W., Dennis, J., Devilee, P., Dieffenbach, A. K., Dunning, A. M., Eiriksdottir, G., Eriksson, J. G., Fasching, P. A., Ferrucci, L., Flesch-Janys, D., Flyger, H., Foroud, T., Franke, L., Garcia, M. E., Garcia-Closas, M., Geller, F., de Geus, E. E., Giles, G. G., Gudbjartsson, D. F., Gudnason, V., Guenel, P., Guo, S., Hall, P., Hamann, U., Haring, R., Hartman, C. A., Heath, A., Hofman, A., Hooning, M. J., Hopper, J. L., Hu, F. B., Hunter, D. J., Karasik, D., Kiel, D. P., Knight, J. A., Kosma, V., Kutalik, Z., Lai, S., Lambrechts, D., Lindblom, A., Maegi, R., Magnusson, P. K., Mannermaa, A., Martin, N. G., Masson, G., McArdle, P. F., McArdle, W. L., Melbye, M., Michailidou, K., Mihailov, E., Milani, L., Milne, R. L., Nevanlinna, H., Neven, P., Nohr, E. A., Oldehinkel, A. J., Oostra, B. A., Palotie, A., Peacock, M., Pedersen, N. L., Peterlongo, P., Peto, J., Pharoah, P. D., Postma, D. S., Pouta, A., Pylkaes, K., Radice, P., Ring, S., Rivadeneira, F., Robino, A., Rose, L. M., Rudolph, A., Salomaa, V., Sanna, S., Schlessinger, D., Schmidt, M. K., Southey, M. C., Sovio, U., Stampfer, M. J., Stoeckl, D., Storniolo, A. M., Timpson, N. J., Tyrer, J., Visser, J. A., Vollenweider, P., Voelzke, H., Waeber, G., Waldenberger, M., Wallaschofski, H., Wang, Q., Willemsen, G., Winqvist, R., Wolffenbuttel, B. H., Wright, M. J., Boomsma, D. I., Econs, M. J., Khaw, K., Loos, R. J., McCarthy, M. I., Montgomery, G. W., Rice, J. P., Streeten, E. A., Thorsteinsdottir, U., van Duijn, C. M., Alizadeh, B. Z., Bergmann, S., Boerwinkle, E., Boyd, H. A., Crisponi, L., Gasparini, P., Gieger, C., Harris, T. B., Ingelsson, E., Jaervelin, M., Kraft, P., Lawlor, D., Metspalu, A., Pennell, C. E., Ridker, P. M., Snieder, H., Sorensen, T. I., Spector, T. D., Strachan, D. P., Uitterlinden, A. G., Wareham, N. J., Widen, E., Zygmunt, M., Murray, A., Easton, D. F., Stefansson, K., Murabito, J. M., Ong, K. K. 2014; 514 (7520): 92-?

    Abstract

    Age at menarche is a marker of timing of puberty in females. It varies widely between individuals, is a heritable trait and is associated with risks for obesity, type 2 diabetes, cardiovascular disease, breast cancer and all-cause mortality. Studies of rare human disorders of puberty and animal models point to a complex hypothalamic-pituitary-hormonal regulation, but the mechanisms that determine pubertal timing and underlie its links to disease risk remain unclear. Here, using genome-wide and custom-genotyping arrays in up to 182,416 women of European descent from 57 studies, we found robust evidence (P < 5 × 10(-8)) for 123 signals at 106 genomic loci associated with age at menarche. Many loci were associated with other pubertal traits in both sexes, and there was substantial overlap with genes implicated in body mass index and various diseases, including rare disorders of puberty. Menarche signals were enriched in imprinted regions, with three loci (DLK1-WDR25, MKRN3-MAGEL2 and KCNK9) demonstrating parent-of-origin-specific associations concordant with known parental expression patterns. Pathway analyses implicated nuclear hormone receptors, particularly retinoic acid and γ-aminobutyric acid-B2 receptor signalling, among novel mechanisms that regulate pubertal timing in humans. Our findings suggest a genetic architecture involving at least hundreds of common variants in the coordinated timing of the pubertal transition.

    View details for DOI 10.1038/nature13545

    View details for Web of Science ID 000342420800042

  • Persistent organic pollutants and liver dysfunction biomarkers in a population-based human sample of men and women ENVIRONMENTAL RESEARCH Kumar, J., Lind, L., Salihovic, S., van Bavel, B., Ingelsson, E., Lind, P. M. 2014; 134: 251-256

    Abstract

    Persistent organic pollutants (POPs) are stable organic compounds generated through different industrial activities. Liver is involved in the metabolism of POPs, and hence exposure to POPs may interfere with liver function. Although a few studies have shown adverse effects of POPs on liver function, large-scale studies involving humans are lacking. We performed this large population-based cross-sectional study to assess the associations between different POPs and liver dysfunction biomarkers.A total of 992 individuals (all aged 70 years, 50% males) were recruited as part of Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) cohort. The total toxic equivalency (TEQ) value was calculated for seven mono-ortho and two non-ortho substituted polychlorinated biphenyls (PCBs) and octachloro-p-dibenzodioxin (OCDD) to assess their toxicological effects. The association of TEQ values, summary measures of 16 PCBs (sum of PCBs) and three organochlorine pesticides (sum of OC pesticides) with liver dysfunction biomarkers (bilirubin; alkaline phosphatase, ALP; alanine aminotransferase, ALT; and gamma-glutamyltransferase, GGT) was analyzed utilizing linear regression analysis.The mono-ortho PCB TEQ values were found to be significantly positively associated with bilirubin (β=0.71, P=0.008), while sum of OC pesticide concentrations was negatively associated with ALP (β=-0.02, P=0.002) after adjusting for various potential confounders. When analyzed individually, a number of different POPs were associated with ALP, ALT and bilirubin. No such association with GGT was observed.Various POPs including PCBs, OCDD and pesticides were associated with the liver dysfunction biomarkers bilirubin, ALT and ALP, suggesting adverse effects on liver function from these environmental pollutants.

    View details for DOI 10.1016/j.envres.2014.07.023

    View details for Web of Science ID 000346817100034

    View details for PubMedID 25173059

  • Influence of persistent organic pollutants on the complement system in a population-based human sample ENVIRONMENT INTERNATIONAL Kumar, J., Lind, P. M., Salihovic, S., van Bavel, B., Ekdahl, K. N., Nilsson, B., Lind, L., Ingelsson, E. 2014; 71: 94-100

    Abstract

    Persistent organic pollutants (POPs) are toxic compounds generated through various industrial activities and have adverse effects on human health. Studies performed in cell cultures and animals have revealed that POPs can alter immune-system functioning. The complement system is part of innate immune system that helps to clear pathogens from the body. We performed a large-scale population-based study to find out associations between summary measures of different POPs and different complement system markers.In this cross-sectional study, 16 polychlorinated biphenyls (PCBs), 3 organochlorine (OC) pesticides, octachloro-p-dibenzodioxin, and 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) were analyzed for their association with levels of protein complement 3 (C3), 3a (C3a), 4 (C4) and C3a/C3 ratio. A total of 992 individuals (all aged 70 years, 50% females) were recruited from the Prospective Investigation of the Vasculature in Uppsala Seniors cohort. Regression analysis adjusting for a variety of confounders was performed to study the associations of different POP exposures (total toxic equivalency value or TEQ and sum of 16 PCBs) with protein complements.The TEQ values were found to be positively associated with C3a (β=0.07, 95% CI=0.017-0.131, p=0.01) and C3a/C3 ratio (β=0.07, 95% CI=0.015-0.126, p=0.01) taking possible confounders into account. The association observed was mainly driven by PCB-126.In this study involving 992 elderly individuals from the general population, we showed that POPs, mainly PCB-126, were associated with levels of complement system markers indicating that the association of these toxic compounds with downstream disease could be mediated by activation of immune system.

    View details for DOI 10.1016/j.envint.2014.06.009

    View details for Web of Science ID 000341745100011

    View details for PubMedID 24996157

  • Association of vitamin D status with arterial blood pressure and hypertension risk: a mendelian randomisation study LANCET DIABETES & ENDOCRINOLOGY Vimaleswaran, K. S., Cavadino, A., Berry, D. J., Jorde, R., Dieffenbach, A. K., Lu, C., Alves, A. C., Heerspink, H. J., Tikkanen, E., Eriksson, J., Wong, A., Mangino, M., Jablonski, K. A., Nolte, I. M., Houston, D. K., Ahluwalia, T. S., van der Most, P. J., Pasko, D., Zgaga, L., Thiering, E., Vitart, V., Fraser, R. M., Huffman, J. E., de Boer, R. A., Schoettker, B., Saum, K., McCarthy, M. I., Dupuis, J., Herzig, K., Sebert, S., Pouta, A., Laitinen, J., Kleber, M. E., Navis, G., Lorentzon, M., Jameson, K., Arden, N., Cooper, J. A., Acharya, J., Hardy, R., Raitakari, O., Ripatti, S., Billings, L. K., Lahti, J., Osmond, C., Penninx, B. W., Rejnmark, L., Lohman, K. K., Paternoster, L., Stolk, R. P., Hernandez, D. G., Byberg, L., Hagstrom, E., Melhus, H., Ingelsson, E., Mellstroem, D., Ljunggren, O., Tzoulaki, I., McLachlan, S., Theodoratou, E., Tiesler, C. M., Jula, A., Navarro, P., Wright, A. F., Polasek, O., Hayward, C., Wilson, J. F., Rudan, I., Salomaa, V., Heinrich, J., Campbell, H., Price, J. F., Karlsson, M., Lind, L., Michaesson, K., Bandinelli, S., Frayling, T. M., Hartman, C. A., Sorensen, T. I., Kritchevsky, S. B., Langdahl, B. L., Eriksson, J. G., Florez, J. C., Spector, T. D., Lehtimaki, T., Kuh, D., Humphries, S. E., Cooper, C., Ohlsson, C., Maerz, W., de Borst, M. H., Kumari, M., Kivimaki, M., Wang, T. J., Power, C., Brenner, H., Grimnes, G., van der Harst, P., Snieder, H., Hingorani, A. D., Pilz, S., Whittaker, J. C., Jarvelin, M., Hypponen, E. 2014; 2 (9): 719-729

    Abstract

    Low plasma 25-hydroxyvitamin D (25[OH]D) concentration is associated with high arterial blood pressure and hypertension risk, but whether this association is causal is unknown. We used a mendelian randomisation approach to test whether 25(OH)D concentration is causally associated with blood pressure and hypertension risk.In this mendelian randomisation study, we generated an allele score (25[OH]D synthesis score) based on variants of genes that affect 25(OH)D synthesis or substrate availability (CYP2R1 and DHCR7), which we used as a proxy for 25(OH)D concentration. We meta-analysed data for up to 108 173 individuals from 35 studies in the D-CarDia collaboration to investigate associations between the allele score and blood pressure measurements. We complemented these analyses with previously published summary statistics from the International Consortium on Blood Pressure (ICBP), the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium, and the Global Blood Pressure Genetics (Global BPGen) consortium.In phenotypic analyses (up to n=49 363), increased 25(OH)D concentration was associated with decreased systolic blood pressure (β per 10% increase, -0·12 mm Hg, 95% CI -0·20 to -0·04; p=0·003) and reduced odds of hypertension (odds ratio [OR] 0·98, 95% CI 0·97-0·99; p=0·0003), but not with decreased diastolic blood pressure (β per 10% increase, -0·02 mm Hg, -0·08 to 0·03; p=0·37). In meta-analyses in which we combined data from D-CarDia and the ICBP (n=146 581, after exclusion of overlapping studies), each 25(OH)D-increasing allele of the synthesis score was associated with a change of -0·10 mm Hg in systolic blood pressure (-0·21 to -0·0001; p=0·0498) and a change of -0·08 mm Hg in diastolic blood pressure (-0·15 to -0·02; p=0·01). When D-CarDia and consortia data for hypertension were meta-analysed together (n=142 255), the synthesis score was associated with a reduced odds of hypertension (OR per allele, 0·98, 0·96-0·99; p=0·001). In instrumental variable analysis, each 10% increase in genetically instrumented 25(OH)D concentration was associated with a change of -0·29 mm Hg in diastolic blood pressure (-0·52 to -0·07; p=0·01), a change of -0·37 mm Hg in systolic blood pressure (-0·73 to 0·003; p=0·052), and an 8·1% decreased odds of hypertension (OR 0·92, 0·87-0·97; p=0·002).Increased plasma concentrations of 25(OH)D might reduce the risk of hypertension. This finding warrants further investigation in an independent, similarly powered study.British Heart Foundation, UK Medical Research Council, and Academy of Finland.

    View details for DOI 10.1016/S2213-8587(14)70113-5

    View details for Web of Science ID 000341800000014

    View details for PubMedID 24974252

  • Dose-Response Relationship of Total and Leisure Time Physical Activity to Risk of Heart Failure A Prospective Cohort Study CIRCULATION-HEART FAILURE Andersen, K., Mariosa, D., Adami, H., Held, C., Ingelsson, E., Lagerros, Y. T., Nyren, O., Ye, W., Bellocco, R., Sundstrom, J. 2014; 7 (5): 701-U37

    Abstract

    The nature of the association between levels of physical activity and risk of heart failure is little known. We investigated nonlinear associations of total and leisure time physical activity with risk of heart failure.In 1997, 39 805 persons without heart failure completed a questionnaire of lifestyle factors and medical history. We used Cox regression models to investigate total (adjusting for education and previous myocardial infarction) and direct (multivariable-adjusted) effects of self-reported total and leisure time physical activity on risk of heart failure of any cause and heart failure of nonischemic origin. Heart failure diagnoses were obtained until December 31, 2010. Higher leisure time physical activity was associated with lower risk of heart failure of any cause; hazard ratio of the total effect of leisure time physical activity was for fifth versus first quintile 0.54; 95% confidence interval was 0.44 to 0.66. The direct effect was similar. High total daily physical activity level was associated with lower risk of heart failure, although the effect was less pronounced than for leisure time physical activity (total effect hazard ratio, 0.81; 95% confidence interval, 0.69-0.95; fifth versus first quintile). A similar direct effect observed.Leisure time physical activity was inversely related to risk of developing heart failure in a dose-response fashion. This was reflected in a similar but less pronounced association of total physical activity with risk of heart failure. Only part of the effects appeared to be mediated by traditional risk factors.

    View details for DOI 10.1161/CIRCHEARTFAILURE.113.001010

    View details for Web of Science ID 000342490900003

    View details for PubMedID 25185250

  • Persistent Organic Pollutants and Inflammatory Markers in a Cross-Sectional Study of Elderly Swedish People: The PIVUS Cohort ENVIRONMENTAL HEALTH PERSPECTIVES Kumar, J., Lind, M., Salihovic, S., van Bavel, B., Ingelsson, E., Lind, L. 2014; 122 (9): 977-983

    Abstract

    Persistent organic pollutants (POPs) are compounds that are generated through various industrial activities and released in the surrounding environment. Different animal studies have shown effects of different POPs on various inflammatory markers.Because very few studies have been conducted in humans, we assessed the associations between different POPs and inflammatory markers in a large population-based sample of elderly men and women (all 70 years of age) from Sweden.This cross-sectional study investigated the concentrations of several polychlorinated biphenyls (PCBs), organochlorine pesticides, polychlorinated dibenzo-p-dioxin, and brominated diphenyl ether congeners and their association with a number of inflammatory markers [vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), E-selectin, C-reactive protein (CRP), total leucocyte count, tumor necrosis factor α (TNF-α), monocyte chemotactic protein 1 (MCP-1), and interleukin 6 (IL-6)] in 992 individuals. These individuals were recruited from the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) cohort. We used a total toxic equivalency (TEQ) value that measures toxicological effects with the relative potencies of various POPs.Following adjustment for potential confounders, the TEQ value (driven mainly by PCB-126) was significantly associated with levels of ICAM-1 (p < 10-5). A similar trend was also observed between sum of PCBs and VCAM-1 (p < 0.001). No significant associations were observed between levels of POPs and other inflammatory markers.TEQ values were associated with levels of ICAM-1, to a lesser degree also with VCAM-1, but not with CRP and several other inflammatory markers. These findings suggest an activation of vascular adhesion molecules by POPs, and particularly by PCB-126.

    View details for DOI 10.1289/ehp.1307613

    View details for Web of Science ID 000341714600027

    View details for PubMedID 24911359

  • Temporal Trends in Incidence of Myocardial Infarction and Ischemic Stroke by Socioeconomic Position in Sweden 1987-2010 PLOS ONE Malki, N., Koupil, I., Eloranta, S., Weibull, C. E., Tiikkaja, S., Ingelsson, E., Sparen, P. 2014; 9 (8)

    Abstract

    We analyzed temporal trends in the incidence of myocardial infarction and ischemic stroke in Sweden by socioeconomic position and investigated whether social inequalities in incidence of these diseases changed over time.We studied a cohort of almost three million Swedish residents born between 1932 and 1960 followed from 1987 until 2010. Incident cases of myocardial infarction and ischemic stroke were identified in the Swedish National Inpatient Register and Cause of Death Register. Socioeconomic position was retrieved from the Population and Housing Censuses. Incidence rates of myocardial infarction and ischemic stroke and incidence rate ratios comparing levels of socioeconomic position were estimated using flexible parametric survival models adjusted for calendar year, attained age, sex, and birth country.The overall incidences of myocardial infarction and ischemic stroke decreased over time among men, but were stable over time among women. With regard to ischemic stroke incidence, socioeconomic inequality increased over time in the age group 55 to 59: the incidence rate ratios for low manual compared to high non-manual increased from 1.3 (95% CI: 1.2-1.4) in 1997 to 1.5 (1.4-1.7) in 2010 among men, and from 1.4 (1.3-1.6) in 1997 to 2.1 (1.8-2.5) in 2010 among women. The socioeconomic inequality in incidence of myocardial infarction was stable over time for both men and women.There was a decrease in myocardial infarction and ischemic stroke incidence over time among men but no significant change for women. Our study highlights existing, and in some cases increasing, social inequalities in the incidence of cardiovascular diseases.

    View details for DOI 10.1371/journal.pone.0105279

    View details for Web of Science ID 000341127500040

    View details for PubMedID 25170919

  • Influence of a prudent diet on circulating cathepsin S in humans NUTRITION JOURNAL Jobs, E., Adamsson, V., Larsson, A., Jobs, M., Nerpin, E., Ingelsson, E., Arnlov, J., Riserus, U. 2014; 13

    Abstract

    Increased circulating cathepsin S levels have been linked to increased risk of cardiometabolic diseases and cancer. However, whether cathepsin S is a modifiable risk factor is unclear. We aimed to investigate the effects of a prudent diet on plasma cathepsin S levels in healthy individuals.Explorative analyses of a randomized study were performed in 88 normal to slightly overweight and hyperlipidemic men and women (aged 25 to 65) that were randomly assigned to ad libitum prudent diet, i.e. healthy Nordic diet (ND) or a control group (habitual Western diet) for 6 weeks. Whereas all foods in the ND were provided, the control group was advised to consume their habitual diet throughout the study. The ND was in line with dietary recommendations, e.g. low in saturated fats, sugars and salt, but high in plant-based foods rich in fibre and unsaturated fats.The ND significantly decreased cathepsin S levels (from 20.1 (+/-4.0 SD) to 19.7 μg/L (+/-4.3 SD)) compared with control group (from 18.2 (+/-2.9 SD) to 19.1 μg/L (+/-3.8 SD)). This difference remained after adjusting for sex and change in insulin sensitivity (P = 0.03), and near significant after adjusting for baseline cathepsin S levels (P = 0.06), but not for change in weight or LDL-C. Changes in cathepsin S levels were directly correlated with change in LDL-C.Compared with a habitual control diet, a provided ad libitum healthy Nordic diet decreased cathepsin S levels in healthy individuals, possibly mediated by weight loss or lowered LDL-C. These differences between groups in cathepsin S were however not robust and therefore need further investigation.

    View details for DOI 10.1186/1475-2891-13-84

    View details for Web of Science ID 000341112000001

    View details for PubMedID 25128296

  • Urinary Kidney Injury Molecule-1 and the Risk of Cardiovascular Mortality in Elderly Men CLINICAL JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Carlsson, A. C., Larsson, A., Helmersson-Karlqvist, J., Lind, L., Ingelsson, E., Larsson, T. E., Bottai, M., Sundstrom, J., Arnlov, J. 2014; 9 (8): 1393-1401

    Abstract

    Kidney injury molecule-1 (KIM-1) has been suggested as a clinically relevant highly specific biomarker of acute kidney tubular damage. However, community-based data on the association between urinary levels of KIM-1 and the risk for cardiovascular mortality are lacking. This study aimed to investigate the association between urinary KIM-1 and cardiovascular mortality.This was a prospective study, using the community-based Uppsala Longitudinal Study of Adult Men (N=590; mean age 77 years; baseline period, 1997-2001; median follow-up 8.1 years; end of follow-up, 2008).During follow-up, 89 participants died of cardiovascular causes (incidence rate, 2.07 per 100 person-years at risk). Models were adjusted for cardiovascular risk factors (age, systolic BP, diabetes, smoking, body mass index, total cholesterol, HDL cholesterol, antihypertensive treatment, lipid-lowering treatment, aspirin treatment, and history of cardiovascular disease) and for markers of kidney dysfunction and damage (cystatin C-based eGFR and urinary albumin/creatinine ratio). Higher urinary KIM-1/creatinine (from 24-hour urine collections) was associated with a higher risk for cardiovascular mortality (hazard ratio per SD increase, 1.27; 95% confidence interval [95% CI], 1.05 to 1.54; P=0.01). Participants with a combination of high KIM-1/creatinine (upper quintile, ≥175 ng/mmol), low eGFR (≤60 ml/min per 1.73 m(2)), and microalbuminuria/macroalbuminuria (albumin/creatinine ratio≥3 g/mol) had a >8-fold increased risk compared with participants with low KIM-1/creatinine (<175 ng/mmol), normal eGFR (>60 ml/min per 1.73 m(2)), and normoalbuminuria (albumin/creatinine ratio<3 g/mol) (hazard ratio, 8.56; 95% CI, 4.17 to 17.56; P<0.001).These findings suggest that higher urinary KIM-1 may predispose to a higher risk of cardiovascular mortality independently of established cardiovascular risk factors, eGFR, and albuminuria. Additional studies are needed to further assess the utility of measuring KIM-1 in the clinical setting.

    View details for DOI 10.2215/CJN.11901113

    View details for Web of Science ID 000339984100010

    View details for PubMedID 24923577

  • Genetic association study of QT interval highlights role for calcium signaling pathways in myocardial repolarization NATURE GENETICS Arking, D. E., Pulit, S. L., Crotti, L., van der Harst, P., Munroe, P. B., Koopmann, T. T., Sotoodehnia, N., Rossin, E. J., Morley, M., Wang, X., Johnson, A. D., Lundby, A., Gudbjartsson, D. F., Noseworthy, P. A., Eijgelsheim, M., Bradford, Y., Tarasov, K. V., Dorr, M., Miiller-Nurasyid, M., Lahtinen, A. M., Nolte, I. M., Smith, A. V., Bis, J. C., Isaacs, A., Newhouse, S. J., Evans, D. S., Post, W. S., Waggott, D., Lyytikainen, L., Hicks, A. A., Eisele, L., Ellinghaus, D., Hayward, C., Navarro, P., Ulivi, S., Tanaka, T., Tester, D. J., Chatel, S., Gustafsson, S., Kumari, M., Morris, R. W., Naluai, A. T., Padmanabhan, S., Kluttig, A., Strohmer, B., Panayiotou, A. G., Torres, M., Knoflach, M., Hubacek, J. A., Slowikowski, K., Raychaudhuri, S., Kumar, R. D., Harris, T. B., Launer, L. J., Shuldiner, A. R., Alonso, A., Bader, J. S., Ehret, G., Huang, H., Kao, W. H., Strait, J. B., Macfarlane, P. W., Brown, M., Caulfield, M. J., Samani, N. J., Kronenberg, F., Willeit, J., Smith, J. G., Greiser, K. H., Schwabedissen, H. M., Werdan, K., Carella, M., Zelante, L., Heckbert, S. R., Psaty, B. M., Rotter, J. I., Kolcic, I., Poagek, O., Wright, A. F., Griffin, M., Daly, M. J., Arnar, D. O., Holm, H., Thorsteinsdottir, U., Denny, J. C., Roden, D. M., Zuvich, R. L., Emilsson, V., Plump, A. S., Larson, M. G., O'Donnell, C. J., Yin, X., Bobboll, M., d'Adamo, A. P., Iorio, A., Sinagra, G., Carracedo, A., Cummings, S. R., Nalls, M. A., Jula, A., Kontula, K. K., Marjamaa, A., Oikarinen, L., Perola, M., Porthan, K., Erbe, R., Hoffmann, P., Jockel, K., Kalsch, H., Nothen, M. M., Den Hoed, M., Loos, R. J., Thelle, D. S., Gieger, C., Meitinger, T., Perz, S., Peters, A., Pruchal, H., Sinner, M. F., Waldenberger, M., de Boer, R. A., Franke, L., van der Vleuten, P. A., Beckmann, B. M., Martens, E., Bardail, A., Hofman, N., Wilde, A. A., Behr, E. R., Dalageorgou, C., Giudicessi, J. R., Medeiros-Domingo, A., Barc, J., Kyndt, F., Probst, V., Ghidoni, A., Insolia, R., Hamilton, R. M., Scherer, S. W., Brandimarto, J., Margulies, K., Moravec, C. E., Del Greco, F., Fuchsberger, C., O'Connell, J. R., Lee, W. K., Watt, G. C., Campbell, H., Wild, S. H., El Mokhtari, N. E., Frey, N., Asselbergs, F. W., Leach, I. M., Navis, G., Van den Berg, M. P., van Veldhuisen, D. J., Kellis, M., Krijthe, B. P., Franco, O. H., Hofman, A., Kors, J. A., Uitterlinden, A. G., Witteman, J. C., Kedenko, L., Lamina, C., Oostra, B. A., Abecasis, G. R., Lakatta, E. G., Mulas, A., Orru, M., Schlessinger, D., Uda, M., Markus, M. R., Volker, U., Snieder, H., Spector, T. D., Arnlov, J., Lind, L., Sundstrom, J., Syvanen, A., Kivimaki, M., Kahonen, M., Mononen, N., Raitakari, I. T., Viikari, J. S., Adamkova, V., Kiech, S., Brion, M., Nicolaides, A. N., Paulweber, B., Haerting, J., Dominiczak, A. F., Nyberg, F., Whincup, P. H., Hingorani, A. D., Schott, J., Bezzina, C. R., Ingelsson, E., Ferrucci, L., Gaspariniin, P., Wilson, J. F., Rudan, I., Franke, A., Miihleisen, T. W., Pramstaller, P. P., Lehtimaki, T. J., Paterson, A. D., Parsa, A., Liu, Y., van Duijn, C. M., Siscovick, D. S., Gudnason, V., Jamshidi, Y., Salomaa, V., Felix, S. B., Sanna, S., Ritchie, M. D., Stricker, B. H., Stefansson, K., Boyer, L. A., Cappola, T. P., Olsen, J. V., Lage, K., Schwartz, P. J., Kaab, S., Chakravarti, A., Ackerman, M. J., Pfeufer, A., de Bakker, P. I., Newton-Cheh, C. 2014; 46 (8): 826-836

    Abstract

    The QT interval, an electrocardiographic measure reflecting myocardial repolarization, is a heritable trait. QT prolongation is a risk factor for ventricular arrhythmias and sudden cardiac death (SCD) and could indicate the presence of the potentially lethal mendelian long-QT syndrome (LQTS). Using a genome-wide association and replication study in up to 100,000 individuals, we identified 35 common variant loci associated with QT interval that collectively explain ∼8-10% of QT-interval variation and highlight the importance of calcium regulation in myocardial repolarization. Rare variant analysis of 6 new QT interval-associated loci in 298 unrelated probands with LQTS identified coding variants not found in controls but of uncertain causality and therefore requiring validation. Several newly identified loci encode proteins that physically interact with other recognized repolarization proteins. Our integration of common variant association, expression and orthogonal protein-protein interaction screens provides new insights into cardiac electrophysiology and identifies new candidate genes for ventricular arrhythmias, LQTS and SCD.

    View details for DOI 10.1038/ng.3014

    View details for Web of Science ID 000339704400010

    View details for PubMedID 24952745

  • Genetic variation in the CYP1A1 gene is related to circulating PCB118 levels in a population-based sample ENVIRONMENTAL RESEARCH Lind, L., Penell, J., Syvaenen, A., Axelsson, T., Ingelsson, E., Morris, A. P., Lindgren, C., Salihovic, S., van Bavel, B., Lind, P. M. 2014; 133: 135-140

    Abstract

    Several of the polychlorinated biphenyls (PCBs), i.e. the dioxin-like PCBs, are known to induce the P450 enzymes CYP1A1, CYP1A2 and CYP1B1 by activating the aryl hydrocarbon receptor (Ah)-receptor. We evaluated if circulating levels of PCBs in a population sample were related to genetic variation in the genes encoding these CYPs. In the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (1016 subjects all aged 70), 21 SNPs in the CYP1A1, CYP1A2 and CYP1B1 genes were genotyped. Sixteen PCB congeners were analysed by high-resolution chromatography coupled to high-resolution mass spectrometry (HRGC/ HRMS). Of the investigated relationships between SNPs in the CYP1A1, CYP1A2 and CYP1B1 and six PCBs (congeners 118, 126, 156, 169, 170 and 206) that captures >80% of the variation of all PCBs measured, only the relationship between CYP1A1 rs2470893 was significantly related to PCB118 levels following strict adjustment for multiple testing (p=0.00011). However, there were several additional SNPs in the CYP1A2 and CYP1B1 that showed nominally significant associations with PCB118 levels (p-values in the 0.003-0.05 range). Further, several SNPs in the CYP1B1 gene were related to both PCB156 and PCB206 with p-values in the 0.005-0.05 range. Very few associations with p<0.05 were seen for PCB126, PCB169 or PCB170. Genetic variation in the CYP1A1 was related to circulating PCB118 levels in the general elderly population. Genetic variation in CYP1A2 and CYP1B1 might also be associated with other PCBs.

    View details for DOI 10.1016/j.envres.2014.05.017

    View details for Web of Science ID 000339705900018

    View details for PubMedID 24926919

  • Genome-wide association analysis identifies six new loci associated with forced vital capacity NATURE GENETICS Loth, D. W., Artigas, M. S., Gharib, S. A., Wain, L. V., Franceschini, N., Koch, B., Pottinger, T. D., Smith, A. V., Duan, Q., Oldmeadow, C., Lee, M. K., Strachan, D. P., James, A. L., Huffman, J. E., Vitart, V., Ramasamy, A., Wareham, N. J., Kaprio, J., Wang, X., Trochet, H., Kaonen, M., Flexeder, C., Albrecht, E., Lopez, L. M., de Jong, K., Thyagarajan, B., Alves, A. C., Enroth, S., Omenaas, E., Joshi, P. K., Fall, T., Vinuela, A., Launer, L. J., Loehr, L. R., Fornage, M., Li, G., Wik, J. B., Tang, W., Manichaikul, A., Lahousse, L., Harris, T. B., North, K. E., Rudnicka, A. R., Hui, J., Gu, X., Lumley, T., Wright, A. F., Hastie, N. D., Campbell, S., Kumar, R., Pin, I., Scott, R. A., Pietilainen, K. H., Surakka, I., Liu, Y., Holliday, E. G., Schulz, H., Heinrich, J., Davies, G., Vonk, J. M., Wojczynski, M., Pouta, A., Johansson, A., Wild, S. H., Ingelsson, E., Rivadeneira, F., Voezke, H., Hysi, P. G., Eiriksdottir, G., Morrison, A. C., Rotter, J. I., Gao, W., Postma, D. S., White, W. B., Rich, S. S., Hofman, A., Aspelund, T., Couper, D., Smith, L. J., Psaty, B. M., Lohman, K., Burchard, E. G., Uitterlinden, A. G., Garcia, M., Joubert, B. R., McArdle, W. L., Musk, A. B., Hansel, N., Heckbert, S. R., Zgaga, L., van Meurs, J. B., Navarro, P., Rudan, I., Oh, Y., Redline, S., Jarvis, D. L., Zhao, J. H., Rantanen, T., O'Connor, G. T., Ripatti, S., Scott, R. J., Karrasch, S., Grallert, H., Gaddis, N. C., Starr, J. M., Wijmenga, C., Minster, R. L., Lederer, D. J., Pekkanen, J., Gyllensten, U., Campbe, H., Morris, A. P., Glaeser, S., Hammond, C. J., Burkart, K. M., Beilby, J., Kritchevsky, S. B., Gucinason, V., Hancock, D. B., Williams, D., Polasek, O., Zemunik, T., Kolcic, I., Petrini, M. F., Wjst, M., Kim, W. J., Porteous, D. J., Scotland, G., Smith, B. H., Villanen, A., Heliovaara, M., Attia, J. R., Sayers, I., Hampel, R., Gieger, C., Deary, I. J., Boezen, H. M., Newman, A., Jarvelin, M., Wilson, J. F., Lind, L., Stricker, B. H., Teumer, A., Spector, T. D., Melen, E., Peters, M. J., Lange, L. A., Barr, R. G., Bracke, K. R., Verhamme, F. M., Sung, J., Hiemstra, P. S., Cassano, P. A., Sood, A., Hayward, C., Dupuis, J., Hall, I. P., Brusselle, G. G., Tobin, M. D., London, S. J. 2014; 46 (7): 669-677

    Abstract

    Forced vital capacity (FVC), a spirometric measure of pulmonary function, reflects lung volume and is used to diagnose and monitor lung diseases. We performed genome-wide association study meta-analysis of FVC in 52,253 individuals from 26 studies and followed up the top associations in 32,917 additional individuals of European ancestry. We found six new regions associated at genome-wide significance (P < 5 × 10(-8)) with FVC in or near EFEMP1, BMP6, MIR129-2-HSD17B12, PRDM11, WWOX and KCNJ2. Two loci previously associated with spirometric measures (GSTCD and PTCH1) were related to FVC. Newly implicated regions were followed up in samples from African-American, Korean, Chinese and Hispanic individuals. We detected transcripts for all six newly implicated genes in human lung tissue. The new loci may inform mechanisms involved in lung development and the pathogenesis of restrictive lung disease.

    View details for DOI 10.1038/ng.3011

    View details for Web of Science ID 000338093800006

    View details for PubMedID 24929828

  • Large-Scale Genome-Wide Association Studies and Meta-Analyses of Longitudinal Change in Adult Lung Function PLOS ONE Tang, W., Kowgier, M., Loth, D. W., Artigas, M. S., Joubert, B. R., Hodge, E., Gharib, S. A., Smith, A. V., Ruczinski, I., Gudnason, V., Mathias, R. A., Harris, T. B., Hansel, N. N., Launer, L. J., Barnes, K. C., Hansen, J. G., Albrecht, E., Aldrich, M. C., Allerhand, M., Barr, R. G., Brusselle, G. G., Couper, D. J., Curjuric, I., Davies, G., Deary, I. J., Dupuis, J., Fall, T., Foy, M., Franceschini, N., Gao, W., Glaeser, S., Gu, X., Hancock, D. B., Heinrich, J., Hofman, A., Imboden, M., Ingelsson, E., James, A., Karrasch, S., Koch, B., Kritchevsky, S. B., Kumar, A., Lahousse, L., Li, G., Lind, L., Lindgren, C., Liu, Y., Lohman, K., Lumley, T., McArdle, W. L., Meibohm, B., Morris, A. P., Morrison, A. C., Musk, B., North, K. E., Palmer, L. J., Probst-Hensch, N. M., Psaty, B. M., Rivadeneira, F., Rotter, J. I., Schulz, H., Smith, L. J., Sood, A., Starr, J. M., Strachan, D. P., Teumer, A., Uitterlinden, A. G., Voelzke, H., Voorman, A., Wain, L. V., Wells, M. T., Wilk, J. B., Williams, O. D., Heckbert, S. R., Stricker, B. H., London, S. J., Fornage, M., Tobin, M. D., O'Connor, G. T., Hall, I. P., Cassano, P. A. 2014; 9 (7)

    Abstract

    Genome-wide association studies (GWAS) have identified numerous loci influencing cross-sectional lung function, but less is known about genes influencing longitudinal change in lung function.We performed GWAS of the rate of change in forced expiratory volume in the first second (FEV1) in 14 longitudinal, population-based cohort studies comprising 27,249 adults of European ancestry using linear mixed effects model and combined cohort-specific results using fixed effect meta-analysis to identify novel genetic loci associated with longitudinal change in lung function. Gene expression analyses were subsequently performed for identified genetic loci. As a secondary aim, we estimated the mean rate of decline in FEV1 by smoking pattern, irrespective of genotypes, across these 14 studies using meta-analysis.The overall meta-analysis produced suggestive evidence for association at the novel IL16/STARD5/TMC3 locus on chromosome 15 (P  =  5.71 × 10(-7)). In addition, meta-analysis using the five cohorts with ≥3 FEV1 measurements per participant identified the novel ME3 locus on chromosome 11 (P  =  2.18 × 10(-8)) at genome-wide significance. Neither locus was associated with FEV1 decline in two additional cohort studies. We confirmed gene expression of IL16, STARD5, and ME3 in multiple lung tissues. Publicly available microarray data confirmed differential expression of all three genes in lung samples from COPD patients compared with controls. Irrespective of genotypes, the combined estimate for FEV1 decline was 26.9, 29.2 and 35.7 mL/year in never, former, and persistent smokers, respectively.In this large-scale GWAS, we identified two novel genetic loci in association with the rate of change in FEV1 that harbor candidate genes with biologically plausible functional links to lung function.

    View details for DOI 10.1371/journal.pone.0100776

    View details for Web of Science ID 000339635000039

    View details for PubMedID 24983941

  • Plasma-Parathyroid Hormone Is Associated With Subclinical and Clinical Atherosclerotic Disease in 2 Community-Based Cohorts ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Hagstrom, E., Michaelsson, K., Melhus, H., Hansen, T., Ahlstrom, H., Johansson, L., Ingelsson, E., Sundstrom, J., Lind, L., Arnlov, J. 2014; 34 (7): 1567-?

    Abstract

    Cardiovascular risk factors have different impact on different arterial territories. Diseases with elevated circulating parathyroid hormone (PTH) such as primary hyperparathyroidism and chronic renal failure have been shown to be associated with an increased risk of cardiovascular disease, predominantly heart or cerebrovascular diseases. However, data on the associations between circulating PTH and peripheral atherosclerosis are limited.Two prospective, community-based studies were used. In 306 men and women, who were 70 years old, from the Prospective investigation of the vasculature in Uppsala seniors (PIVUS) study, cross-sectional relations between PTH and atherosclerotic burden assessed by whole-body magnetic resonance angiography were investigated. In 998 men, who were 71 years old, from the Uppsala longitudinal study of adult men (ULSAM) study, the association between PTH concentration and risk of subsequent nonfatal atherosclerotic disease (excluding coronary or cerebrovascular disease) was investigated. Adjusting for established vascular risk factors, PTH was associated with burden of atherosclerosis (increase in total atherosclerotic score per SD PTH increase: 0.04, 0.003-0.08; P=0.03) in the PIVUS study. During follow-up in the ULSAM study (median 16.7 years), 89 men were diagnosed with nonfatal atherosclerotic disease. In Cox-regression analyses adjusting for established vascular risk factors and mineral metabolism, higher PTH was associated with an increased risk of nonfatal atherosclerotic disease (hazard ratio for 1 SD increase of PTH: 1.55, 1.33-1.88; P<0.0001). Results were similar when including fatal atherosclerotic disease in the outcome.In 2 independent community-based cohorts, PTH was associated to the degree of atherosclerosis and risk of clinically overt atherosclerotic disease, respectively. Our data confirm and extend previous studies supporting a role for PTH in the development of atherosclerotic disease.

    View details for DOI 10.1161/ATVBAHA.113.303062

    View details for Web of Science ID 000337732900033

    View details for PubMedID 24626438

  • Serum selenium in relation to measures of glucose metabolism and incidence of Type 2 diabetes in an older Swedish population DIABETIC MEDICINE Gao, H., Hagg, S., Sjogren, P., Lambert, P. C., Ingelsson, E., van Dam, R. M. 2014; 31 (7): 787-793

    Abstract

    The relation between selenium status and risk of Type 2 diabetes is controversial. We aimed to evaluate associations of serum selenium, a marker of dietary selenium, with measures of glucose metabolism and risk of diabetes.We used data from a population-based, longitudinal cohort of 1925 Swedish men who were 50 years old and did not have diabetes at baseline in the 1970s. At baseline, an intravenous glucose tolerance test was performed and, at a follow-up examination after 20 years, an oral glucose tolerance test and a hyperinsulinaemic euglycaemic clamp for the assessment of insulin sensitivity were conducted.At baseline, the mean (standard deviation) selenium concentration was 75.6 (14.3) μg/l. During 20 years of follow-up, 88 incident cases of diabetes occurred in 1024 participants with follow-up data. Baseline serum selenium levels were not associated with risk of diabetes (odds ratio 1.06; 95% CI 0.83-1.38). Higher selenium levels were associated with lower early insulin response (standardized β -0.08; 95% CI -0.14 to -0.03) at baseline after adjusting for potential confounders, but not with any other measures of β-cell function or insulin sensitivity at baseline or follow-up. The association with early insulin response was non-significant after taking multiple testing into account.Our results do not support a role of dietary selenium in the development of disturbances in glucose metabolism or diabetes in older individuals.

    View details for DOI 10.1111/dme.12429

    View details for Web of Science ID 000337621100006

    View details for PubMedID 24606531

  • Genome Wide Association Identifies Common Variants at the SERPINA6/SERPINA1 Locus Influencing Plasma Cortisol and Corticosteroid Binding Globulin PLOS GENETICS Bolton, J. L., Hayward, C., Direk, N., Lewis, J. G., Hammond, G. L., Hill, L. A., Anderson, A., Huffman, J., Wilson, J. F., Campbell, H., Rudan, I., Wright, A., Hastie, N., Wild, S. H., Velders, F. P., Hofman, A., Uitterlinden, A. G., Lahti, J., Raikkonen, K., Kajantie, E., Widen, E., Palotie, A., Eriksson, J. G., Kaakinen, M., Jarvelin, M., Timpson, N. J., Smith, G. D., Ring, S. M., Evans, D. M., St Pourcain, B., Tanaka, T., Milaneschi, Y., Bandinelli, S., Ferrucci, L., van der Harst, P., Rosmalen, J. G., Bakker, S. J., Verweij, N., Dullaart, R. P., Mahajan, A., Lindgren, C. M., Morris, A., Lind, L., Ingelsson, E., Anderson, L. N., Pennell, C. E., Lye, S. J., Matthews, S. G., Eriksson, J., Mellstrom, D., Ohlsson, C., Price, J. F., Strachan, M. W., Reynolds, R. M., Tiemeier, H., Walker, B. R. 2014; 10 (7)

    Abstract

    Variation in plasma levels of cortisol, an essential hormone in the stress response, is associated in population-based studies with cardio-metabolic, inflammatory and neuro-cognitive traits and diseases. Heritability of plasma cortisol is estimated at 30-60% but no common genetic contribution has been identified. The CORtisol NETwork (CORNET) consortium undertook genome wide association meta-analysis for plasma cortisol in 12,597 Caucasian participants, replicated in 2,795 participants. The results indicate that <1% of variance in plasma cortisol is accounted for by genetic variation in a single region of chromosome 14. This locus spans SERPINA6, encoding corticosteroid binding globulin (CBG, the major cortisol-binding protein in plasma), and SERPINA1, encoding α1-antitrypsin (which inhibits cleavage of the reactive centre loop that releases cortisol from CBG). Three partially independent signals were identified within the region, represented by common SNPs; detailed biochemical investigation in a nested sub-cohort showed all these SNPs were associated with variation in total cortisol binding activity in plasma, but some variants influenced total CBG concentrations while the top hit (rs12589136) influenced the immunoreactivity of the reactive centre loop of CBG. Exome chip and 1000 Genomes imputation analysis of this locus in the CROATIA-Korcula cohort identified missense mutations in SERPINA6 and SERPINA1 that did not account for the effects of common variants. These findings reveal a novel common genetic source of variation in binding of cortisol by CBG, and reinforce the key role of CBG in determining plasma cortisol levels. In turn this genetic variation may contribute to cortisol-associated degenerative diseases.

    View details for DOI 10.1371/journal.pgen.1004474

    View details for Web of Science ID 000339902600028

    View details for PubMedID 25010111

  • Mosaic loss of chromosome Y in peripheral blood is associated with shorter survival and higher risk of cancer NATURE GENETICS Forsberg, L. A., Rasi, C., Malmqvist, N., Davies, H., Pasupulati, S., Pakalapati, G., Sandgren, J., de Stahl, T. D., Zaghlool, A., Giedraitis, V., Lannfelt, L., Score, J., Cross, N. C., Absher, D., Janson, E. T., Lindgren, C. M., Morris, A. P., Ingelsson, E., Lind, L., Dumanski, J. P. 2014; 46 (6): 624-628

    Abstract

    Incidence and mortality for sex-unspecific cancers are higher among men, a fact that is largely unexplained. Furthermore, age-related loss of chromosome Y (LOY) is frequent in normal hematopoietic cells, but the phenotypic consequences of LOY have been elusive. From analysis of 1,153 elderly men, we report that LOY in peripheral blood was associated with risks of all-cause mortality (hazards ratio (HR) = 1.91, 95% confidence interval (CI) = 1.17-3.13; 637 events) and non-hematological cancer mortality (HR = 3.62, 95% CI = 1.56-8.41; 132 events). LOY affected at least 8.2% of the subjects in this cohort, and median survival times among men with LOY were 5.5 years shorter. Association of LOY with risk of all-cause mortality was validated in an independent cohort (HR = 3.66) in which 20.5% of subjects showed LOY. These results illustrate the impact of post-zygotic mosaicism on disease risk, could explain why males are more frequently affected by cancer and suggest that chromosome Y is important in processes beyond sex determination. LOY in blood could become a predictive biomarker of male carcinogenesis.

    View details for DOI 10.1038/ng.2966

    View details for Web of Science ID 000336870700021

    View details for PubMedID 24777449

  • Genetic factors may play a prominent role in the development of coronary heart disease dependent on important environmental factors JOURNAL OF INTERNAL MEDICINE Song, C., Chang, Z., Magnusson, P. K., Ingelsson, E., Pedersen, N. L. 2014; 275 (6): 631-639

    Abstract

    The aim of the study was to examine whether various lifestyle factors modify genetic influences on coronary heart disease (CHD).The effect of lifestyle factors [including smoking, sedentary lifestyle, alcohol intake and body mass index (BMI)] on risk of CHD was evaluated via Cox regression models in a twin study of gene-environment interaction. Using structure equation modelling, we estimated genetic variance of CHD dependent on lifestyle factors.In total, 51 065 same-sex twins from 25 715 twin pairs born before 1958 and registered in the Swedish Twin Registry were eligible for this study. During the 40-year follow-up, 7264 incident CHD events were recorded.Smoking, sedentary lifestyle and above average BMI were significantly associated with increased CHD incidence. The heritability of CHD decreased with increasing age, as well as with increasing levels of BMI, in both men and women.The difference in the genetic component of CHD as a function of BMI suggests that genetic factors may play a more prominent role for disease development in the absence of important environmental factors. Increased knowledge of gene-environment interactions will be important for a full understanding of the aetiology of CHD.

    View details for DOI 10.1111/joim.12177

    View details for Web of Science ID 000337787300010

    View details for PubMedID 24330166

  • Impact of type 2 diabetes susceptibility variants on quantitative glycemic traits reveals mechanistic heterogeneity. Diabetes Dimas, A. S., Lagou, V., Barker, A., Knowles, J. W., Mägi, R., Hivert, M., Benazzo, A., Rybin, D., Jackson, A. U., Stringham, H. M., Song, C., Fischer-Rosinsky, A., Boesgaard, T. W., Grarup, N., Abbasi, F. A., Assimes, T. L., Hao, K., Yang, X., Lecoeur, C., Barroso, I., Bonnycastle, L. L., Böttcher, Y., Bumpstead, S., Chines, P. S., Erdos, M. R., Graessler, J., Kovacs, P., Morken, M. A., Narisu, N., Payne, F., Stancakova, A., Swift, A. J., Tönjes, A., Bornstein, S. R., Cauchi, S., Froguel, P., Meyre, D., Schwarz, P. E., Häring, H., Smith, U., Boehnke, M., Bergman, R. N., Collins, F. S., Mohlke, K. L., Tuomilehto, J., Quertemous, T., Lind, L., Hansen, T., Pedersen, O., Walker, M., Pfeiffer, A. F., Spranger, J., Stumvoll, M., Meigs, J. B., Wareham, N. J., Kuusisto, J., Laakso, M., Langenberg, C., Dupuis, J., Watanabe, R. M., Florez, J. C., Ingelsson, E., McCarthy, M. I., Prokopenko, I. 2014; 63 (6): 2158-2171

    Abstract

    Patients with established type 2 diabetes display both beta-cell dysfunction and insulin resistance. To define fundamental processes leading to the diabetic state, we examined the relationship between type 2 diabetes risk variants at 37 established susceptibility loci and indices of proinsulin processing, insulin secretion and insulin sensitivity. We included data from up to 58,614 non-diabetic subjects with basal measures, and 17,327 with dynamic measures. We employed additive genetic models with adjustment for sex, age and BMI, followed by fixed-effects inverse variance meta-analyses. Cluster analyses grouped risk loci into five major categories based on their relationship to these continuous glycemic phenotypes. The first cluster (PPARG, KLF14, IRS1, GCKR) was characterized by primary effects on insulin sensitivity. The second (MTNR1B, GCK) featured risk alleles associated with reduced insulin secretion and fasting hyperglycemia. ARAP1 constituted a third cluster characterized by defects in insulin processing. A fourth cluster (including TCF7L2, SLC30A8, HHEX/IDE, CDKAL1, CDKN2A/2B) was defined by loci influencing insulin processing and secretion without detectable change in fasting glucose. The final group contained twenty risk loci with no clear-cut associations to continuous glycemic traits. By assembling extensive data on continuous glycemic traits, we have exposed the diverse mechanisms whereby type 2 diabetes risk variants impact disease predisposition.

    View details for DOI 10.2337/db13-0949

    View details for PubMedID 24296717

  • Genetic variation in the CYP2B6 Gene is related to circulating 2,2 ',4,4 '-tetrabromodiphenyl ether (BDE-47) concentrations: an observational population-based study ENVIRONMENTAL HEALTH Penell, J., Lind, L., Fall, T., Syvanen, A., Axelsson, T., Lundmark, P., Morris, A. P., Lindgren, C., Mahajan, A., Salihovic, S., van Bavel, B., Ingelsson, E., Lind, P. M. 2014; 13

    Abstract

    Since human CYP2B6 has been identified as the major CYP enzyme involved in the metabolism of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) and that human 2B6 is a highly polymorphic CYP, with known functional variants, we evaluated if circulating concentrations of a major brominated flame retardant, BDE-47, were related to genetic variation in the CYP2B6 gene in a population sample.In the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (men and women all aged 70), 25 single nucleotide polymorphisms (SNPs) in the CYP2B6 gene were genotyped. Circulating concentrations of BDE-47 were analyzed by high-resolution gas chromatography coupled to high-resolution mass spectrometry (HRGC/ HRMS).Several SNPs in the CYP2B6 gene were associated with circulating concentrations of BDE-47 (P = 10-4 to 10-9). The investigated SNPs came primarily from two haplotypes, although the correlation between the haplotypes was rather high. Conditional analyses adjusting for the SNP with the strongest association with the exposure (rs2014141) did not provide evidence for independent signals.Circulating concentrations of BDE-47 were related to genetic variation in the CYP2B6 gene in an elderly population.

    View details for DOI 10.1186/1476-069X-13-34

    View details for Web of Science ID 000336098900001

    View details for PubMedID 24885815

  • DNA mismatch repair gene MSH6 implicated in determining age at natural menopause HUMAN MOLECULAR GENETICS Perry, J. R., Hsu, Y., Chasman, D. I., Johnson, A. D., Elks, C., Albrecht, E., Andrulis, I. L., Beesley, J., Berenson, G. S., Bergmann, S., Bojesen, S. E., Bolla, M. K., Brown, J., Buring, J. E., Campbell, H., Chang-Claude, J., Chenevix-Trench, G., Corre, T., Couch, F. J., Cox, A., Czene, K., D'Adamo, A. P., Davies, G., Deary, I. J., Dennis, J., Easton, D. F., Engelhardt, E. G., Eriksson, J. G., Esko, T., Fasching, P. A., Figueroa, J. D., Flyger, H., Fraser, A., Garcia-Closas, M., Gasparini, P., Gieger, C., Giles, G., Guenel, P., Haegg, S., Hall, P., Hayward, C., Hopper, J., Ingelsson, E., Kardia, L. R., Kasiman, K., Knight, J. A., Lahti, J., Lawlor, D. A., Magnusson, P. K., Margolin, S., Marsh, J. A., Metspalu, A., Olson, J. E., Pennell, C. E., Polasek, O., Rahman, I., Ridker, P. M., Robino, A., Rudan, I., Rudolph, A., Salumets, A., Schmidt, M. K., Schoemaker, M. J., Smith, E. N., Smith, J. A., Southey, M., Stoeckl, D., Swerdlow, A. J., Thompson, D. J., Truong, T., Ulivi, S., Waldenberger, M., Wang, Q., Wild, S., Wilson, J. F., Wright, A. F., Zgaga, L., Ong, K. K., Murabito, J. M., Karasik, D., Murray, A. 2014; 23 (9): 2490-2497

    Abstract

    The length of female reproductive lifespan is associated with multiple adverse outcomes, including breast cancer, cardiovascular disease and infertility. The biological processes that govern the timing of the beginning and end of reproductive life are not well understood. Genetic variants are known to contribute to ∼50% of the variation in both age at menarche and menopause, but to date the known genes explain <15% of the genetic component. We have used genome-wide association in a bivariate meta-analysis of both traits to identify genes involved in determining reproductive lifespan. We observed significant genetic correlation between the two traits using genome-wide complex trait analysis. However, we found no robust statistical evidence for individual variants with an effect on both traits. A novel association with age at menopause was detected for a variant rs1800932 in the mismatch repair gene MSH6 (P = 1.9 × 10(-9)), which was also associated with altered expression levels of MSH6 mRNA in multiple tissues. This study contributes to the growing evidence that DNA repair processes play a key role in ovarian ageing and could be an important therapeutic target for infertility.

    View details for DOI 10.1093/hmg/ddt620

    View details for Web of Science ID 000334359100022

    View details for PubMedID 24357391

  • A Central Role for GRB10 in Regulation of Islet Function in Man PLOS GENETICS Prokopenko, I., Poon, W., Maegi, R., Prasad, R. B., Salehi, S. A., Almgren, P., Osmark, P., Bouatia-Naji, N., Wierup, N., Fall, T., Stancakova, A., Barker, A., Lagou, V., Osmond, C., Xie, W., Lahti, J., Jackson, A. U., Cheng, Y., Liu, J., O'Connell, J. R., Blomstedt, P. A., Fadista, J., Alkayyali, S., Dayeh, T., Ahlqvist, E., Taneera, J., Lecoeur, C., Kumar, A., Hansson, O., Hansson, K., Voight, B. F., Kang, H. M., Levy-Marchal, C., Vatin, V., Palotie, A., Syvanen, A., Mari, A., Weedon, M. N., Loos, R. J., Ong, K. K., Nilsson, P., Isomaa, B., Tuomi, T., Wareham, N. J., Stumvoll, M., Widen, E., Lakka, T. A., Langenberg, C., Tonjes, A., Rauramaa, R., Kuusisto, J., Frayling, T. M., Froguel, P., Walker, M., Eriksson, J. G., Ling, C., Kovacs, P., Ingelsson, E., McCarthy, M. I., Shuldiner, A. R., Silver, K. D., Laakso, M., Groop, L., Lyssenko, V. 2014; 10 (4)

    Abstract

    Variants in the growth factor receptor-bound protein 10 (GRB10) gene were in a GWAS meta-analysis associated with reduced glucose-stimulated insulin secretion and increased risk of type 2 diabetes (T2D) if inherited from the father, but inexplicably reduced fasting glucose when inherited from the mother. GRB10 is a negative regulator of insulin signaling and imprinted in a parent-of-origin fashion in different tissues. GRB10 knock-down in human pancreatic islets showed reduced insulin and glucagon secretion, which together with changes in insulin sensitivity may explain the paradoxical reduction of glucose despite a decrease in insulin secretion. Together, these findings suggest that tissue-specific methylation and possibly imprinting of GRB10 can influence glucose metabolism and contribute to T2D pathogenesis. The data also emphasize the need in genetic studies to consider whether risk alleles are inherited from the mother or the father.

    View details for DOI 10.1371/journal.pgen.1004235

    View details for Web of Science ID 000335499600017

    View details for PubMedID 24699409

  • Loss-of-function mutations in SLC30A8 protect against type 2 diabetes NATURE GENETICS Flannick, J., Thorleifsson, G., Beer, N. L., Jacobs, S. B., Grarup, N., Burtt, N. P., Mahajan, A., Fuchsberger, C., Atzmon, G., Benediktsson, R., Blangero, J., Bowden, D. W., Brandslund, I., Brosnan, J., Burslem, F., Chambers, J., Cho, Y. S., Christensen, C., Douglas, D. A., Duggirala, R., Dymek, Z., Farjoun, Y., Fennell, T., Fontanillas, P., Forsen, T., Gabriel, S., Glaser, B., Gudbjartsson, D. F., Hanis, C., Hansen, T., Hreidarsson, A. B., Hveem, K., Ingelsson, E., Isomaa, B., Johansson, S., Jorgensen, T., Jorgensen, M. E., Kathiresan, S., Kong, A., Kooner, J., Kravic, J., Laakso, M., Lee, J., Lind, L., Lindgren, C. M., Linneberg, A., Masson, G., Meitinger, T., Mohlke, K. L., Molven, A., Morris, A. P., Potluri, S., Rauramaa, R., Ribel-Madsen, R., Richard, A., Rolph, T., Salomaa, V., Segre, A. V., Skaerstrand, H., Steinthorsdottir, V., Stringham, H. M., Sulem, P., Tai, E. S., Teo, Y. Y., Teslovich, T., Thorsteinsdottir, U., Trimmer, J. K., Tuomi, T., Tuomilehto, J., Vaziri-Sani, F., Voight, B. F., Wilson, J. G., Boehnke, M., McCarthy, M. I., Njolstad, P. R., Pedersen, O., Groop, L., Cox, D. R., Stefansson, K., Altshuler, D. 2014; 46 (4): 357-?

    Abstract

    Loss-of-function mutations protective against human disease provide in vivo validation of therapeutic targets, but none have yet been described for type 2 diabetes (T2D). Through sequencing or genotyping of ~150,000 individuals across 5 ancestry groups, we identified 12 rare protein-truncating variants in SLC30A8, which encodes an islet zinc transporter (ZnT8) and harbors a common variant (p.Trp325Arg) associated with T2D risk and glucose and proinsulin levels. Collectively, carriers of protein-truncating variants had 65% reduced T2D risk (P = 1.7 × 10(-6)), and non-diabetic Icelandic carriers of a frameshift variant (p.Lys34Serfs*50) demonstrated reduced glucose levels (-0.17 s.d., P = 4.6 × 10(-4)). The two most common protein-truncating variants (p.Arg138* and p.Lys34Serfs*50) individually associate with T2D protection and encode unstable ZnT8 proteins. Previous functional study of SLC30A8 suggested that reduced zinc transport increases T2D risk, and phenotypic heterogeneity was observed in mouse Slc30a8 knockouts. In contrast, loss-of-function mutations in humans provide strong evidence that SLC30A8 haploinsufficiency protects against T2D, suggesting ZnT8 inhibition as a therapeutic strategy in T2D prevention.

    View details for DOI 10.1038/ng.2915

    View details for Web of Science ID 000334510100011

    View details for PubMedID 24584071

  • Interplay of overweight and insulin resistance on hypertension development JOURNAL OF HYPERTENSION Lytsy, P., Ingelsson, E., Lind, L., Arnlov, J., Sundstrom, J. 2014; 32 (4): 834-839

    Abstract

    Obesity and hypertension are associated, possibly through causal pathways involving insulin resistance and metabolic derangements. We aimed to investigate in a whites sample if overweight or obese persons without insulin resistance are at risk of developing hypertension or blood pressure progression.In a meta-analysis, using multivariable-adjusted mixed-effects logistic regression models, we investigated the risks of hypertension development and blood pressure progression by combinations of relative weight classes and presence or absence of insulin resistance (defined as highest vs. lower three quartiles using the homeostatic model assessment method) in the Uppsala Longitudinal Study of Adult Men (n = 2322) and the Prospective Investigation of the Vasculature in Uppsala Seniors studies (n = 1066). These two samples, consisting mainly of middle-aged and elderly men, provided 1846 observations for the development of hypertension in normotensive individuals and 4223 observations for progressing to a higher blood pressure stage.During a median of 10 years of follow-up, 884 (47.9%) developed hypertension and 1639 (38.8%) progressed to a higher blood pressure stage. Overweight or obese persons without insulin resistance had an increased risk of hypertension development [odds ratio (OR) 1.46, 95% confidence interval 1.14-1.88] and blood pressure progression (OR 1.32, 1.10-1.59) compared with normal-weight persons without insulin resistance.According to this study, being overweight or obese without insulin resistance increases the risk of hypertension and blood pressure progression. This adds to the evidence that overweight and obesity may be harmful per se, and that overweight and obesity without glucometabolic derangements are not benign conditions.

    View details for DOI 10.1097/HJH.0000000000000081

    View details for Web of Science ID 000333301000019

    View details for PubMedID 24370898

  • Cerebrovascular and ischemic heart disease in young adults born preterm: a population-based Swedish cohort study EUROPEAN JOURNAL OF EPIDEMIOLOGY Ueda, P., Cnattingius, S., Stephansson, O., Ingelsson, E., Ludvigsson, J. F., Bonamy, A. E. 2014; 29 (4): 253-260

    Abstract

    Preterm birth is associated with overall cardiovascular mortality in young adulthood, but which specific conditions that underlie this association is unknown. We studied mortality and morbidity from cerebrovascular and ischemic heart disease in individuals born preterm. In a nationwide Swedish study, we included 1,306,943 individuals without congenital malformations born between 1983 and 1995, followed from 15 years of age to December 31st, 2010. Of these, 73,489 (5.6 %) were born preterm (<37 weeks of gestation). Cox proportional hazards regression analysis was used to calculate hazard ratios (HR) with 95 % confidence intervals (CI), after adjusting for maternal characteristics and birth weight for gestational age. Of 955 incident cases of cerebrovascular disease, 58 (6.1 %) occurred in preterm born subjects. The corresponding numbers of ischemic heart disease cases were 180 and 13 (7.2 %), respectively. Birth before 32 weeks was associated with a nearly twofold increased risk of cerebrovascular disease; adjusted HR, (95 % CI) = 1.89 (1.01-3.54) compared to term born individuals, whereas individuals born at 32-36 weeks were not at increased risk. Preterm birth was not associated with later ischemic heart disease; no cases of ischemic heart disease were recorded among those born before 32 weeks and the HR (95 % CI) for those born at 32-36 weeks of gestation was 1.45 (0.81-2.57), compared to term-born individuals. Birth before 32 weeks is associated with increased risk of cerebrovascular disease in young adulthood. Our data suggest that cardiovascular health promotion in follow-up programs after very preterm birth may be beneficial.

    View details for DOI 10.1007/s10654-014-9892-5

    View details for Web of Science ID 000336801600004

    View details for PubMedID 24687624

  • Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility NATURE GENETICS Mahajan, A., Go, M. J., Zhang, W., Below, J. E., Gaulton, K. J., Ferreira, T., Horikoshi, M., Johnson, A. D., Ng, M. C., Prokopenko, I., Saleheen, D., Wang, X., Zeggini, E., Abecasis, G. R., Adair, L. S., Almgren, P., Atalay, M., Aung, T., Baldassarre, D., Balkau, B., Bao, Y., Barnett, A. H., Barroso, I., Basit, A., Been, L. F., Beilby, J., Bell, G. I., Benediktsson, R., Bergman, R. N., Boehm, B. O., Boerwinkle, E., Bonnycastle, L. L., Burtt, N., Cai, Q., Campbell, H., Carey, J., Cauchi, S., Caulfield, M., Chan, J. C., Chang, L., Chang, T., Chang, Y., Charpentier, G., Chen, C., Chen, H., Chen, Y., Chia, K., Chidambaram, M., Chines, P. S., Cho, N. H., Cho, Y. M., Chuang, L., Collins, F. S., Cornelis, M. C., Couper, D. J., Crenshaw, A. T., van Dam, R. M., Danesh, J., Das, D., de Faire, U., Dedoussis, G., Deloukas, P., Dimas, A. S., Dina, C., Doney, A. S., Donnelly, P. J., Dorkhan, M., Van Duijn, C., Dupuis, J., Edkins, S., Elliott, P., Emilsson, V., Erbel, R., Eriksson, J. G., Escobedo, J., Esko, T., Eury, E., Florez, J. C., Fontanillas, P., Forouhi, N. G., Forsen, T., Fox, C., Fraser, R. M., Frayling, T. M., Froguel, P., Frossard, P., Gao, Y., Gertow, K., Gieger, C., Gigante, B., Grallert, H., Grant, G. B., Groop, L. C., Groves, C. J., Grundberg, E., Guiducci, C., Hamsten, A., Han, B., Hara, K., Hassanali, N., Hattersley, A. T., Hayward, C., Hedman, A. K., Herder, C., Hofman, A., Holmen, O. L., Hovingh, K., Hreidarsson, A. B., Hu, C., Hu, F. B., Hui, J., Humphries, S. E., Hunt, S. E., Hunter, D. J., Hveem, K., Hydrie, Z. I., Ikegami, H., Illig, T., Ingelsson, E., Islam, M., Isomaa, B., Jackson, A. U., Jafar, T., James, A., Jia, W., Joeckel, K., Jonsson, A., Jowett, J. B., Kadowaki, T., Kang, H. M., Kanoni, S., Kao, W. H., Kathiresan, S., Kato, N., Katulanda, P., Keinanen-Kiukaanniemi, S. M., Kelly, A. M., Khan, H., Khaw, K., Khor, C., Kim, H., Kim, S., Kim, Y. J., Kinnunen, L., Klopp, N., Kong, A., Korpi-Hyovalti, E., Kowlessur, S., Kraft, P., Kravic, J., Kristensen, M. M., Krithika, S., Kumar, A., Kumate, J., Kuusisto, J., Kwak, S. H., Laakso, M., Lagou, V., Lakka, T. A., Langenberg, C., Langford, C., Lawrence, R., Leander, K., Lee, J., Lee, N. R., Li, M., Li, X., Li, Y., Liang, J., Liju, S., Lim, W., Lind, L., Lindgren, C. M., Lindholm, E., Liu, C., Liu, J. J., Lobbens, S., Long, J., Loos, R. J., Lu, W., Luan, J., Lyssenko, V., Ma, R. C., Maeda, S., Maegi, R., Mannisto, S., Matthews, D. R., Meigs, J. B., Melander, O., Metspalu, A., Meyer, J., Mirza, G., Mihailov, E., Moebus, S., Mohan, V., Mohlke, K. L., Morris, A. D., Muehleisen, T. W., Mueller-Nurasyid, M., Musk, B., Nakamura, J., Nakashima, E., Navarro, P., Peng-Keat Ng, P. K., Nica, A. C., Nilsson, P. M., Njolstad, I., Noethen, M. M., Ohnaka, K., Ong, T. H., Owen, K. R., Palmer, C. N., Pankow, J. S., Park, K. S., Parkin, M., Pechlivanis, S., Pedersen, N. L., Peltonen, L., Perry, J. R., Peters, A., Pinidiyapathirage, J. M., Platou, C. G., Potter, S., Price, J. F., Qi, L., Radha, V., Rallidis, L., Rasheed, A., Rathmann, W., Rauramaa, R., Raychaudhuri, S., Rayner, N. W., Rees, S. D., Rehnberg, E., Ripatti, S., Robertson, N., Roden, M., Rossin, E. J., Rudan, I., Rybin, D., Saaristo, T. E., Salomaa, V., Saltevo, J., Samuel, M., Sanghera, D. K., Saramies, J., Scott, J., Scott, L. J., Scott, R. A., Segre, A. V., Sehmi, J., Sennblad, B., Shah, N., Shah, S., Shera, A. S., Shu, X. O., Shuldiner, A. R., Sigurdsson, G., Sijbrands, E., Silveira, A., Sim, X., Sivapalaratnam, S., Small, K. S., So, W. Y., Stancakova, A., Stefansson, K., Steinbach, G., Steinthorsdottir, V., Stirrups, K., Strawbridge, R. J., Stringham, H. M., Sun, Q., Suo, C., Syvanen, A., Takayanagi, R., Takeuchi, F., Tay, W. T., Teslovich, T. M., Thorand, B., Thorleifsson, G., Thorsteinsdottir, U., Tikkanen, E., Trakalo, J., Tremoli, E., Trip, M. D., Tsai, F. J., Tuomi, T., Tuomilehto, J., Uitterlinden, A. G., Valladares-Salgado, A., Vedantam, S., Veglia, F., Voight, B. F., Wang, C., Wareham, N. J., Wennauer, R., Wickremasinghe, A. R., Wilsgaard, T., Wilson, J. F., Wiltshire, S., Winckler, W., Wong, T. Y., Wood, A. R., Wu, J., Wu, Y., Yamamoto, K., Yamauchi, T., Yang, M., Yengo, L., Yokota, M., Young, R., Zabaneh, D., Zhang, F., Zhang, R., Zheng, W., Zimmet, P. Z., Altshuler, D., Bowden, D. W., Cho, Y. S., Cox, N. J., Cruz, M., Hanis, C. L., Kooner, J., Lee, J., Seielstad, M., Teo, Y. Y., Boehnke, M., Parra, E. J., Chambers, J. C., Tai, E. S., McCarthy, M. I., Morris, A. P. 2014; 46 (3): 234-?

    Abstract

    To further understanding of the genetic basis of type 2 diabetes (T2D) susceptibility, we aggregated published meta-analyses of genome-wide association studies (GWAS), including 26,488 cases and 83,964 controls of European, east Asian, south Asian and Mexican and Mexican American ancestry. We observed a significant excess in the directional consistency of T2D risk alleles across ancestry groups, even at SNPs demonstrating only weak evidence of association. By following up the strongest signals of association from the trans-ethnic meta-analysis in an additional 21,491 cases and 55,647 controls of European ancestry, we identified seven new T2D susceptibility loci. Furthermore, we observed considerable improvements in the fine-mapping resolution of common variant association signals at several T2D susceptibility loci. These observations highlight the benefits of trans-ethnic GWAS for the discovery and characterization of complex trait loci and emphasize an exciting opportunity to extend insight into the genetic architecture and pathogenesis of human diseases across populations of diverse ancestry.

    View details for DOI 10.1038/ng.2897

    View details for Web of Science ID 000332036700007

    View details for PubMedID 24509480

  • FTO Genetic Variants and Risk of Obesity and Type 2 Diabetes: A Meta-Analysis of 28,394 Indians OBESITY Vasan, S. K., Karpe, F., Gu, H. F., Brismar, K., Fall, C. H., Ingelsson, E., Fall, T. 2014; 22 (3): 964-970

    Abstract

    To investigate the magnitude of association of FTO variants with obesity, type 2 diabetes (T2DM), and related traits among Asian Indians.Random-effect meta-analysis was performed on pooled data from eight studies (n = 28,394) for obesity and related traits and six studies (n = 24,987) for assessment of T2DM risk in Indians where FTO variants were reported.The minor A-allele of the FTO variant rs9939609 was associated with increased risk of obesity (OR 1.15, 95% CI 1.08-1.21, p = 2.14 × 10(-) (5) ), BMI (β = 0.30 kg/m2, 95% CI 0.21-0.38, p = 4.78 × 10(-) (11) ) and other regional adiposity measurements [waist (β = 0.74 cm, 95% CI 0.49-0.99), HC (β = 0.52, 95% CI 0.26-0.78), and waist-hip ratio (WHR) (β = 0.002, 95% CI 0.001-0.004)] in Indians (p ≤ 0.001). An increased risk for T2DM (OR 1.11; 95% CI 1.04-1.19, p = 0.002) was observed, which attenuated when adjusted for age, gender, and BMI (OR 1.09; 95%CI 1.02-1.16, p = 0.01).Our study provides evidence of association between common FTO variant and obesity risk among Indians with comparable effect sizes as in Caucasians. The attenuation of FTO-T2DM risk on BMI adjustment reinforces that BMI does not fully account for the adiposity effects among Asian Indians who are more centrally obese.

    View details for DOI 10.1002/oby.20606

    View details for Web of Science ID 000332224800049

    View details for PubMedID 23963770

  • Genome-wide association studies of obesity and metabolic syndrome MOLECULAR AND CELLULAR ENDOCRINOLOGY Fall, T., Ingelsson, E. 2014; 382 (1): 740-757

    Abstract

    Until just a few years ago, the genetic determinants of obesity and metabolic syndrome were largely unknown, with the exception of a few forms of monogenic extreme obesity. Since genome-wide association studies (GWAS) became available, large advances have been made. The first single nucleotide polymorphism robustly associated with increased body mass index (BMI) was in 2007 mapped to a gene with for the time unknown function. This gene, now known as fat mass and obesity associated (FTO) has been repeatedly replicated in several ethnicities and is affecting obesity by regulating appetite. Since the first report from a GWAS of obesity, an increasing number of markers have been shown to be associated with BMI, other measures of obesity or fat distribution and metabolic syndrome. This systematic review of obesity GWAS will summarize genome-wide significant findings for obesity and metabolic syndrome and briefly give a few suggestions of what is to be expected in the next few years.

    View details for DOI 10.1016/j.mce.2012.08.018

    View details for Web of Science ID 000330421600078

    View details for PubMedID 22963884

  • Common Familial Effects on Ischemic Stroke and Myocardial Infarction: A Prospective Population-Based Cohort Study. Frontiers in cardiovascular medicine Kasiman, K., Lundholm, C., Sandin, S., Malki, N., Sparén, P., Ingelsson, E. 2014; 1: 3-?

    Abstract

    Recent genome-wide association studies suggest some overlap of genetic determinants of ischemic stroke (IS) and myocardial infarction (MI). This study aimed to assess shared familial risk between IS and MI in a large, population-wide cohort study.Study participants free of IS and MI and their affected siblings were extracted from the Swedish Hospital Discharge and Cause of Death Registers between 1987 and 2007, forming an exposed sib-pair. They were matched by birth year of both siblings and calendar period to up to five unexposed sib-pairs. Stratified Cox regression analyses were used to assess familial risk of MI and IS in those exposed to having a sibling with IS (n = 31,659) and MI (n = 62,766), respectively, compared to unexposed (n = 143,728 and 265,974).The overall risk of MI when exposed to having a sibling with IS was statistically significantly increased (RR, 1.44; 95% CI, 1.34-1.55, p < 0.001) to a similar extent as risk of IS when exposed to having a sibling with MI (RR, 1.41; 95% CI, 1.32-1.50, p < 0.001). The familial risks were similar in full siblings for both groups (RR for MI, 1.46; 95% CI, 1.35-1.58, p < 0.001; and RR for IS, 1.40; 95% CI, 1.30-1.40, p < 0.001) and half siblings (RR for MI, 1.29; 95% CI, 1.05-1.59, p < 0.001; and RR for IS, 1.38; 95% CI, 1.16-1.65, p < 0.001).This large, population-wide study indicates that there is considerable overlap of familial risk between IS and MI.

    View details for DOI 10.3389/fcvm.2014.00003

    View details for PubMedID 26664855

  • Utilizing Twins as Controls for Non-Twin Case-Materials in Genome Wide Association Studies PLOS ONE Ganna, A., Ortega-Alonso, A., Havulinna, A., Salomaa, V., Kaprio, J., Pedersen, N. L., Sullivan, P. F., Ingelsson, E., Hultman, C. M., Magnusson, P. K. 2013; 8 (12)

    Abstract

    Twin registries around the globe have collected DNA samples from large numbers of monozygotic and dizygotic twins. The twin sample collections are frequently used as controls in disease-specific studies together with non-twins. This approach is unbiased under the hypothesis that twins and singletons are comparable in terms of allele frequencies; i.e. there are no genetic variants associated with being a twin per se. To test this hypothesis we performed a genome-wide association study comparing the allele frequency of 572,352 single nucleotide polymorphisms (SNPs) in 1,413 monozygotic (MZ) and 5,451 dizygotic (DZ) twins with 3,720 healthy singletons. Twins and singletons have been genotyped using the same platform. SNPs showing association with being a twin at P-value < 1 × 10(-5) were selected for replication analysis in 1,492 twins (463 MZ and 1,029 DZ) and 1,880 singletons from Finland. No SNPs reached genome-wide significance (P-value < 5 × 10(-8)) in the main analysis combining MZ and DZ twins. In a secondary analysis including only DZ twins two SNPs (rs2033541 close to ADAMTSL1 and rs4149283 close to ABCA1) were genome-wide significant after meta-analysis with the Finnish population. The estimated proportion of variance on the liability scale explained by all SNPs was 0.08 (P-value=0.003) when MZ and DZ were considered together and smaller for MZ (0.06, P-value=0.10) compared to DZ (0.09, P-value=0.003) when analyzed separately. In conclusion, twins and singletons can be used in genetic studies together with general population samples without introducing large bias. Further research is needed to explore genetic variances associated with DZ twinning.

    View details for DOI 10.1371/journal.pone.0083101

    View details for Web of Science ID 000328707400121

    View details for PubMedID 24340086

  • Association Between Circulating Endostatin, Hypertension Duration, and Hypertensive Target-Organ Damage HYPERTENSION Carlsson, A. C., Ruge, T., Sundstrom, J., Ingelsson, E., Larsson, A., Lind, L., Arnlov, J. 2013; 62 (6): 1146-1151

    Abstract

    Our aim is to study associations between circulating endostatin, hypertension duration, and hypertensive target-organ damage. Long-term hypertension induces cardiovascular and renal remodeling. Circulating endostatin, a biologically active derivate of collagen XVIII, has been suggested to be a relevant marker for extracellular matrix turnover and remodeling in various diseases. However, the role of endostatin in hypertension and hypertensive target-organ damage is unclear. Serum endostatin was measured in 2 independent community-based cohorts: the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS; women 51%; n = 812; mean age, 75 years) and the Uppsala Longitudinal Study of Adult Men (ULSAM; n=785; mean age, 77.6 years). Retrospective data on blood pressure measurements and antihypertensive medication (PIVUS >5 years, ULSAM >27 years), and cross-sectional data on echocardiographic left ventricular mass, endothelial function (endothelium-dependent vasodilation assessed by the invasive forearm model), and urinary albumin/creatinine ratio were available. In PIVUS, participants with ≥ 5 years of history of hypertension portrayed 0.42 SD (95% confidence interval, 0.23-0.61; P<0.001) higher serum endostatin, compared with that of normotensives. This association was replicated in ULSAM, in which participants with 27 years hypertension duration had the highest endostatin (0.57 SD higher; 95% confidence interval, 0.35-0.80; P<0.001). In addition, higher endostatin was associated with higher left ventricular mass, worsened endothelial function, and higher urinary albumin/creatinine ratio (P<0.03 for all) in participants with prevalent hypertension. Circulating endostatin is associated with the duration of hypertension, and vascular, myocardial, and renal indices of hypertensive target-organ damage. Further studies are warranted to assess the prognostic role of endostatin in individuals with hypertension.

    View details for DOI 10.1161/HYPERTENSIONAHA.113.02250

    View details for Web of Science ID 000326919300036

    View details for PubMedID 24082055

  • Inference of the Genetic Architecture Underlying BMI and Height with the Use of 20,240 Sibling Pairs AMERICAN JOURNAL OF HUMAN GENETICS Hemani, G., Yang, J., Vinkhuyzen, A., Powell, J. E., Willemsen, G., Hottenga, J., Abdellaoui, A., Mangino, M., Valdes, A. M., Medland, S. E., Madden, P. A., Heath, A. C., Henders, A. K., Nyholt, D. R., de Geus, E. J., Magnusson, P. K., Ingelsson, E., Montgomery, G. W., Spector, T. D., Boomsma, D. I., Pedersen, N. L., Martin, N. G., Visscher, P. M. 2013; 93 (5): 865-875

    Abstract

    Evidence that complex traits are highly polygenic has been presented by population-based genome-wide association studies (GWASs) through the identification of many significant variants, as well as by family-based de novo sequencing studies indicating that several traits have a large mutational target size. Here, using a third study design, we show results consistent with extreme polygenicity for body mass index (BMI) and height. On a sample of 20,240 siblings (from 9,570 nuclear families), we used a within-family method to obtain narrow-sense heritability estimates of 0.42 (SE = 0.17, p = 0.01) and 0.69 (SE = 0.14, p = 6 × 10(-)(7)) for BMI and height, respectively, after adjusting for covariates. The genomic inflation factors from locus-specific linkage analysis were 1.69 (SE = 0.21, p = 0.04) for BMI and 2.18 (SE = 0.21, p = 2 × 10(-10)) for height. This inflation is free of confounding and congruent with polygenicity, consistent with observations of ever-increasing genomic-inflation factors from GWASs with large sample sizes, implying that those signals are due to true genetic signals across the genome rather than population stratification. We also demonstrate that the distribution of the observed test statistics is consistent with both rare and common variants underlying a polygenic architecture and that previous reports of linkage signals in complex traits are probably a consequence of polygenic architecture rather than the segregation of variants with large effects. The convergent empirical evidence from GWASs, de novo studies, and within-family segregation implies that family-based sequencing studies for complex traits require very large sample sizes because the effects of causal variants are small on average.

    View details for DOI 10.1016/j.ajhg.2013.10.005

    View details for Web of Science ID 000326996600007

    View details for PubMedID 24183453

  • Common variants associated with plasma triglycerides and risk for coronary artery disease. Nature genetics Do, R., Willer, C. J., Schmidt, E. M., Sengupta, S., Gao, C., Peloso, G. M., Gustafsson, S., Kanoni, S., Ganna, A., Chen, J., Buchkovich, M. L., Mora, S., Beckmann, J. S., Bragg-Gresham, J. L., Chang, H., Demirkan, A., den Hertog, H. M., Donnelly, L. A., Ehret, G. B., Esko, T., Feitosa, M. F., Ferreira, T., Fischer, K., Fontanillas, P., Fraser, R. M., Freitag, D. F., Gurdasani, D., Heikkilä, K., Hyppönen, E., Isaacs, A., Jackson, A. U., Johansson, A., Johnson, T., Kaakinen, M., Kettunen, J., Kleber, M. E., Li, X., Luan, J., Lyytikäinen, L., Magnusson, P. K., Mangino, M., Mihailov, E., Montasser, M. E., Müller-Nurasyid, M., Nolte, I. M., O'Connell, J. R., Palmer, C. D., Perola, M., Petersen, A., Sanna, S., Saxena, R., Service, S. K., Shah, S., Shungin, D., Sidore, C., Song, C., Strawbridge, R. J., Surakka, I., Tanaka, T., Teslovich, T. M., Thorleifsson, G., van den Herik, E. G., Voight, B. F., Volcik, K. A., Waite, L. L., Wong, A., Wu, Y., Zhang, W., Absher, D., Asiki, G., Barroso, I., Been, L. F., Bolton, J. L., Bonnycastle, L. L., Brambilla, P., Burnett, M. S., Cesana, G., Dimitriou, M., Doney, A. S., Döring, A., Elliott, P., Epstein, S. E., Eyjolfsson, G. I., Gigante, B., Goodarzi, M. O., Grallert, H., Gravito, M. L., Groves, C. J., Hallmans, G., Hartikainen, A., Hayward, C., Hernandez, D., Hicks, A. A., Holm, H., Hung, Y., Illig, T., Jones, M. R., Kaleebu, P., Kastelein, J. J., Khaw, K., Kim, E., Klopp, N., Komulainen, P., Kumari, M., Langenberg, C., Lehtimäki, T., Lin, S., Lindström, J., Loos, R. J., Mach, F., McArdle, W. L., Meisinger, C., Mitchell, B. D., Müller, G., Nagaraja, R., Narisu, N., Nieminen, T. V., Nsubuga, R. N., Olafsson, I., Ong, K. K., Palotie, A., Papamarkou, T., Pomilla, C., Pouta, A., Rader, D. J., Reilly, M. P., Ridker, P. M., Rivadeneira, F., Rudan, I., Ruokonen, A., Samani, N., Scharnagl, H., Seeley, J., Silander, K., Stancáková, A., Stirrups, K., Swift, A. J., Tiret, L., Uitterlinden, A. G., van Pelt, L. J., Vedantam, S., Wainwright, N., Wijmenga, C., Wild, S. H., Willemsen, G., Wilsgaard, T., Wilson, J. F., Young, E. H., Zhao, J. H., Adair, L. S., Arveiler, D., Assimes, T. L., Bandinelli, S., Bennett, F., Bochud, M., Boehm, B. O., Boomsma, D. I., Borecki, I. B., Bornstein, S. R., Bovet, P., Burnier, M., Campbell, H., Chakravarti, A., Chambers, J. C., Chen, Y. I., Collins, F. S., Cooper, R. S., Danesh, J., Dedoussis, G., de Faire, U., Feranil, A. B., Ferrières, J., Ferrucci, L., Freimer, N. B., Gieger, C., Groop, L. C., Gudnason, V., Gyllensten, U., Hamsten, A., Harris, T. B., Hingorani, A., Hirschhorn, J. N., Hofman, A., Hovingh, G. K., Hsiung, C. A., Humphries, S. E., Hunt, S. C., Hveem, K., Iribarren, C., Järvelin, M., Jula, A., Kähönen, M., Kaprio, J., Kesäniemi, A., Kivimaki, M., Kooner, J. S., Koudstaal, P. J., Krauss, R. M., Kuh, D., Kuusisto, J., Kyvik, K. O., Laakso, M., Lakka, T. A., Lind, L., Lindgren, C. M., Martin, N. G., März, W., McCarthy, M. I., McKenzie, C. A., Meneton, P., Metspalu, A., Moilanen, L., Morris, A. D., Munroe, P. B., Njølstad, I., Pedersen, N. L., Power, C., Pramstaller, P. P., Price, J. F., Psaty, B. M., Quertermous, T., Rauramaa, R., Saleheen, D., Salomaa, V., Sanghera, D. K., Saramies, J., Schwarz, P. E., Sheu, W. H., Shuldiner, A. R., Siegbahn, A., Spector, T. D., Stefansson, K., Strachan, D. P., Tayo, B. O., Tremoli, E., Tuomilehto, J., Uusitupa, M., van Duijn, C. M., Vollenweider, P., Wallentin, L., Wareham, N. J., Whitfield, J. B., Wolffenbuttel, B. H., Altshuler, D., Ordovas, J. M., Boerwinkle, E., Palmer, C. N., Thorsteinsdottir, U., Chasman, D. I., Rotter, J. I., Franks, P. W., Ripatti, S., Cupples, L. A., Sandhu, M. S., Rich, S. S., Boehnke, M., Deloukas, P., Mohlke, K. L., Ingelsson, E., Abecasis, G. R., Daly, M. J., Neale, B. M., Kathiresan, S. 2013; 45 (11): 1345-1352

    Abstract

    Triglycerides are transported in plasma by specific triglyceride-rich lipoproteins; in epidemiological studies, increased triglyceride levels correlate with higher risk for coronary artery disease (CAD). However, it is unclear whether this association reflects causal processes. We used 185 common variants recently mapped for plasma lipids (P < 5 × 10(-8) for each) to examine the role of triglycerides in risk for CAD. First, we highlight loci associated with both low-density lipoprotein cholesterol (LDL-C) and triglyceride levels, and we show that the direction and magnitude of the associations with both traits are factors in determining CAD risk. Second, we consider loci with only a strong association with triglycerides and show that these loci are also associated with CAD. Finally, in a model accounting for effects on LDL-C and/or high-density lipoprotein cholesterol (HDL-C) levels, the strength of a polymorphism's effect on triglyceride levels is correlated with the magnitude of its effect on CAD risk. These results suggest that triglyceride-rich lipoproteins causally influence risk for CAD.

    View details for DOI 10.1038/ng.2795

    View details for PubMedID 24097064

  • Discovery and refinement of loci associated with lipid levels. Nature genetics Willer, C. J., Schmidt, E. M., Sengupta, S., Peloso, G. M., Gustafsson, S., Kanoni, S., Ganna, A., Chen, J., Buchkovich, M. L., Mora, S., Beckmann, J. S., Bragg-Gresham, J. L., Chang, H., Demirkan, A., den Hertog, H. M., Do, R., Donnelly, L. A., Ehret, G. B., Esko, T., Feitosa, M. F., Ferreira, T., Fischer, K., Fontanillas, P., Fraser, R. M., Freitag, D. F., Gurdasani, D., Heikkilä, K., Hyppönen, E., Isaacs, A., Jackson, A. U., Johansson, A., Johnson, T., Kaakinen, M., Kettunen, J., Kleber, M. E., Li, X., Luan, J., Lyytikäinen, L., Magnusson, P. K., Mangino, M., Mihailov, E., Montasser, M. E., Müller-Nurasyid, M., Nolte, I. M., O'Connell, J. R., Palmer, C. D., Perola, M., Petersen, A., Sanna, S., Saxena, R., Service, S. K., Shah, S., Shungin, D., Sidore, C., Song, C., Strawbridge, R. J., Surakka, I., Tanaka, T., Teslovich, T. M., Thorleifsson, G., van den Herik, E. G., Voight, B. F., Volcik, K. A., Waite, L. L., Wong, A., Wu, Y., Zhang, W., Absher, D., Asiki, G., Barroso, I., Been, L. F., Bolton, J. L., Bonnycastle, L. L., Brambilla, P., Burnett, M. S., Cesana, G., Dimitriou, M., Doney, A. S., Döring, A., Elliott, P., Epstein, S. E., Eyjolfsson, G. I., Gigante, B., Goodarzi, M. O., Grallert, H., Gravito, M. L., Groves, C. J., Hallmans, G., Hartikainen, A., Hayward, C., Hernandez, D., Hicks, A. A., Holm, H., Hung, Y., Illig, T., Jones, M. R., Kaleebu, P., Kastelein, J. J., Khaw, K., Kim, E., Klopp, N., Komulainen, P., Kumari, M., Langenberg, C., Lehtimäki, T., Lin, S., Lindström, J., Loos, R. J., Mach, F., McArdle, W. L., Meisinger, C., Mitchell, B. D., Müller, G., Nagaraja, R., Narisu, N., Nieminen, T. V., Nsubuga, R. N., Olafsson, I., Ong, K. K., Palotie, A., Papamarkou, T., Pomilla, C., Pouta, A., Rader, D. J., Reilly, M. P., Ridker, P. M., Rivadeneira, F., Rudan, I., Ruokonen, A., Samani, N., Scharnagl, H., Seeley, J., Silander, K., Stancáková, A., Stirrups, K., Swift, A. J., Tiret, L., Uitterlinden, A. G., van Pelt, L. J., Vedantam, S., Wainwright, N., Wijmenga, C., Wild, S. H., Willemsen, G., Wilsgaard, T., Wilson, J. F., Young, E. H., Zhao, J. H., Adair, L. S., Arveiler, D., Assimes, T. L., Bandinelli, S., Bennett, F., Bochud, M., Boehm, B. O., Boomsma, D. I., Borecki, I. B., Bornstein, S. R., Bovet, P., Burnier, M., Campbell, H., Chakravarti, A., Chambers, J. C., Chen, Y. I., Collins, F. S., Cooper, R. S., Danesh, J., Dedoussis, G., de Faire, U., Feranil, A. B., Ferrières, J., Ferrucci, L., Freimer, N. B., Gieger, C., Groop, L. C., Gudnason, V., Gyllensten, U., Hamsten, A., Harris, T. B., Hingorani, A., Hirschhorn, J. N., Hofman, A., Hovingh, G. K., Hsiung, C. A., Humphries, S. E., Hunt, S. C., Hveem, K., Iribarren, C., Järvelin, M., Jula, A., Kähönen, M., Kaprio, J., Kesäniemi, A., Kivimaki, M., Kooner, J. S., Koudstaal, P. J., Krauss, R. M., Kuh, D., Kuusisto, J., Kyvik, K. O., Laakso, M., Lakka, T. A., Lind, L., Lindgren, C. M., Martin, N. G., März, W., McCarthy, M. I., McKenzie, C. A., Meneton, P., Metspalu, A., Moilanen, L., Morris, A. D., Munroe, P. B., Njølstad, I., Pedersen, N. L., Power, C., Pramstaller, P. P., Price, J. F., Psaty, B. M., Quertermous, T., Rauramaa, R., Saleheen, D., Salomaa, V., Sanghera, D. K., Saramies, J., Schwarz, P. E., Sheu, W. H., Shuldiner, A. R., Siegbahn, A., Spector, T. D., Stefansson, K., Strachan, D. P., Tayo, B. O., Tremoli, E., Tuomilehto, J., Uusitupa, M., van Duijn, C. M., Vollenweider, P., Wallentin, L., Wareham, N. J., Whitfield, J. B., Wolffenbuttel, B. H., Ordovas, J. M., Boerwinkle, E., Palmer, C. N., Thorsteinsdottir, U., Chasman, D. I., Rotter, J. I., Franks, P. W., Ripatti, S., Cupples, L. A., Sandhu, M. S., Rich, S. S., Boehnke, M., Deloukas, P., Kathiresan, S., Mohlke, K. L., Ingelsson, E., Abecasis, G. R. 2013; 45 (11): 1274-1283

    Abstract

    Levels of low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, triglycerides and total cholesterol are heritable, modifiable risk factors for coronary artery disease. To identify new loci and refine known loci influencing these lipids, we examined 188,577 individuals using genome-wide and custom genotyping arrays. We identify and annotate 157 loci associated with lipid levels at P < 5 × 10(-8), including 62 loci not previously associated with lipid levels in humans. Using dense genotyping in individuals of European, East Asian, South Asian and African ancestry, we narrow association signals in 12 loci. We find that loci associated with blood lipid levels are often associated with cardiovascular and metabolic traits, including coronary artery disease, type 2 diabetes, blood pressure, waist-hip ratio and body mass index. Our results demonstrate the value of using genetic data from individuals of diverse ancestry and provide insights into the biological mechanisms regulating blood lipids to guide future genetic, biological and therapeutic research.

    View details for DOI 10.1038/ng.2797

    View details for PubMedID 24097068

  • Serum Endostatin and Risk of Mortality in the Elderly Findings From 2 Community-Based Cohorts ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Arnlov, J., Ruge, T., Ingelsson, E., Larsson, A., Sundstrom, J., Lind, L. 2013; 33 (11): 2689-2695

    Abstract

    Experimental data imply that endostatin, a proteolytically cleaved fragment of collagen XVIII, could be involved in the development of cardiovascular disease and cancer. Prospective data concerning the relation between circulating endostatin and mortality are lacking. Accordingly, we aimed to study associations between circulating endostatin and mortality risk.Serum endostatin was analyzed in 2 community-based cohorts: the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS; women 50%, n=931; mean age, 70 years; median follow-up, 7.9 years) and the Uppsala Longitudinal Study of Adult Men (ULSAM; n=748; mean age, 77 years; median follow-up, 9.7 years). During follow-up, 90 participants died in PIVUS (1.28/100 person-years at risk), and 417 participants died in ULSAM (6.7/100 person-years at risk). In multivariable Cox regression models adjusted for age and established cardiovascular risk factors, 1 SD higher ln(serum endostatin level) was associated with a hazard ratio of mortality of 1.39 and 95% confidence interval, 1.26 to 1.53, on average in both cohorts. In the ULSAM cohort, serum endostatin was also associated with cardiovascular mortality (177 deaths; hazard ratio per SD of ln[endostatin] 1.45, 95% confidence interval [1.25-1.71]) and cancer mortality (115 deaths; hazard ratio per SD of ln[endostatin] 1.35, 95% confidence interval [1.10-1.66]).High serum endostatin was associated with increased mortality risk in 2 independent community-based cohorts of the elderly. Our observational data support the importance of extracellular matrix remodeling in the underlying pathophysiology of cardiovascular disease and cancer.

    View details for DOI 10.1161/ATVBAHA.113.301704

    View details for Web of Science ID 000326418700033

    View details for PubMedID 24030549

  • Discovery and refinement of loci associated with lipid levels. Nature genetics Willer, C. J., Schmidt, E. M., Sengupta, S., Peloso, G. M., Gustafsson, S., Kanoni, S., Ganna, A., Chen, J., Buchkovich, M. L., Mora, S., Beckmann, J. S., Bragg-Gresham, J. L., Chang, H., Demirkan, A., den Hertog, H. M., Do, R., Donnelly, L. A., Ehret, G. B., Esko, T., Feitosa, M. F., Ferreira, T., Fischer, K., Fontanillas, P., Fraser, R. M., Freitag, D. F., Gurdasani, D., Heikkilä, K., Hyppönen, E., Isaacs, A., Jackson, A. U., Johansson, A., Johnson, T., Kaakinen, M., Kettunen, J., Kleber, M. E., Li, X., Luan, J., Lyytikäinen, L., Magnusson, P. K., Mangino, M., Mihailov, E., Montasser, M. E., Müller-Nurasyid, M., Nolte, I. M., O'Connell, J. R., Palmer, C. D., Perola, M., Petersen, A., Sanna, S., Saxena, R., Service, S. K., Shah, S., Shungin, D., Sidore, C., Song, C., Strawbridge, R. J., Surakka, I., Tanaka, T., Teslovich, T. M., Thorleifsson, G., van den Herik, E. G., Voight, B. F., Volcik, K. A., Waite, L. L., Wong, A., Wu, Y., Zhang, W., Absher, D., Asiki, G., Barroso, I., Been, L. F., Bolton, J. L., Bonnycastle, L. L., Brambilla, P., Burnett, M. S., Cesana, G., Dimitriou, M., Doney, A. S., Döring, A., Elliott, P., Epstein, S. E., Eyjolfsson, G. I., Gigante, B., Goodarzi, M. O., Grallert, H., Gravito, M. L., Groves, C. J., Hallmans, G., Hartikainen, A., Hayward, C., Hernandez, D., Hicks, A. A., Holm, H., Hung, Y., Illig, T., Jones, M. R., Kaleebu, P., Kastelein, J. J., Khaw, K., Kim, E., Klopp, N., Komulainen, P., Kumari, M., Langenberg, C., Lehtimäki, T., Lin, S., Lindström, J., Loos, R. J., Mach, F., McArdle, W. L., Meisinger, C., Mitchell, B. D., Müller, G., Nagaraja, R., Narisu, N., Nieminen, T. V., Nsubuga, R. N., Olafsson, I., Ong, K. K., Palotie, A., Papamarkou, T., Pomilla, C., Pouta, A., Rader, D. J., Reilly, M. P., Ridker, P. M., Rivadeneira, F., Rudan, I., Ruokonen, A., Samani, N., Scharnagl, H., Seeley, J., Silander, K., Stancáková, A., Stirrups, K., Swift, A. J., Tiret, L., Uitterlinden, A. G., van Pelt, L. J., Vedantam, S., Wainwright, N., Wijmenga, C., Wild, S. H., Willemsen, G., Wilsgaard, T., Wilson, J. F., Young, E. H., Zhao, J. H., Adair, L. S., Arveiler, D., Assimes, T. L., Bandinelli, S., Bennett, F., Bochud, M., Boehm, B. O., Boomsma, D. I., Borecki, I. B., Bornstein, S. R., Bovet, P., Burnier, M., Campbell, H., Chakravarti, A., Chambers, J. C., Chen, Y. I., Collins, F. S., Cooper, R. S., Danesh, J., Dedoussis, G., de Faire, U., Feranil, A. B., Ferrières, J., Ferrucci, L., Freimer, N. B., Gieger, C., Groop, L. C., Gudnason, V., Gyllensten, U., Hamsten, A., Harris, T. B., Hingorani, A., Hirschhorn, J. N., Hofman, A., Hovingh, G. K., Hsiung, C. A., Humphries, S. E., Hunt, S. C., Hveem, K., Iribarren, C., Järvelin, M., Jula, A., Kähönen, M., Kaprio, J., Kesäniemi, A., Kivimaki, M., Kooner, J. S., Koudstaal, P. J., Krauss, R. M., Kuh, D., Kuusisto, J., Kyvik, K. O., Laakso, M., Lakka, T. A., Lind, L., Lindgren, C. M., Martin, N. G., März, W., McCarthy, M. I., McKenzie, C. A., Meneton, P., Metspalu, A., Moilanen, L., Morris, A. D., Munroe, P. B., Njølstad, I., Pedersen, N. L., Power, C., Pramstaller, P. P., Price, J. F., Psaty, B. M., Quertermous, T., Rauramaa, R., Saleheen, D., Salomaa, V., Sanghera, D. K., Saramies, J., Schwarz, P. E., Sheu, W. H., Shuldiner, A. R., Siegbahn, A., Spector, T. D., Stefansson, K., Strachan, D. P., Tayo, B. O., Tremoli, E., Tuomilehto, J., Uusitupa, M., van Duijn, C. M., Vollenweider, P., Wallentin, L., Wareham, N. J., Whitfield, J. B., Wolffenbuttel, B. H., Ordovas, J. M., Boerwinkle, E., Palmer, C. N., Thorsteinsdottir, U., Chasman, D. I., Rotter, J. I., Franks, P. W., Ripatti, S., Cupples, L. A., Sandhu, M. S., Rich, S. S., Boehnke, M., Deloukas, P., Kathiresan, S., Mohlke, K. L., Ingelsson, E., Abecasis, G. R. 2013; 45 (11): 1274-1283

    View details for DOI 10.1038/ng.2797

    View details for PubMedID 24097068

  • Valvular osteoclasts in calcification and aortic valve stenosis severity INTERNATIONAL JOURNAL OF CARDIOLOGY Nagy, E., Eriksson, P., Yousry, M., Caidahl, K., Ingelsson, E., Hansson, G. K., Franco-Cereceda, A., Back, M. 2013; 168 (3): 2264-2271

    Abstract

    Bone remodeling in calcified aortic valves is thought to originate from microfractures at multiple sites of the valve, at which osteoclasts and osteoblasts are recruited. The aim of the present study was to assess circulating mediators of bone homeostasis, correlate them to the severity of stenosis and explore the spatio-temporal distribution of bone turnover in different parts of calcified aortic valve tissue.Plasma and explanted aortic valves were obtained from 46 patients undergoing aortic valve replacement surgery. Plasma levels of tartrate-resistant acid phosphatase (TRAP), receptor activator of nuclear-κB (RANK) ligand and Runt-related transcription factor 2 (Runx2/Cbfa1) exhibited a significant correlation to the severity of aortic stenosis. mRNA levels in normal, thickened and calcified parts of aortic valves assessed by quantitative real-time PCR were significantly elevated in calcified parts of valves for TRAP (5.08 ± 1.6-fold, P<0.001) RANK ligand (8.6 ± 4.2-fold, P<0.001) and RANK (1.98 ± 0.78-fold, P=0.015). In an age, gender and aortic valve anatomy-adjusted multivariable regression analysis the local transcript levels of TRAP correlated significantly with echocardiographic parameters quantifying stenosis severity in early stages, whereas the expression level of Runx2/Cbfa1 was a predictor of the stenosis severity in advanced stages.These findings suggest a critical role of bone turnover as a determinant of aortic stenosis severity.

    View details for DOI 10.1016/j.ijcard.2013.01.207

    View details for Web of Science ID 000326184400088

    View details for PubMedID 23452891

  • Mendelian randomization studies do not support a causal role for reduced circulating adiponectin levels in insulin resistance and type 2 diabetes. Diabetes Yaghootkar, H., Lamina, C., Scott, R. A., Dastani, Z., Hivert, M., Warren, L. L., Stancáková, A., Buxbaum, S. G., Lyytikäinen, L., Henneman, P., Wu, Y., Cheung, C. Y., Pankow, J. S., Jackson, A. U., Gustafsson, S., Zhao, J. H., Ballantyne, C. M., Xie, W., Bergman, R. N., Boehnke, M., El Bouazzaoui, F., Collins, F. S., Dunn, S. H., Dupuis, J., Forouhi, N. G., Gillson, C., Hattersley, A. T., Hong, J., Kähönen, M., Kuusisto, J., Kedenko, L., Kronenberg, F., Doria, A., Assimes, T. L., Ferrannini, E., Hansen, T., Hao, K., Häring, H., Knowles, J. W., Lindgren, C. M., Nolan, J. J., Paananen, J., Pedersen, O., Quertermous, T., Smith, U., Lehtimäki, T., Liu, C., Loos, R. J., McCarthy, M. I., Morris, A. D., Vasan, R. S., Spector, T. D., Teslovich, T. M., Tuomilehto, J., Van Dijk, K. W., Viikari, J. S., Zhu, N., Langenberg, C., Ingelsson, E., Semple, R. K., Sinaiko, A. R., Palmer, C. N., Walker, M., Lam, K. S., Paulweber, B., Mohlke, K. L., Van Duijn, C., Raitakari, O. T., Bidulescu, A., Wareham, N. J., Laakso, M., Waterworth, D. M., Lawlor, D. A., Meigs, J. B., Richards, J. B., Frayling, T. M. 2013; 62 (10): 3589-3598

    Abstract

    Adiponectin is strongly inversely associated with insulin resistance and type 2 diabetes, but its causal role remains controversial. We used a Mendelian randomization approach to test the hypothesis that adiponectin causally influences insulin resistance and type 2 diabetes. We used genetic variants at the ADIPOQ gene as instruments to calculate a regression slope between adiponectin levels and metabolic traits (up to 31,000 individuals) and a combination of instrumental variables and summary statistics-based genetic risk scores to test the associations with gold-standard measures of insulin sensitivity (2,969 individuals) and type 2 diabetes (15,960 case subjects and 64,731 control subjects). In conventional regression analyses, a 1-SD decrease in adiponectin levels was correlated with a 0.31-SD (95% CI 0.26-0.35) increase in fasting insulin, a 0.34-SD (0.30-0.38) decrease in insulin sensitivity, and a type 2 diabetes odds ratio (OR) of 1.75 (1.47-2.13). The instrumental variable analysis revealed no evidence of a causal association between genetically lower circulating adiponectin and higher fasting insulin (0.02 SD; 95% CI -0.07 to 0.11; N = 29,771), nominal evidence of a causal relationship with lower insulin sensitivity (-0.20 SD; 95% CI -0.38 to -0.02; N = 1,860), and no evidence of a relationship with type 2 diabetes (OR 0.94; 95% CI 0.75-1.19; N = 2,777 case subjects and 13,011 control subjects). Using the ADIPOQ summary statistics genetic risk scores, we found no evidence of an association between adiponectin-lowering alleles and insulin sensitivity (effect per weighted adiponectin-lowering allele: -0.03 SD; 95% CI -0.07 to 0.01; N = 2,969) or type 2 diabetes (OR per weighted adiponectin-lowering allele: 0.99; 95% CI 0.95-1.04; 15,960 case subjects vs. 64,731 control subjects). These results do not provide any consistent evidence that interventions aimed at increasing adiponectin levels will improve insulin sensitivity or risk of type 2 diabetes.

    View details for DOI 10.2337/db13-0128

    View details for PubMedID 23835345

    View details for PubMedCentralID PMC3781444

  • Multilocus Genetic Risk Scores for Coronary Heart Disease Prediction ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Ganna, A., Magnusson, P. K., Pedersen, N. L., de Faire, U., Reilly, M., Arnloe, J., Sundstrom, J., Hamsten, A., Ingelsson, E. 2013; 33 (9): 2267-2272

    Abstract

    Current guidelines do not support the use of genetic profiles in risk assessment of coronary heart disease (CHD). However, new single nucleotide polymorphisms associated with CHD and intermediate cardiovascular traits have recently been discovered. We aimed to compare several multilocus genetic risk score (MGRS) in terms of association with CHD and to evaluate clinical use.We investigated 6 Swedish prospective cohort studies with 10 612 participants free of CHD at baseline. We developed 1 overall MGRS based on 395 single nucleotide polymorphisms reported as being associated with cardiovascular traits, 1 CHD-specific MGRS, including 46 single nucleotide polymorphisms, and 6 trait-specific MGRS for each established CHD risk factors. Both the overall and the CHD-specific MGRS were significantly associated with CHD risk (781 incident events; hazard ratios for fourth versus first quartile, 1.54 and 1.52; P<0.001) and improved risk classification beyond established risk factors (net reclassification improvement, 4.2% and 4.9%; P=0.006 and 0.017). Discrimination improvement was modest (C-index improvement, 0.004). A polygene MGRS performed worse than the CHD-specific MGRS. We estimate that 1 additional CHD event for every 318 people screened at intermediate risk could be saved by measuring the CHD-specific genetic score in addition to the established risk factors.Our results indicate that genetic information could be of some clinical value for prediction of CHD, although further studies are needed to address aspects, such as feasibility, ethics, and cost efficiency of genetic profiling in the primary prevention setting.

    View details for DOI 10.1161/ATVBAHA.113.301218

    View details for Web of Science ID 000323320700032

    View details for PubMedID 23685553

  • Global DNA hypermethylation is associated with high serum levels of persistent organic pollutants in an elderly population ENVIRONMENT INTERNATIONAL Lind, L., Penell, J., Luttropp, K., Nordfors, L., Syvanen, A., Axelsson, T., Salihovic, S., van Bavel, B., Fall, T., Ingelsson, E., Lind, P. M. 2013; 59: 456-461

    Abstract

    Dioxin exposure has experimentally been associated with changes in DNA methylation, an epigenetic change that is associated with disease. The present study aims to investigate if serum levels of dioxin and other persistent environmental pollutants are related to global DNA methylation in a human sample. In the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (all aged 70), global DNA methylation was measured by the Luminometric Methylation Assay in 524 subjects. Twenty-three different POPs, including 16 PCBs, five pesticides, one dioxin (OCDD) and one brominated flame retardant (BDE47) were analysed by HRGC/HRMS. Ten single nucleotide polymorphisms (SNPs) in the Aryl hydrocarbon (Ah)-receptor were analysed by mini-sequencing. High levels of toxic equivalency (TEQ) for PCBs and dioxin were associated with DNA hypermethylation (p=0.030). This was mainly attributed to coplanar non-ortho PCBs. While no significant associations were found between DNA methylation and SNPs in the Ah-receptor, an interaction was found between the SNP rs2237297 and TEQ so that TEQ was associated with hypermethylation (p=0.009) only in subjects with one G-allele (n=103). Also high levels of the PCB126 congener, the OCDD, and the pesticide metabolite p,p'-DDE were related to DNA hypermethylation (p=0.01, 0.03 and 0.003, respectively). In conclusion, in a sample of elderly subjects, high TEQ including PCBs and the dioxin OCDD and high serum levels of PCB126, OCDD, and p,p'-DDE were related to global DNA hypermethylation in a cross-sectional analysis.

    View details for DOI 10.1016/j.envint.2013.07.008

    View details for Web of Science ID 000324901000049

    View details for PubMedID 23933504

  • Meta-analysis of Gene-Level Associations for Rare Variants Based on Single-Variant Statistics AMERICAN JOURNAL OF HUMAN GENETICS Hu, Y., Berndt, S. I., Gustafsson, S., Ganna, A., Hirschhorn, J., North, K. E., Ingelsson, E., Lin, D. 2013; 93 (2): 236-248

    Abstract

    Meta-analysis of genome-wide association studies (GWASs) has led to the discoveries of many common variants associated with complex human diseases. There is a growing recognition that identifying "causal" rare variants also requires large-scale meta-analysis. The fact that association tests with rare variants are performed at the gene level rather than at the variant level poses unprecedented challenges in the meta-analysis. First, different studies may adopt different gene-level tests, so the results are not compatible. Second, gene-level tests require multivariate statistics (i.e., components of the test statistic and their covariance matrix), which are difficult to obtain. To overcome these challenges, we propose to perform gene-level tests for rare variants by combining the results of single-variant analysis (i.e., p values of association tests and effect estimates) from participating studies. This simple strategy is possible because of an insight that multivariate statistics can be recovered from single-variant statistics, together with the correlation matrix of the single-variant test statistics, which can be estimated from one of the participating studies or from a publicly available database. We show both theoretically and numerically that the proposed meta-analysis approach provides accurate control of the type I error and is as powerful as joint analysis of individual participant data. This approach accommodates any disease phenotype and any study design and produces all commonly used gene-level tests. An application to the GWAS summary results of the Genetic Investigation of ANthropometric Traits (GIANT) consortium reveals rare and low-frequency variants associated with human height. The relevant software is freely available.

    View details for DOI 10.1016/j.ajhg.2013.06.011

    View details for Web of Science ID 000323186200004

    View details for PubMedID 23891470

  • Genetic variation in the dimethylarginine dimethylaminohydrolase 1 gene (DDAH1) is related to asymmetric dimethylarginine (ADMA) levels, but not to endothelium-dependent vasodilation VASCULAR MEDICINE Lind, L., Ingelsson, E., Kumar, J., Syvanen, A., Axelsson, T., Teerlink, T. 2013; 18 (4): 192-199

    Abstract

    Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthase. The breakdown of ADMA is mainly governed by the activity of dimethylarginine dimethylaminohydrolases (DDAHs). We investigated if genetic variation in the DDAH1 and DDAH2 genes were related to ADMA and l-arginine levels, as well as measures of endothelium-dependent vasodilation.In 1016 70-year-old participants of the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (50% women), we measured endothelium-dependent vasodilation (EDV) using the invasive forearm technique with acetylcholine given in the brachial artery and the brachial artery ultrasound technique with measurement of flow-mediated dilatation (FMD). Plasma l-arginine and ADMA levels were measured by high-performance liquid chromatography and 55 single nucleotide polymorphisms (SNPs) in the DDAH1 and DDAH2 genes were genotyped.Several of the genotypes in the DDAH1 gene were highly significantly related to ADMA levels (p = 10(-7) at best), but not to the l-arginine levels. No relationships between the genotypes in the DDAH2 gene and ADMA or l-arginine levels were found. None of the DDAH1 genotypes being closely related to ADMA levels were significantly related to EDV or FMD. Neither were any of the DDAH2 genotypes closely related to any of the measurements of vasoreactivity.A close relationship was seen between SNPs in the DDAH1, but not DDAH2, gene and ADMA levels. However, variation in those genes was not related to measures of EDV in this elderly population.

    View details for DOI 10.1177/1358863X13496488

    View details for Web of Science ID 000323328500003

    View details for PubMedID 23892448

  • Oxidative Stress and Inflammatory Markers in Relation to Circulating Levels of Adiponectin OBESITY Gustafsson, S., Lind, L., Soderberg, S., Zilmer, M., Hulthe, J., Ingelsson, E. 2013; 21 (7): 1467-1473

    Abstract

    Previous epidemiological studies together with animal studies have suggested an association between adiponectin and oxidative stress and inflammation, but community-based studies are lacking. Our objective was to investigate the relative importance of oxidative stress and inflammatory markers, representing different pathways in relation to adiponectin.In a cross-sectional sample of 929 70-year-old individuals (50% women) of the Prospective Investigation of the Vasculature in Uppsala Seniors study, relations between serum adiponectin and oxidative stress [conjugated dienes (CD), homocysteine, total antioxidant capacity, oxidized low-density lipoprotein (OxLDL), OxLDL antibodies, baseline CD of LDL, glutathione (GSH), total glutathione (TGSH), glutathione disulfide], circulation interleukins (IL-6, IL-8), other cytokines [tumor necrosis factor α, monocyte chemotactic protein-1 (MCP-1), epidermal growth factor (EGF), vascular endothelial growth factor], cell adhesion molecules (vascular cell adhesion molecule-1, intercellular adhesion molecule-1, E-selectin, P-selectin, L-selectin), and systemic inflammatory markers [C-reactive protein (CRP), leukocyte count] in separate models were investigated.In age- and sex-adjusted, as well as multivariable-adjusted models, adiponectin was significantly and positively associated with GSH, log TGSH, whereas an inverse association was observed for CD and log EGF. An inverse association between adiponectin and MCP-1, log E-selectin, and log CRP was significant in age- and sex-adjusted models, but not in multivariable-adjusted models.Our results imply that higher levels of adiponectin are associated with a more beneficial oxidative stress profile, with higher levels of principal anti-oxidative GSH and total GSH together with lower levels of lipid peroxidation, possibly through shared pathways. Further studies are needed to investigate whether changes in the oxidative stress profile may be a mechanism linking adiponectin with type 2 diabetes and/or cardiovascular disease.

    View details for DOI 10.1002/oby.20097

    View details for Web of Science ID 000323351400022

    View details for PubMedID 23585283

  • Gene x Physical Activity Interactions in Obesity: Combined Analysis of 111,421 Individuals of European Ancestry PLOS GENETICS Ahmad, S., Rukh, G., Varga, T. V., Ali, A., Kurbasic, A., Shungin, D., Ericson, U., Koivula, R. W., Chu, A. Y., Rose, L. M., Ganna, A., Qi, Q., Stancakova, A., Sandholt, C. H., Elks, C. E., Curhan, G., Jensen, M. K., Tamimi, R. M., Allin, K. H., Jorgensen, T., Brage, S., Langenberg, C., Aadahl, M., Grarup, N., Linneberg, A., Pare, G., Magnusson, P. K., Pedersen, N. L., Boehnke, M., Hamsten, A., Mohlke, K. L., Pasquale, L. T., Pedersen, O., Scott, R. A., Ridker, P. M., Ingelsson, E., Laakso, M., Hansen, T., Qi, L., Wareham, N. J., Chasman, D. I., Hallmans, G., Hu, F. B., Renstrom, F., Orho-Melander, M., Franks, P. W. 2013; 9 (7)

    Abstract

    Numerous obesity loci have been identified using genome-wide association studies. A UK study indicated that physical activity may attenuate the cumulative effect of 12 of these loci, but replication studies are lacking. Therefore, we tested whether the aggregate effect of these loci is diminished in adults of European ancestry reporting high levels of physical activity. Twelve obesity-susceptibility loci were genotyped or imputed in 111,421 participants. A genetic risk score (GRS) was calculated by summing the BMI-associated alleles of each genetic variant. Physical activity was assessed using self-administered questionnaires. Multiplicative interactions between the GRS and physical activity on BMI were tested in linear and logistic regression models in each cohort, with adjustment for age, age(2), sex, study center (for multicenter studies), and the marginal terms for physical activity and the GRS. These results were combined using meta-analysis weighted by cohort sample size. The meta-analysis yielded a statistically significant GRS × physical activity interaction effect estimate (Pinteraction  = 0.015). However, a statistically significant interaction effect was only apparent in North American cohorts (n = 39,810, Pinteraction  = 0.014 vs. n = 71,611, Pinteraction  = 0.275 for Europeans). In secondary analyses, both the FTO rs1121980 (Pinteraction  = 0.003) and the SEC16B rs10913469 (Pinteraction  = 0.025) variants showed evidence of SNP × physical activity interactions. This meta-analysis of 111,421 individuals provides further support for an interaction between physical activity and a GRS in obesity disposition, although these findings hinge on the inclusion of cohorts from North America, indicating that these results are either population-specific or non-causal.

    View details for DOI 10.1371/journal.pgen.1003607

    View details for Web of Science ID 000322321100013

    View details for PubMedID 23935507

  • Clinical depression, antidepressant use and risk of future cardiovascular disease EUROPEAN JOURNAL OF EPIDEMIOLOGY Rahman, I., Humphreys, K., Bennet, A. M., Ingelsson, E., Pedersen, N. L., Magnusson, P. K. 2013; 28 (7): 589-595

    Abstract

    Many studies have shown that depression contributes to a higher risk of developing cardiovascular disease (CVD). Use of antidepressants and its association with CVD development has also been investigated previously but the results have been conflicting. Further, depression and use of antidepressants have been more widely studied in relation to coronary heart disease rather than stroke. A population-based cohort study consisting of 36,654 Swedish elderly twins was conducted with a follow-up of maximum 4 years. Information on exposures, outcomes and covariates were collected from the Swedish national patient registers, the Swedish prescribed drug registry and the Swedish twin registry. Depression and antidepressant use were both associated with CVD development. The risk was most pronounced among depressed patients who did not use antidepressants (HR 1. 48, CI 1.10-2.00). When assessing the two main CVD outcomes coronary heart disease and ischemic stroke separately, the predominant association was found for ischemic stroke while it was absent for coronary heart disease. The association between depression and stroke also remained significant when restricting to depression diagnoses occurring at least 10 years before baseline. The study supports that depression is a possible risk factor for development of CVD. Moreover, the hazard rate for CVD outcomes was highest among depressed patients who had not used antidepressants. The association with clinical depression is more marked in relation to stroke and disappears in relation to development of coronary heart disease.

    View details for DOI 10.1007/s10654-013-9821-z

    View details for Web of Science ID 000323836900007

    View details for PubMedID 23836399

  • GWAS of 126,559 Individuals Identifies Genetic Variants Associated with Educational Attainment SCIENCE Rietveld, C. A., Medland, S. E., Derringer, J., Yang, J., Esko, T., Martin, N. W., Westra, H., Shakhbazov, K., Abdellaoui, A., Agrawal, A., Albrecht, E., Alizadeh, B. Z., Amin, N., Bamard, J., Baumeister, S. E., Benke, K. S., Bielak, L. F., Boatman, J. A., Boyle, P. A., Davies, G., de Leeuw, C., Eklund, N., Evans, D. S., Ferhmann, R., Fischer, K., Gieger, C., Gjessing, H. K., Haegg, S., Harris, J. R., Hayward, C., Holzapfel, C., Ibrahim-Verbaas, C. A., Ingelsson, E., Jacobsson, B., Joshi, P. K., Jugessur, A., Kaakinen, M., Kanoni, S., Karjalainen, J., Kolcic, I., Kristiansson, K., Kutalik, Z., Lahti, J., Lee, S. H., Lin, P., Lind, P. A., Liu, Y., Lohman, K., Loitfelder, M., McMahon, G., Vidal, P. M., Meirelles, O., Milani, L., Myhre, R., Nuotio, M., Oldmeadow, C. J., Petrovic, K. E., Peyrot, W. J., Polasek, O., Quaye, L., Reinmaa, E., Rice, J. P., Rizzi, T. S., Schmidt, H., Schmidt, R., Smith, A. V., Smith, J. A., Tanaka, T., Terracciano, A., van der Loos, M. J., Vitart, V., Voelzke, H., Wellmann, J., Yu, L., Zhao, W., Allik, J., Attia, J. R., Bandinelli, S., Bastardot, F., Beauchamp, J., Bennett, D. A., Berger, K., Bierut, L. J., Boomsma, D. I., Bueltmann, U., Campbell, H., Chabris, C. F., Cherkas, L., Chung, M. K., Cucca, F., de Andrade, M., De Jager, P. L., De Neve, J., Deary, I. J., Dedoussis, G. V., Deloukas, P., Dimitriou, M., Eiriksdottir, G., Elderson, M. F., Eriksson, J. G., Evans, D. M., Faul, J. D., Ferrucci, L., Garcia, M. E., Groenberg, H., Guonason, V., Hall, P., Harris, J. M., Harris, T. B., Hastie, N. D., Heath, A. C., Hernandez, D. G., Hoffmann, W., Hofman, A., Holle, R., Holliday, E. G., Hottenga, J., Iacono, W. G., Illig, T., Jaervelin, M., Kaehoenen, M., Kaprio, J., Kirkpatrick, R. M., Kowgier, M., Latvala, A., Launer, L. J., Lawlor, D. A., Lehtimaeki, T., Li, J., Lichtenstein, P., Lichtner, P., Liewald, D. C., Madden, P. A., Magnusson, P. K., Maekinen, T. E., Masala, M., McGue, M., Metspalu, A., Mielck, A., Miller, M. B., Montgomery, G. W., Mukherjee, S., Nyholt, D. R., Oostra, B. A., Palmer, L. J., Palotie, A., Penninx, B. W., Perola, M., Peyser, P. A., Preisig, M., Raeikkoenen, K., Raitakari, O. T., Realo, A., Ring, S. M., Ripatti, S., Rivadeneira, F., Rudan, I., Rustichini, A., Salomaa, V., Sarin, A., Schlessinger, D., Scott, R. J., Snieder, H., St Pourcain, B., Starr, J. M., Sul, J. H., Surakka, I., Svento, R., Teumer, A., Tiemeier, H., van Rooij, F. J., Van Wagoner, D. R., Vartiainen, E., Viikari, J., Vollenweider, P., Vonk, J. M., Waeber, G., Weir, D. R., Wichmann, H., Widen, E., Willemsen, G., Wilson, J. F., Wright, A. F., Conley, D., Davey-Smith, G., Franke, L., Groenen, P. J., Hofman, A., Johannesson, M., Kardia, S. L., Krueger, R. F., Laibson, D., Martin, N. G., Meyer, M. N., Posthuma, D., Thurik, A. R., Timpson, N. J., Uitterlinden, A. G., van Duijn, C. M., Visscher, P. M., Benjamin, D. J., Cesarini, D., Koellinger, P. D. 2013; 340 (6139): 1467-1471

    Abstract

    A genome-wide association study (GWAS) of educational attainment was conducted in a discovery sample of 101,069 individuals and a replication sample of 25,490. Three independent single-nucleotide polymorphisms (SNPs) are genome-wide significant (rs9320913, rs11584700, rs4851266), and all three replicate. Estimated effects sizes are small (coefficient of determination R(2) ≈ 0.02%), approximately 1 month of schooling per allele. A linear polygenic score from all measured SNPs accounts for ≈2% of the variance in both educational attainment and cognitive function. Genes in the region of the loci have previously been associated with health, cognitive, and central nervous system phenotypes, and bioinformatics analyses suggest the involvement of the anterior caudate nucleus. These findings provide promising candidate SNPs for follow-up work, and our effect size estimates can anchor power analyses in social-science genetics.

    View details for DOI 10.1126/science.1235488

    View details for Web of Science ID 000320647000046

    View details for PubMedID 23722424

  • Sex-stratified genome-wide association studies including 270,000 individuals show sexual dimorphism in genetic loci for anthropometric traits. PLoS genetics Randall, J. C., Winkler, T. W., Kutalik, Z., Berndt, S. I., Jackson, A. U., Monda, K. L., Kilpeläinen, T. O., Esko, T., Mägi, R., Li, S., Workalemahu, T., Feitosa, M. F., Croteau-Chonka, D. C., Day, F. R., Fall, T., Ferreira, T., Gustafsson, S., Locke, A. E., Mathieson, I., Scherag, A., Vedantam, S., Wood, A. R., Liang, L., Steinthorsdottir, V., Thorleifsson, G., Dermitzakis, E. T., Dimas, A. S., Karpe, F., Min, J. L., Nicholson, G., Clegg, D. J., Person, T., Krohn, J. P., Bauer, S., Buechler, C., Eisinger, K., Bonnefond, A., Froguel, P., Hottenga, J., Prokopenko, I., Waite, L. L., Harris, T. B., Smith, A. V., Shuldiner, A. R., McArdle, W. L., Caulfield, M. J., Munroe, P. B., Grönberg, H., Chen, Y. I., Li, G., Beckmann, J. S., Johnson, T., Thorsteinsdottir, U., Teder-Laving, M., Khaw, K., Wareham, N. J., Zhao, J. H., Amin, N., Oostra, B. A., Kraja, A. T., Province, M. A., Cupples, L. A., Heard-Costa, N. L., Kaprio, J., Ripatti, S., Surakka, I., Collins, F. S., Saramies, J., Tuomilehto, J., Jula, A., Salomaa, V., Erdmann, J., Hengstenberg, C., Loley, C., Schunkert, H., Lamina, C., Wichmann, H. E., Albrecht, E., Gieger, C., Hicks, A. A., Johansson, A., Pramstaller, P. P., Kathiresan, S., Speliotes, E. K., Penninx, B., Hartikainen, A., Jarvelin, M., Gyllensten, U., Boomsma, D. I., Campbell, H., Wilson, J. F., Chanock, S. J., Farrall, M., Goel, A., Medina-Gomez, C., Rivadeneira, F., Estrada, K., Uitterlinden, A. G., Hofman, A., Zillikens, M. C., den Heijer, M., Kiemeney, L. A., Maschio, A., Hall, P., Tyrer, J., Teumer, A., Völzke, H., Kovacs, P., Tönjes, A., Mangino, M., Spector, T. D., Hayward, C., Rudan, I., Hall, A. S., Samani, N. J., Attwood, A. P., Sambrook, J. G., Hung, J., Palmer, L. J., Lokki, M., Sinisalo, J., Boucher, G., Huikuri, H., Lorentzon, M., Ohlsson, C., Eklund, N., Eriksson, J. G., Barlassina, C., Rivolta, C., Nolte, I. M., Snieder, H., van der Klauw, M. M., van Vliet-Ostaptchouk, J. V., Gejman, P. V., Shi, J., Jacobs, K. B., Wang, Z., Bakker, S. J., Mateo Leach, I., Navis, G., van der Harst, P., Martin, N. G., Medland, S. E., Montgomery, G. W., Yang, J., Chasman, D. I., Ridker, P. M., Rose, L. M., Lehtimäki, T., Raitakari, O., Absher, D., Iribarren, C., Basart, H., Hovingh, K. G., Hyppönen, E., Power, C., Anderson, D., Beilby, J. P., Hui, J., Jolley, J., Sager, H., Bornstein, S. R., Schwarz, P. E., Kristiansson, K., Perola, M., Lindström, J., Swift, A. J., Uusitupa, M., Atalay, M., Lakka, T. A., Rauramaa, R., Bolton, J. L., Fowkes, G., Fraser, R. M., Price, J. F., Fischer, K., Krjutå Kov, K., Metspalu, A., Mihailov, E., Langenberg, C., Luan, J., Ong, K. K., Chines, P. S., Keinanen-Kiukaanniemi, S. M., Saaristo, T. E., Edkins, S., Franks, P. W., Hallmans, G., Shungin, D., Morris, A. D., Palmer, C. N., Erbel, R., Moebus, S., Nöthen, M. M., Pechlivanis, S., Hveem, K., Narisu, N., Hamsten, A., Humphries, S. E., Strawbridge, R. J., Tremoli, E., Grallert, H., Thorand, B., Illig, T., Koenig, W., Müller-Nurasyid, M., Peters, A., Boehm, B. O., Kleber, M. E., März, W., Winkelmann, B. R., Kuusisto, J., Laakso, M., Arveiler, D., Cesana, G., Kuulasmaa, K., Virtamo, J., Yarnell, J. W., Kuh, D., Wong, A., Lind, L., de Faire, U., Gigante, B., Magnusson, P. K., Pedersen, N. L., Dedoussis, G., Dimitriou, M., Kolovou, G., Kanoni, S., Stirrups, K., Bonnycastle, L. L., Njølstad, I., Wilsgaard, T., Ganna, A., Rehnberg, E., Hingorani, A., Kivimaki, M., Kumari, M., Assimes, T. L., Barroso, I., Boehnke, M., Borecki, I. B., Deloukas, P., Fox, C. S., Frayling, T., Groop, L. C., Haritunians, T., Hunter, D., Ingelsson, E., Kaplan, R., Mohlke, K. L., O'Connell, J. R., Schlessinger, D., Strachan, D. P., Stefansson, K., van Duijn, C. M., Abecasis, G. R., McCarthy, M. I., Hirschhorn, J. N., Qi, L., Loos, R. J., Lindgren, C. M., North, K. E., Heid, I. M. 2013; 9 (6): e1003500

    Abstract

    Given the anthropometric differences between men and women and previous evidence of sex-difference in genetic effects, we conducted a genome-wide search for sexually dimorphic associations with height, weight, body mass index, waist circumference, hip circumference, and waist-to-hip-ratio (133,723 individuals) and took forward 348 SNPs into follow-up (additional 137,052 individuals) in a total of 94 studies. Seven loci displayed significant sex-difference (FDR<5%), including four previously established (near GRB14/COBLL1, LYPLAL1/SLC30A10, VEGFA, ADAMTS9) and three novel anthropometric trait loci (near MAP3K1, HSD17B4, PPARG), all of which were genome-wide significant in women (P<5×10(-8)), but not in men. Sex-differences were apparent only for waist phenotypes, not for height, weight, BMI, or hip circumference. Moreover, we found no evidence for genetic effects with opposite directions in men versus women. The PPARG locus is of specific interest due to its role in diabetes genetics and therapy. Our results demonstrate the value of sex-specific GWAS to unravel the sexually dimorphic genetic underpinning of complex traits.

    View details for DOI 10.1371/journal.pgen.1003500

    View details for PubMedID 23754948

    View details for PubMedCentralID PMC3674993

  • Sortilin receptor 1 predicts longitudinal cognitive change NEUROBIOLOGY OF AGING Reynolds, C. A., Zavala, C., Gatz, M., Vie, L., Johansson, B., Malmberg, B., Ingelsson, E., Prince, J. A., Pedersen, N. L. 2013; 34 (6)

    Abstract

    The gene encoding sortilin receptor 1 (SORL1) has been associated with Alzheimer's disease risk. We examined 15 SORL1 variants and single nucleotide polymorphism (SNP) set risk scores in relation to longitudinal verbal, spatial, memory, and perceptual speed performance, testing for age trends and sex-specific effects. Altogether, 1609 individuals from 3 population-based Swedish twin studies were assessed up to 5 times across 16 years. Controlling for apolipoprotein E genotype (APOE), multiple simple and sex-moderated associations were observed for spatial, episodic memory, and verbal trajectories (p = 1.25E-03 to p = 4.83E-02). Five variants (rs11600875, rs753780, rs7105365, rs11820794, rs2070045) were associated across domains. Notably, in those homozygous for the rs2070045 risk allele, men demonstrated initially favorable performance but accelerating declines, and women showed overall lower performance. SNP set risk scores predicted spatial (Card Rotations, p = 5.92E-03) and episodic memory trajectories (Thurstone Picture Memory, p = 3.34E-02), where higher risk scores benefited men's versus women's performance up to age 75 but with accelerating declines. SORL1 is associated with cognitive aging, and might contribute differentially to change in men and women.

    View details for DOI 10.1016/j.neurobiolaging.2012.12.006

    View details for Web of Science ID 000317417100030

    View details for PubMedID 23318115

  • The Role of Adiposity in Cardiometabolic Traits: A Mendelian Randomization Analysis PLOS MEDICINE Fall, T., Hagg, S., Maegi, R., Ploner, A., Fischer, K., Horikoshi, M., Sarin, A., Thorleifsson, G., Ladenvall, C., Kals, M., Kuningas, M., Draisma, H. H., Ried, J. S., Van Zuydam, N. R., Huikari, V., Mangino, M., Sonestedt, E., Benyamin, B., Nelson, C. P., Rivera, N. V., Kristiansson, K., Shen, H., Havulinna, A. S., Dehghan, A., Donnelly, L. A., Kaakinen, M., Nuotio, M., Robertson, N., de Bruijn, R. F., Ikram, M. A., Amin, N., Balmforth, A. J., Braund, P. S., Doney, A. S., Doering, A., Elliott, P., Esko, T., Franco, O. H., Gretarsdottir, S., Hartikainen, A., Heikkila, K., Herzig, K., Holm, H., Hottenga, J. J., Hypponen, E., Illig, T., Isaacs, A., Isomaa, B., Karssen, L. C., Kettunen, J., Koenig, W., Kuulasmaa, K., Laatikainen, T., Laitinen, J., Lindgren, C., Lyssenko, V., Laara, E., Rayner, N. W., Mannisto, S., Pouta, A., Rathmann, W., Rivadeneira, F., Ruokonen, A., Savolainen, M. J., Sijbrands, E. J., Small, K. S., Smit, J. H., Steinthorsdottir, V., Syvanen, A., Taanila, A., Tobin, M. D., Uitterlinden, A. G., Willems, S. M., Willemsen, G., Witteman, J., Perola, M., Evans, A., Ferrieres, J., Virtamo, J., Kee, F., Tregouet, D., Arveiler, D., Amouyel, P., Ferrario, M. M., Brambilla, P., Hall, A. S., Heath, A., Madden, P. A., Martin, N. G., Montgomery, G. W., Whitfield, J. B., Jula, A., Knekt, P., Oostra, B., van Duijn, C. M., Penninx, B. W., Smith, G. D., Kaprio, J., Samani, N. J., Gieger, C., Peters, A., Wichmann, H., Boomsma, D. I., de Geus, E. J., Tuomi, T., Power, C., Hammond, C. J., Spector, T. D., Lind, L., Orho-Melander, M., Palmer, C. N., Morris, A. D., Groop, L., Jarvelin, M., Salomaa, V., Vartiainen, E., Hofman, A., Ripatti, S., Metspalu, A., Thorsteinsdottir, U., Stefansson, K., Pedersen, N. L., McCarthy, M. I., Ingelsson, E., Prokopenko, I. 2013; 10 (6)

    Abstract

    The association between adiposity and cardiometabolic traits is well known from epidemiological studies. Whilst the causal relationship is clear for some of these traits, for others it is not. We aimed to determine whether adiposity is causally related to various cardiometabolic traits using the Mendelian randomization approach.We used the adiposity-associated variant rs9939609 at the FTO locus as an instrumental variable (IV) for body mass index (BMI) in a Mendelian randomization design. Thirty-six population-based studies of individuals of European descent contributed to the analyses. Age- and sex-adjusted regression models were fitted to test for association between (i) rs9939609 and BMI (n  =  198,502), (ii) rs9939609 and 24 traits, and (iii) BMI and 24 traits. The causal effect of BMI on the outcome measures was quantified by IV estimators. The estimators were compared to the BMI-trait associations derived from the same individuals. In the IV analysis, we demonstrated novel evidence for a causal relationship between adiposity and incident heart failure (hazard ratio, 1.19 per BMI-unit increase; 95% CI, 1.03-1.39) and replicated earlier reports of a causal association with type 2 diabetes, metabolic syndrome, dyslipidemia, and hypertension (odds ratio for IV estimator, 1.1-1.4; all p < 0.05). For quantitative traits, our results provide novel evidence for a causal effect of adiposity on the liver enzymes alanine aminotransferase and gamma-glutamyl transferase and confirm previous reports of a causal effect of adiposity on systolic and diastolic blood pressure, fasting insulin, 2-h post-load glucose from the oral glucose tolerance test, C-reactive protein, triglycerides, and high-density lipoprotein cholesterol levels (all p < 0.05). The estimated causal effects were in agreement with traditional observational measures in all instances except for type 2 diabetes, where the causal estimate was larger than the observational estimate (p  =  0.001).We provide novel evidence for a causal relationship between adiposity and heart failure as well as between adiposity and increased liver enzymes.

    View details for DOI 10.1371/journal.pmed.1001474

    View details for Web of Science ID 000321034800017

    View details for PubMedID 23824655

  • A common variant in the FTO locus is associated with waist-hip ratio in Indian adolescents PEDIATRIC OBESITY Vasan, S. K., Fall, T., Job, V., Gu, H. F., Ingelsson, E., Brismar, K., Karpe, F., Thomas, N. 2013; 8 (3): e45-e49

    Abstract

    Common variants in the FTO locus, and near MC4R locus, have been shown to have a robust association with obesity in children and adults among various ethnic groups. Associations with obesity traits among Indian adolescents have not been determined.To study the association of rs9939609 (FTO) and rs17782313 (MC4R) to obesity related anthropometric traits in Indian adolescents.Subjects for the current study were recruited from a cross-sectional cohort of 1,230 adolescents (age mean ± SD: 17.1 ± 1.9 years) from South India.The variant at the FTO locus was found to be associated with waist-hip ratio (WHR) but not with overall obesity in this population. No significant association was observed for obesity-traits and Mc4R variant rs17782313.The common variant of FTO (rs9939609) is associated with body fat distribution during early growth in Indian adolescents and may predispose to obesity and metabolic consequences in adulthood.

    View details for DOI 10.1111/j.2047-6310.2013.00118.x

    View details for Web of Science ID 000319405000003

    View details for PubMedID 23447422

  • Serum FGF23 and Risk of Cardiovascular Events in Relation to Mineral Metabolism and Cardiovascular Pathology CLINICAL JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Arnlov, J., Carlsson, A. C., Sundstrom, J., Ingelsson, E., Larsson, A., Lind, L., Larsson, T. E. 2013; 8 (5): 781-786

    Abstract

    Circulating fibroblast growth factor-23 is associated with adverse cardiovascular outcomes in CKD and non-CKD individuals, but the underlying mechanism remains unclear. This study tested whether this association is independent of mineral metabolism and indices of subclinical cardiovascular pathology.The prospective association between fibroblast growth factor-23 and major cardiovascular events (a composite of hospital-treated myocardial infarction, hospital-treated stroke, or all-cause mortality) was investigated in the community-based Prospective Investigation of the Vasculature in Uppsala Seniors (n=973; mean age=70 years, 50% women) using multivariate logistic regression. Subjects were recruited between January of 2001 and June of 2004.During follow-up (median=5.1 years), 112 participants suffered a major cardiovascular event. In logistic regression models adjusted for age, sex, and estimated GFR, higher fibroblast growth factor-23 was associated with increased risk for major cardiovascular events (odds ratio for tertiles 2 and 3 versus tertile 1=1.92, 95% confidence interval=1.19-3.09, P<0.01). After additional adjustments in the model, adding established cardiovascular risk factors, confounders of mineral metabolism (calcium, phosphate, parathyroid hormone, and 25(OH)-vitamin D), and indices of subclinical pathology (flow-mediated vasodilation, endothelial-dependent and -independent vasodilation, arterial stiffness, and atherosclerosis and left ventricular mass) attenuated this relationship, but it remained significant (odds ratio for tertiles 2 and 3 versus tertile 1=1.69, 95% confidence interval=1.01-2.82, P<0.05).Fibroblast growth factor-23 is an independent predictor of cardiovascular events in the community, even after accounting for mineral metabolism abnormalities and subclinical cardiovascular damage. Circulating fibroblast growth factor-23 may reflect novel and important aspects of cardiovascular risk yet to be unraveled.

    View details for DOI 10.2215/CJN.09570912

    View details for Web of Science ID 000318531800013

    View details for PubMedID 23335040

  • Genetic determinants of mortality. Can findings from genome-wide association studies explain variation in human mortality? HUMAN GENETICS Ganna, A., Rivadeneira, F., Hofman, A., Uitterlinden, A. G., Magnusson, P. K., Pedersen, N. L., Ingelsson, E., Tiemeier, H. 2013; 132 (5): 553-561

    Abstract

    Twin studies have estimated the heritability of longevity to be approximately 20-30 %. Genome-wide association studies (GWAS) have revealed a large number of determinants of morbidity, but so far, no new polymorphisms have been discovered to be associated with longevity per se in GWAS. We aim to determine whether the genetic architecture of mortality can be explained by single nucleotide polymorphisms (SNPs) associated with common traits and diseases related to mortality. By extensive quality control of published GWAS we created a genetic score from 707 common SNPs associated with 125 diseases or risk factors related with overall mortality. We prospectively studied the association of the genetic score with: (1) time-to-death; (2) incidence of the first of nine major diseases (coronary heart disease, stroke, heart failure, diabetes, dementia, lung, breast, colon and prostate cancers) in two population-based cohorts of Dutch and Swedish individuals (N = 15,039; age range 47-99 years). During a median follow-up of 6.3 years (max 22.2 years), we observed 4,318 deaths and 2,132 incident disease events. The genetic score was significantly associated with time-to-death [hazard ratio (HR) per added risk allele = 1.003, P value = 0.006; HR 4th vs. 1st quartile = 1.103]. The association between the genetic score and incidence of major diseases was stronger (HR per added risk allele = 1.004, P value = 0.002; HR 4th vs. 1st quartile = 1.160). Associations were stronger for individuals dying at older ages. Our findings are compatible with the view of mortality as a complex and highly polygenetic trait, not easily explainable by common genetic variants related to diseases and physiological traits.

    View details for DOI 10.1007/s00439-013-1267-6

    View details for Web of Science ID 000317691100006

    View details for PubMedID 23354976

  • Genome-wide meta-analysis identifies 11 new loci for anthropometric traits and provides insights into genetic architecture. Nature genetics Berndt, S. I., Gustafsson, S., Mägi, R., Ganna, A., Wheeler, E., Feitosa, M. F., Justice, A. E., Monda, K. L., Croteau-Chonka, D. C., Day, F. R., Esko, T., Fall, T., Ferreira, T., Gentilini, D., Jackson, A. U., Luan, J., Randall, J. C., Vedantam, S., Willer, C. J., Winkler, T. W., Wood, A. R., Workalemahu, T., Hu, Y., Lee, S. H., Liang, L., Lin, D., Min, J. L., Neale, B. M., Thorleifsson, G., Yang, J., Albrecht, E., Amin, N., Bragg-Gresham, J. L., Cadby, G., den Heijer, M., Eklund, N., Fischer, K., Goel, A., Hottenga, J., Huffman, J. E., Jarick, I., Johansson, A., Johnson, T., Kanoni, S., Kleber, M. E., König, I. R., Kristiansson, K., Kutalik, Z., Lamina, C., Lecoeur, C., Li, G., Mangino, M., McArdle, W. L., Medina-Gomez, C., Müller-Nurasyid, M., Ngwa, J. S., Nolte, I. M., Paternoster, L., Pechlivanis, S., Perola, M., Peters, M. J., Preuss, M., Rose, L. M., Shi, J., Shungin, D., Smith, A. V., Strawbridge, R. J., Surakka, I., Teumer, A., Trip, M. D., Tyrer, J., van Vliet-Ostaptchouk, J. V., Vandenput, L., Waite, L. L., Zhao, J. H., Absher, D., Asselbergs, F. W., Atalay, M., Attwood, A. P., Balmforth, A. J., Basart, H., Beilby, J., Bonnycastle, L. L., Brambilla, P., Bruinenberg, M., Campbell, H., Chasman, D. I., Chines, P. S., Collins, F. S., Connell, J. M., Cookson, W. O., de Faire, U., de Vegt, F., Dei, M., Dimitriou, M., Edkins, S., Estrada, K., Evans, D. M., Farrall, M., Ferrario, M. M., Ferrières, J., Franke, L., Frau, F., Gejman, P. V., Grallert, H., Grönberg, H., Gudnason, V., Hall, A. S., Hall, P., Hartikainen, A., Hayward, C., Heard-Costa, N. L., Heath, A. C., Hebebrand, J., Homuth, G., Hu, F. B., Hunt, S. E., Hyppönen, E., Iribarren, C., Jacobs, K. B., Jansson, J., Jula, A., Kähönen, M., Kathiresan, S., Kee, F., Khaw, K., Kivimäki, M., Koenig, W., Kraja, A. T., Kumari, M., Kuulasmaa, K., Kuusisto, J., Laitinen, J. H., Lakka, T. A., Langenberg, C., Launer, L. J., Lind, L., Lindström, J., Liu, J., Liuzzi, A., Lokki, M., Lorentzon, M., Madden, P. A., Magnusson, P. K., Manunta, P., Marek, D., März, W., Mateo Leach, I., McKnight, B., Medland, S. E., Mihailov, E., Milani, L., Montgomery, G. W., Mooser, V., Mühleisen, T. W., Munroe, P. B., Musk, A. W., Narisu, N., Navis, G., Nicholson, G., Nohr, E. A., Ong, K. K., Oostra, B. A., Palmer, C. N., Palotie, A., Peden, J. F., Pedersen, N., Peters, A., Polasek, O., Pouta, A., Pramstaller, P. P., Prokopenko, I., Pütter, C., Radhakrishnan, A., Raitakari, O., Rendon, A., Rivadeneira, F., Rudan, I., Saaristo, T. E., Sambrook, J. G., Sanders, A. R., Sanna, S., Saramies, J., Schipf, S., Schreiber, S., Schunkert, H., Shin, S., Signorini, S., Sinisalo, J., Skrobek, B., Soranzo, N., Stancáková, A., Stark, K., Stephens, J. C., Stirrups, K., Stolk, R. P., Stumvoll, M., Swift, A. J., Theodoraki, E. V., Thorand, B., Tregouet, D., Tremoli, E., van der Klauw, M. M., van Meurs, J. B., Vermeulen, S. H., Viikari, J., Virtamo, J., Vitart, V., Waeber, G., Wang, Z., Widén, E., Wild, S. H., Willemsen, G., Winkelmann, B. R., Witteman, J. C., Wolffenbuttel, B. H., Wong, A., Wright, A. F., Zillikens, M. C., Amouyel, P., Boehm, B. O., Boerwinkle, E., Boomsma, D. I., Caulfield, M. J., Chanock, S. J., Cupples, L. A., Cusi, D., Dedoussis, G. V., Erdmann, J., Eriksson, J. G., Franks, P. W., Froguel, P., Gieger, C., Gyllensten, U., Hamsten, A., Harris, T. B., Hengstenberg, C., Hicks, A. A., Hingorani, A., Hinney, A., Hofman, A., Hovingh, K. G., Hveem, K., Illig, T., Jarvelin, M., Jöckel, K., Keinanen-Kiukaanniemi, S. M., Kiemeney, L. A., Kuh, D., Laakso, M., Lehtimäki, T., Levinson, D. F., Martin, N. G., Metspalu, A., Morris, A. D., Nieminen, M. S., Njølstad, I., Ohlsson, C., Oldehinkel, A. J., Ouwehand, W. H., Palmer, L. J., Penninx, B., Power, C., Province, M. A., Psaty, B. M., Qi, L., Rauramaa, R., Ridker, P. M., Ripatti, S., Salomaa, V., Samani, N. J., Snieder, H., Sørensen, T. I., Spector, T. D., Stefansson, K., Tönjes, A., Tuomilehto, J., Uitterlinden, A. G., Uusitupa, M., van der Harst, P., Vollenweider, P., Wallaschofski, H., Wareham, N. J., Watkins, H., Wichmann, H., Wilson, J. F., Abecasis, G. R., Assimes, T. L., Barroso, I., Boehnke, M., Borecki, I. B., Deloukas, P., Fox, C. S., Frayling, T., Groop, L. C., Haritunian, T., Heid, I. M., Hunter, D., Kaplan, R. C., Karpe, F., Moffatt, M. F., Mohlke, K. L., O'Connell, J. R., Pawitan, Y., Schadt, E. E., Schlessinger, D., Steinthorsdottir, V., Strachan, D. P., Thorsteinsdottir, U., van Duijn, C. M., Visscher, P. M., Di Blasio, A. M., Hirschhorn, J. N., Lindgren, C. M., Morris, A. P., Meyre, D., Scherag, A., McCarthy, M. I., Speliotes, E. K., North, K. E., Loos, R. J., Ingelsson, E. 2013; 45 (5): 501-512

    Abstract

    Approaches exploiting trait distribution extremes may be used to identify loci associated with common traits, but it is unknown whether these loci are generalizable to the broader population. In a genome-wide search for loci associated with the upper versus the lower 5th percentiles of body mass index, height and waist-to-hip ratio, as well as clinical classes of obesity, including up to 263,407 individuals of European ancestry, we identified 4 new loci (IGFBP4, H6PD, RSRC1 and PPP2R2A) influencing height detected in the distribution tails and 7 new loci (HNF4G, RPTOR, GNAT2, MRPS33P4, ADCY9, HS6ST3 and ZZZ3) for clinical classes of obesity. Further, we find a large overlap in genetic structure and the distribution of variants between traits based on extremes and the general population and little etiological heterogeneity between obesity subgroups.

    View details for DOI 10.1038/ng.2606

    View details for PubMedID 23563607

    View details for PubMedCentralID PMC3973018

  • Identification of heart rate-associated loci and their effects on cardiac conduction and rhythm disorders. Nature genetics Den Hoed, M., Eijgelsheim, M., Esko, T., Brundel, B. J., Peal, D. S., Evans, D. M., Nolte, I. M., Segrè, A. V., Holm, H., Handsaker, R. E., Westra, H., Johnson, T., Isaacs, A., Yang, J., Lundby, A., Zhao, J. H., Kim, Y. J., Go, M. J., Almgren, P., Bochud, M., Boucher, G., Cornelis, M. C., Gudbjartsson, D., Hadley, D., van der Harst, P., Hayward, C., den Heijer, M., Igl, W., Jackson, A. U., Kutalik, Z., Luan, J., Kemp, J. P., Kristiansson, K., Ladenvall, C., Lorentzon, M., Montasser, M. E., Njajou, O. T., O'Reilly, P. F., Padmanabhan, S., St Pourcain, B., Rankinen, T., Salo, P., Tanaka, T., Timpson, N. J., Vitart, V., Waite, L., Wheeler, W., Zhang, W., Draisma, H. H., Feitosa, M. F., Kerr, K. F., Lind, P. A., Mihailov, E., Onland-Moret, N. C., Song, C., Weedon, M. N., Xie, W., Yengo, L., Absher, D., Albert, C. M., Alonso, A., Arking, D. E., de Bakker, P. I., Balkau, B., Barlassina, C., Benaglio, P., Bis, J. C., Bouatia-Naji, N., Brage, S., Chanock, S. J., Chines, P. S., Chung, M., Darbar, D., Dina, C., Dörr, M., Elliott, P., Felix, S. B., Fischer, K., Fuchsberger, C., de Geus, E. J., Goyette, P., Gudnason, V., Harris, T. B., Hartikainen, A., Havulinna, A. S., Heckbert, S. R., Hicks, A. A., Hofman, A., Holewijn, S., Hoogstra-Berends, F., Hottenga, J., Jensen, M. K., Johansson, A., Junttila, J., Kääb, S., Kanon, B., Ketkar, S., Khaw, K., Knowles, J. W., Kooner, A. S., Kors, J. A., Kumari, M., Milani, L., Laiho, P., Lakatta, E. G., Langenberg, C., Leusink, M., Liu, Y., Luben, R. N., Lunetta, K. L., Lynch, S. N., Markus, M. R., Marques-Vidal, P., Mateo Leach, I., McArdle, W. L., McCarroll, S. A., Medland, S. E., Miller, K. A., Montgomery, G. W., Morrison, A. C., Müller-Nurasyid, M., Navarro, P., Nelis, M., O'Connell, J. R., O'Donnell, C. J., Ong, K. K., Newman, A. B., Peters, A., Polasek, O., Pouta, A., Pramstaller, P. P., Psaty, B. M., Rao, D. C., Ring, S. M., Rossin, E. J., Rudan, D., Sanna, S., Scott, R. A., Sehmi, J. S., Sharp, S., Shin, J. T., Singleton, A. B., Smith, A. V., Soranzo, N., Spector, T. D., Stewart, C., Stringham, H. M., Tarasov, K. V., Uitterlinden, A. G., Vandenput, L., Hwang, S., Whitfield, J. B., Wijmenga, C., Wild, S. H., Willemsen, G., Wilson, J. F., Witteman, J. C., Wong, A., Wong, Q., Jamshidi, Y., Zitting, P., Boer, J. M., Boomsma, D. I., Borecki, I. B., van Duijn, C. M., Ekelund, U., Forouhi, N. G., Froguel, P., Hingorani, A., Ingelsson, E., Kivimaki, M., Kronmal, R. A., Kuh, D., Lind, L., Martin, N. G., Oostra, B. A., Pedersen, N. L., Quertermous, T., Rotter, J. I., van der Schouw, Y. T., Verschuren, W. M., Walker, M., Albanes, D., Arnar, D. O., Assimes, T. L., Bandinelli, S., Boehnke, M., de Boer, R. A., Bouchard, C., Caulfield, W. L., Chambers, J. C., Curhan, G., Cusi, D., Eriksson, J., Ferrucci, L., van Gilst, W. H., Glorioso, N., de Graaf, J., Groop, L., Gyllensten, U., Hsueh, W., Hu, F. B., Huikuri, H. V., Hunter, D. J., Iribarren, C., Isomaa, B., Jarvelin, M., Jula, A., Kähönen, M., Kiemeney, L. A., van der Klauw, M. M., Kooner, J. S., Kraft, P., Iacoviello, L., Lehtimäki, T., Lokki, M. L., Mitchell, B. D., Navis, G., Nieminen, M. S., Ohlsson, C., Poulter, N. R., Qi, L., Raitakari, O. T., Rimm, E. B., Rioux, J. D., Rizzi, F., Rudan, I., Salomaa, V., Sever, P. S., Shields, D. C., Shuldiner, A. R., Sinisalo, J., Stanton, A. V., Stolk, R. P., Strachan, D. P., Tardif, J., Thorsteinsdottir, U., Tuomilehto, J., van Veldhuisen, D. J., Virtamo, J., Viikari, J., Vollenweider, P., Waeber, G., Widen, E., Cho, Y. S., Olsen, J. V., Visscher, P. M., Willer, C., Franke, L., Erdmann, J., Thompson, J. R., Pfeufer, A., Sotoodehnia, N., Newton-Cheh, C., Ellinor, P. T., Stricker, B. H., Metspalu, A., Perola, M., Beckmann, J. S., Smith, G. D., Stefansson, K., Wareham, N. J., Munroe, P. B., Sibon, O. C., Milan, D. J., Snieder, H., Samani, N. J., Loos, R. J. 2013; 45 (6): 621-631

    Abstract

    Elevated resting heart rate is associated with greater risk of cardiovascular disease and mortality. In a 2-stage meta-analysis of genome-wide association studies in up to 181,171 individuals, we identified 14 new loci associated with heart rate and confirmed associations with all 7 previously established loci. Experimental downregulation of gene expression in Drosophila melanogaster and Danio rerio identified 20 genes at 11 loci that are relevant for heart rate regulation and highlight a role for genes involved in signal transmission, embryonic cardiac development and the pathophysiology of dilated cardiomyopathy, congenital heart failure and/or sudden cardiac death. In addition, genetic susceptibility to increased heart rate is associated with altered cardiac conduction and reduced risk of sick sinus syndrome, and both heart rate-increasing and heart rate-decreasing variants associate with risk of atrial fibrillation. Our findings provide fresh insights into the mechanisms regulating heart rate and identify new therapeutic targets.

    View details for DOI 10.1038/ng.2610

    View details for PubMedID 23583979

  • Evidence of a Causal Relationship Between Adiponectin Levels and Insulin Sensitivity DIABETES Gao, H., Fall, T., van Dam, R. M., Flyvbjerg, A., Zethelius, B., Ingelsson, E., Hagg, S. 2013; 62 (4): 1338-1344

    View details for DOI 10.2337/db12-0935

    View details for Web of Science ID 000316526000044

  • Urinary neutrophil gelatinase-associated lipocalin (NGAL) is associated with mortality in a community-based cohort of older Swedish men ATHEROSCLEROSIS Helmersson-Karlqvist, J., Larsson, A., Carlsson, A. C., Venge, P., Sundstrom, J., Ingelsson, E., Lind, L., Arnlov, J. 2013; 227 (2): 408-413

    Abstract

    Neutrophil gelatinase-associated lipocalin (NGAL) indicates tubular kidney damage, neutrophil activation and possibly atherogenesis, however the prospective association between urinary NGAL (u-NGAL) and cardiovascular death in the community is not known.This study evaluates the association between urinary and serum NGAL and mortality in a Swedish population of 597 men aged 78 years. During the study (median follow-up 8.1 years) 261 men died, 90 of cardiovascular causes.U-NGAL was associated with increased all-cause and cardiovascular mortality (HR 2.0 for quartile 4 vs. quartile 1, 95% CI 1.0-4.0, P < 0.05) in Cox regression models independently of cardiovascular risk factors, CRP and cystatin C estimated glomerular filtration rate (eGFRCysC) but not urinary Albumin (u-Alb). A combination of low eGFRCysC (≤60 mL/min), high u-Alb (≥3 mg/mmol Cr) and high u-NGAL (≥1.19 μg/mmol Cr) was associated with a 9-fold increased cardiovascular mortality (P < 0.001) and a 3-fold increased all-cause mortality (P < 0.001). Serum NGAL was associated with increased all-cause mortality risk independent of other cardiovascular risk factors (HR 1.4 for quartile 4 vs.1, 95% CI 1.0-1.9, P < 0.05) but not after adjustment with CRP, eGFRCysC or u-Alb.This community study is the first to show that the tubular kidney biomarker u-NGAL associated with increased cardiovascular and all-cause mortality independent of cardiovascular risk factors and glomerular filtration. Additional research is needed to evaluate the utility of NGAL in clinical practice.

    View details for DOI 10.1016/j.atherosclerosis.2013.01.009

    View details for Web of Science ID 000316603100034

    View details for PubMedID 23375682

  • Urinary kidney injury molecule 1 and incidence of heart failure in elderly men EUROPEAN JOURNAL OF HEART FAILURE Carlsson, A. C., Larsson, A., Helmersson-Karlqvist, J., Lind, L., Ingelsson, E., Larsson, T. E., Sundstrom, J., Arnlov, J. 2013; 15 (4): 441-446

    Abstract

    There is growing recognition of the clinical importance of cardiorenal syndrome-the bidirectional interplay between kidney and cardiac dysfunction. Yet, the role of kidney tubular damage in the development of heart failure is less studied. The objective of this study was to investigate whether urinary kidney injury molecule (KIM)-1, a specific marker of tubular damage, predisposes to an increased heart failure risk.This was a community-based cohort study [Uppsala Longitudinal study of Adult Men (ULSAM)] of 565, 77-year-old men free from heart failure at baseline. Heart failure hospitalizations were used as outcome. During follow-up (median 8.0 years), 73 participants were hospitalized for heart failure. In models adjusted for cardiovascular risk factors (age, systolic blood pressure, diabetes, smoking, body mass index, LDL/HDL ratio, antihypertensive treatment, lipid-lowering treatment, aspirin treatment, LV hypertrophy, and prevalent cardiovascular disease) and markers of kidney dysfunction and damage [cystatin C-based glomerular filtration rate (GFR) and urinary albumin/creatinine ratio], a higher urinary KIM-1/creatinine ratio was associated with higher risk for heart failure (hazard ratio upper vs. lower tertile, 1.81; 95% confidence interval 1.01-3.29; P < 0.05). Participants with a combination of low GFR (<60 mL/min/1.72 m(2)) and high KIM-1/creatinine (>128 ng/mmol) had a 3-fold increase in heart failure risk compared with participants with normal GFR and KIM-1 (P < 0.001).Our findings suggest that kidney tubular damage predisposes to an increased risk for heart failure in the community. Further studies are needed to clarify the causal role of KIM-1 in the development of heart failure, and to evaluate the clinical utility of urinary KIM-1 measurements.

    View details for DOI 10.1093/eurjhf/hfs187

    View details for Web of Science ID 000316955400012

    View details for PubMedID 23220287

  • Evidence of a causal relationship between adiponectin levels and insulin sensitivity: a Mendelian randomization study. Diabetes Gao, H., Fall, T., van Dam, R. M., Flyvbjerg, A., Zethelius, B., Ingelsson, E., Hägg, S. 2013; 62 (4): 1338-1344

    Abstract

    The adipocyte-secreted protein adiponectin is associated with insulin sensitivity in observational studies. We aimed to evaluate whether this relationship is causal using a Mendelian randomization approach. In a sample of Swedish men aged 71 years (n = 942) from the Uppsala Longitudinal Study of Adult Men (ULSAM), insulin sensitivity (M/I ratio) was measured by the euglycemic insulin clamp. We used three genetic variants in the ADIPOQ locus as instrumental variables (IVs) to estimate the potential causal effect of adiponectin on insulin sensitivity and compared these with results from conventional linear regression. The three ADIPOQ variants, rs17300539, rs3774261, and rs6444175, were strongly associated with serum adiponectin levels (all P ≤ 5.3 × 10(-9)) and were also significantly associated with M/I ratio in the expected direction (all P ≤ 0.022). IV analysis confirmed that genetically determined adiponectin increased insulin sensitivity (β = 0.47-0.81, all P ≤ 0.014) comparable with observational estimates (β = 0.50, all P(difference) ≥ 0.136). Adjustment for BMI and waist circumference partly explained the association of both genetically determined and observed adiponectin levels with insulin sensitivity. The observed association between higher adiponectin levels and increased insulin sensitivity is likely to represent a causal relationship partly mediated by reduced adiposity.

    View details for DOI 10.2337/db12-0935

    View details for PubMedID 23274890

  • Genetic Variants from Lipid-Related Pathways and Risk for Incident Myocardial Infarction PLOS ONE Song, C., Pedersen, N. L., Reynolds, C. A., Sabater-Lleal, M., Kanoni, S., Willenborg, C., Syvanen, A., Watkins, H., Hamsten, A., Prince, J. A., Ingelsson, E. 2013; 8 (3)

    Abstract

    Circulating lipids levels, as well as several familial lipid metabolism disorders, are strongly associated with initiation and progression of atherosclerosis and incidence of myocardial infarction (MI).We hypothesized that genetic variants associated with circulating lipid levels would also be associated with MI incidence, and have tested this in three independent samples.Using age- and sex-adjusted additive genetic models, we analyzed 554 single nucleotide polymorphisms (SNPs) in 41 candidate gene regions proposed to be involved in lipid-related pathways potentially predisposing to incidence of MI in 2,602 participants of the Swedish Twin Register (STR; 57% women). All associations with nominal P<0.01 were further investigated in the Uppsala Longitudinal Study of Adult Men (ULSAM; N = 1,142).In the present study, we report associations of lipid-related SNPs with incident MI in two community-based longitudinal studies with in silico replication in a meta-analysis of genome-wide association studies. Overall, there were 9 SNPs in STR with nominal P-value <0.01 that were successfully genotyped in ULSAM. rs4149313 located in ABCA1 was associated with MI incidence in both longitudinal study samples with nominal significance (hazard ratio, 1.36 and 1.40; P-value, 0.004 and 0.015 in STR and ULSAM, respectively). In silico replication supported the association of rs4149313 with coronary artery disease in an independent meta-analysis including 173,975 individuals of European descent from the CARDIoGRAMplusC4D consortium (odds ratio, 1.03; P-value, 0.048).rs4149313 is one of the few amino acid changing variants in ABCA1 known to associate with reduced cholesterol efflux. Our results are suggestive of a weak association between this variant and the development of atherosclerosis and MI.

    View details for DOI 10.1371/journal.pone.0060454

    View details for Web of Science ID 000317263900073

    View details for PubMedID 23555974

  • A Genome-Wide Assessment of Variability in Human Serum Metabolism HUMAN MUTATION Hong, M., Karlsson, R., Magnusson, P. K., Lewis, M. R., Isaacs, W., Zheng, L. S., Xu, J., Gronberg, H., Ingelsson, E., Pawitan, Y., Broeckling, C., Prenni, J. E., Wiklund, F., Prince, J. A. 2013; 34 (3): 515-524

    Abstract

    The study of the genetic regulation of metabolism in human serum samples can contribute to a better understanding of the intermediate biological steps that lead from polymorphism to disease. Here, we conducted a genome-wide association study (GWAS) to discover metabolic quantitative trait loci (mQTLs) utilizing samples from a study of prostate cancer in Swedish men, consisting of 402 individuals (214 cases and 188 controls) in a discovery set and 489 case-only samples in a replication set. A global nontargeted metabolite profiling approach was utilized resulting in the detection of 6,138 molecular features followed by targeted identification of associated metabolites. Seven replicating loci were identified (PYROXD2, FADS1, PON1, CYP4F2, UGT1A8, ACADL, and LIPC) with associated sequence variants contributing significantly to trait variance for one or more metabolites (P = 10(-13) -10(-91)). Regional mQTL enrichment analyses implicated two loci that included FADS1 and a novel locus near PDGFC. Biological pathway analysis implicated ACADM, ACADS, ACAD8, ACAD10, ACAD11, and ACOXL, reflecting significant enrichment of genes with acyl-CoA dehydrogenase activity. mQTL SNPs and mQTL-harboring genes were over-represented across GWASs conducted to date, suggesting that these data may have utility in tracing the molecular basis of some complex disease associations.

    View details for DOI 10.1002/humu.22267

    View details for Web of Science ID 000315186400014

    View details for PubMedID 23281178

  • Higher Magnesium Intake Is Associated with Lower Fasting Glucose and Insulin, with No Evidence of Interaction with Select Genetic Loci, in a Meta-Analysis of 15 CHARGE Consortium Studies JOURNAL OF NUTRITION Hruby, A., Ngwa, J. S., Renstrom, F., Wojczynski, M. K., Ganna, A., Hallmans, G., Houston, D. K., Jacques, P. F., Kanoni, S., Lehtimaki, T., Lemaitre, R. N., Manichaikul, A., North, K. E., Ntalla, I., Sonestedt, E., Tanaka, T., van Rooij, F. J., Bandinelli, S., Djousse, L., Grigoriou, E., Johansson, I., Lohman, K. K., Pankow, J. S., Raitakari, O. T., Riserus, U., Yannakoulia, M., Zillikens, M. C., Hassanali, N., Liu, Y., Mozaffarian, D., Papoutsakis, C., Syvanen, A., Uitterlinden, A. G., Viikari, J., Groves, C. J., Hofman, A., Lind, L., McCarthy, M. I., Mikkila, V., Mukamal, K., Franco, O. H., Borecki, I. B., Cupples, L. A., Dedoussis, G. V., Ferrucci, L., Hu, F. B., Ingelsson, E., Kahonen, M., Kao, W. H., Kritchevsky, S. B., Orho-Melander, M., Prokopenko, I., Rotter, J. I., Siscovick, D. S., Witteman, J. C., Franks, P. W., Meigs, J. B., McKeown, N. M., Nettleton, J. A. 2013; 143 (3): 345-353

    Abstract

    Favorable associations between magnesium intake and glycemic traits, such as fasting glucose and insulin, are observed in observational and clinical studies, but whether genetic variation affects these associations is largely unknown. We hypothesized that single nucleotide polymorphisms (SNPs) associated with either glycemic traits or magnesium metabolism affect the association between magnesium intake and fasting glucose and insulin. Fifteen studies from the CHARGE (Cohorts for Heart and Aging Research in Genomic Epidemiology) Consortium provided data from up to 52,684 participants of European descent without known diabetes. In fixed-effects meta-analyses, we quantified 1) cross-sectional associations of dietary magnesium intake with fasting glucose (mmol/L) and insulin (ln-pmol/L) and 2) interactions between magnesium intake and SNPs related to fasting glucose (16 SNPs), insulin (2 SNPs), or magnesium (8 SNPs) on fasting glucose and insulin. After adjustment for age, sex, energy intake, BMI, and behavioral risk factors, magnesium (per 50-mg/d increment) was inversely associated with fasting glucose [β = -0.009 mmol/L (95% CI: -0.013, -0.005), P < 0.0001] and insulin [-0.020 ln-pmol/L (95% CI: -0.024, -0.017), P < 0.0001]. No magnesium-related SNP or interaction between any SNP and magnesium reached significance after correction for multiple testing. However, rs2274924 in magnesium transporter-encoding TRPM6 showed a nominal association (uncorrected P = 0.03) with glucose, and rs11558471 in SLC30A8 and rs3740393 near CNNM2 showed a nominal interaction (uncorrected, both P = 0.02) with magnesium on glucose. Consistent with other studies, a higher magnesium intake was associated with lower fasting glucose and insulin. Nominal evidence of TRPM6 influence and magnesium interaction with select loci suggests that further investigation is warranted.

    View details for DOI 10.3945/jn.112.172049

    View details for Web of Science ID 000315173500014

    View details for PubMedID 23343670

  • Assigning precursor-product ion relationships in indiscriminant MS/MS data from non-targeted metabolite profiling studies METABOLOMICS Broeckling, C. D., Heuberger, A. L., Prince, J. A., Ingelsson, E., Prenni, J. E. 2013; 9 (1): 33-43
  • The Swedish Twin Registry: Establishment of a Biobank and Other Recent Developments TWIN RESEARCH AND HUMAN GENETICS Magnusson, P. K., Almqvist, C., Rahman, I., Ganna, A., Viktorin, A., Walum, H., Halldner, L., Lundstrom, S., Ullen, F., Langstrom, N., Larsson, H., Nyman, A., Gumpert, C. H., Rastam, M., Anckarsater, H., Cnattingius, S., Johannesson, M., Ingelsson, E., Klareskog, L., de Faire, U., Pedersen, N. L., Lichtenstein, P. 2013; 16 (1): 317-329

    Abstract

    The Swedish Twin Registry (STR) today contains more than 194,000 twins and more than 75,000 pairs have zygosity determined by an intra-pair similarity algorithm, DNA, or by being of opposite sex. Of these, approximately 20,000, 25,000, and 30,000 pairs are monozygotic, same-sex dizygotic, and opposite-sex dizygotic pairs, respectively. Since its establishment in the late 1950s, the STR has been an important epidemiological resource for the study of genetic and environmental influences on a multitude of traits, behaviors, and diseases. Following large investments in the collection of biological specimens in the past 10 years we have now established a Swedish twin biobank with DNA from 45,000 twins and blood serum from 15,000 twins, which effectively has also transformed the registry into a powerful resource for molecular studies. We here describe the main projects within which the new collections of both biological samples as well as phenotypic measures have been collected. Coverage by year of birth, zygosity determination, ethnic heterogeneity, and influences of in vitro fertilization are also described.

    View details for DOI 10.1017/thg.2012.104

    View details for Web of Science ID 000314799700048

    View details for PubMedID 23137839

  • Causal Relationship between Obesity and Vitamin D Status: Bi-Directional Mendelian Randomization Analysis of Multiple Cohorts PLOS MEDICINE Vimaleswaran, K. S., Berry, D. J., Lu, C., Tikkanen, E., Pilz, S., Hiraki, L. T., Cooper, J. D., Dastani, Z., Li, R., Houston, D. K., Wood, A. R., Michaelsson, K., Vandenput, L., Zgaga, L., Yerges-Armstrong, L. M., McCarthy, M. I., Dupuis, J., Kaakinen, M., Kleber, M. E., Jameson, K., Arden, N., Raitakari, O., Viikari, J., Lohman, K. K., Ferrucci, L., Melhus, H., Ingelsson, E., Byberg, L., Lind, L., Lorentzon, M., Salomaa, V., Campbell, H., Dunlop, M., Mitchell, B. D., Herzig, K., Pouta, A., Hartikainen, A., Streeten, E. A., Theodoratou, E., Jula, A., Wareham, N. J., Ohlsson, C., Frayling, T. M., Kritchevsky, S. B., Spector, T. D., Richards, J. B., Lehtimaki, T., Ouwehand, W. H., Kraft, P., Cooper, C., Maerz, W., Power, C., Loos, R. J., Wang, T. J., Jaervelin, M., Whittaker, J. C., Hingorani, A. D., Hyppoenen, E. 2013; 10 (2)

    Abstract

    Obesity is associated with vitamin D deficiency, and both are areas of active public health concern. We explored the causality and direction of the relationship between body mass index (BMI) and 25-hydroxyvitamin D [25(OH)D] using genetic markers as instrumental variables (IVs) in bi-directional Mendelian randomization (MR) analysis.We used information from 21 adult cohorts (up to 42,024 participants) with 12 BMI-related SNPs (combined in an allelic score) to produce an instrument for BMI and four SNPs associated with 25(OH)D (combined in two allelic scores, separately for genes encoding its synthesis or metabolism) as an instrument for vitamin D. Regression estimates for the IVs (allele scores) were generated within-study and pooled by meta-analysis to generate summary effects. Associations between vitamin D scores and BMI were confirmed in the Genetic Investigation of Anthropometric Traits (GIANT) consortium (n = 123,864). Each 1 kg/m(2) higher BMI was associated with 1.15% lower 25(OH)D (p = 6.52×10⁻²⁷). The BMI allele score was associated both with BMI (p = 6.30×10⁻⁶²) and 25(OH)D (-0.06% [95% CI -0.10 to -0.02], p = 0.004) in the cohorts that underwent meta-analysis. The two vitamin D allele scores were strongly associated with 25(OH)D (p≤8.07×10⁻⁵⁷ for both scores) but not with BMI (synthesis score, p = 0.88; metabolism score, p = 0.08) in the meta-analysis. A 10% higher genetically instrumented BMI was associated with 4.2% lower 25(OH)D concentrations (IV ratio: -4.2 [95% CI -7.1 to -1.3], p = 0.005). No association was seen for genetically instrumented 25(OH)D with BMI, a finding that was confirmed using data from the GIANT consortium (p≥0.57 for both vitamin D scores).On the basis of a bi-directional genetic approach that limits confounding, our study suggests that a higher BMI leads to lower 25(OH)D, while any effects of lower 25(OH)D increasing BMI are likely to be small. Population level interventions to reduce BMI are expected to decrease the prevalence of vitamin D deficiency.

    View details for DOI 10.1371/journal.pmed.1001383

    View details for Web of Science ID 000315592800005

    View details for PubMedID 23393431

  • Meta-Analysis Investigating Associations Between Healthy Diet and Fasting Glucose and Insulin Levels and Modification by Loci Associated With Glucose Homeostasis in Data From 15 Cohorts AMERICAN JOURNAL OF EPIDEMIOLOGY Nettleton, J. A., Hivert, M., Lemaitre, R. N., McKeown, N. M., Mozaffarian, D., Tanaka, T., Wojczynski, M. K., Hruby, A., Djousse, L., Ngwa, J. S., Follis, J. L., Dimitriou, M., Ganna, A., Houston, D. K., Kanoni, S., Mikkila, V., Manichaikul, A., Ntalla, I., Renstrom, F., Sonestedt, E., van Rooij, F. J., Bandinelli, S., de Koning, L., Ericson, U., Hassanali, N., Kiefte-de Jong, J. C., Lohman, K. K., Raitakari, O., Papoutsakis, C., Sjogren, P., Stirrups, K., Ax, E., Deloukas, P., Groves, C. J., Jacques, P. F., Johansson, I., Liu, Y., McCarthy, M. I., North, K., Viikari, J., Zillikens, M. C., Dupuis, J., Hofman, A., Kolovou, G., Mukamal, K., Prokopenko, I., Rolandsson, O., Seppala, I., Cupples, L. A., Hu, F. B., Kahonen, M., Uitterlinden, A. G., Borecki, I. B., Ferrucci, L., Jacobs, D. R., Kritchevsky, S. B., Orho-Melander, M., Pankow, J. S., Lehtimaki, T., Witteman, J. C., Ingelsson, E., Siscovick, D. S., Dedoussis, G., Meigs, J. B., Franks, P. W. 2013; 177 (2): 103-115

    Abstract

    Whether loci that influence fasting glucose (FG) and fasting insulin (FI) levels, as identified by genome-wide association studies, modify associations of diet with FG or FI is unknown. We utilized data from 15 U.S. and European cohort studies comprising 51,289 persons without diabetes to test whether genotype and diet interact to influence FG or FI concentration. We constructed a diet score using study-specific quartile rankings for intakes of whole grains, fish, fruits, vegetables, and nuts/seeds (favorable) and red/processed meats, sweets, sugared beverages, and fried potatoes (unfavorable). We used linear regression within studies, followed by inverse-variance-weighted meta-analysis, to quantify 1) associations of diet score with FG and FI levels and 2) interactions of diet score with 16 FG-associated loci and 2 FI-associated loci. Diet score (per unit increase) was inversely associated with FG (β = -0.004 mmol/L, 95% confidence interval: -0.005, -0.003) and FI (β = -0.008 ln-pmol/L, 95% confidence interval: -0.009, -0.007) levels after adjustment for demographic factors, lifestyle, and body mass index. Genotype variation at the studied loci did not modify these associations. Healthier diets were associated with lower FG and FI concentrations regardless of genotype at previously replicated FG- and FI-associated loci. Studies focusing on genomic regions that do not yield highly statistically significant associations from main-effect genome-wide association studies may be more fruitful in identifying diet-gene interactions.

    View details for DOI 10.1093/aje/kws297

    View details for Web of Science ID 000313528700001

    View details for PubMedID 23255780

  • Serum Cathepsin S Is Associated With Decreased Insulin Sensitivity and the Development of Type 2 Diabetes in a Community-Based Cohort of Elderly Men DIABETES CARE Jobs, E., Riserus, U., Ingelsson, E., Sundstrom, J., Jobs, M., Nerpin, E., Iggman, D., Basu, S., Larsson, A., Lind, L., Arnlov, J. 2013; 36 (1): 163-165

    Abstract

    To investigate associations between serum cathepsin S, impaired insulin sensitivity, defective insulin secretion, and diabetes risk in a community-based sample of elderly men without diabetes.Serum cathepsin S, insulin sensitivity (euglycemic-hyperinsulinemic clamp), and insulin secretion (early insulin response during an oral glucose tolerance test) were measured in 905 participants of the Uppsala Longitudinal Study of Adult Men (mean age, 71 years). Thirty participants developed diabetes during 6 years of follow-up.After adjustment for age, anthropometric variables, and inflammatory markers, higher cathepsin S was associated with decreased insulin sensitivity (regression coefficient per SD increase -0.09 [95% CI -0.14 to -0.04], P = 0.001), but no association with early insulin response was found. Moreover, higher cathepsin S was associated with a higher risk for developing diabetes (odds ratio per SD increase 1.48 [1.08-2.01], P = 0.01).Cathepsin S activity appears to be involved in the early dysregulation of glucose and insulin metabolism.

    View details for DOI 10.2337/dc12-0494

    View details for Web of Science ID 000314465700048

    View details for PubMedID 22923671

  • Large-scale association analysis identifies new risk loci for coronary artery disease NATURE GENETICS Deloukas, P., Kanoni, S., Willenborg, C., Farrall, M., Assimes, T. L., Thompson, J. R., Ingelsson, E., Saleheen, D., Erdmann, J., Goldstein, B. A., Stirrups, K., Koenig, I. R., Cazier, J., Johansson, A., Hall, A. S., Lee, J., Willer, C. J., Chambers, J. C., Esko, T., Folkersen, L., Goel, A., Grundberg, E., Havulinna, A. S., Ho, W. K., Hopewell, J. C., Eriksson, N., Kleber, M. E., Kristiansson, K., Lundmark, P., Lyytikainen, L., Rafelt, S., Shungin, D., Strawbridge, R. J., Thorleifsson, G., Tikkanen, E., Van Zuydam, N., Voight, B. F., Waite, L. L., Zhang, W., Ziegler, A., Absher, D., Altshuler, D., Balmforth, A. J., Barroso, I., Braund, P. S., Burgdorf, C., Claudi-Boehm, S., Cox, D., Dimitriou, M., Do, R., Doney, A. S., El Mokhtari, N., Eriksson, P., Fischer, K., Fontanillas, P., Franco-Cereceda, A., Gigante, B., Groop, L., Gustafsson, S., Hager, J., Hallmans, G., Han, B., Hunt, S. E., Kang, H. M., Illig, T., Kessler, T., Knowles, J. W., Kolovou, G., Kuusisto, J., Langenberg, C., Langford, C., Leander, K., Lokki, M., Lundmark, A., McCarthy, M. I., Meisinger, C., Melander, O., Mihailov, E., Maouche, S., Morris, A. D., Mueller-Nurasyid, M., Nikus, K., Peden, J. F., Rayner, N. W., Rasheed, A., Rosinger, S., Rubin, D., Rumpf, M. P., Schaefer, A., Sivananthan, M., Song, C., Stewart, A. F., Tan, S., Thorgeirsson, G., van der Schoot, C. E., Wagner, P. J., Wells, G. A., Wild, P. S., Yang, T., Amouyel, P., Arveiler, D., Basart, H., Boehnke, M., Boerwinkle, E., Brambilla, P., Cambien, F., Cupples, A. L., de Faire, U., Dehghan, A., Diemert, P., Epstein, S. E., Evans, A., Ferrario, M. M., Ferrieres, J., Gauguier, D., Go, A. S., Goodall, A. H., Gudnason, V., Hazen, S. L., Holm, H., Iribarren, C., Jang, Y., Kahonen, M., Kee, F., Kim, H., Klopp, N., Koenig, W., Kratzer, W., Kuulasmaa, K., Laakso, M., Laaksonen, R., Lee, J., Lind, L., Ouwehand, W. H., Parish, S., Park, J. E., Pedersen, N. L., Peters, A., Quertermous, T., Rader, D. J., Salomaa, V., Schadt, E., Shah, S. H., Sinisalo, J., Stark, K., Stefansson, K., Tregouet, D., Virtamo, J., Wallentin, L., Wareham, N., Zimmermann, M. E., Nieminen, M. S., Hengstenberg, C., Sandhu, M. S., Pastinen, T., Syvanen, A., Hovingh, G. K., Dedoussis, G., Franks, P. W., Lehtimaki, T., Metspalu, A., Zalloua, P. A., Siegbahn, A., Schreiber, S., Ripatti, S., Blankenberg, S. S., Perola, M., Clarke, R., Boehm, B. O., O'Donnell, C., Reilly, M. P., Maerz, W., Collins, R., Kathiresan, S., Hamsten, A., Kooner, J. S., Thorsteinsdottir, U., Danesh, J., Palmer, C. N., Roberts, R., Watkins, H., Schunkert, H., Samani, N. J. 2013; 45 (1): 25-U52

    Abstract

    Coronary artery disease (CAD) is the commonest cause of death. Here, we report an association analysis in 63,746 CAD cases and 130,681 controls identifying 15 loci reaching genome-wide significance, taking the number of susceptibility loci for CAD to 46, and a further 104 independent variants (r(2) < 0.2) strongly associated with CAD at a 5% false discovery rate (FDR). Together, these variants explain approximately 10.6% of CAD heritability. Of the 46 genome-wide significant lead SNPs, 12 show a significant association with a lipid trait, and 5 show a significant association with blood pressure, but none is significantly associated with diabetes. Network analysis with 233 candidate genes (loci at 10% FDR) generated 5 interaction networks comprising 85% of these putative genes involved in CAD. The four most significant pathways mapping to these networks are linked to lipid metabolism and inflammation, underscoring the causal role of these activities in the genetic etiology of CAD. Our study provides insights into the genetic basis of CAD and identifies key biological pathways.

    View details for DOI 10.1038/ng.2480

    View details for Web of Science ID 000312838800009

    View details for PubMedID 23202125

  • Higher fibroblast growth factor-23 increases the risk of all-cause and cardiovascular mortality in the community KIDNEY INTERNATIONAL Arnlov, J., Carlsson, A. C., Sundstrom, J., Ingelsson, E., Larsson, A., Lind, L., Larsson, T. E. 2013; 83 (1): 160-166

    Abstract

    Fibroblast growth factor-23 (FGF23), a regulator of mineral metabolism, has been linked to cardiovascular disease in chronic kidney disease. As community-based data of the longitudinal association between FGF23 and cardiovascular events are lacking, we investigated a possible relationship in 727 men of the Uppsala Longitudinal Study of Adult Men population-based cohort (mean age 77 years). During a median follow-up of 9.7 years, 110 participants died of cardiovascular causes. In Cox regression models adjusted for age and established cardiovascular risk factors, higher serum FGF23 was associated with a significantly increased risk for cardiovascular mortality (hazard ratio (HR) per increased s.d. of 1.36). This relationship remained significant, albeit attenuated, after adjustment for glomerular filtration rate (GFR) (HR 1.21). FGF23 was also associated with all-cause mortality, although the association was weaker than that with cardiovascular mortality, and it was nonsignificant in fully adjusted multivariate models. Spline analysis suggested a log-linear relationship between FGF23 and outcome. Participants with a combination of high FGF23 (>60 pg/ml), low GFR (<60 ml/min), and micro-/macro-albuminuria (albumin/creatinine ratio above 3 mg/ml) had an almost eightfold increased risk compared with participants without these abnormalities. Thus, a higher FGF23 level is associated with an increased cardiovascular mortality risk in the community. Clinical trials are needed to determine whether FGF23 is a modifiable risk factor.

    View details for DOI 10.1038/ki.2012.327

    View details for Web of Science ID 000313387200023

    View details for PubMedID 22951890

  • Associations of Variants in FTO and Near MC4R With Obesity Traits in South Asian Indians OBESITY Vasan, S. K., Fall, T., Neville, M. J., Antonisamy, B., Fall, C. H., Geethanjali, F. S., Gu, H. F., Raghupathy, P., Samuel, P., Thomas, N., Brismar, K., Ingelsson, E., Karpe, F. 2012; 20 (11): 2268-2277

    Abstract

    Recent genome-wide association studies show that loci in FTO and melanocortin 4 receptor (MC4R) associate with obesity-related traits. Outside Western populations the associations between these variants have not always been consistent and in Indians it has been suggested that FTO relates to diabetes without an obvious intermediary obesity phenotype. We investigated the association between genetic variants in FTO (rs9939609) and near MC4R (rs17782313) with obesity- and type 2 diabetes (T2DM)-related traits in a longitudinal birth cohort of 2,151 healthy individuals from the Vellore birth cohort in South India. The FTO locus displayed significant associations with several conventional obesity-related anthropometric traits. The per allele increase is about 1% for BMI, waist circumference (WC), hip circumference (HC), and waist-hip ratio. Consistent associations were observed for adipose tissue-specific measurements such as skinfold thickness reinforcing the association with obesity-related traits. Obesity associations for the MC4R locus were weak or nonsignificant but a signal for height (P < 0.001) was observed. The effect on obesity-related traits for FTO was seen in adulthood, but not at younger ages. The loci also showed nominal associations with increased blood glucose but these associations were lost on BMI adjustment. The effect of FTO on obesity-related traits was driven by an urban environmental influence. We conclude that rs9939609 variant in the FTO locus is associated with measures of adiposity and metabolic consequences in South Indians with an enhanced effect associated with urban living. The detection of these associations in Indians is challenging because conventional anthropometric obesity measures work poorly in the Indian "thin-fat" phenotype.

    View details for DOI 10.1038/oby.2012.64

    View details for Web of Science ID 000310492200015

    View details for PubMedID 22421923

  • Clinical and Genetic Correlates of Growth Differentiation Factor 15 in the Community CLINICAL CHEMISTRY Ho, J. E., Mahajan, A., Chen, M., Larson, M. G., McCabe, E. L., Ghorbani, A., Cheng, S., Johnson, A. D., Lindgren, C. M., Kempf, T., Lind, L., Ingelsson, E., Vasan, R. S., Januzzi, J., Wollert, K. C., Morris, A. P., Wang, T. J. 2012; 58 (11): 1582-1591

    Abstract

    Growth differentiation factor 15 (GDF15), a stress-responsive cytokine produced in cardiovascular cells under conditions of inflammation and oxidative stress, is emerging as an important prognostic marker in individuals with and without existing cardiovascular disease (CVD). We therefore examined the clinical and genetic correlates of circulating GDF15 concentrations, which have not been investigated collectively.Plasma GDF15 concentrations were measured in 2991 participants in the Framingham Offspring Study who were free of clinically overt CVD (mean age, 59 years; 56% women). Clinical correlates of GDF15 were examined in multivariable analyses. We then conducted a genomewide association study of the GDF15 concentration that included participants in the Framingham Offspring Study and participants in the PIVUS (Prospective Investigation of the Vasculature in Uppsala Seniors) study.GDF15 was positively associated with age, smoking, antihypertensive treatment, diabetes, worse kidney function, and use of nonsteroidal antiinflammatory drugs (NSAIDs), but it was negatively associated with total cholesterol and HDL cholesterol. Clinical correlates accounted for 38% of interindividual variation in the circulating GDF15 concentration, whereas genetic factors accounted for up to 38% of the residual variability (h(2) = 0.38; P = 2.5 × 10(-11)). We identified 1 locus of genomewide significance. This locus, which is on chromosome 19p13.11 and includes the GDF15 gene, is associated with GDF15 concentration (smallest P = 2.74 × 10(-32) for rs888663). Conditional analyses revealed 2 independent association signals at this locus (rs888663 and rs1054564), which were associated with altered cis gene expression in blood cell lines.In ambulatory individuals, both cardiometabolic risk factors and genetic factors play important roles in determining circulating GDF15 concentrations and contribute similarly to the overall variation.

    View details for DOI 10.1373/clinchem.2012.190322

    View details for Web of Science ID 000310489300014

    View details for PubMedID 22997280

  • FTO genotype is associated with phenotypic variability of body mass index NATURE Yang, J., Loos, R. J., Powell, J. E., Medland, S. E., Speliotes, E. K., Chasman, D. I., Rose, L. M., Thorleifsson, G., Steinthorsdottir, V., Maegi, R., Waite, L., Smith, A. V., Yerges-Armstrong, L. M., Monda, K. L., Hadley, D., Mahajan, A., Li, G., Kapur, K., Vitart, V., Huffman, J. E., Wang, S. R., Palmer, C., Esko, T., Fischer, K., Zhao, J. H., Demirkan, A., Isaacs, A., Feitosa, M. F., Luan, J., Heard-Costa, N. L., White, C., Jackson, A. U., Preuss, M., Ziegler, A., Eriksson, J., Kutalik, Z., Frau, F., Nolte, I. M., van Vliet-Ostaptchouk, J. V., Hottenga, J., Jacobs, K. B., Verweij, N., Goel, A., Medina-Gomez, C., Estrada, K., Bragg-Gresham, J. L., Sanna, S., Sidore, C., Tyrer, J., Teumer, A., Prokopenko, I., Mangino, M., Lindgren, C. M., Assimes, T. L., Shuldiner, A. R., Hui, J., Beilby, J. P., McArdle, W. L., Hall, P., Haritunians, T., Zgaga, L., Kolcic, I., Polasek, O., Zemunik, T., Oostra, B. A., Junttila, M. J., Groenberg, H., Schreiber, S., Peters, A., Hicks, A. A., Stephens, J., Foad, N. S., Laitinen, J., Pouta, A., Kaakinen, M., Willemsen, G., Vink, J. M., Wild, S. H., Navis, G., Asselbergs, F. W., Homuth, G., John, U., Iribarren, C., Harris, T., Launer, L., Gudnason, V., O'Connell, J. R., Boerwinkle, E., Cadby, G., Palmer, L. J., James, A. L., Musk, A. W., Ingelsson, E., Psaty, B. M., Beckmann, J. S., Waeber, G., Vollenweider, P., Hayward, C., Wright, A. F., Rudan, I., Groop, L. C., Metspalu, A., Khaw, K. T., van Duijn, C. M., Borecki, I. B., Province, M. A., Wareham, N. J., Tardif, J., Huikuri, H. V., Cupples, L. A., Atwood, L. D., Fox, C. S., Boehnke, M., Collins, F. S., Mohlke, K. L., Erdmann, J., Schunkert, H., Hengstenberg, C., Stark, K., Lorentzon, M., Ohlsson, C., Cusi, D., Staessen, J. A., van der Klauw, M. M., Pramstaller, P. P., Kathiresan, S., Jolley, J. D., Ripatti, S., Jarvelin, M., de Geus, E. J., Boomsma, D. I., Penninx, B., Wilson, J. F., Campbell, H., Chanock, S. J., van der Harst, P., Hamsten, A., Watkins, H., Hofman, A., Witteman, J. C., Zillikens, M. C., Uitterlinden, A. G., Rivadeneira, F., Zillikens, M. C., Kiemeney, L. A., Vermeulen, S. H., Abecasis, G. R., Schlessinger, D., Schipf, S., Stumvoll, M., Toenjes, A., Spector, T. D., North, K. E., Lettre, G., McCarthy, M. I., Berndt, S. I., Heath, A. C., Madden, P. A., Nyholt, D. R., Montgomery, G. W., Martin, N. G., McKnight, B., Strachan, D. P., Hill, W. G., Snieder, H., Ridker, P. M., Thorsteinsdottir, U., Stefansson, K., Frayling, T. M., Hirschhorn, J. N., Goddard, M. E., Visscher, P. M. 2012; 490 (7419): 267-?

    Abstract

    There is evidence across several species for genetic control of phenotypic variation of complex traits, such that the variance among phenotypes is genotype dependent. Understanding genetic control of variability is important in evolutionary biology, agricultural selection programmes and human medicine, yet for complex traits, no individual genetic variants associated with variance, as opposed to the mean, have been identified. Here we perform a meta-analysis of genome-wide association studies of phenotypic variation using ∼170,000 samples on height and body mass index (BMI) in human populations. We report evidence that the single nucleotide polymorphism (SNP) rs7202116 at the FTO gene locus, which is known to be associated with obesity (as measured by mean BMI for each rs7202116 genotype), is also associated with phenotypic variability. We show that the results are not due to scale effects or other artefacts, and find no other experiment-wise significant evidence for effects on variability, either at loci other than FTO for BMI or at any locus for height. The difference in variance for BMI among individuals with opposite homozygous genotypes at the FTO locus is approximately 7%, corresponding to a difference of ∼0.5 kilograms in the standard deviation of weight. Our results indicate that genetic variants can be discovered that are associated with variability, and that between-person variability in obesity can partly be explained by the genotype at the FTO locus. The results are consistent with reported FTO by environment interactions for BMI, possibly mediated by DNA methylation. Our BMI results for other SNPs and our height results for all SNPs suggest that most genetic variants, including those that influence mean height or mean BMI, are not associated with phenotypic variance, or that their effects on variability are too small to detect even with samples sizes greater than 100,000.

    View details for DOI 10.1038/nature11401

    View details for Web of Science ID 000309733300051

    View details for PubMedID 22982992

    View details for PubMedCentralID PMC3564953

  • C-Reactive Protein, Fibrinogen, and Cardiovascular Disease Prediction NEW ENGLAND JOURNAL OF MEDICINE Kaptoge, S., Di Angelantonio, E., Pennells, L., Wood, A. M., White, I. R., Gao, P., Walker, M., Thompson, A., Sarwar, N., Caslake, M., Butterworth, A. S., Amouyel, P., Assmann, G., Bakker, S. J., Barr, E. L., Barrett-Connor, E., Benjamin, E. J., Bjorkelund, C., Brenner, H., Brunner, E., Clarke, R., Cooper, J. A., Cremer, P., Cushman, M., Dagenais, G. R., D'Agostino, R. B., Dankner, R., Davey-Smith, G., Deeg, D., Dekker, J. M., Engstrom, G., Folsom, A. R., Fowkes, F. G., Gallacher, J., Gaziano, J. M., Giampaoli, S., Gillum, R. F., Hofman, A., Howard, B. V., Ingelsson, E., Iso, H., Jorgensen, T., Kiechl, S., Kitamura, A., Kiyohara, Y., Koenig, W., Kromhout, D., Kuller, L. H., Lawlor, D. A., Meade, T. W., Nissinen, A., Nordestgaard, B. G., Onat, A., Panagiotakos, D. B., Psaty, B. M., Rodriguez, B., Rosengren, A., Salomaa, V., Kauhanen, J., Salonen, J. T., Shaffer, J. A., Shea, S., Ford, I., Stehouwer, C. D., Strandberg, T. E., Tipping, R. W., Tosetto, A., Wassertheil-Smoller, S., Wennberg, P., Westendorp, R. G., Whincup, P. H., Wilhelmsen, L., Woodward, M., Lowe, G. D., Wareham, N. J., Khaw, K., Sattar, N., Packard, C. J., Gudnason, V., Ridker, P. M., Pepys, M. B., Thompson, S. G., Danesh, J. 2012; 367 (14): 1310-1320

    Abstract

    There is debate about the value of assessing levels of C-reactive protein (CRP) and other biomarkers of inflammation for the prediction of first cardiovascular events.We analyzed data from 52 prospective studies that included 246,669 participants without a history of cardiovascular disease to investigate the value of adding CRP or fibrinogen levels to conventional risk factors for the prediction of cardiovascular risk. We calculated measures of discrimination and reclassification during follow-up and modeled the clinical implications of initiation of statin therapy after the assessment of CRP or fibrinogen.The addition of information on high-density lipoprotein cholesterol to a prognostic model for cardiovascular disease that included age, sex, smoking status, blood pressure, history of diabetes, and total cholesterol level increased the C-index, a measure of risk discrimination, by 0.0050. The further addition to this model of information on CRP or fibrinogen increased the C-index by 0.0039 and 0.0027, respectively (P<0.001), and yielded a net reclassification improvement of 1.52% and 0.83%, respectively, for the predicted 10-year risk categories of "low" (<10%), "intermediate" (10% to <20%), and "high" (≥20%) (P<0.02 for both comparisons). We estimated that among 100,000 adults 40 years of age or older, 15,025 persons would initially be classified as being at intermediate risk for a cardiovascular event if conventional risk factors alone were used to calculate risk. Assuming that statin therapy would be initiated in accordance with Adult Treatment Panel III guidelines (i.e., for persons with a predicted risk of ≥20% and for those with certain other risk factors, such as diabetes, irrespective of their 10-year predicted risk), additional targeted assessment of CRP or fibrinogen levels in the 13,199 remaining participants at intermediate risk could help prevent approximately 30 additional cardiovascular events over the course of 10 years.In a study of people without known cardiovascular disease, we estimated that under current treatment guidelines, assessment of the CRP or fibrinogen level in people at intermediate risk for a cardiovascular event could help prevent one additional event over a period of 10 years for every 400 to 500 people screened. (Funded by the British Heart Foundation and others.).

    View details for DOI 10.1056/NEJMoa1107477

    View details for Web of Science ID 000309406100007

    View details for PubMedID 23034020

  • Smokeless tobacco (snus) and risk of heart failure: results from two Swedish cohorts EUROPEAN JOURNAL OF PREVENTIVE CARDIOLOGY Arefalk, G., Hergens, M., Ingelsson, E., Arnlov, J., Michaelsson, K., Lind, L., Ye, W., Nyren, O., Lambe, M., Sundstrom, J. 2012; 19 (5): 1120-1127

    Abstract

    Oral moist snuff (snus) is discussed as a safer alternative to smoking, and its use is increasing. Based on its documented effect on blood pressure, we hypothesized that use of snus increases the risk of heart failure.Two independent Swedish prospective cohorts; the Uppsala Longitudinal Study of Adult Men (ULSAM), a community-based sample of 1076 elderly men, and the Construction Workers Cohort (CWC), a sample of 118,425 never-smoking male construction workers.Cox proportional hazards models were used to investigate possible associations of snus use with risk of a first hospitalization for heart failure.In ULSAM, 95 men were hospitalized for heart failure, during a median follow up of 8.9 years. In a model adjusted for established risk factors including past and present smoking exposure, current snus use was associated with a higher risk of heart failure [hazard ratio (HR) 2.08, 95% confidence interval (CI) 1.03-4.22] relative to non-use. Snus use was particularly associated with risk of non-ischaemic heart failure (HR 2.55, 95% CI 1.12-5.82). In CWC, 545 men were hospitalized for heart failure, during a median follow up of 18 years. In multivariable-adjusted models, current snus use was moderately associated with a higher risk of heart failure (HR 1.28, 95% CI 1.00-1.64) and non-ischaemic heart failure (HR 1.28, 95% CI 0.97-1.68) relative to never tobacco use.Data from two independent cohorts suggest that use of snus may be associated with a higher risk of heart failure.

    View details for DOI 10.1177/1741826711420003

    View details for Web of Science ID 000309527700022

    View details for PubMedID 21828223

  • Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes NATURE GENETICS Morris, A. P., Voight, B. F., Teslovich, T. M., Ferreira, T., Segre, A. V., Steinthorsdottir, V., Strawbridge, R. J., Khan, H., Grallert, H., Mahajan, A., Prokopenko, I., Kang, H. M., Dina, C., Esko, T., Fraser, R. M., Kanoni, S., Kumar, A., Lagou, V., Langenberg, C., Luan, J., Lindgren, C. M., Mueller-Nurasyid, M., Pechlivanis, S., Rayner, N. W., Scott, L. J., Wiltshire, S., Yengo, L., Kinnunen, L., Rossin, E. J., Raychaudhuri, S., Johnson, A. D., Dimas, A. S., Loos, R. J., Vedantam, S., Chen, H., Florez, J. C., Fox, C., Liu, C., Rybin, D., Couper, D. J., Kao, W. H., Li, M., Cornelis, M. C., Kraft, P., Sun, Q., van Dam, R. M., Stringham, H. M., Chines, P. S., Fischer, K., Fontanillas, P., Holmen, O. L., Hunt, S. E., Jackson, A. U., Kong, A., Lawrence, R., Meyer, J., Perry, J. R., Platou, C. G., Potter, S., Rehnberg, E., Robertson, N., Sivapalaratnam, S., Stancakova, A., Stirrups, K., Thorleifsson, G., Tikkanen, E., Wood, A. R., Almgren, P., Atalay, M., Benediktsson, R., Bonnycastle, L. L., Burtt, N., Carey, J., Charpentier, G., Crenshaw, A. T., Doney, A. S., Dorkhan, M., Edkins, S., Emilsson, V., Eury, E., Forsen, T., Gertow, K., Gigante, B., Grant, G. B., Groves, C. J., Guiducci, C., Herder, C., Hreidarsson, A. B., Hui, J., James, A., Jonsson, A., Rathmann, W., Klopp, N., Kravic, J., Krjutskov, K., Langford, C., Leander, K., Lindholm, E., Lobbens, S., Mannisto, S., Mirza, G., Muehleisen, T. W., Musk, B., Parkin, M., Rallidis, L., Saramies, J., Sennblad, B., Shah, S., Sigurdsson, G., Silveira, A., Steinbach, G., Thorand, B., Trakalo, J., Veglia, F., Wennauer, R., Winckler, W., Zabaneh, D., Campbell, H., Van Duijn, C., Uitterlinden, A. G., Hofman, A., Sijbrands, E., Abecasis, G. R., Owen, K. R., Zeggini, E., Trip, M. D., Forouhi, N. G., Syvanen, A., Eriksson, J. G., Peltonen, L., Noethen, M. M., Balkau, B., Palmer, C. N., Lyssenko, V., Tuomi, T., Isomaa, B., Hunter, D. J., Qi, L., Shuldiner, A. R., Roden, M., Barroso, I., Wilsgaard, T., Beilby, J., Hovingh, K., Price, J. F., Wilson, J. F., Rauramaa, R., Lakka, T. A., Lind, L., Dedoussis, G., Njolstad, I., Pedersen, N. L., Khaw, K., Wareham, N. J., Keinanen-Kiukaanniemi, S. M., Saaristo, T. E., Korpi-Hyovalti, E., Saltevo, J., Laakso, M., Kuusisto, J., Metspalu, A., Collins, F. S., Mohlke, K. L., Bergman, R. N., Tuomilehto, J., Boehm, B. O., Gieger, C., Hveem, K., Cauchi, S., Froguel, P., Baldassarre, D., Tremoli, E., Humphries, S. E., Saleheen, D., Danesh, J., Ingelsson, E., Ripatti, S., Salomaa, V., Erbel, R., Joeckel, K., Moebus, S., Peters, A., Illig, T., de Faire, U., Hamsten, A., Morris, A. D., Donnelly, P. J., Frayling, T. M., Hattersley, A. T., Boerwinkle, E., Melander, O., Kathiresan, S., Nilsson, P. M., Deloukas, P., Thorsteinsdottir, U., Groop, L. C., Stefansson, K., Hu, F., Pankow, J. S., Dupuis, J., Meigs, J. B., Altshuler, D., Boehnke, M., McCarthy, M. I. 2012; 44 (9): 981-?

    Abstract

    To extend understanding of the genetic architecture and molecular basis of type 2 diabetes (T2D), we conducted a meta-analysis of genetic variants on the Metabochip, including 34,840 cases and 114,981 controls, overwhelmingly of European descent. We identified ten previously unreported T2D susceptibility loci, including two showing sex-differentiated association. Genome-wide analyses of these data are consistent with a long tail of additional common variant loci explaining much of the variation in susceptibility to T2D. Exploration of the enlarged set of susceptibility loci implicates several processes, including CREBBP-related transcription, adipocytokine signaling and cell cycle regulation, in diabetes pathogenesis.

    View details for DOI 10.1038/ng.2383

    View details for Web of Science ID 000308491200009

    View details for PubMedID 22885922

  • Association between glomerular filtration rate and endothelial function in an elderly community cohort ATHEROSCLEROSIS Nerpin, E., Ingelsson, E., Riserus, U., Helmersson-Karlqvist, J., Sundstrom, J., Jobs, E., Larsson, A., Lind, L., Arnlov, J. 2012; 224 (1): 242-246

    Abstract

    Endothelial dysfunction is prevalent among individuals with chronic kidney disease. However, the association between glomerular filtration rate and endothelial function in the community is unclear and needs to be investigated in the general population.In the community-based Prospective Investigation of the Vasculature of Uppsala Seniors study (PIVUS, n = 952, mean age 70, women 49.3%), we investigated cross-sectional associations between estimated cystatin C-based glomerular filtration rate (eGFR), and 3 measures representing different aspects of endothelial function (endothelial-dependent vasodilation [EDV], endothelial independent vasodilatation [EIDV], and flow-mediated dilatation [FMD]). We also performed pre-specified sub-group analyses in participants with normal eGFR (>60 ml/min/1.73 m(2)).In the whole cohort, 10 ml/min/1.73 m(2) higher eGFR was associated with 3% higher EDV (p = 0.001) and 2% higher EIDV (p = 0.007), adjusted for age and sex. The associations were attenuated and no longer statistically significant after adjusting for established cardiovascular risk factors. In participants with eGFR >60 ml/min/1.73 m(2), 10 ml higher eGFR was associated with 2% higher EDV (p = 0.04) after adjusting for sex and age. eGFR was not associated to FMD in any model or sub-sample.This community-based study suggests that eGFR is associated with endothelial function also in persons with normal kidney function, but that this association is largely explained by confounding by established cardiovascular risk factors. Thus, our data do not support the notion of a direct causal interplay between renal and vascular function prior to the development of CKD.

    View details for DOI 10.1016/j.atherosclerosis.2012.07.008

    View details for Web of Science ID 000308078000038

    View details for PubMedID 22841608

  • Relations of Circulating Resistin and Adiponectin and Cardiac Structure and Function: The Framingham Offspring Study OBESITY McManus, D. D., Lyass, A., Ingelsson, E., Massaro, J. M., Meigs, J. B., Aragam, J., Benjamin, E. J., Vasan, R. S. 2012; 20 (9): 1882-1886

    Abstract

    Obesity is associated with pathological cardiac remodeling and risk of heart failure (HF). Adipocytokines (ADKs) may mediate the increased risk of cardiovascular disease associated with excess adiposity. Yet data relating ADKs to cardiac remodeling phenotypes are sparse. We related two circulating ADKs, resistin and adiponectin, to three important echocardiographic markers of cardiac remodeling, left ventricular mass (LVM), left atrial diameter (LAD), and LV fractional shortening (LVFS) in 2,615 participants (mean age 61 years, 55% women) in the Framingham Offspring Study. Adiponectin concentrations were inversely related to LVM in multivariable linear regression models adjusting for key clinical correlates including BMI (regression coefficient per s.d.-increment in ln-adiponectin = -3.37, P = 0.02; P for trend across quartiles = 0.02). Adiponectin was not associated with LAD or LVFS (P > 0.56). Resistin concentrations were inversely related to LVFS (regression coefficient per s.d.-increment in ln-resistin = -0.01, P = 0.03; P for trend across quartiles = 0.04). Resistin was not associated with LVM or LAD (P > 0.05). In our moderate-sized, community-based sample, higher circulating concentrations of adiponectin and resistin were associated with lower LVM and lower LVFS, respectively. In conclusion, these associations identify potential mechanisms by which excess adiposity may mediate adverse cardiac remodeling and HF risk.

    View details for DOI 10.1038/oby.2011.32

    View details for Web of Science ID 000308279100020

    View details for PubMedID 21350435

  • Relations of circulating vitamin D concentrations with left ventricular geometry and function. European journal of heart failure Fall, T., Shiue, I., Bergeå Af Geijerstam, P., Sundström, J., Ärnlöv, J., Larsson, A., Melhus, H., Lind, L., Ingelsson, E. 2012; 14 (9): 985-991

    Abstract

    Vitamin D deficiency has been associated with risk of overt cardiovascular disease (CVD), but associations with subclinical disease are not well characterized. Hence, we examined associations of circulating vitamin D concentrations and left ventricular (LV) geometry and function by echocardiography at baseline and after 5 years in a community-based study.In the PIVUS study, we measured serum 25-dihydroxyvitamin-D (25-OH D) at age 70 and performed echocardiography including LV mass, wall thickness, end-diastolic diameter, end-systolic diameter (LVESD), left atrial diameter, fractional shortening, ejection fraction, isovolumic relaxation time, and E/A ratio at both age 70 and 75. We included 870 participants (52% women) without prior myocardial infarctions, heart failure, or prevalent valvular disease. After adjusting for potential confounders, 25-OH D at baseline was found to be significantly associated with LVESD, fractional shortening, and ejection fraction (β, -0.42 mm, P = 0.03; β, 0.70%, P = 0.03; and β, 0.91% P = 0.01, respectively), per 1 SD increase in 25-OH D (SD = 20 nmol/L) at baseline. In longitudinal analyses, vitamin D levels at baseline were not significantly associated with change in LV geometry and function after 5 years.In our community-based study among the elderly, we found higher circulating vitamin D concentrations to be associated cross-sectionally with better LV systolic function and smaller LVESD at baseline. The association persisted after adjusting for several potential confounders, including cardiovascular risk factors and calcium, phosphate, and parathyroid hormone levels. Randomized clinical trials are needed to establish firmly or refute a causal relationship between vitamin D levels and changes in LV geometry and function.

    View details for DOI 10.1093/eurjhf/hfs091

    View details for PubMedID 22723659

  • Relations of circulating vitamin D concentrations with left ventricular geometry and function EUROPEAN JOURNAL OF HEART FAILURE Fall, T., Shiue, I., af Geijerstam, P. B., Sundstrom, J., Arnlov, J., Larsson, A., Melhus, H., Lind, L., Ingelsson, E. 2012; 14 (9): 985-991
  • Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways NATURE GENETICS Scott, R. A., Lagou, V., Welch, R. P., Wheeler, E., Montasser, M. E., Luan, J., Maegi, R., Strawbridge, R. J., Rehnberg, E., Gustafsson, S., Kanoni, S., Rasmussen-Torvik, L. J., Yengo, L., Lecoeur, C., Shungin, D., Sanna, S., Sidore, C., Johnson, P. C., Jukema, J. W., Johnson, T., Mahajan, A., Verweij, N., Thorleifsson, G., Hottenga, J., Shah, S., Smith, A. V., Sennblad, B., Gieger, C., Salo, P., Perola, M., Timpson, N. J., Evans, D. M., St Pourcain, B., Wu, Y., Andrews, J. S., Hui, J., Bielak, L. F., Zhao, W., Horikoshi, M., Navarro, P., Isaacs, A., O'Connell, J. R., Stirrups, K., Vitart, V., Hayward, C., Esko, T., Mihailov, E., Fraser, R. M., Fall, T., Voight, B. F., Raychaudhuri, S., Chen, H., Lindgren, C. M., Morris, A. P., Rayner, N. W., Robertson, N., Rybin, D., Liu, C., Beckmann, J. S., Willems, S. M., Chines, P. S., Jackson, A. U., Kang, H. M., Stringham, H. M., Song, K., Tanaka, T., Peden, J. F., Goel, A., Hicks, A. A., An, P., Mueller-Nurasyid, M., Franco-Cereceda, A., Folkersen, L., Marullo, L., Jansen, H., Oldehinkel, A. J., Bruinenberg, M., Pankow, J. S., North, K. E., Forouhi, N. G., Loos, R. J., Edkins, S., Varga, T. V., Hallmans, G., Oksa, H., Antonella, M., Nagaraja, R., Trompet, S., Ford, I., Bakker, S. J., Kong, A., Kumari, M., Gigante, B., Herder, C., Munroe, P. B., Caulfield, M., Antti, J., Mangino, M., Small, K., Miljkovic, I., Liu, Y., Atalay, M., Kiess, W., James, A. L., Rivadeneira, F., Uitterlinden, A. G., Palmer, C. N., Doney, A. S., Willemsen, G., Smit, J. H., Campbell, S., Polasek, O., Bonnycastle, L. L., Hercberg, S., Dimitriou, M., Bolton, J. L., Fowkes, G. R., Kovacs, P., Lindstrom, J., Zemunik, T., Bandinelli, S., Wild, S. H., Basart, H. V., Rathmann, W., Grallert, H., Maerz, W., Kleber, M. E., Boehm, B. O., Peters, A., Pramstaller, P. P., Province, M. A., Borecki, I. B., Hastie, N. D., Rudan, I., Campbell, H., Watkins, H., Farrall, M., Stumvoll, M., Ferrucci, L., Waterworth, D. M., Bergman, R. N., Collins, F. S., Tuomilehto, J., Watanabe, R. M., de Geus, E. J., Penninx, B. W., Hofman, A., Oostra, B. A., Psaty, B. M., Vollenweider, P., Wilson, J. F., Wright, A. F., Hovingh, G. K., Metspalu, A., Uusitupa, M., Magnusson, P. K., Kyvik, K. O., Kaprio, J., Price, J. F., Dedoussis, G. V., Deloukas, P., Meneton, P., Lind, L., Boehnke, M., Shuldiner, A. R., van Duijn, C. M., Morris, A. D., Toenjes, A., Peyser, P. A., Beilby, J. P., Koerner, A., Kuusisto, J., Laakso, M., Bornstein, S. R., Schwarz, P. E., Lakka, T. A., Rauramaa, R., Adair, L. S., Smith, G. D., Spector, T. D., Illig, T., de Faire, U., Hamsten, A., Gudnason, V., Kivimaki, M., Hingorani, A., Keinanen-Kiukaanniemi, S. M., Saaristo, T. E., Boomsma, D. I., Stefansson, K., van der Harst, P., Dupuis, J., Pedersen, N. L., Sattar, N., Harris, T. B., Cucca, F., Ripatti, S., Salomaa, V., Mohlke, K. L., Balkau, B., Froguel, P., Pouta, A., Jarvelin, M., Wareham, N. J., Bouatia-Naji, N., McCarthy, M. I., Franks, P. W., Meigs, J. B., Teslovich, T. M., Florez, J. C., Langenberg, C., Ingelsson, E., Prokopenko, I., Barroso, I. 2012; 44 (9): 991-?

    Abstract

    Through genome-wide association meta-analyses of up to 133,010 individuals of European ancestry without diabetes, including individuals newly genotyped using the Metabochip, we have increased the number of confirmed loci influencing glycemic traits to 53, of which 33 also increase type 2 diabetes risk (q < 0.05). Loci influencing fasting insulin concentration showed association with lipid levels and fat distribution, suggesting impact on insulin resistance. Gene-based analyses identified further biologically plausible loci, suggesting that additional loci beyond those reaching genome-wide significance are likely to represent real associations. This conclusion is supported by an excess of directionally consistent and nominally significant signals between discovery and follow-up studies. Functional analysis of these newly discovered loci will further improve our understanding of glycemic control.

    View details for DOI 10.1038/ng.2385

    View details for Web of Science ID 000308491200010

    View details for PubMedID 22885924

  • Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study LANCET Voight, B. F., Peloso, G. M., Orho-Melander, M., Frikke-Schmidt, R., Barbalic, M., Jensen, M. K., Hindy, G., Holm, H., Ding, E. L., Johnson, T., Schunkert, H., Samani, N. J., Clarke, R., Hopewell, J. C., Thompson, J. F., Li, M., Thorleifsson, G., Newton-Cheh, C., Musunuru, K., Pirruccello, J. P., Saleheen, D., Chen, L., Stewart, A. F., Schillert, A., Thorsteinsdottir, U., Thorgeirsson, G., Anand, S., Engert, J. C., Morgan, T., Spertus, J., Stoll, M., Berger, K., Martinelli, N., Girelli, D., McKeown, P. P., Patterson, C. C., Epstein, S. E., Devaney, J., Burnett, M., Mooser, V., Ripatti, S., Surakka, I., Nieminen, M. S., Sinisalo, J., Lokki, M., Perola, M., Havulinna, A., de Faire, U., Gigante, B., Ingelsson, E., Zeller, T., Wild, P., de Bakker, P. I., Klungel, O. H., Maitland-van der Zee, A., Peters, B. J., de Boer, A., Grobbee, D. E., Kamphuisen, P. W., Deneer, V. H., Elbers, C. C., Onland-Moret, N. C., Hofker, M. H., Wijmenga, C., Verschuren, W. M., Boer, J. M., van der Schouw, Y. T., Rasheed, A., Frossard, P., Demissie, S., Willer, C., Do, R., Ordovas, J. M., Abecasis, G. R., Boehnke, M., Mohlke, K. L., Daly, M. J., Guiducci, C., Burtt, N. P., Surti, A., Gonzalez, E., Purcell, S., Gabriel, S., Marrugat, J., Peden, J., Erdmann, J., Diemert, P., Willenborg, C., Koenig, I. R., Fischer, M., Hengstenberg, C., Ziegler, A., Buysschaert, I., Lambrechts, D., Van de Werf, F., Fox, K. A., El Mokhtari, N. E., Rubin, D., Schrezenmeir, J., Schreiber, S., Schaefer, A., Danesh, J., Blankenberg, S., Roberts, R., McPherson, R., Watkins, H., Hall, A. S., Overvad, K., Rimm, E., Boerwinkle, E., Tybjaerg-Hansen, A., Cupples, L. A., Reilly, M. P., Melander, O., Mannucci, P. M., Ardissino, D., Siscovick, D., Elosua, R., Stefansson, K., O'Donnell, C. J., Salomaa, V., Rader, D. J., Peltonen, L., Schwartz, S. M., Altshuler, D., Kathiresan, S. 2012; 380 (9841): 572-580

    Abstract

    High plasma HDL cholesterol is associated with reduced risk of myocardial infarction, but whether this association is causal is unclear. Exploiting the fact that genotypes are randomly assigned at meiosis, are independent of non-genetic confounding, and are unmodified by disease processes, mendelian randomisation can be used to test the hypothesis that the association of a plasma biomarker with disease is causal.We performed two mendelian randomisation analyses. First, we used as an instrument a single nucleotide polymorphism (SNP) in the endothelial lipase gene (LIPG Asn396Ser) and tested this SNP in 20 studies (20,913 myocardial infarction cases, 95,407 controls). Second, we used as an instrument a genetic score consisting of 14 common SNPs that exclusively associate with HDL cholesterol and tested this score in up to 12,482 cases of myocardial infarction and 41,331 controls. As a positive control, we also tested a genetic score of 13 common SNPs exclusively associated with LDL cholesterol.Carriers of the LIPG 396Ser allele (2·6% frequency) had higher HDL cholesterol (0·14 mmol/L higher, p=8×10(-13)) but similar levels of other lipid and non-lipid risk factors for myocardial infarction compared with non-carriers. This difference in HDL cholesterol is expected to decrease risk of myocardial infarction by 13% (odds ratio [OR] 0·87, 95% CI 0·84-0·91). However, we noted that the 396Ser allele was not associated with risk of myocardial infarction (OR 0·99, 95% CI 0·88-1·11, p=0·85). From observational epidemiology, an increase of 1 SD in HDL cholesterol was associated with reduced risk of myocardial infarction (OR 0·62, 95% CI 0·58-0·66). However, a 1 SD increase in HDL cholesterol due to genetic score was not associated with risk of myocardial infarction (OR 0·93, 95% CI 0·68-1·26, p=0·63). For LDL cholesterol, the estimate from observational epidemiology (a 1 SD increase in LDL cholesterol associated with OR 1·54, 95% CI 1·45-1·63) was concordant with that from genetic score (OR 2·13, 95% CI 1·69-2·69, p=2×10(-10)).Some genetic mechanisms that raise plasma HDL cholesterol do not seem to lower risk of myocardial infarction. These data challenge the concept that raising of plasma HDL cholesterol will uniformly translate into reductions in risk of myocardial infarction.US National Institutes of Health, The Wellcome Trust, European Union, British Heart Foundation, and the German Federal Ministry of Education and Research.

    View details for DOI 10.1016/S0140-6736(12)60312-2

    View details for Web of Science ID 000307511400030

    View details for PubMedID 22607825

  • Cyclooxygenase-2 inhibitors and cardiovascular risk in a nation-wide cohort study after the withdrawal of rofecoxib EUROPEAN HEART JOURNAL Back, M., Yin, L., Ingelsson, E. 2012; 33 (15): 1928-1933

    Abstract

    The use of selective cyclooxygenase (COX)-2 inhibitors (coxibs) has been associated with an increased cardiovascular risk. The aim of the present study was to evaluate the association of coxib use and future risk of cardiovascular events in a population-based cohort followed after the warnings concerning the cardiovascular safety of this class of drugs were issued.A nation-wide, population-based cohort of 7 million subjects, integrating data from the Prescribed Drug, Patient, Cause of Death, Income, Educational and Emigration Registers, was followed from 1 July 2005 to 31 December 2008. Analyses were performed for different cardiovascular outcomes in the whole population after exclusion of individuals with prior cardiovascular diagnosis (incident primary cardiovascular events; sample size, n = 6 991 645). Cox proportional hazard ratios (HRs) revealed no significant association of coxib use with risk for myocardial infarction, ischaemic stroke, or heart failure. In contrast to these findings, coxib use was associated with an increased risk for a first episode of atrial fibrillation [HR 1.16; 95% confidence interval (CI) 1.05-1.29]. A post hoc analysis for different coxibs revealed a significant association with incident atrial fibrillation for etoricoxib (HR 1.35; 95% CI 1.19-1.54) but not for celecoxib (HR 0.94; 95% CI 0.79-1.11).Whereas safety measures appear to have limited serious cardiovascular consequences of COX-2 inhibitors, the risk of developing atrial fibrillation may have been overlooked and may necessitate consideration and precautions.

    View details for DOI 10.1093/eurheartj/ehr421

    View details for Web of Science ID 000307172300019

    View details for PubMedID 22108833

  • The role of obesity-related genetic loci in insulin sensitivity DIABETIC MEDICINE Fall, T., Arnlov, J., Berne, C., Ingelsson, E. 2012; 29 (7): E62-E66

    Abstract

    Despite rapid advancements and many new diabetes susceptibility loci found in the past few years, few genetic variants associated with insulin sensitivity have been described, potentially attributable to the lack of larger cohorts examined with gold standard methods for insulin sensitivity assessment. There is a strong link between obesity and insulin sensitivity, and we hypothesized that known obesity susceptibility loci may act via effects on insulin sensitivity.A cohort of 71-year-old men without diabetes (Uppsala Longitudinal Study of Adult Men) underwent a euglycaemic-hyperinsulinaemic clamp and genotyping for genetic variants representing 32 loci recently reported to be associated with BMI (n = 926). The effect of these loci on the insulin sensitivity index (M/I ratio) was examined using linear regression. An in silico replication was performed in publically available data for the three top single-nucleotide polymorphisms from the Meta-Analyses of Glucose and Insulin-related traits Consortium analyses of homeostasis model assessment of insulin resistance (n = 37,037).Three loci (SH2B1, MTCH2 and NEGR1) were associated with decreased insulin sensitivity at a nominal significance (P ≤ 0.05) after adjustment for BMI, but did not hold for multiple comparison correction. SH2B1 rs7359397 was also associated with homeostasis model assessment of insulin resistance in the Meta-Analyses of Glucose and Insulin-related traits Consortium data set (P = 3.9 × 10(-3)).Our study supports earlier reports of SH2B1 to be of importance in insulin sensitivity and, in addition, suggests potential roles of NEGR1 and MTCH2.

    View details for DOI 10.1111/j.1464-5491.2012.03665.x

    View details for Web of Science ID 000305458500009

    View details for PubMedID 22443470

  • A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance NATURE GENETICS Manning, A. K., Hivert, M., Scott, R. A., Grimsby, J. L., Bouatia-Naji, N., Chen, H., Rybin, D., Liu, C., Bielak, L. F., Prokopenko, I., Amin, N., Barnes, D., Cadby, G., Hottenga, J., Ingelsson, E., Jackson, A. U., Johnson, T., Kanoni, S., Ladenvall, C., Lagou, V., Lahti, J., Lecoeur, C., Liu, Y., Martinez-Larrad, M. T., Montasser, M. E., Navarro, P., Perry, J. R., Rasmussen-Torvik, L. J., Salo, P., Sattar, N., Shungin, D., Strawbridge, R. J., Tanaka, T., van Duijn, C. M., An, P., de Andrade, M., Andrews, J. S., Aspelund, T., Atalay, M., Aulchenko, Y., Balkau, B., Bandinelli, S., Beckmann, J. S., Beilby, J. P., Bellis, C., Bergman, R. N., Blangero, J., Boban, M., Boehnke, M., Boerwinkle, E., Bonnycastle, L. L., Boomsma, D. I., Borecki, I. B., Boettcher, Y., Bouchard, C., Brunner, E., Budimir, D., Campbell, H., Carlson, O., Chines, P. S., Clarke, R., Collins, F. S., Corbaton-Anchuelo, A., Couper, D., de Faire, U., Dedoussis, G. V., Deloukas, P., Dimitriou, M., Egan, J. M., Eiriksdottir, G., Erdos, M. R., Eriksson, J. G., Eury, E., Ferrucci, L., Ford, I., Forouhi, N. G., Fox, C. S., Franzosi, M. G., Franks, P. W., Frayling, T. M., Froguel, P., Galan, P., de Geus, E., Gigante, B., Glazer, N. L., Goel, A., Groop, L., Gudnason, V., Hallmans, G., Hamsten, A., Hansson, O., Harris, T. B., Hayward, C., Heath, S., Hercberg, S., Hicks, A. A., Hingorani, A., Hofman, A., Hui, J., Hung, J., Jarvelin, M., Jhun, M. A., Johnson, P. C., Jukema, J. W., Jula, A., Kao, W. H., Kaprio, J., Kardia, S. L., Keinanen-Kiukaanniemi, S., Kivimaki, M., Kolcic, I., Kovacs, P., Kumari, M., Kuusisto, J., Kyvik, K. O., Laakso, M., Lakka, T., Lannfelt, L., Lathrop, G. M., Launer, L. J., Leander, K., Li, G., Lind, L., Lindstrom, J., Lobbens, S., Loos, R. J., Luan, J., Lyssenko, V., Magi, R., Magnusson, P. K., Marmot, M., Meneton, P., Mohlke, K. L., Mooser, V., Morken, M. A., Miljkovic, I., Narisu, N., O'Connell, J., Ong, K. K., Oostra, B. A., Palmer, L. J., Palotie, A., Pankow, J. S., Peden, J. F., Pedersen, N. L., Pehlic, M., Peltonen, L., Penninx, B., Pericic, M., Perola, M., Perusse, L., Peyser, P. A., Polasek, O., Pramstaller, P. P., Province, M. A., Raikkonen, K., Rauramaa, R., Rehnberg, E., Rice, K., Rotter, J. I., Rudan, I., Ruokonen, A., Saaristo, T., Sabater-Lleal, M., Salomaa, V., Savage, D. B., Saxena, R., Schwarz, P., Seedorf, U., Sennblad, B., Serrano-Rios, M., Shuldiner, A. R., Sijbrands, E. J., Siscovick, D. S., Smit, J. H., Small, K. S., Smith, N. L., Smith, A. V., Stancakova, A., Stirrups, K., Stumvoll, M., Sun, Y. V., Swift, A. J., Toenjes, A., Tuomilehto, J., Trompet, S., Uitterlinden, A. G., Uusitupa, M., Vikstrom, M., Vitart, V., Vohl, M., Voight, B. F., Vollenweider, P., Waeber, G., Waterworth, D. M., Watkins, H., Wheeler, E., Widen, E., Wild, S. H., Willems, S. M., Willemsen, G., Wilson, J. F., Witteman, J. C., Wright, A. F., Yaghootkar, H., Zelenika, D., Zemunik, T., Zgaga, L., Wareham, N. J., McCarthy, M. I., Barroso, I., Watanabe, R. M., Florez, J. C., Dupuis, J., Meigs, J. B., Langenberg, C. 2012; 44 (6): 659-U81

    Abstract

    Recent genome-wide association studies have described many loci implicated in type 2 diabetes (T2D) pathophysiology and β-cell dysfunction but have contributed little to the understanding of the genetic basis of insulin resistance. We hypothesized that genes implicated in insulin resistance pathways might be uncovered by accounting for differences in body mass index (BMI) and potential interactions between BMI and genetic variants. We applied a joint meta-analysis approach to test associations with fasting insulin and glucose on a genome-wide scale. We present six previously unknown loci associated with fasting insulin at P < 5 × 10(-8) in combined discovery and follow-up analyses of 52 studies comprising up to 96,496 non-diabetic individuals. Risk variants were associated with higher triglyceride and lower high-density lipoprotein (HDL) cholesterol levels, suggesting a role for these loci in insulin resistance pathways. The discovery of these loci will aid further characterization of the role of insulin resistance in T2D pathophysiology.

    View details for DOI 10.1038/ng.2274

    View details for Web of Science ID 000304551100012

    View details for PubMedID 22581228

  • No Interactions Between Previously Associated 2-Hour Glucose Gene Variants and Physical Activity or BMI on 2-Hour Glucose Levels DIABETES Scott, R. A., Chu, A. Y., Grarup, N., Manning, A. K., Hivert, M., Shungin, D., Toenjes, A., Yesupriya, A., Barnes, D., Bouatia-Naji, N., Glazer, N. L., Jackson, A. U., Kutalik, Z., Lagou, V., Marek, D., Rasmussen-Torvik, L. J., Stringham, H. M., Tanaka, T., Aadahl, M., Arking, D. E., Bergmann, S., Boerwinkle, E., Bonnycastle, L. L., Bornstein, S. R., Brunner, E., Bumpstead, S. J., Brage, S., Carlson, O. D., Chen, H., Chen, Y. I., Chines, P. S., Collins, F. S., Couper, D. J., Dennison, E. M., Dowling, N. F., Egan, J. S., Ekelund, U., Erdos, M. R., Forouhi, N. G., Fox, C. S., Goodarzi, M. O., Graessler, J., Gustafsson, S., Hallmans, G., Hansen, T., Hingorani, A., Holloway, J. W., Hu, F. B., Isomaa, B., Jameson, K. A., Johansson, I., Jonsson, A., Jorgensen, T., Kivimaki, M., Kovacs, P., Kumari, M., Kuusisto, J., Laakso, M., Lecoeur, C., Levy-Marchal, C., Li, G., Loos, R. J., Lyssenko, V., Marmot, M., Marques-Vidal, P., Morken, M. A., Mueller, G., North, K. E., Pankow, J. S., Payne, F., Prokopenko, I., Psaty, B. M., Renstrom, F., Rice, K., Rotter, J. I., Rybin, D., Sandholt, C. H., Sayer, A. A., Shrader, P., Schwarz, P. E., Siscovick, D. S., Stancakova, A., Stumvoll, M., Teslovich, T. M., Waeber, G., Williams, G. H., Witte, D. R., Wood, A. R., Xie, W., Boehnke, M., Cooper, C., Ferrucci, L., Froguel, P., Groop, L., Kao, W. H., Vollenweider, P., Walker, M., Watanabe, R. M., Pedersen, O., Meigs, J. B., Ingelsson, E., Barroso, I., Florez, J. C., Franks, P. W., Dupuis, J., Wareham, N. J., Langenberg, C. 2012; 61 (5): 1291-1296

    Abstract

    Gene-lifestyle interactions have been suggested to contribute to the development of type 2 diabetes. Glucose levels 2 h after a standard 75-g glucose challenge are used to diagnose diabetes and are associated with both genetic and lifestyle factors. However, whether these factors interact to determine 2-h glucose levels is unknown. We meta-analyzed single nucleotide polymorphism (SNP) × BMI and SNP × physical activity (PA) interaction regression models for five SNPs previously associated with 2-h glucose levels from up to 22 studies comprising 54,884 individuals without diabetes. PA levels were dichotomized, with individuals below the first quintile classified as inactive (20%) and the remainder as active (80%). BMI was considered a continuous trait. Inactive individuals had higher 2-h glucose levels than active individuals (β = 0.22 mmol/L [95% CI 0.13-0.31], P = 1.63 × 10(-6)). All SNPs were associated with 2-h glucose (β = 0.06-0.12 mmol/allele, P ≤ 1.53 × 10(-7)), but no significant interactions were found with PA (P > 0.18) or BMI (P ≥ 0.04). In this large study of gene-lifestyle interaction, we observed no interactions between genetic and lifestyle factors, both of which were associated with 2-h glucose. It is perhaps unlikely that top loci from genome-wide association studies will exhibit strong subgroup-specific effects, and may not, therefore, make the best candidates for the study of interactions.

    View details for DOI 10.2337/db11-0973

    View details for Web of Science ID 000303179100042

    View details for PubMedID 22415877

  • Risk Prediction Measures for Case-Cohort and Nested Case-Control Designs: An Application to Cardiovascular Disease AMERICAN JOURNAL OF EPIDEMIOLOGY Ganna, A., Reilly, M., de Faire, U., Pedersen, N., Magnusson, P., Ingelsson, E. 2012; 175 (7): 715-724

    Abstract

    Case-cohort and nested case-control designs are often used to select an appropriate subsample of individuals from prospective cohort studies. Despite the great attention that has been given to the calculation of association estimators, no formal methods have been described for estimating risk prediction measures from these 2 sampling designs. Using real data from the Swedish Twin Registry (2004-2009), the authors sampled unstratified and stratified (matched) case-cohort and nested case-control subsamples and compared them with the full cohort (as "gold standard"). The real biomarker (high density lipoprotein cholesterol) and simulated biomarkers (BIO1 and BIO2) were studied in terms of association with cardiovascular disease, individual risk of cardiovascular disease at 3 years, and main prediction metrics. Overall, stratification improved efficiency, with stratified case-cohort designs being comparable to matched nested case-control designs. Individual risks and prediction measures calculated by using case-cohort and nested case-control designs after appropriate reweighting could be assessed with good efficiency, except for the finely matched nested case-control design, where matching variables could not be included in the individual risk estimation. In conclusion, the authors have shown that case-cohort and nested case-control designs can be used in settings where the research aim is to evaluate the prediction ability of new markers and that matching strategies for nested case-control designs may lead to biased prediction measures.

    View details for DOI 10.1093/aje/kwr374

    View details for Web of Science ID 000302483500016

    View details for PubMedID 22396388

  • Familial Effects on Ischemic Stroke The Role of Sibling Kinship, Sex, and Age of Onset CIRCULATION-CARDIOVASCULAR GENETICS Kasiman, K., Lundholm, C., Sandin, S., Malki, N., Sparen, P., Ingelsson, E. 2012; 5 (2): 226-233

    Abstract

    Previous studies on familial risk of ischemic stroke have supported genetic influence on the disease incidence. This study aimed to characterize these familial effects in a nationwide population-based study by taking into account sibling relations, sex of siblings, and age of onset, with respect to ischemic stroke incidence.Incident ischemic stroke cases identified from the Swedish Hospital Discharge and Cause of Death Registers between 1987 and 2007 were linked to their stroke-free siblings (study participants), forming an exposed sib-pair. Each exposed sib-pair was matched up to 5 unexposed sib-pairs from the Multi-Generation Registry by birth and calendar years. Incident ischemic stroke risk was assessed using hazard estimates obtained from stratified Cox regression analyses. A total of 30 735 exposed and 152 391 unexposed study participants were included in the analyses. The overall risk of incident ischemic stroke when exposed was significantly increased (relative risk, 1.61; 95% confidence interval, 1.48-1.75; P<0.001). Familial risk was higher in full (relative risk, 1.64; 95% confidence interval, 1.50-1.81; P<0.001) than in half (relative risk, 1.41; 95% confidence interval, 1.10-1.82; P=0.007) siblings. Familial risk of early ischemic stroke almost doubled when exposed to early ischemic stroke (relative risk, 1.94; 95% confidence interval, 1.41-2.67; P<0.001).There was a 60% increased risk for ischemic stroke in individuals having a sibling with prior stroke. The familial effect was even higher for full-sibling relations. Familial effects were observed in both male and female individuals, and no differential effects depending on the sex of either of the siblings were found.

    View details for DOI 10.1161/CIRCGENETICS.111.962241

    View details for Web of Science ID 000309884300013

    View details for PubMedID 22407453

  • Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies LANCET Sarwar, N., Butterworth, A. S., Freitag, D. F., Gregson, J., Willeit, P., Gorman, D. N., Gao, P., Saleheen, D., Rendon, A., Nelson, C. P., Braund, P. S., Hall, A. S., Chasman, D. I., Tybjaerg-Hansen, A., Chambers, J. C., Benjamin, E. J., Franks, P. W., Clarke, R., Wilde, A. A., Trip, M. D., Steri, M., Witteman, J. C., Qi, L., van der Schoot, C. E., de Faire, U., Erdmann, J., Stringham, H. M., Koenig, W., Rader, D. J., Melzer, D., Reich, D., Psaty, B. M., Kleber, M. E., Panagiotakos, D. B., Willeit, J., Wennberg, P., Woodward, M., Adamovic, S., Rimm, E. B., Meade, T. W., Gillum, R. F., Shaffer, J. A., Hofman, A., Onat, A., Sundstrom, J., Wassertheil-Smoller, S., Mellstrom, D., Gallacher, J., Cushman, M., Tracy, R. P., Kauhanen, J., Karlsson, M., Salonen, J. T., Wilhelmsen, L., Amouyel, P., Cantin, B., Best, L. G., Ben-Shlomo, Y., Manson, J. E., Davey-Smith, G., de Bakker, P. I., O'Donnell, C. J., Wilson, J. F., Wilson, A. G., Assimes, T. L., Jansson, J., Ohlsson, C., Tivesten, A., Ljunggren, O., Reilly, M. P., Hamsten, A., Ingelsson, E., Cambien, F., Hung, J., Thomas, G. N., Boehnke, M., Schunkert, H., Asselbergs, F. W., Kastelein, J. J., Gudnason, V., Salomaa, V., Harris, T. B., Kooner, J. S., Allin, K. H., Nordestgaard, B. G., Hopewell, J. C., Goodall, A. H., Ridker, P. M., Holm, H., Watkins, H., Ouwehand, W. H., Samani, N. J., Kaptoge, S., Di Angelantonio, E., Harari, O., Danesh, J. 2012; 379 (9822): 1205-1213

    Abstract

    Persistent inflammation has been proposed to contribute to various stages in the pathogenesis of cardiovascular disease. Interleukin-6 receptor (IL6R) signalling propagates downstream inflammation cascades. To assess whether this pathway is causally relevant to coronary heart disease, we studied a functional genetic variant known to affect IL6R signalling.In a collaborative meta-analysis, we studied Asp358Ala (rs2228145) in IL6R in relation to a panel of conventional risk factors and inflammation biomarkers in 125,222 participants. We also compared the frequency of Asp358Ala in 51,441 patients with coronary heart disease and in 136,226 controls. To gain insight into possible mechanisms, we assessed Asp358Ala in relation to localised gene expression and to postlipopolysaccharide stimulation of interleukin 6.The minor allele frequency of Asp358Ala was 39%. Asp358Ala was not associated with lipid concentrations, blood pressure, adiposity, dysglycaemia, or smoking (p value for association per minor allele ≥0·04 for each). By contrast, for every copy of 358Ala inherited, mean concentration of IL6R increased by 34·3% (95% CI 30·4-38·2) and of interleukin 6 by 14·6% (10·7-18·4), and mean concentration of C-reactive protein was reduced by 7·5% (5·9-9·1) and of fibrinogen by 1·0% (0·7-1·3). For every copy of 358Ala inherited, risk of coronary heart disease was reduced by 3·4% (1·8-5·0). Asp358Ala was not related to IL6R mRNA levels or interleukin-6 production in monocytes.Large-scale human genetic and biomarker data are consistent with a causal association between IL6R-related pathways and coronary heart disease.British Heart Foundation; UK Medical Research Council; UK National Institute of Health Research, Cambridge Biomedical Research Centre; BUPA Foundation.

    View details for DOI 10.1016/S0140-6736(11)61931-4

    View details for Web of Science ID 000302230400033

    View details for PubMedID 22421339

  • Meta-analyses identify 13 loci associated with age at menopause and highlight DNA repair and immune pathways NATURE GENETICS Stolk, L., Perry, J. R., Chasman, D. I., He, C., Mangino, M., Sulem, P., Barbalic, M., Broer, L., Byrne, E. M., Ernst, F., Esko, T., Franceschini, N., Gudbjartsson, D. F., Hottenga, J., Kraft, P., McArdle, P. F., Porcu, E., Shin, S., Smith, A. V., van Wingerden, S., Zhai, G., Zhuang, W. V., Albrecht, E., Alizadeh, B. Z., Aspelund, T., Bandinelli, S., Lauc, L. B., Beckmann, J. S., Boban, M., Boerwinkle, E., Broekmans, F. J., Burri, A., Campbell, H., Chanock, S. J., Chen, C., Cornelis, M. C., Corre, T., Coviello, A. D., D'Adamo, P., Davies, G., de Faire, U., de Geus, E. J., Deary, I. J., Dedoussis, G. V., Deloukas, P., Ebrahim, S., Eiriksdottir, G., Emilsson, V., Eriksson, J. G., Fauser, B. C., Ferreli, L., Ferrucci, L., Fischer, K., Folsom, A. R., Garcia, M. E., Gasparini, P., Gieger, C., Glazer, N., Grobbee, D. E., Hall, P., Haller, T., Hankinson, S. E., Hass, M., Hayward, C., Heath, A. C., Hofman, A., Ingelsson, E., Janssens, A. C., Johnson, A. D., Karasik, D., Kardia, S. L., Keyzer, J., Kiel, D. P., Kolcic, I., Kutalik, Z., Lahti, J., Lai, S., Laisk, T., Laven, J. S., Lawlor, D. A., Liu, J., Lopez, L. M., Louwers, Y. V., Magnusson, P. K., Marongiu, M., Martin, N. G., Klaric, I. M., Masciullo, C., McKnight, B., Medland, S. E., Melzer, D., Mooser, V., Navarro, P., Newman, A. B., Nyholt, D. R., Onland-Moret, N. C., Palotie, A., Pare, G., Parker, A. N., Pedersen, N. L., Peeters, P. H., Pistis, G., Plump, A. S., Polasek, O., Pop, V. J., Psaty, B. M., Raikkonen, K., Rehnberg, E., Rotter, J. I., Rudan, I., Sala, C., Salumets, A., Scuteri, A., Singleton, A., Smith, J. A., Snieder, H., Soranzo, N., Stacey, S. N., Starr, J. M., Stathopoulou, M. G., Stirrups, K., Stolk, R. P., Styrkarsdottir, U., Sun, Y. V., Tenesa, A., Thorand, B., Toniolo, D., Tryggvadottir, L., Tsui, K., Ulivi, S., van Dam, R. M., van der Schouw, Y. T., van Gils, C. H., van Nierop, P., Vink, J. M., Visscher, P. M., Voorhuis, M., Waeber, G., Wallaschofski, H., Wichmann, H. E., Widen, E., Wijnands-van Gent, C. J., Willemsen, G., Wilson, J. F., Wolffenbuttel, B. H., Wright, A. F., Yerges-Armstrong, L. M., Zemunik, T., Zgaga, L., Zillikens, M. C., Zygmunt, M., Arnold, A. M., Boomsma, D. I., Buring, J. E., Crisponi, L., Demerath, E. W., Gudnason, V., Harris, T. B., Hu, F. B., Hunter, D. J., Launer, L. J., Metspalu, A., Montgomery, G. W., Oostra, B. A., Ridker, P. M., Sanna, S., Schlessinger, D., Spector, T. D., Stefansson, K., Streeten, E. A., Thorsteinsdottir, U., Uda, M., Uitterlinden, A. G., van Duijn, C. M., Voelzke, H., Murray, A., Murabito, J. M., Visser, J. A., Lunetta, K. L. 2012; 44 (3): 260-U55

    Abstract

    To newly identify loci for age at natural menopause, we carried out a meta-analysis of 22 genome-wide association studies (GWAS) in 38,968 women of European descent, with replication in up to 14,435 women. In addition to four known loci, we identified 13 loci newly associated with age at natural menopause (at P < 5 × 10(-8)). Candidate genes located at these newly associated loci include genes implicated in DNA repair (EXO1, HELQ, UIMC1, FAM175A, FANCI, TLK1, POLG and PRIM1) and immune function (IL11, NLRP11 and PRRC2A (also known as BAT2)). Gene-set enrichment pathway analyses using the full GWAS data set identified exoDNase, NF-κB signaling and mitochondrial dysfunction as biological processes related to timing of menopause.

    View details for DOI 10.1038/ng.1051

    View details for Web of Science ID 000300843600010

    View details for PubMedID 22267201

  • Genome-wide and gene-based association implicates FRMD6 in alzheimer disease HUMAN MUTATION Hong, M., Reynolds, C. A., Feldman, A. L., Kallin, M., Lambert, J., Amouyel, P., Ingelsson, E., Pedersen, N. L., Prince, J. A. 2012; 33 (3): 521-529

    Abstract

    Genome-wide association studies (GWAS) that allow for allelic heterogeneity may facilitate the discovery of novel genes not detectable by models that require replication of a single variant site. One strategy to accomplish this is to focus on genes rather than markers as units of association, and so potentially capture a spectrum of causal alleles that differ across populations. Here, we conducted a GWAS of Alzheimer disease (AD) in 2,586 Swedes and performed gene-based meta-analysis with three additional studies from France, Canada, and the United States, in total encompassing 4,259 cases and 8,284 controls. Implementing a newly designed gene-based algorithm, we identified two loci apart from the region around APOE that achieved study-wide significance in combined samples, the strongest finding being for FRMD6 on chromosome 14q (P = 2.6 × 10(-14)) and a weaker signal for NARS2 that is immediately adjacent to GAB2 on chromosome 11q (P = 7.8 × 10(-9)). Ontology-based pathway analyses revealed significant enrichment of genes involved in glycosylation. Results suggest that gene-based approaches that accommodate allelic heterogeneity in GWAS can provide a complementary avenue for gene discovery and may help to explain a portion of the missing heritability not detectable with single nucleotide polymorphisms (SNPs) derived from marker-specific meta-analysis.

    View details for DOI 10.1002/humu.22009

    View details for Web of Science ID 000300706000013

    View details for PubMedID 22190428

  • Nationwide cohort study of the leukotriene receptor antagonist montelukast and incident or recurrent cardiovascular disease JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY Ingelsson, E., Yin, L., Back, M. 2012; 129 (3): 702-U153

    Abstract

    The leukotriene pathway has been associated with an increased cardiovascular risk. However, the effects of the antileukotriene treatment used in asthmatic patients on cardiovascular outcomes have remained largely unexplored.We sought to examine a potential protective role of the leukotriene receptor antagonist montelukast on future risk of incident and recurrent myocardial infarction and ischemic stroke.A nationwide population-based cohort of approximately 7 million persons integrating data from the Prescribed Drug, Patient, Cause of Death, Income, Educational, and Emigration Registers was followed from July 1, 2005, to December 31, 2008. Analyses were performed in the whole population after exclusion of subjects with a prior cardiovascular diagnosis (incident events; sample size, n = 6,910,923 for myocardial infarction and n = 6,932,578 for stroke) and in subjects with a prior diagnosis (recurrent events; n = 153,937 and n = 132,291 for stroke and myocardial infarction, respectively).Cox proportional hazard ratios (HRs) did not reveal an association of montelukast use with incident events. In contrast to these findings, montelukast use was associated with a lower risk for recurrent stroke (HR, 0.62; 95% CI, 0.38-0.99) accounting for age, sex, education level, and yearly income. Adjusting the latter finding also for respiratory and cardiovascular medications and diagnoses revealed similar point estimates (HR, 0.62; 95% CI, 0.39-1.0). Post hoc analyses revealed a significant association of montelukast use with a lower risk for recurrent myocardial infarction in male subjects (HR, 0.65; 95% CI, 0.43-0.99).These data provide a first indication for a potential role of the antiasthma drug montelukast for secondary prevention of cardiovascular disease.

    View details for DOI 10.1016/j.jaci.2011.11.052

    View details for Web of Science ID 000301189300014

    View details for PubMedID 22244598

  • Subfertility and risk of later life maternal cardiovascular disease HUMAN REPRODUCTION Parikh, N. I., Cnattingius, S., Mittleman, M. A., Ludvigsson, J. F., Ingelsson, E. 2012; 27 (2): 568-575

    Abstract

    Subfertility shares common pathways with cardiovascular disease (CVD), including polycystic ovarian syndrome, obesity and thyroid disorders. Women with prior 0-1 pregnancies are at an increased risk of incident CVD when compared with women with two pregnancies. It is uncertain whether history of subfertility among women eventually giving birth is a risk factor for CVD.Among Swedish women with self-reported data on subfertility in the Swedish Medical Birth Register (n = 863 324), we used Cox proportional hazards models to relate a history of subfertility to CVD risk after adjustment for age, birth year, highest income, education, birth country, hypertension, diabetes, preterm birth, small for gestational age (SGA), smoking and for BMI in separate models. In additional analyses, we excluded women with: (i) pregnancy-related or non-pregnancy-related hypertension and/or diabetes; and (ii) preterm births and/or SGA babies.Among nulliparous women eventually having a childbirth (between 1983 and 2005, the median follow-up time 11.9; 0-23 years and 9 906 621 person-years of follow-up), there was an increased risk of CVD among women reporting ≥ 5 years of subfertility versus 0 years (hazard ratio 1.19, 95% confidence interval 1.02-1.39). There were not significantly elevated CVD risks for women with 1-2 or 3-4 years of subfertility versus 0 years. Accounting for BMI did not change results. Excluding women with hypertension and/or diabetes attenuated associations, whereas exclusion of women with preterm and/or SGA births did not change findings. CONCLUSIONS Subfertility among women eventually having a childbirth is a risk factor for CVD even upon accounting for cardiovascular risk factors and adverse pregnancy outcomes. Future studies should explore the mechanisms underlying this association.

    View details for DOI 10.1093/humrep/der400

    View details for Web of Science ID 000299131800032

    View details for PubMedID 22131387

  • Inflammation, oxidative stress, glomerular filtration rate, and albuminuria in elderly men: a cross-sectional study. BMC research notes Nerpin, E., Helmersson-Karlqvist, J., Risérus, U., Sundström, J., Larsson, A., Jobs, E., Basu, S., Ingelsson, E., Arnlöv, J. 2012; 5: 537-?

    Abstract

    The role of inflammation and oxidative stress in mild renal impairment in the elderly is not well studied. Accordingly, we aimed at investigating the associations between estimated glomerular filtration rate (eGFR), albumin/creatinine ratio (ACR), and markers of different inflammatory pathways and oxidative stress in a community based cohort of elderly men.Cystatin C-based GFR, ACR, and biomarkers of cytokine-mediated inflammation (interleukin-6, high-sensitivity C-reactive protein[CRP], serum amyloid A[SAA]), cyclooxygenase-mediated inflammation (urinary prostaglandin F2α [PGF2α]), and oxidative stress (urinary F2 isoprostanes) were assessed in the Uppsala Longitudinal Study of Adult Men(n = 647, mean age 77 years).In linear regression models adjusting for age, BMI, smoking, blood pressure, LDL-cholesterol, HDL-cholesterol, triglycerides, and treatment with statins, ACE-inhibitors, ASA, and anti-inflammatory agents, eGFR was inversely associated with CRP, interleukin-6, and SAA (β-coefficient -0.13 to -0.19, p < 0.001 for all), and positively associated with urinary F2-isoprostanes (β-coefficient 0.09, p = 0.02). In line with this, ACR was positively associated with CRP, interleukin-6, and SAA (β- coefficient 0.09-0.12, p < 0.02 for all), and negatively associated with urinary F2-isoprostanes (β-coefficient -0.12, p = 0.002). The associations were similar but with lower regression coefficients in a sub-sample with normal eGFR (>60 ml/min/1.73 m2, n = 514), with the exception that F2-isoprostane and SAA were no longer associated with eGFR.Our data indicate that cytokine-mediated inflammation is involved in the early stages of impaired kidney function in the elderly, but that cyclooxygenase-mediated inflammation does not play a role at this stage. The unexpected association between higher eGFR/lower albuminuria and increased F2-isoprostanes in urine merits further studies.

    View details for DOI 10.1186/1756-0500-5-537

    View details for PubMedID 23016573

  • Novel loci for adiponectin levels and their influence on type 2 diabetes and metabolic traits: a multi-ethnic meta-analysis of 45,891 individuals. PLoS genetics Dastani, Z., Hivert, M., Timpson, N., Perry, J. R., Yuan, X., Scott, R. A., Henneman, P., Heid, I. M., Kizer, J. R., Lyytikäinen, L., Fuchsberger, C., Tanaka, T., Morris, A. P., Small, K., Isaacs, A., Beekman, M., Coassin, S., Lohman, K., Qi, L., Kanoni, S., Pankow, J. S., Uh, H., Wu, Y., Bidulescu, A., Rasmussen-Torvik, L. J., Greenwood, C. M., Ladouceur, M., Grimsby, J., Manning, A. K., Liu, C., Kooner, J., Mooser, V. E., Vollenweider, P., Kapur, K. A., Chambers, J., Wareham, N. J., Langenberg, C., Frants, R., Willems-Vandijk, K., Oostra, B. A., Willems, S. M., Lamina, C., Winkler, T. W., Psaty, B. M., Tracy, R. P., Brody, J., Chen, I., Viikari, J., Kähönen, M., Pramstaller, P. P., Evans, D. M., St Pourcain, B., Sattar, N., Wood, A. R., Bandinelli, S., Carlson, O. D., Egan, J. M., Böhringer, S., van Heemst, D., Kedenko, L., Kristiansson, K., Nuotio, M., Loo, B., Harris, T., Garcia, M., Kanaya, A., Haun, M., Klopp, N., Wichmann, H., Deloukas, P., Katsareli, E., Couper, D. J., Duncan, B. B., Kloppenburg, M., Adair, L. S., Borja, J. B., Wilson, J. G., Musani, S., Guo, X., Johnson, T., Semple, R., Teslovich, T. M., Allison, M. A., Redline, S., Buxbaum, S. G., Mohlke, K. L., Meulenbelt, I., Ballantyne, C. M., Dedoussis, G. V., Hu, F. B., Liu, Y., Paulweber, B., Spector, T. D., Slagboom, P. E., Ferrucci, L., Jula, A., Perola, M., Raitakari, O., Florez, J. C., Salomaa, V., Eriksson, J. G., Frayling, T. M., Hicks, A. A., Lehtimäki, T., Smith, G. D., Siscovick, D. S., Kronenberg, F., Van Duijn, C., Loos, R. J., Waterworth, D. M., Meigs, J. B., Dupuis, J., Richards, J. B., Voight, B. F., Scott, L. J., Steinthorsdottir, V., Dina, C., Welch, R. P., Zeggini, E., Huth, C., Aulchenko, Y. S., Thorleifsson, G., McCulloch, L. J., Ferreira, T., Grallert, H., Amin, N., Wu, G., Willer, C. J., Raychaudhuri, S., McCarroll, S. A., Hofmann, O. M., Segrè, A. V., van Hoek, M., Navarro, P., Ardlie, K., Balkau, B., Benediktsson, R., Bennett, A. J., Blagieva, R., Boerwinkle, E., Bonnycastle, L. L., Boström, K. B., Bravenboer, B., Bumpstead, S., Burtt, N. P., Charpentier, G., Chines, P. S., Cornelis, M., Crawford, G., Doney, A. S., Elliott, K. S., Elliott, A. L., Erdos, M. R., Fox, C. S., Franklin, C. S., Ganser, M., Gieger, C., Grarup, N., Green, T., Griffin, S., Groves, C. J., Guiducci, C., Hadjadj, S., Hassanali, N., Herder, C., Isomaa, B., Jackson, A. U., Johnson, P. R., Jørgensen, T., Kao, W. H., Kong, A., Kraft, P., Kuusisto, J., Lauritzen, T., Li, M., Lieverse, A., Lindgren, C. M., Lyssenko, V., Marre, M., Meitinger, T., Midthjell, K., Morken, M. A., Narisu, N., Nilsson, P., Owen, K. R., Payne, F., Petersen, A., Platou, C., Proença, C., Prokopenko, I., Rathmann, W., Rayner, N. W., Robertson, N. R., Rocheleau, G., Roden, M., Sampson, M. J., Saxena, R., Shields, B. M., Shrader, P., Sigurdsson, G., Sparsø, T., Strassburger, K., Stringham, H. M., Sun, Q., Swift, A. J., Thorand, B., Tichet, J., Tuomi, T., van Dam, R. M., Van Haeften, T. W., van Herpt, T., van Vliet-Ostaptchouk, J. V., Walters, G. B., Weedon, M. N., Wijmenga, C., Witteman, J., Bergman, R. N., Cauchi, S., Collins, F. S., Gloyn, A. L., Gyllensten, U., Hansen, T., Hide, W. A., Hitman, G. A., Hofman, A., Hunter, D. J., Hveem, K., Laakso, M., Morris, A. D., Palmer, C. N., Rudan, I., Sijbrands, E., Stein, L. D., Tuomilehto, J., Uitterlinden, A., Walker, M., Watanabe, R. M., Abecasis, G. R., Boehm, B. O., Campbell, H., Daly, M. J., Hattersley, A. T., Pedersen, O., Barroso, I., Groop, L., Sladek, R., Thorsteinsdottir, U., Wilson, J. F., Illig, T., Froguel, P., van Duijn, C. M., Stefansson, K., Altshuler, D., Boehnke, M., McCarthy, M. I., Soranzo, N., Wheeler, E., Glazer, N. L., Bouatia-Naji, N., Mägi, R., Randall, J., Elliott, P., Rybin, D., Dehghan, A., Hottenga, J. J., Song, K., Goel, A., Lajunen, T., Doney, A., Cavalcanti-Proença, C., Kumari, M., Timpson, N. J., Zabena, C., Ingelsson, E., An, P., O'Connell, J., Luan, J., Elliott, A., McCarroll, S. A., Roccasecca, R. M., Pattou, F., Sethupathy, P., Ariyurek, Y., Barter, P., Beilby, J. P., Ben-Shlomo, Y., Bergmann, S., Bochud, M., Bonnefond, A., Borch-Johnsen, K., Böttcher, Y., Brunner, E., Bumpstead, S. J., Chen, Y. I., Chines, P., Clarke, R., Coin, L. J., Cooper, M. N., Crisponi, L., Day, I. N., de Geus, E. J., Delplanque, J., Fedson, A. C., Fischer-Rosinsky, A., Forouhi, N. G., Franzosi, M. G., Galan, P., Goodarzi, M. O., Graessler, J., Grundy, S., Gwilliam, R., Hallmans, G., Hammond, N., Han, X., Hartikainen, A., Hayward, C., Heath, S. C., Hercberg, S., Hillman, D. R., Hingorani, A. D., Hui, J., Hung, J., Kaakinen, M., Kaprio, J., Kesaniemi, Y. A., Kivimaki, M., Knight, B., Koskinen, S., Kovacs, P., Kyvik, K. O., Lathrop, G. M., Lawlor, D. A., Le Bacquer, O., Lecoeur, C., Li, Y., Mahley, R., Mangino, M., Martínez-Larrad, M. T., McAteer, J. B., McPherson, R., Meisinger, C., Melzer, D., Meyre, D., Mitchell, B. D., Mukherjee, S., Naitza, S., Neville, M. J., Orrù, M., Pakyz, R., Paolisso, G., Pattaro, C., Pearson, D., Peden, J. F., Pedersen, N. L., Pfeiffer, A. F., Pichler, I., Polasek, O., Posthuma, D., Potter, S. C., Pouta, A., Province, M. A., Rayner, N. W., Rice, K., Ripatti, S., Rivadeneira, F., Rolandsson, O., Sandbaek, A., Sandhu, M., Sanna, S., Sayer, A. A., Scheet, P., Seedorf, U., Sharp, S. J., Shields, B., Sigurðsson, G., Sijbrands, E. J., Silveira, A., Simpson, L., Singleton, A., Smith, N. L., Sovio, U., Swift, A., Syddall, H., Syvänen, A., Tönjes, A., Uitterlinden, A. G., Van Dijk, K. W., Varma, D., Visvikis-Siest, S., Vitart, V., Vogelzangs, N., Waeber, G., Wagner, P. J., Walley, A., Ward, K. L., Watkins, H., Wild, S. H., Willemsen, G., Witteman, J. C., Yarnell, J. W., Zelenika, D., Zethelius, B., Zhai, G., Zhao, J. H., Zillikens, M. C., Borecki, I. B., Meneton, P., Magnusson, P. K., Nathan, D. M., Williams, G. H., Silander, K., Bornstein, S. R., Schwarz, P., Spranger, J., Karpe, F., Shuldiner, A. R., Cooper, C., Serrano-Ríos, M., Lind, L., Palmer, L. J., Hu, F. B., Franks, P. W., Ebrahim, S., Marmot, M., Kao, W. H., Pramstaller, P. P., Wright, A. F., Stumvoll, M., Hamsten, A., Buchanan, T. A., Valle, T. T., Rotter, J. I., Penninx, B. W., Boomsma, D. I., Cao, A., Scuteri, A., Schlessinger, D., Uda, M., Ruokonen, A., Jarvelin, M., Peltonen, L., Mooser, V., Sladek, R., Musunuru, K., Smith, A. V., Edmondson, A. C., Stylianou, I. M., Koseki, M., Pirruccello, J. P., Chasman, D. I., Johansen, C. T., Fouchier, S. W., Peloso, G. M., Barbalic, M., Ricketts, S. L., Bis, J. C., Feitosa, M. F., Orho-Melander, M., Melander, O., Li, X., Li, M., Cho, Y. S., Go, M. J., Kim, Y. J., Lee, J., Park, T., Kim, K., Sim, X., Ong, R. T., Croteau-Chonka, D. C., Lange, L. A., Smith, J. D., Ziegler, A., Zhang, W., Zee, R. Y., Whitfield, J. B., Thompson, J. R., Surakka, I., Spector, T. D., Smit, J. H., Sinisalo, J., Scott, J., Saharinen, J., Sabatti, C., Rose, L. M., Roberts, R., Rieder, M., Parker, A. N., Pare, G., O'Donnell, C. J., Nieminen, M. S., Nickerson, D. A., Montgomery, G. W., McArdle, W., Masson, D., Martin, N. G., Marroni, F., Lucas, G., Luben, R., Lokki, M., Lettre, G., Launer, L. J., Lakatta, E. G., Laaksonen, R., Kyvik, K. O., König, I. R., Khaw, K., Kaplan, L. M., Johansson, Å., Janssens, A. C., Igl, W., Hovingh, G. K., Hengstenberg, C., Havulinna, A. S., Hastie, N. D., Harris, T. B., Haritunians, T., Hall, A. S., Groop, L. C., Gonzalez, E., Freimer, N. B., Erdmann, J., Ejebe, K. G., Döring, A., Dominiczak, A. F., Demissie, S., Deloukas, P., de Faire, U., Crawford, G., Chen, Y. I., Caulfield, M. J., Boekholdt, S. M., Assimes, T. L., Quertermous, T., Seielstad, M., Wong, T. Y., Tai, E., Feranil, A. B., Kuzawa, C. W., Taylor, H. A., Gabriel, S. B., Holm, H., Gudnason, V., Krauss, R. M., Ordovas, J. M., Munroe, P. B., Kooner, J. S., Tall, A. R., Hegele, R. A., Kastelein, J. J., Schadt, E. E., Strachan, D. P., Reilly, M. P., Samani, N. J., Schunkert, H., Cupples, L. A., Sandhu, M. S., Ridker, P. M., Rader, D. J., Kathiresan, S. 2012; 8 (3)

    Abstract

    Circulating levels of adiponectin, a hormone produced predominantly by adipocytes, are highly heritable and are inversely associated with type 2 diabetes mellitus (T2D) and other metabolic traits. We conducted a meta-analysis of genome-wide association studies in 39,883 individuals of European ancestry to identify genes associated with metabolic disease. We identified 8 novel loci associated with adiponectin levels and confirmed 2 previously reported loci (P = 4.5×10(-8)-1.2×10(-43)). Using a novel method to combine data across ethnicities (N = 4,232 African Americans, N = 1,776 Asians, and N = 29,347 Europeans), we identified two additional novel loci. Expression analyses of 436 human adipocyte samples revealed that mRNA levels of 18 genes at candidate regions were associated with adiponectin concentrations after accounting for multiple testing (p<3×10(-4)). We next developed a multi-SNP genotypic risk score to test the association of adiponectin decreasing risk alleles on metabolic traits and diseases using consortia-level meta-analytic data. This risk score was associated with increased risk of T2D (p = 4.3×10(-3), n = 22,044), increased triglycerides (p = 2.6×10(-14), n = 93,440), increased waist-to-hip ratio (p = 1.8×10(-5), n = 77,167), increased glucose two hours post oral glucose tolerance testing (p = 4.4×10(-3), n = 15,234), increased fasting insulin (p = 0.015, n = 48,238), but with lower in HDL-cholesterol concentrations (p = 4.5×10(-13), n = 96,748) and decreased BMI (p = 1.4×10(-4), n = 121,335). These findings identify novel genetic determinants of adiponectin levels, which, taken together, influence risk of T2D and markers of insulin resistance.

    View details for DOI 10.1371/journal.pgen.1002607

    View details for PubMedID 22479202

  • A genome-wide association search for type 2 diabetes genes in African Americans. PloS one Palmer, N. D., McDonough, C. W., Hicks, P. J., Roh, B. H., Wing, M. R., An, S. S., Hester, J. M., Cooke, J. N., Bostrom, M. A., Rudock, M. E., Talbert, M. E., Lewis, J. P., Ferrara, A., Lu, L., Ziegler, J. T., Sale, M. M., Divers, J., Shriner, D., Adeyemo, A., Rotimi, C. N., Ng, M. C., Langefeld, C. D., Freedman, B. I., Bowden, D. W., Voight, B. F., Scott, L. J., Steinthorsdottir, V., Morris, A. P., Dina, C., Welch, R. P., Zeggini, E., Huth, C., Aulchenko, Y. S., Thorleifsson, G., McCulloch, L. J., Ferreira, T., Grallert, H., Amin, N., Wu, G., Willer, C. J., Raychaudhuri, S., McCarroll, S. A., Langenberg, C., Hofmann, O. M., Dupuis, J., Qi, L., Segrè, A. V., van Hoek, M., Navarro, P., Ardlie, K., Balkau, B., Benediktsson, R., Bennett, A. J., Blagieva, R., Boerwinkle, E., Bonnycastle, L. L., Boström, K. B., Bravenboer, B., Bumpstead, S., Burtt, N. P., Charpentier, G., Chines, P. S., Cornelis, M., Couper, D. J., Crawford, G., Doney, A. S., Elliott, K. S., Elliott, A. L., Erdos, M. R., Fox, C. S., Franklin, C. S., Ganser, M., Gieger, C., Grarup, N., Green, T., Griffin, S., Groves, C. J., Guiducci, C., Hadjadj, S., Hassanali, N., Herder, C., Isomaa, B., Jackson, A. U., Johnson, P. R., Jørgensen, T., Kao, W. H., Klopp, N., Kong, A., Kraft, P., Kuusisto, J., Lauritzen, T., Li, M., Lieverse, A., Lindgren, C. M., Lyssenko, V., Marre, M., Meitinger, T., Midthjell, K., Morken, M. A., Narisu, N., Nilsson, P., Owen, K. R., Payne, F., Perry, J. R., Petersen, A., Platou, C., Proença, C., Prokopenko, I., Rathmann, W., Rayner, N. W., Robertson, N. R., Rocheleau, G., Roden, M., Sampson, M. J., Saxena, R., Shields, B. M., Shrader, P., Sigurdsson, G., Sparsø, T., Strassburger, K., Stringham, H. M., Sun, Q., Swift, A. J., Thorand, B., Tichet, J., Tuomi, T., van Dam, R. M., Van Haeften, T. W., van Herpt, T., van Vliet-Ostaptchouk, J. V., Walters, G. B., Weedon, M. N., Wijmenga, C., Witteman, J., Bergman, R. N., Cauchi, S., Collins, F. S., Gloyn, A. L., Gyllensten, U., Hansen, T., Hide, W. A., Hitman, G. A., Hofman, A., Hunter, D. J., Hveem, K., Laakso, M., Mohlke, K. L., Morris, A. D., Palmer, C. N., Pramstaller, P. P., Rudan, I., Sijbrands, E., Stein, L. D., Tuomilehto, J., Uitterlinden, A., Walker, M., Wareham, N. J., Watanabe, R. M., Abecasis, G. R., Boehm, B. O., Campbell, H., Daly, M. J., Hattersley, A. T., Hu, F. B., Meigs, J. B., Pankow, J. S., Pedersen, O., Wichmann, H., Barroso, I., Florez, J. C., Frayling, T. M., Groop, L., Sladek, R., Thorsteinsdottir, U., Wilson, J. F., Illig, T., Froguel, P., van Duijn, C. M., Stefansson, K., Altshuler, D., Boehnke, M., McCarthy, M. I., Soranzo, N., Wheeler, E., Glazer, N. L., Bouatia-Naji, N., Mägi, R., Randall, J., Johnson, T., Elliott, P., Rybin, D., Henneman, P., Dehghan, A., Hottenga, J. J., Song, K., Goel, A., Egan, J. M., Lajunen, T., Doney, A., Kanoni, S., Cavalcanti-Proença, C., Kumari, M., Timpson, N. J., Zabena, C., Ingelsson, E., An, P., O'Connell, J., Luan, J., Elliott, A., McCarroll, S. A., Roccasecca, R. M., Pattou, F., Sethupathy, P., Ariyurek, Y., Barter, P., Beilby, J. P., Ben-Shlomo, Y., Bergmann, S., Bochud, M., Bonnefond, A., Borch-Johnsen, K., Böttcher, Y., Brunner, E., Bumpstead, S. J., Chen, Y. I., Chines, P., Clarke, R., Coin, L. J., Cooper, M. N., Crisponi, L., Day, I. N., de Geus, E. J., Delplanque, J., Fedson, A. C., Fischer-Rosinsky, A., Forouhi, N. G., Frants, R., Franzosi, M. G., Galan, P., Goodarzi, M. O., Graessler, J., Grundy, S., Gwilliam, R., Hallmans, G., Hammond, N., Han, X., Hartikainen, A., Hayward, C., Heath, S. C., Hercberg, S., Hicks, A. A., Hillman, D. R., Hingorani, A. D., Hui, J., Hung, J., Jula, A., Kaakinen, M., Kaprio, J., Kesaniemi, Y. A., Kivimaki, M., Knight, B., Koskinen, S., Kovacs, P., Kyvik, K. O., Lathrop, G. M., Lawlor, D. A., Le Bacquer, O., Lecoeur, C., Li, Y., Mahley, R., Mangino, M., Manning, A. K., Martínez-Larrad, M. T., McAteer, J. B., McPherson, R., Meisinger, C., Melzer, D., Meyre, D., Mitchell, B. D., Mukherjee, S., Naitza, S., Neville, M. J., Oostra, B. A., Orrù, M., Pakyz, R., Paolisso, G., Pattaro, C., Pearson, D., Peden, J. F., Pedersen, N. L., Perola, M., Pfeiffer, A. F., Pichler, I., Polasek, O., Posthuma, D., Potter, S. C., Pouta, A., Province, M. A., Psaty, B. M., Rayner, N. W., Rice, K., Ripatti, S., Rivadeneira, F., Rolandsson, O., Sandbaek, A., Sandhu, M., Sanna, S., Sayer, A. A., Scheet, P., Seedorf, U., Sharp, S. J., Shields, B., Sijbrands, E. J., Silveira, A., Simpson, L., Singleton, A., Smith, N. L., Sovio, U., Swift, A., Syddall, H., Syvänen, A., Tanaka, T., Tönjes, A., Uitterlinden, A. G., Van Dijk, K. W., Varma, D., Visvikis-Siest, S., Vitart, V., Vogelzangs, N., Waeber, G., Wagner, P. J., Walley, A., Ward, K. L., Watkins, H., Wild, S. H., Willemsen, G., Witteman, J. C., Yarnell, J. W., Zelenika, D., Zethelius, B., Zhai, G., Zhao, J. H., Zillikens, M. C., Borecki, I. B., Loos, R. J., Meneton, P., Magnusson, P. K., Nathan, D. M., Williams, G. H., Silander, K., Salomaa, V., Smith, G. D., Bornstein, S. R., Schwarz, P., Spranger, J., Karpe, F., Shuldiner, A. R., Cooper, C., Dedoussis, G. V., Serrano-Ríos, M., Lind, L., Palmer, L. J., Franks, P. W., Ebrahim, S., Marmot, M., Kao, W. H., Pramstaller, P. P., Wright, A. F., Stumvoll, M., Hamsten, A., Buchanan, T. A., Valle, T. T., Rotter, J. I., Siscovick, D. S., Penninx, B. W., Boomsma, D. I., Deloukas, P., Spector, T. D., Ferrucci, L., Cao, A., Scuteri, A., Schlessinger, D., Uda, M., Ruokonen, A., Jarvelin, M., Waterworth, D. M., Vollenweider, P., Peltonen, L., Mooser, V., Sladek, R. 2012; 7 (1)

    Abstract

    African Americans are disproportionately affected by type 2 diabetes (T2DM) yet few studies have examined T2DM using genome-wide association approaches in this ethnicity. The aim of this study was to identify genes associated with T2DM in the African American population. We performed a Genome Wide Association Study (GWAS) using the Affymetrix 6.0 array in 965 African-American cases with T2DM and end-stage renal disease (T2DM-ESRD) and 1029 population-based controls. The most significant SNPs (n = 550 independent loci) were genotyped in a replication cohort and 122 SNPs (n = 98 independent loci) were further tested through genotyping three additional validation cohorts followed by meta-analysis in all five cohorts totaling 3,132 cases and 3,317 controls. Twelve SNPs had evidence of association in the GWAS (P<0.0071), were directionally consistent in the Replication cohort and were associated with T2DM in subjects without nephropathy (P<0.05). Meta-analysis in all cases and controls revealed a single SNP reaching genome-wide significance (P<2.5×10(-8)). SNP rs7560163 (P = 7.0×10(-9), OR (95% CI) = 0.75 (0.67-0.84)) is located intergenically between RND3 and RBM43. Four additional loci (rs7542900, rs4659485, rs2722769 and rs7107217) were associated with T2DM (P<0.05) and reached more nominal levels of significance (P<2.5×10(-5)) in the overall analysis and may represent novel loci that contribute to T2DM. We have identified novel T2DM-susceptibility variants in the African-American population. Notably, T2DM risk was associated with the major allele and implies an interesting genetic architecture in this population. These results suggest that multiple loci underlie T2DM susceptibility in the African-American population and that these loci are distinct from those identified in other ethnic populations.

    View details for DOI 10.1371/journal.pone.0029202

    View details for PubMedID 22238593

  • Birth Characteristics and Subsequent Risks of Maternal Cardiovascular Disease Effects of Gestational Age and Fetal Growth CIRCULATION Bonamy, A. E., Parikh, N. I., Cnattingius, S., Ludvigsson, J. F., Ingelsson, E. 2011; 124 (25): 2839-U174

    Abstract

    Prior studies showing an inverse relationship between low birth weight in offspring and maternal risks of cardiovascular diseases (CVD) are limited by lack of information on gestational age and/or insufficient adjustment for confounders.In a nationwide Swedish study, we included information on 923 686 women and their first singleton births between 1983 and 2005. Cox proportional hazards models were used to study associations between gestational length, fetal growth, and maternal incident hospitalization or death from CVD (coronary heart disease, cerebrovascular events, and heart failure). Multivariable adjusted models accounted for birth year, income, education, country of birth, smoking, diabetes mellitus, hypertension, and preeclampsia. The risk of maternal CVD increased with decreasing gestational age whereas the risk increase related to fetal growth appeared to be restricted to very small-for-gestational-age (SGA) infants. Compared with mothers of non-SGA infants born at term, the hazard ratio of CVD ranged from 1.39 (95% confidence interval 1.22-1.58) to 2.57 (95% confidence interval 1.97-3.34) among mothers to moderately and very preterm infants, respectively. There was a significant interaction between preterm birth and fetal growth with respect to mothers' risk of CVD (P<0.001). Among mothers to very SGA infants, the hazard ratio of CVD ranged from 1.38 (95% confidence interval 1.15-1.65) to 3.40 (95% confidence interval 2.26-5.11) in mothers to term and very preterm infants, respectively.Delivery of a preterm or SGA infant is associated with later life maternal hospitalization or death from CVD even after accounting for socioeconomic factors, smoking, and pregnancy-related complications.

    View details for DOI 10.1161/CIRCULATIONAHA.111.034884

    View details for Web of Science ID 000298414900014

    View details for PubMedID 22124377

  • Genome-Wide Association Identifies Nine Common Variants Associated With Fasting Proinsulin Levels and Provides New Insights Into the Pathophysiology of Type 2 Diabetes DIABETES Strawbridge, R. J., Dupuis, J., Prokopenko, I., Barker, A., Ahlqvist, E., Rybin, D., Petrie, J. R., Travers, M. E., Bouatia-Naji, N., Dimas, A. S., Nica, A., Wheeler, E., Chen, H., Voight, B. F., Taneera, J., Kanoni, S., Peden, J. F., Turrini, F., Gustafsson, S., Zabena, C., Almgren, P., Barker, D. J., Barnes, D., Dennison, E. M., Eriksson, J. G., Eriksson, P., Eury, E., Folkersen, L., Fox, C. S., Frayling, T. M., Goel, A., Gu, H. F., Horikoshi, M., Isomaa, B., Jackson, A. U., Jameson, K. A., Kajantie, E., Kerr-Conte, J., Kuulasmaa, T., Kuusisto, J., Loos, R. J., Luan, J., Makrilakis, K., Manning, A. K., Teresa Martinez-Larrad, M., Narisu, N., Mannila, M. N., Ohrvik, J., Osmond, C., Pascoe, L., Payne, F., Sayer, A. A., Sennblad, B., Silveira, A., Stancakova, A., Stirrups, K., Swift, A. J., Syvanen, A., Tuomi, T., van 't Hooft, F. M., Walker, M., Weedon, M. N., Xie, W., Zethelius, B., Ongen, H., Malarstig, A., Hopewell, J. C., Saleheen, D., Chambers, J., Parish, S., Danesh, J., Kooner, J., Ostenson, C., Lind, L., Cooper, C. C., Serrano-Rios, M., Ferrannini, E., Forsen, T. J., Clarke, R., Franzosi, M. G., Seedorf, U., Watkins, H., Froguel, P., Johnson, P., Deloukas, P., Collins, F. S., Laakso, M., Dermitzakis, E. T., Boehnke, M., McCarthy, M. I., Wareham, N. J., Groop, L., Pattou, F., Gloyn, A. L., Dedoussis, G. V., Lyssenko, V., Meigs, J. B., Barroso, I., Watanabe, R. M., Ingelsson, E., Langenberg, C., Hamsten, A., Florez, J. C. 2011; 60 (10): 2624-2634

    Abstract

    Proinsulin is a precursor of mature insulin and C-peptide. Higher circulating proinsulin levels are associated with impaired β-cell function, raised glucose levels, insulin resistance, and type 2 diabetes (T2D). Studies of the insulin processing pathway could provide new insights about T2D pathophysiology.We have conducted a meta-analysis of genome-wide association tests of ∼2.5 million genotyped or imputed single nucleotide polymorphisms (SNPs) and fasting proinsulin levels in 10,701 nondiabetic adults of European ancestry, with follow-up of 23 loci in up to 16,378 individuals, using additive genetic models adjusted for age, sex, fasting insulin, and study-specific covariates.Nine SNPs at eight loci were associated with proinsulin levels (P < 5 × 10(-8)). Two loci (LARP6 and SGSM2) have not been previously related to metabolic traits, one (MADD) has been associated with fasting glucose, one (PCSK1) has been implicated in obesity, and four (TCF7L2, SLC30A8, VPS13C/C2CD4A/B, and ARAP1, formerly CENTD2) increase T2D risk. The proinsulin-raising allele of ARAP1 was associated with a lower fasting glucose (P = 1.7 × 10(-4)), improved β-cell function (P = 1.1 × 10(-5)), and lower risk of T2D (odds ratio 0.88; P = 7.8 × 10(-6)). Notably, PCSK1 encodes the protein prohormone convertase 1/3, the first enzyme in the insulin processing pathway. A genotype score composed of the nine proinsulin-raising alleles was not associated with coronary disease in two large case-control datasets.We have identified nine genetic variants associated with fasting proinsulin. Our findings illuminate the biology underlying glucose homeostasis and T2D development in humans and argue against a direct role of proinsulin in coronary artery disease pathogenesis.

    View details for DOI 10.2337/db11-0415

    View details for Web of Science ID 000295998700022

    View details for PubMedID 21873549

  • Association Between Serum Cathepsin S and Mortality in Older Adults JAMA-JOURNAL OF THE AMERICAN MEDICAL ASSOCIATION Jobs, E., Ingelsson, E., Riserus, U., Nerpin, E., Jobs, M., Sundstrom, J., Basu, S., Larsson, A., Lind, L., Arnlov, J. 2011; 306 (10): 1113-1121

    Abstract

    Experimental data suggest that cathepsin S, a cysteine protease, is involved in the complex pathways leading to cardiovascular disease and cancer. However, prospective data concerning a potential association between circulating cathepsin S levels and mortality are lacking.To investigate associations between circulating cathepsin S levels and mortality in 2 independent cohorts of elderly men and women.Prospective study using 2 community-based cohorts, the Uppsala Longitudinal Study of Adult Men (ULSAM; n = 1009; mean age: 71 years; baseline period: 1991-1995; median follow-up: 12.6 years; end of follow-up: 2006) and the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS; n = 987; 50% women; mean age: 70 years; baseline period: 2001-2004; median follow-up: 7.9 years; end of follow-up: 2010). Serum samples were used to measure cathepsin S.Total mortality.During follow-up, 413 participants died in the ULSAM cohort (incidence rate: 3.59/100 person-years at risk) and 100 participants died in the PIVUS cohort (incidence rate: 1.32/100 person-years at risk). In multivariable Cox regression models adjusted for age, systolic blood pressure, diabetes, smoking status, body mass index, total cholesterol, high-density lipoprotein cholesterol, antihypertensive treatment, lipid-lowering treatment, and history of cardiovascular disease, higher serum cathepsin S was associated with an increased risk for mortality (ULSAM cohort: hazard ratio [HR] for 1-unit increase of cathepsin S, 1.04 [95% CI, 1.01-1.06], P = .009; PIVUS cohort: HR for 1-unit increase of cathepsin S, 1.03 [95% CI, 1.00-1.07], P = .04). In the ULSAM cohort, serum cathepsin S also was associated with cardiovascular mortality (131 deaths; HR for quintile 5 vs quintiles 1-4, 1.62 [95% CI, 1.11-2.37]; P = .01) and cancer mortality (148 deaths; HR for 1-unit increase of cathepsin S, 1.05 [95% CI, 1.01-1.10]; P = .01).Among elderly individuals in 2 independent cohorts, higher serum cathepsin S levels were associated with increased mortality risk. Additional research is needed to delineate the role of cathepsin S and whether its measurement might have clinical utility.

    View details for DOI 10.1001/jama.2011.1246

    View details for Web of Science ID 000294806200025

    View details for PubMedID 21878432

  • The combined contribution of albuminuria and glomerular filtration rate to the prediction of cardiovascular mortality in elderly men NEPHROLOGY DIALYSIS TRANSPLANTATION Nerpin, E., Ingelsson, E., Riserus, U., Sundstrom, J., Larsson, A., Jobs, E., Jobs, M., Hallan, S., Zethelius, B., Berglund, L., Basu, S., Arnlov, J. 2011; 26 (9): 2820-U1504

    Abstract

    Cardiovascular risk prediction is particularly important in the primary prevention of cardiovascular disease (CVD). Yet, data on whether the combined addition of albuminuria and estimated glomerular filtration rate (eGFR) improves cardiovascular risk prediction in individuals without CVD in the community is scarce.We investigated associations between urinary albumin excretion rate (UAER), cystatin C-based eGFR and cardiovascular mortality in a community-based cohort of elderly men (ULSAM study; n = 1113, mean age 71 years, 208 cardiovascular deaths, median follow-up 12.9 years) with prespecified analyses in participants without CVD (n = 649, 86 cardiovascular deaths).Using multivariable Cox regression, higher UAER and lower eGFR were associated with increased risk for cardiovascular mortality independently of established cardiovascular risk factors in the whole sample and in men without CVD at baseline [subsample without CVD: UAER; hazard ratio (HR) per 1 SD 1.26, 95% confidence interval (CI) 1.05-1.51, P = 0.01; eGFR: HR per 1 SD 0.74, 95% CI 0.59-0.92, P = 0.007]. Analyses of model discrimination, calibration, reclassification and global fit suggested that UAER and eGFR also add relevant prognostic information beyond established cardiovascular risk factors in participants without prevalent CVD. Interestingly, established cutoffs used to diagnose microalbuminuria (UAER > 20 μg/min) and chronic kidney disease Stage 3 (eGFR < 60 mL/min/1.73 m(2)), appeared less suitable for cardiovascular risk prediction [integrated discrimination improvement (IDI) 0.006, P = 0.11], while cutoffs UAER > 6 μg/min and eGFR < 45 mL/min/1.73 m(2) significantly improved IDI (0.047, P < 0.001).UAER and eGFR improved cardiovascular risk prediction beyond established cardiovascular risk factors, suggesting that these kidney biomarkers may be useful in predicting cardiovascular death in elderly men.

    View details for DOI 10.1093/ndt/gfq848

    View details for Web of Science ID 000295231600017

    View details for PubMedID 21335440

  • Total Zinc Intake May Modify the Glucose-Raising Effect of a Zinc Transporter (SLC30A8) Variant A 14-Cohort Meta-analysis DIABETES Kanoni, S., Nettleton, J. A., Hivert, M., Ye, Z., van Rooij, F. J., Shungin, D., Sonestedt, E., Ngwa, J. S., Wojczynski, M. K., Lemaitre, R. N., Gustafsson, S., Anderson, J. S., Tanaka, T., Hindy, G., Saylor, G., Renstrom, F., Bennett, A. J., van Duijn, C. M., Florez, J. C., Fox, C. S., Hofman, A., Hoogeveen, R. C., Houston, D. K., Hu, F. B., Jacques, P. F., Johansson, I., Lind, L., Liu, Y., McKeown, N., Ordovas, J., Pankow, J. S., Sijbrands, E. J., Syvanen, A., Uitterlinden, A. G., Yannakoulia, M., Zillikens, M. C., Wareham, N. J., Prokopenko, I., Bandinelli, S., Forouhi, N. G., Cupples, L. A., Loos, R. J., Hallmans, G., Dupuis, J., Langenberg, C., Ferrucci, L., Kritchevsky, S. B., McCarthy, M. I., Ingelsson, E., Borecki, I. B., Witteman, J. C., Orho-Melander, M., Siscovick, D. S., Meigs, J. B., Franks, P. W., Dedoussis, G. V. 2011; 60 (9): 2407-2416

    Abstract

    Many genetic variants have been associated with glucose homeostasis and type 2 diabetes in genome-wide association studies. Zinc is an essential micronutrient that is important for β-cell function and glucose homeostasis. We tested the hypothesis that zinc intake could influence the glucose-raising effect of specific variants.We conducted a 14-cohort meta-analysis to assess the interaction of 20 genetic variants known to be related to glycemic traits and zinc metabolism with dietary zinc intake (food sources) and a 5-cohort meta-analysis to assess the interaction with total zinc intake (food sources and supplements) on fasting glucose levels among individuals of European ancestry without diabetes.We observed a significant association of total zinc intake with lower fasting glucose levels (β-coefficient ± SE per 1 mg/day of zinc intake: -0.0012 ± 0.0003 mmol/L, summary P value = 0.0003), while the association of dietary zinc intake was not significant. We identified a nominally significant interaction between total zinc intake and the SLC30A8 rs11558471 variant on fasting glucose levels (β-coefficient ± SE per A allele for 1 mg/day of greater total zinc intake: -0.0017 ± 0.0006 mmol/L, summary interaction P value = 0.005); this result suggests a stronger inverse association between total zinc intake and fasting glucose in individuals carrying the glucose-raising A allele compared with individuals who do not carry it. None of the other interaction tests were statistically significant.Our results suggest that higher total zinc intake may attenuate the glucose-raising effect of the rs11558471 SLC30A8 (zinc transporter) variant. Our findings also support evidence for the association of higher total zinc intake with lower fasting glucose levels.

    View details for DOI 10.2337/db11-0176

    View details for Web of Science ID 000294699600024

    View details for PubMedID 21810599

  • Identification of cis- and trans-Acting Genetic Variants Explaining Up to Half the Variation in Circulating Vascular Endothelial Growth Factor Levels CIRCULATION RESEARCH Debette, S., Visvikis-Siest, S., Chen, M., Ndiaye, N., Song, C., DeStefano, A., Safa, R., Nezhad, M. A., Sawyer, D., Marteau, J., Xanthakis, V., Siest, G., Sullivan, L., Pfister, M., Smith, H., Choi, S., Lamont, J., Lind, L., Yang, Q., Fitzgerald, P., Ingelsson, E., Vasan, R. S., Seshadri, S. 2011; 109 (5): 554-U245

    Abstract

    Vascular endothelial growth factor (VEGF) affects angiogenesis, atherosclerosis, and cancer. Although the heritability of circulating VEGF levels is high, little is known about its genetic underpinnings.Our aim was to identify genetic variants associated with circulating VEGF levels, using an unbiased genome-wide approach, and to explore their functional significance with gene expression and pathway analysis.We undertook a genome-wide association study of serum VEGF levels in 3527 participants of the Framingham Heart Study, with preplanned replication in 1727 participants from 2 independent samples, the STANISLAS Family Study and the Prospective Investigation of the Vasculature in Uppsala Seniors study. One hundred forty single nucleotide polymorphism (SNPs) reached genome-wide significance (P<5×10(-8)). We found evidence of replication for the most significant associations in both replication datasets. In a conditional genome-wide association study, 4 SNPs mapping to 3 chromosomal regions were independently associated with circulating VEGF levels: rs6921438 and rs4416670 (6p21.1, P=6.11×10(-506) and P=1.47×10(-12)), rs6993770 (8q23.1, P=2.50×10(-16)), and rs10738760 (9p24.2, P=1.96×10(-34)). A genetic score including these 4 SNPs explained 48% of the heritability of serum VEGF levels. Six of the SNPs that reached genome-wide significance in the genome-wide association study were significantly associated with VEGF messenger RNA levels in peripheral blood mononuclear cells. Ingenuity pathway analyses showed found plausible biological links between VEGF and 2 novel genes in these loci (ZFPM2 and VLDLR).Genetic variants explaining up to half the heritability of serum VEGF levels were identified. These new insights provide important clues to the pathways regulating circulating VEGF levels.

    View details for DOI 10.1161/CIRCRESAHA.111.243790

    View details for Web of Science ID 000294006800012

    View details for PubMedID 21757650

  • Genetic variation near IRS1 associates with reduced adiposity and an impaired metabolic profile NATURE GENETICS Kilpelainen, T. O., Zillikens, M. C., Stancakova, A., Finucane, F. M., Ried, J. S., Langenberg, C., Zhang, W., Beckmann, J. S., Luan, J., Vandenput, L., Styrkarsdottir, U., Zhou, Y., Smith, A. V., Zhao, J., Amin, N., Vedantam, S., Shin, S., Haritunians, T., Fu, M., Feitosa, M. F., Kumari, M., Halldorsson, B. V., Tikkanen, E., Mangino, M., Hayward, C., Song, C., Arnold, A. M., Aulchenko, Y. S., Oostra, B. A., Campbell, H., Cupples, L. A., Davis, K. E., Doering, A., Eiriksdottir, G., Estrada, K., Manuel Fernandez-Real, J., Garcia, M., Gieger, C., Glazer, N. L., Guiducci, C., Hofman, A., Humphries, S. E., Isomaa, B., Jacobs, L. C., Jula, A., Karasik, D., Karlsson, M. K., Khaw, K., Kim, L. J., Kivimaeki, M., Klopp, N., Kuehnel, B., Kuusisto, J., Liu, Y., Ljunggren, O., Lorentzon, M., Luben, R. N., McKnight, B., Mellstrom, D., Mitchell, B. D., Mooser, V., Maria Moreno, J., Mannisto, S., O'Connell, J. R., Pascoe, L., Peltonen, L., Peral, B., Perola, M., Psaty, B. M., Salomaa, V., Savage, D. B., Semple, R. K., Skaric-Juric, T., Sigurdsson, G., Song, K. S., Spector, T. D., Syvanen, A., Talmud, P. J., Thorleifsson, G., Thorsteinsdottir, U., Uitterlinden, A. G., van Duijn, C. M., Vidal-Puig, A., Wild, S. H., Wright, A. F., Clegg, D. J., Schadt, E., Wilson, J. F., Rudan, I., Ripatti, S., Borecki, I. B., Shuldiner, A. R., Ingelsson, E., Jansson, J., Kaplan, R. C., Gudnason, V., Harris, T. B., Groop, L., Kiel, D. P., Rivadeneira, F., Walker, M., Barroso, I., Vollenweider, P., Waeber, G., Chambers, J. C., Kooner, J. S., Soranzo, N., Hirschhorn, J. N., Stefansson, K., Wichmann, H., Ohlsson, C., O'Rahilly, S., Wareham, N. J., Speliotes, E. K., Fox, C. S., Laakso, M., Loos, R. J. 2011; 43 (8): 753-U58

    Abstract

    Genome-wide association studies have identified 32 loci influencing body mass index, but this measure does not distinguish lean from fat mass. To identify adiposity loci, we meta-analyzed associations between ∼2.5 million SNPs and body fat percentage from 36,626 individuals and followed up the 14 most significant (P < 10(-6)) independent loci in 39,576 individuals. We confirmed a previously established adiposity locus in FTO (P = 3 × 10(-26)) and identified two new loci associated with body fat percentage, one near IRS1 (P = 4 × 10(-11)) and one near SPRY2 (P = 3 × 10(-8)). Both loci contain genes with potential links to adipocyte physiology. Notably, the body-fat-decreasing allele near IRS1 is associated with decreased IRS1 expression and with an impaired metabolic profile, including an increased visceral to subcutaneous fat ratio, insulin resistance, dyslipidemia, risk of diabetes and coronary artery disease and decreased adiponectin levels. Our findings provide new insights into adiposity and insulin resistance.

    View details for DOI 10.1038/ng.866

    View details for Web of Science ID 000293178300011

    View details for PubMedID 21706003

  • A Detailed Cardiovascular Characterization of Obesity Without the Metabolic Syndrome ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Lind, L., Siegbahn, A., Ingelsson, E., Sundstrom, J., Arnlov, J. 2011; 31 (8): E27-U24

    Abstract

    Although obesity without metabolic disturbances has been regarded as harmless, we have recently shown that obese subjects without the metabolic syndrome (MetS) has an increased risk of cardiovascular (CV) disorders and mortality during long-term follow-up. To investigate the basis for that increased risk, we studied the impact of obesity without MetS on multiple markers of subclinical CV disease.At age 70, 1016 subjects were investigated in the Prospective Investigation of the Vasculature in Uppsala Seniors study. According to body mass index (BMI)/MetS status, they were categorized as normal weight (BMI <25 kg/m(2)) without MetS (National Cholesterol Education Program criteria, n=319), normal weight with MetS (n=19), overweight (BMI 25 to 29.9 kg/m(2)) without MetS (n=333), overweight with MetS (n=94), obese (BMI ≥30 kg/m(2)) without MetS (n=102), and obese with MetS (n=118). Several different measurements of endothelial reactivity, arterial compliance (plethysmography and ultrasound), carotid artery atherosclerosis, and echocardiography were performed, and 7 markers of coagulation/fibrinolysis were measured. Subjects with obesity without MetS showed impaired vasoreactivity, a more echolucent carotid artery wall, increased left ventricular mass and function together with impaired coagulation/fibrinolysis compared with normal-weight subjects without the MetS (P<0.05 to 0.001). The majority of these disturbances were also seen in overweight subjects without the MetS.In contrast to some previous studies, our data do not support that obesity without MetS is a benign condition, because obesity without MetS was associated with impairments in multiple markers of subclinical CV disease. This was also the case for overweight subjects without the MetS.

    View details for DOI 10.1161/ATVBAHA.110.221572

    View details for Web of Science ID 000292918500002

    View details for PubMedID 21546604

  • Genomic inflation factors under polygenic inheritance EUROPEAN JOURNAL OF HUMAN GENETICS Yang, J., Weedon, M. N., Purcell, S., Lettre, G., Estrada, K., Willer, C. J., Smith, A. V., Ingelsson, E., O'Connell, J. R., Mangino, M., Maegi, R., Madden, P. A., Heath, A. C., Nyholt, D. R., Martin, N. G., Montgomery, G. W., Frayling, T. M., Hirschhorn, J. N., McCarthy, M. I., Goddard, M. E., Visscher, P. M. 2011; 19 (7): 807-812

    Abstract

    Population structure, including population stratification and cryptic relatedness, can cause spurious associations in genome-wide association studies (GWAS). Usually, the scaled median or mean test statistic for association calculated from multiple single-nucleotide-polymorphisms across the genome is used to assess such effects, and 'genomic control' can be applied subsequently to adjust test statistics at individual loci by a genomic inflation factor. Published GWAS have clearly shown that there are many loci underlying genetic variation for a wide range of complex diseases and traits, implying that a substantial proportion of the genome should show inflation of the test statistic. Here, we show by theory, simulation and analysis of data that in the absence of population structure and other technical artefacts, but in the presence of polygenic inheritance, substantial genomic inflation is expected. Its magnitude depends on sample size, heritability, linkage disequilibrium structure and the number of causal variants. Our predictions are consistent with empirical observations on height in independent samples of ~4000 and ~133,000 individuals.

    View details for DOI 10.1038/ejhg.2011.39

    View details for Web of Science ID 000291678400017

    View details for PubMedID 21407268

  • CUBN Is a Gene Locus for Albuminuria JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Boeger, C. A., Chen, M., Tin, A., Olden, M., Koettgen, A., de Boer, I. H., Fuchsberger, C., O'Seaghdha, C. M., Pattaro, C., Teumer, A., Liu, C., Glazer, N. L., Li, M., O'Conne, J. R., Tanaka, T., Peralta, C. A., Kutalik, Z., Luan, J., Zhao, J. H., Hwang, S., Akylbekova, E., Kramer, H., van der Harst, P., Smith, A. V., Lohman, K., de Andrade, M., Hayward, C., Kollerits, B., Toenjes, A., Aspelund, T., Ingelsson, E., Eiriksdottir, G., Launer, L. J., Harris, T. B., Shuldiner, A. R., Mitchell, B. D., Arking, D. E., Franceschini, N., Boerwinkle, E., Egan, J., Hernandez, D., Reilly, M., Townsend, R. R., Lumley, T., Siscovick, D. S., Psaty, B. M., Kestenbaum, B., Haritunians, T., Bergmann, S., Vollenweider, P., Waeber, G., Mooser, V., Waterworth, D., Johnson, A. D., Florez, J. C., Meigs, J. B., Lu, X., Turner, S. T., Atkinson, E. J., Leak, T. S., Aasarod, K., Skorpen, F., Syvaenen, A., Illig, T., Baumert, J., Koenig, W., Kraemer, B. K., Devuyst, O., Mychaleckyj, J. C., Minelli, C., Bakker, S. J., Kedenko, L., Paulweber, B., Coassin, S., Endlich, K., Kroemer, H. K., Biffar, R., Stracke, S., Voelzke, H., Stumvol, M., Maegi, R., Campbell, H., Vitart, V., Hastie, N. D., Gudnason, V., Kardia, S. L., Liu, Y., Polasek, O., Curhan, G., Kronenberg, F., Prokopenko, I., Rudan, I., Aernloev, J., Hallan, S., Navis, G., Parsa, A., Ferrucci, L., Coresh, J., Shlipak, M. G., Bul, S. B., Paterson, A. D., Wichmann, H., Wareham, N. J., Loos, R. J., Rotter, J. I., Pramstaller, P. P., Cupples, L. A., Beckmann, J. S., Yang, Q., Heid, I. M., Rettig, R., Dreisbach, A. W., Bochud, M., Fox, C. S., Kao, W. H. 2011; 22 (3): 555-570

    Abstract

    Identification of genetic risk factors for albuminuria may alter strategies for early prevention of CKD progression, particularly among patients with diabetes. Little is known about the influence of common genetic variants on albuminuria in both general and diabetic populations. We performed a meta-analysis of data from 63,153 individuals of European ancestry with genotype information from genome-wide association studies (CKDGen Consortium) and from a large candidate gene study (CARe Consortium) to identify susceptibility loci for the quantitative trait urinary albumin-to-creatinine ratio (UACR) and the clinical diagnosis microalbuminuria. We identified an association between a missense variant (I2984V) in the CUBN gene, which encodes cubilin, and both UACR (P = 1.1 × 10(-11)) and microalbuminuria (P = 0.001). We observed similar associations among 6981 African Americans in the CARe Consortium. The associations between this variant and both UACR and microalbuminuria were significant in individuals of European ancestry regardless of diabetes status. Finally, this variant associated with a 41% increased risk for the development of persistent microalbuminuria during 20 years of follow-up among 1304 participants with type 1 diabetes in the prospective DCCT/EDIC Study. In summary, we identified a missense CUBN variant that associates with levels of albuminuria in both the general population and in individuals with diabetes.

    View details for DOI 10.1681/ASN.2010060598

    View details for Web of Science ID 000288778800023

    View details for PubMedID 21355061

  • Hysterectomy and risk of cardiovascular disease: a population-based cohort study EUROPEAN HEART JOURNAL Ingelsson, E., Lundholm, C., Johansson, A. L., Altman, D. 2011; 32 (6): 745-750

    Abstract

    Hysterectomy for benign indications is one of the commonest surgical procedures in women, but the association between the procedure and cardiovascular disease (CVD) is not fully understood. In this population-based cohort study, we studied the effects of hysterectomy, with or without oopherectomy, on the risk of later life CVD.Using nationwide healthcare registers, we identified all Swedish women having a hysterectomy on benign indications between 1973 and 2003 (n = 184,441), and non-hysterectomized controls (n = 640,043). Main outcome measure was the first hospitalization or death of incident CVD (coronary heart disease, stroke, or heart failure). Occurrence of CVD was determined by individual linkage to the Inpatient Register. In women below age 50 at study entry, hysterectomy was associated with a significantly increased risk of CVD during follow-up [hazard ratio (HR), 1.18, 95% confidence interval (CI), 1.13-1.23; HR, 2.22, 95% CI, 1.01-4.83; and HR, 1.25, 95% CI, 1.06-1.48; in women without oopherectomy, with oopherectomy before or at study entry, respectively, using women without hysterectomy or oopherectomy as reference]. In women aged 50 or above at study entry, there were no significant associations between hysterectomy and incident CVD.Hysterectomy in women aged 50 years or younger substantially increases the risk for CVD later in life and oopherectomy further adds to the risk of both coronary heart disease and stroke.

    View details for DOI 10.1093/eurheartj/ehq477

    View details for Web of Science ID 000288549100019

    View details for PubMedID 21186237

  • Plasma levels of glucagon like peptide-1 associate with diastolic function in elderly men DIABETIC MEDICINE Nathanson, D., Zethelius, B., Berne, C., Lind, L., Andren, B., Ingelsson, E., Holst, J. J., Nystroem, T. 2011; 28 (3): 301-305

    Abstract

    Congestive heart failure is a major cause of morbidity and mortality in diabetes. Besides the glycaemic effects of glucagon-like peptide 1 (GLP-1) mimetics, their effects on the heart are of interest.We aimed to investigate longitudinal relationships between plasma levels of fasting GLP-1 (fGLP-1), 60-min oral glucose tolerance test-stimulated GLP-1 levels (60GLP-1), and the dynamic GLP-1 response after oral glucose tolerance test (ΔGLP-1 = 60GLP-1 - fGLP-1) and incidence of hospitalized congestive heart failure, during a follow-up time of a maximum of 9.8 years in 71-year-old men. We also investigated, cross-sectionally, the association between GLP-1 and left ventricular function as estimated by echocardiography. R: During the follow-up period, 16 of 290 participants with normal glucose tolerance experienced a congestive heart failure event (rate 0.7/100 person-years at risk), as did eight of 136 participants (rate 0.8/100 person-years at risk) with impaired glucose tolerance and nine of 72 participants (rate 1.7/100 person-years at risk) with Type 2 diabetes mellitus. Although GLP-1 concentrations did not predict congestive heart failure (fGLP-1: HR 0.98, 95% CI 0.4-2.4; 60GLP-1: HR 1.1, 95% CI 0.4-2.6; ΔGLP-1: HR 0.9, 95% CI 0.3-2.3), there was an association between left ventricular diastolic function (E/A ratio) and fGLP-1 (r = 0.19, P = 0.001), 60GLP-1 (r = 0.20, P < 0.001) and ΔGLP-1 (r = 0.18, P = 0.004). There was a lack of differences in plasma levels of GLP-1 between the groups with Type 2 diabetes and normal glucose tolerance.There were no longitudinal associations between GLP-1 levels and incidence of hospitalization for congestive heart failure. However, without any causality proven, GLP-1 levels did correlate, cross-sectionally, with left ventricular diastolic function in this cohort, suggesting that pathways including GLP-1 might be involved in the regulation of cardiac diastolic function.

    View details for DOI 10.1111/j.1464-5491.2010.03207.x

    View details for Web of Science ID 000287243300007

    View details for PubMedID 21309838

  • Nationwide Cohort Study of Risk of Ischemic Heart Disease in Patients With Celiac Disease CIRCULATION Ludvigsson, J. F., James, S., Askling, J., Stenestrand, U., Ingelsson, E. 2011; 123 (5): 483-U239

    Abstract

    Studies on ischemic heart disease (IHD) incidence in individuals with celiac disease (CD) are contradictory and do not take small intestinal pathology into account.In this Swedish population-based cohort study, we examined the risk of IHD in patients with CD based on small intestinal histopathology. We defined IHD as death or incident disease in myocardial infarction or angina pectoris in Swedish national registers. In 2006 to 2008, we collected duodenal/jejunal biopsy data on CD (equal to villous atrophy; Marsh 3; n=28 190 unique individuals) and inflammation without villous atrophy (Marsh 1 to 2; n=12 598) from all 28 pathology departments in Sweden. A third cohort consisted of 3658 individuals with normal mucosa but positive CD serology (Marsh 0, latent CD). We found an increased risk of incident IHD in patients undergoing small intestinal biopsy that was independent of small intestinal histopathology (CD: hazard ratio [HR], 1.19; 95% confidence interval [CI], 1.11 to 1.28; 991 events; inflammation: HR, 1.28; 95% CI, 1.19 to 1.39; 809 events; and latent CD: HR, 1.14; 95% CI, 0.87 to 1.50; 62 events). Celiac disease (HR, 1.22; 95% CI, 1.06 to 1.40) and inflammation (HR, 1.32; 95% CI, 1.14 to 1.52) were both associated with death resulting from IHD, whereas latent CD was not (HR, 0.71; 95% CI, 0.34 to 1.50).Individuals with CD or small intestinal inflammation are at increased risk of incident IHD. We were unable to show a positive association between latent CD and incident IHD.

    View details for DOI 10.1161/CIRCULATIONAHA.110.965624

    View details for Web of Science ID 000287021300011

    View details for PubMedID 21262996

  • Biomarkers of Extracellular Matrix Metabolism (MMP-9 and TIMP-1) and Risk of Stroke, Myocardial Infarction, and Cause-Specific Mortality: Cohort Study PLOS ONE Hansson, J., Vasan, R. S., Arnlov, J., Ingelsson, E., Lind, L., Larsson, A., Michaelsson, K., Sundstrom, J. 2011; 6 (1)

    Abstract

    Turnover of the extracellular matrix in all solid organs is governed mainly by a balance between the degrading matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs). An altered extracellular matrix metabolism has been implicated in a variety of diseases. We investigated relations of serum levels of MMP-9 and TIMP-1 to mortality risk from an etiological perspective.The prospective Uppsala Longitudinal Study of Adult Men (ULSAM) cohort, followed from 1991-1995 for up to 18.1 years. A random population-based sample of 1,082 71-year-old men, no loss to follow-up. Endpoints were all-cause (n = 628), cardiovascular (n = 230), non-cardiovascular (n = 398) and cancer mortality (n = 178), and fatal or non-fatal myocardial infarction (n = 138) or stroke (n = 163).Serum MMP-9 and TIMP-1 levels were associated with risk of all-cause mortality (Cox proportional hazard ratio [HR] per standard deviation 1.10, 95% confidence interval [CI] 1.03-1.19; and 1.11, 1.02-1.20; respectively). TIMP-1 levels were mainly related to risks of cardiovascular mortality and stroke (HR per standard deviation 1.22, 95% CI 1.09-1.37; and 1.18, 1.04-1.35; respectively). All relations except those of TIMP-1 to stroke risk were attenuated by adjustment for cardiovascular disease risk factors. Relations in a subsample without cardiovascular disease or cancer were similar to those in the total sample.In this community-based cohort of elderly men, serum MMP-9 and TIMP-1 levels were related to mortality risk. An altered extracellular matrix metabolism may be involved in several detrimental pathways, and circulating MMP-9 or TIMP-1 levels may be relevant markers thereof.

    View details for DOI 10.1371/journal.pone.0016185

    View details for Web of Science ID 000286520600027

    View details for PubMedID 21283828

  • LifeGene-a large prospective population-based study of global relevance EUROPEAN JOURNAL OF EPIDEMIOLOGY Almqvist, C., Adami, H., Franks, P. W., Groop, L., Ingelsson, E., Kere, J., Lissner, L., Litton, J., Maeurer, M., Michaelsson, K., Palmgren, J., Pershagen, G., Ploner, A., Sullivan, P. F., Tybring, G., Pedersen, N. L. 2011; 26 (1): 67-77

    Abstract

    Studying gene-environment interactions requires that the amount and quality of the lifestyle data is comparable to what is available for the corresponding genomic data. Sweden has several crucial prerequisites for comprehensive longitudinal biomedical research, such as the personal identity number, the universally available national health care system, continuously updated population and health registries and a scientifically motivated population. LifeGene builds on these strengths to bridge the gap between basic research and clinical applications with particular attention to populations, through a unique design in a research-friendly setting. LifeGene is designed both as a prospective cohort study and an infrastructure with repeated contacts of study participants approximately every 5 years. Index persons aged 18-45 years old will be recruited and invited to include their household members (partner and any children). A comprehensive questionnaire addressing cutting-edge research questions will be administered through the web with short follow-ups annually. Biosamples and physical measurements will also be collected at baseline, and re-administered every 5 years thereafter. Event-based sampling will be a key feature of LifeGene. The household-based design will give the opportunity to involve young couples prior to and during pregnancy, allowing for the first study of children born into cohort with complete pre-and perinatal data from both the mother and father. Questions and sampling schemes will be tailored to the participants' age and life events. The target of LifeGene is to enroll 500,000 Swedes and follow them longitudinally for at least 20 years.

    View details for DOI 10.1007/s10654-010-9521-x

    View details for Web of Science ID 000286104700009

    View details for PubMedID 21104112

  • Impact of BMI and the Metabolic Syndrome on the Risk of Diabetes in Middle-Aged Men DIABETES CARE Arnlov, J., Sundstrom, J., Ingelsson, E., Lind, L. 2011; 34 (1): 61-65

    Abstract

    The existence of an obese subgroup with a healthy metabolic profile and low diabetes risk has been proposed; yet long-term data are lacking. We aimed to investigate associations between combinations of BMI categories and metabolic syndrome and risk of type 2 diabetes in middle-aged men.At age 50, cardiovascular risk factors were assessed in 1,675 participants without diabetes in the community-based Uppsala Longitudinal Study of Adult Men (ULSAM) study. According to BMI/metabolic syndrome status, they were categorized as normal weight (BMI <25 kg/m²) without metabolic syndrome (National Cholesterol Education Program criteria, n = 853), normal weight with metabolic syndrome (n = 60), overweight (BMI 25-30 kg/m²) without metabolic syndrome (n = 557), overweight with metabolic syndrome (n = 117), obese (BMI >30 kg/m²) without metabolic syndrome (n = 28), and obese with metabolic syndrome (n = 60). We investigated the associations between BMI/metabolic syndrome categories at baseline and diabetes incidence.After 20 years, 160 participants had developed diabetes. In logistic regression models adjusting for age, smoking, and physical activity, increased risks for diabetes were observed in the normal weight with metabolic syndrome (odds ratio 3.28 [95% CI] 1.38-7.81; P = 0.007), overweight without metabolic syndrome (3.49 [2.26-5.42]; P < 0.001), overweight with metabolic syndrome (7.77 [4.44-13.62]; P < 0.001), obese without metabolic syndrome (11.72 [4.88-28.16]; P < 0.001), and obese with metabolic syndrome (10.06 [5.19-19.51]; P < 0.001) categories compared with the normal weight without metabolic syndrome category.Overweight or obese men without metabolic syndrome were at increased risk for diabetes. Our data provide further evidence that overweight and obesity in the absence of the metabolic syndrome should not be considered a harmless condition.

    View details for DOI 10.2337/dc10-0955

    View details for Web of Science ID 000286497000014

    View details for PubMedID 20852030

  • Thirty new loci for age at menarche identified by a meta-analysis of genome-wide association studies NATURE GENETICS Elks, C. E., Perry, J. R., Sulem, P., Chasman, D. I., Franceschini, N., He, C., Lunetta, K. L., Visser, J. A., Byrne, E. M., Cousminer, D. L., Gudbjartsson, D. F., Esko, T., Feenstra, B., Hottenga, J., Koller, D. L., Kutalik, Z., Lin, P., Mangino, M., Marongiu, M., McArdle, P. F., Smith, A. V., Stolk, L., Van Wingerden, S. H., Zhao, J. H., Albrecht, E., Corre, T., Ingelsson, E., Hayward, C., Magnusson, P. K., Smith, E. N., Ulivi, S., Warrington, N. M., Zgaga, L., Alavere, H., Amin, N., Aspelund, T., Bandinelli, S., Barroso, I., Berenson, G. S., Bergmann, S., Blackburn, H., Boerwinkle, E., Buring, J. E., Busonero, F., Campbell, H., Chanock, S. J., Chen, W., Cornelis, M. C., Couper, D., Coviello, A. D., D'Adamo, P., de Faire, U., de Geus, E. J., Deloukas, P., Doering, A., Smith, G. D., Easton, D. F., Eiriksdottir, G., Emilsson, V., Eriksson, J., Ferrucci, L., Folsom, A. R., Foroud, T., Garcia, M., Gasparini, P., Geller, F., Gieger, C., Gudnason, V., Hall, P., Hankinson, S. E., Ferreli, L., Heath, A. C., Hernandez, D. G., Hofman, A., Hu, F. B., Illig, T., Jaervelin, M., Johnson, A. D., Karasik, D., Khaw, K., Kiel, D. P., Kilpelaeinen, T. O., Kolcic, I., Kraft, P., Launer, L. J., Laven, J. S., Li, S., Liu, J., Levy, D., Martin, N. G., McArdle, W. L., Melbye, M., Mooser, V., Murray, J. C., Murray, S. S., Nalls, M. A., Navarro, P., Nelis, M., Ness, A. R., Northstone, K., Oostra, B. A., Peacock, M., Palmer, L. J., Palotie, A., Pare, G., Parker, A. N., Pedersen, N. L., Peltonen, L., Pennell, C. E., Pharoah, P., Polasek, O., Plump, A. S., Pouta, A., Porcu, E., Rafnar, T., Rice, J. P., Ring, S. M., Rivadeneira, F., Rudan, I., Sala, C., Salomaa, V., Sanna, S., Schlessinger, D., Schork, N. J., Scuteri, A., Segre, A. V., Shuldiner, A. R., Soranzo, N., Sovio, U., Srinivasan, S. R., Strachan, D. P., Tammesoo, M., Tikkanen, E., Toniolo, D., Tsui, K., Tryggvadottir, L., Tyrer, J., Uda, M., van Dam, R. M., van Meurs, J. B., Vollenweider, P., Waeber, G., Wareham, N. J., Waterworth, D. M., Weedon, M. N., Wichmann, H. E., Willemsen, G., Wilson, J. F., Wright, A. F., Young, L., Zhai, G., Zhuang, W. V., Bierut, L. J., Boomsma, D. I., Boyd, H. A., Crisponi, L., Demerath, E. W., van Duijn, C. M., Econs, M. J., Harris, T. B., Hunter, D. J., Loos, R. J., Metspalu, A., Montgomery, G. W., Ridker, P. M., Spector, T. D., Streeten, E. A., Stefansson, K., Thorsteinsdottir, U., Uitterlinden, A. G., Widen, E., Murabito, J. M., Ong, K. K., Murray, A. 2010; 42 (12): 1077-U73

    Abstract

    To identify loci for age at menarche, we performed a meta-analysis of 32 genome-wide association studies in 87,802 women of European descent, with replication in up to 14,731 women. In addition to the known loci at LIN28B (P = 5.4 × 10⁻⁶⁰) and 9q31.2 (P = 2.2 × 10⁻³³), we identified 30 new menarche loci (all P < 5 × 10⁻⁸) and found suggestive evidence for a further 10 loci (P < 1.9 × 10⁻⁶). The new loci included four previously associated with body mass index (in or near FTO, SEC16B, TRA2B and TMEM18), three in or near other genes implicated in energy homeostasis (BSX, CRTC1 and MCHR2) and three in or near genes implicated in hormonal regulation (INHBA, PCSK2 and RXRG). Ingenuity and gene-set enrichment pathway analyses identified coenzyme A and fatty acid biosynthesis as biological processes related to menarche timing.

    View details for DOI 10.1038/ng.714

    View details for Web of Science ID 000284578800010

    View details for PubMedID 21102462

    View details for PubMedCentralID PMC3140055

  • Plasma parathyroid hormone and risk of congestive heart failure in the community EUROPEAN JOURNAL OF HEART FAILURE Hagstrom, E., Ingelsson, E., Sundstrom, J., Hellman, P., Larsson, T. E., Berglund, L., Melhus, H., Held, C., Michaelsson, K., Lind, L., Arnlov, J. 2010; 12 (11): 1186-1192

    Abstract

    In experimental studies parathyroid hormone (PTH) has been associated with underlying causes of heart failure (HF) such as atherosclerosis, left ventricular hypertrophy, and myocardial fibrosis. Individuals with increased levels of PTH, such as primary or secondary hyperparathyroidism patients, have increased risk of ischaemic heart disease and HF. Moreover, increasing PTH is associated with worse prognosis in patients with overt HF. However, the association between PTH and the development HF in the community has not been reported.In a prospective, community-based study of 864 elderly men without HF or valvular disease at baseline (mean age 71 years, the ULSAM study) the association between plasma (P)-PTH and HF hospitalization was investigated adjusted for established HF risk factors (myocardial infarction, hypertension, diabetes, electrocardiographic left ventricular hypertrophy, smoking, and hypercholesterolaemia) and variables reflecting mineral metabolism (S-calcium, S-phosphate, P-vitamin D, S-albumin, dietary calcium and vitamin D intake, physical activity, glomerular filtration rate, and blood draw season). During follow-up (median 8 years), 75 individuals were hospitalized due to HF. In multivariable Cox-regression analyses, higher P-PTH was associated with increased HF hospitalization (hazard ratio for 1-SD increase of PTH, 1.41, 95% CI 1.12-1.77, P = 0.003). Parathyroid hormone also predicted hospitalization in participants without apparent ischaemic HF and in participants with normal P-PTH.In a large community-based sample of elderly men, PTH predicted HF hospitalizations, also after accounting for established risk factors and mineral metabolism variables.  Our data suggest a role for PTH in the development of HF even in the absence of overt hyperparathyroidism.

    View details for DOI 10.1093/eurjhf/hfq134

    View details for Web of Science ID 000283674300008

    View details for PubMedID 20797986

  • Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index NATURE GENETICS Speliotes, E. K., Willer, C. J., Berndt, S. I., Monda, K. L., Thorleifsson, G., Jackson, A. U., Allen, H. L., Lindgren, C. M., Luan, J., Maegi, R., Randall, J. C., Vedantam, S., Winkler, T. W., Qi, L., Workalemahu, T., Heid, I. M., Steinthorsdottir, V., Stringham, H. M., Weedon, M. N., Wheeler, E., Wood, A. R., Ferreira, T., Weyant, R. J., Segre, A. V., Estrada, K., Liang, L., Nemesh, J., Park, J., Gustafsson, S., Kilpelaenen, T. O., Yang, J., Bouatia-Naji, N., Esko, T., Feitosa, M. F., Kutalik, Z., Mangino, M., Raychaudhuri, S., Scherag, A., Smith, A. V., Welch, R., Zhao, J. H., Aben, K. K., Absher, D. M., Amin, N., Dixon, A. L., Fisher, E., Glazer, N. L., Goddard, M. E., Heard-Costa, N. L., Hoesel, V., Hottenga, J., Johansson, A., Johnson, T., Ketkar, S., Lamina, C., Li, S., Moffatt, M. F., Myers, R. H., Narisu, N., Perry, J. R., Peters, M. J., Preuss, M., Ripatti, S., Rivadeneira, F., Sandholt, C., Scott, L. J., Timpson, N. J., Tyrer, J. P., van Wingerden, S., Watanabe, R. M., White, C. C., Wiklund, F., Barlassina, C., Chasman, D. I., Cooper, M. N., Jansson, J., Lawrence, R. W., Pellikka, N., Prokopenko, I., Shi, J., Thiering, E., Alavere, H., Alibrandi, M. T., Almgren, P., Arnold, A. M., Aspelund, T., Atwood, L. D., Balkau, B., Balmforth, A. J., Bennett, A. J., Ben-Shlomo, Y., Bergman, R. N., Bergmann, S., Biebermann, H., Blakemore, A. I., Boes, T., Bonnycastle, L. L., Bornstein, S. R., Brown, M. J., Buchanan, T. A., Busonero, F., Campbell, H., Cappuccio, F. P., Cavalcanti-Proenca, C., Chen, Y. I., Chen, C., Chines, P. S., Clarke, R., Coin, L., Connell, J., Day, I. N., den Heijer, M., Duan, J., Ebrahim, S., Elliott, P., Elosua, R., Eiriksdottir, G., Erdos, M. R., Eriksson, J. G., Facheris, M. F., Felix, S. B., Fischer-Posovszky, P., Folsom, A. R., Friedrich, N., Freimer, N. B., Fu, M., Gaget, S., Gejman, P. V., Geus, E. J., Gieger, C., Gjesing, A. P., Goel, A., Goyette, P., Grallert, H., Graessler, J., Greenawalt, D. M., Groves, C. J., Gudnason, V., Guiducci, C., Hartikainen, A., Hassanali, N., Hall, A. S., Havulinna, A. S., Hayward, C., Heath, A. C., Hengstenberg, C., Hicks, A. A., Hinney, A., Hofman, A., Homuth, G., Hui, J., Igl, W., Iribarren, C., Isomaa, B., Jacobs, K. B., Jarick, I., Jewell, E., John, U., Jorgensen, T., Jousilahti, P., Jula, A., Kaakinen, M., Kajantie, E., Kaplan, L. M., Kathiresan, S., Kettunen, J., Kinnunen, L., Knowles, J. W., Kolcic, I., Koenig, I. R., Koskinen, S., Kovacs, P., Kuusisto, J., Kraft, P., Kvaloy, K., Laitinen, J., Lantieri, O., Lanzani, C., Launer, L. J., Lecoeur, C., Lehtimaeki, T., Lettre, G., Liu, J., Lokki, M., Lorentzon, M., Luben, R. N., Ludwig, B., Manunta, P., Marek, D., Marre, M., Martin, N. G., McArdle, W. L., McCarthy, A., McKnight, B., Meitinger, T., Melander, O., Meyre, D., Midthjell, K., Montgomery, G. W., Morken, M. A., Morris, A. P., Mulic, R., Ngwa, J. S., Nelis, M., Neville, M. J., Nyholt, D. R., O'Donnell, C. J., O'Rahilly, S., Ong, K. K., Oostra, B., Pare, G., Parker, A. N., Perola, M., Pichler, I., Pietilaeinen, K. H., Platou, C. G., Polasek, O., Pouta, A., Rafelt, S., Raitakari, O., Rayner, N. W., Ridderstrale, M., Rief, W., Ruokonen, A., Robertson, N. R., Rzehak, P., Salomaa, V., Sanders, A. R., Sandhu, M. S., Sanna, S., Saramies, J., Savolainen, M. J., Scherag, S., Schipf, S., Schreiber, S., Schunkert, H., Silander, K., Sinisalo, J., Siscovick, D. S., Smit, J. H., Soranzo, N., Sovio, U., Stephens, J., Surakka, I., Swift, A. J., Tammesoo, M., Tardif, J., Teder-Laving, M., Teslovich, T. M., Thompson, J. R., Thomson, B., Toenjes, A., Tuomi, T., van Meurs, J. B., van Ommen, G., Vatin, V., Viikari, J., Visvikis-Siest, S., Vitart, V., Vogel, C. I., Voight, B. F., Waite, L. L., Wallaschofski, H., Walters, G. B., Widen, E., Wiegand, S., Wild, S. H., Willemsen, G., Witte, D. R., Witteman, J. C., Xu, J., Zhang, Q., Zgaga, L., Ziegler, A., Zitting, P., Beilby, J. P., Farooqi, I. S., Hebebrand, J., Huikuri, H. V., James, A. L., Kaehoenen, M., Levinson, D. F., Macciardi, F., Nieminen, M. S., Ohlsson, C., Palmer, L. J., Ridker, P. M., Stumvoll, M., Beckmann, J. S., Boeing, H., Boerwinkle, E., Boomsma, D. I., Caulfield, M. J., Chanock, S. J., Collins, F. S., Cupples, L. A., Smith, G. D., Erdmann, J., Froguel, P., Greonberg, H., Gyllensten, U., Hall, P., Hansen, T., Harris, T. B., Hattersley, A. T., Hayes, R. B., Heinrich, J., Hu, F. B., Hveem, K., Illig, T., Jarvelin, M., Kaprio, J., Karpe, F., Khaw, K., Kiemeney, L. A., Krude, H., Laakso, M., Lawlor, D. A., Metspalu, A., Munroe, P. B., Ouwehand, W. H., Pedersen, O., Penninx, B. W., Peters, A., Pramstaller, P. P., Quertermous, T., Reinehr, T., Rissanen, A., Rudan, I., Samani, N. J., Schwarz, P. E., Shuldiner, A. R., Spector, T. D., Tuomilehto, J., Uda, M., Uitterlinden, A., Valle, T. T., Wabitsch, M., Waeber, G., Wareham, N. J., Watkins, H., Wilson, J. F., Wright, A. F., Zillikens, M. C., Chatterjee, N., McCarroll, S. A., Purcell, S., Schadt, E. E., Visscher, P. M., Assimes, T. L., Borecki, I. B., Deloukas, P., Fox, C. S., Groop, L. C., Haritunians, T., Hunter, D. J., Kaplan, R. C., Mohlke, K. L., O'Connell, J. R., Peltonen, L., Schlessinger, D., Strachan, D. P., van Duijn, C. M., Wichmann, H., Frayling, T. M., Thorsteinsdottir, U., Abecasis, G. R., Barroso, I., Boehnke, M., Stefansson, K., North, K. E., McCarthy, M. I., Hirschhorn, J. N., Ingelsson, E., Loos, R. J. 2010; 42 (11): 937-U53

    Abstract

    Obesity is globally prevalent and highly heritable, but its underlying genetic factors remain largely elusive. To identify genetic loci for obesity susceptibility, we examined associations between body mass index and ∼ 2.8 million SNPs in up to 123,865 individuals with targeted follow up of 42 SNPs in up to 125,931 additional individuals. We confirmed 14 known obesity susceptibility loci and identified 18 new loci associated with body mass index (P < 5 × 10⁻⁸), one of which includes a copy number variant near GPRC5B. Some loci (at MC4R, POMC, SH2B1 and BDNF) map near key hypothalamic regulators of energy balance, and one of these loci is near GIPR, an incretin receptor. Furthermore, genes in other newly associated loci may provide new insights into human body weight regulation.

    View details for DOI 10.1038/ng.686

    View details for Web of Science ID 000283540500010

    View details for PubMedID 20935630

  • Meta-analysis identifies 13 new loci associated with waist-hip ratio and reveals sexual dimorphism in the genetic basis of fat distribution NATURE GENETICS Heid, I. M., Jackson, A. U., Randall, J. C., Winkler, T. W., Qi, L., Steinthorsdottir, V., Thorleifsson, G., Zillikens, M. C., Speliotes, E. K., Maegi, R., Workalemahu, T., White, C. C., Bouatia-Naji, N., Harris, T. B., Berndt, S. I., Ingelsson, E., Willer, C. J., Weedon, M. N., Luan, J., Vedantam, S., Esko, T., Kilpelaeinen, T. O., Kutalik, Z., Li, S., Monda, K. L., Dixon, A. L., Holmes, C. C., Kaplan, L. M., Liang, L., Min, J. L., Moffatt, M. F., Molony, C., Nicholson, G., Schadt, E. E., Zondervan, K. T., Feitosa, M. F., Ferreira, T., Allen, H. L., Weyant, R. J., Wheeler, E., Wood, A. R., Estrada, K., Goddard, M. E., Lettre, G., Mangino, M., Nyholt, D. R., Purcell, S., Smith, A. V., Visscher, P. M., Yang, J., McCarroll, S. A., Nemesh, J., Voight, B. F., Absher, D., Amin, N., Aspelund, T., Coin, L., Glazer, N. L., Hayward, C., Heard-Costa, N. L., Hottenga, J., Johansson, A., Johnson, T., Kaakinen, M., Kapur, K., Ketkar, S., Knowles, J. W., Kraft, P., Kraja, A. T., Lamina, C., Leitzmann, M. F., McKnight, B., Morris, A. P., Ong, K. K., Perry, J. R., Peters, M. J., Polasek, O., Prokopenko, I., Rayner, N. W., Ripatti, S., Rivadeneira, F., Robertson, N. R., Sanna, S., Sovio, U., Surakka, I., Teumer, A., van Wingerden, S., Vitart, V., Zhao, J. H., Cavalcanti-Proenca, C., Chines, P. S., Fisher, E., Kulzer, J. R., Lecoeur, C., Narisu, N., Sandholt, C., Scott, L. J., Silander, K., Stark, K., Tammesoo, M., Teslovich, T. M., Timpson, N. J., Watanabe, R. M., Welch, R., Chasman, D. I., Cooper, M. N., Jansson, J., Kettunen, J., Lawrence, R. W., Pellikka, N., Perola, M., Vandenput, L., Alavere, H., Almgren, P., Atwood, L. D., Bennett, A. J., Biffar, R., Bonnycastle, L. L., Bornstein, S. R., Buchanan, T. A., Campbell, H., Day, I. N., Dei, M., Doerr, M., Elliott, P., Erdos, M. R., Eriksson, J. G., Freimer, N. B., Fu, M., Gaget, S., Geus, E. J., Gjesing, A. P., Grallert, H., Graessler, J., Groves, C. J., Guiducci, C., Hartikainen, A., Hassanali, N., Havulinna, A. S., Herzig, K., Hicks, A. A., Hui, J., Igl, W., Jousilahti, P., Jula, A., Kajantie, E., Kinnunen, L., Kolcic, I., Koskinen, S., Kovacs, P., Kroemer, H. K., Krzelj, V., Kuusisto, J., Kvaloy, K., Laitinen, J., Lantieri, O., Lathrop, G. M., Lokki, M., Luben, R. N., Ludwig, B., McArdle, W. L., McCarthy, A., Morken, M. A., Nelis, M., Neville, M. J., Pare, G., Parker, A. N., Peden, J. F., Pichler, I., Pietilainen, K. H., Platou, C. G., Pouta, A., Ridderstrale, M., Samani, N. J., Saramies, J., Sinisalo, J., Smit, J. H., Strawbridge, R. J., Stringham, H. M., Swift, A. J., Teder-Laving, M., Thomson, B., Usala, G., van Meurs, J. B., van Ommen, G., Vatin, V., Volpato, C. B., Wallaschofski, H., Walters, G. B., Widen, E., Wild, S. H., Willemsen, G., Witte, D. R., Zgaga, L., Zitting, P., Beilby, J. P., James, A. L., Kahonen, M., Lehtimaki, T., Nieminen, M. S., Ohlsson, C., Palmer, L. J., Raitakari, O., Ridker, P. M., Stumvoll, M., Toenjes, A., Viikari, J., Balkau, B., Ben-Shlomo, Y., Bergman, R. N., Boeing, H., Smith, G. D., Ebrahim, S., Froguel, P., Hansen, T., Hengstenberg, C., Hveem, K., Isomaa, B., Jorgensen, T., Karpe, F., Khaw, K., Laakso, M., Lawlor, D. A., Marre, M., Meitinger, T., Metspalu, A., Midthjell, K., Pedersen, O., Salomaa, V., Schwarz, P. E., Tuomi, T., Tuomilehto, J., Valle, T. T., Wareham, N. J., Arnold, A. M., Beckmann, J. S., Bergmann, S., Boerwinkle, E., Boomsma, D. I., Caulfield, M. J., Collins, F. S., Eiriksdottir, G., Gudnason, V., Gyllensten, U., Hamsten, A., Hattersley, A. T., Hofman, A., Hu, F. B., Illig, T., Iribarren, C., Jarvelin, M., Kao, W. H., Kaprio, J., Launer, L. J., Munroe, P. B., Oostra, B., Penninx, B. W., Pramstaller, P. P., Psaty, B. M., Quertermous, T., Rissanen, A., Rudan, I., Shuldiner, A. R., Soranzo, N., Spector, T. D., Syvanen, A., Uda, M., Uitterlinden, A., Voelzke, H., Vollenweider, P., Wilson, J. F., Witteman, J. C., Wright, A. F., Abecasis, G. R., Boehnke, M., Borecki, I. B., Deloukas, P., Frayling, T. M., Groop, L. C., Haritunians, T., Hunter, D. J., Kaplan, R. C., North, K. E., O'Connell, J. R., Peltonen, L., Schlessinger, D., Strachan, D. P., Hirschhorn, J. N., Assimes, T. L., Wichmann, H., Thorsteinsdottir, U., van Duijn, C. M., Stefansson, K., Cupples, L. A., Loos, R. J., Barroso, I., McCarthy, M. I., Fox, C. S., Mohlke, K. L., Lindgren, C. M. 2010; 42 (11): 949-U160

    Abstract

    Waist-hip ratio (WHR) is a measure of body fat distribution and a predictor of metabolic consequences independent of overall adiposity. WHR is heritable, but few genetic variants influencing this trait have been identified. We conducted a meta-analysis of 32 genome-wide association studies for WHR adjusted for body mass index (comprising up to 77,167 participants), following up 16 loci in an additional 29 studies (comprising up to 113,636 subjects). We identified 13 new loci in or near RSPO3, VEGFA, TBX15-WARS2, NFE2L3, GRB14, DNM3-PIGC, ITPR2-SSPN, LY86, HOXC13, ADAMTS9, ZNRF3-KREMEN1, NISCH-STAB1 and CPEB4 (P = 1.9 × 10⁻⁹ to P = 1.8 × 10⁻⁴⁰) and the known signal at LYPLAL1. Seven of these loci exhibited marked sexual dimorphism, all with a stronger effect on WHR in women than men (P for sex difference = 1.9 × 10⁻³ to P = 1.2 × 10⁻¹³). These findings provide evidence for multiple loci that modulate body fat distribution independent of overall adiposity and reveal strong gene-by-sex interactions.

    View details for DOI 10.1038/ng.685

    View details for Web of Science ID 000283540500011

    View details for PubMedID 20935629

  • Differential White Blood Cell Count and Type 2 Diabetes: Systematic Review and Meta-Analysis of Cross-Sectional and Prospective Studies PLOS ONE Gkrania-Klotsas, E., Ye, Z., Cooper, A. J., Sharp, S. J., Luben, R., Biggs, M. L., Chen, L., Gokulakrishnan, K., Hanefeld, M., Ingelsson, E., Lai, W., Lin, S., Lind, L., Lohsoonthorn, V., Mohan, V., Muscari, A., Nilsson, G., Ohrvik, J., Qiang, J. C., Jenny, N. S., Tamakoshi, K., Temelkova-Kurktschiev, T., Wang, Y., Yajnik, C. S., Zoli, M., Khaw, K., Forouhi, N. G., Wareham, N. J., Langenberg, C. 2010; 5 (10)

    Abstract

    Biological evidence suggests that inflammation might induce type 2 diabetes (T2D), and epidemiological studies have shown an association between higher white blood cell count (WBC) and T2D. However, the association has not been systematically investigated.Studies were identified through computer-based and manual searches. Previously unreported studies were sought through correspondence. 20 studies were identified (8,647 T2D cases and 85,040 non-cases). Estimates of the association of WBC with T2D were combined using random effects meta-analysis; sources of heterogeneity as well as presence of publication bias were explored.The combined relative risk (RR) comparing the top to bottom tertile of the WBC count was 1.61 (95% CI: 1.45; 1.79, p = 1.5*10(-18)). Substantial heterogeneity was present (I(2) = 83%). For granulocytes the RR was 1.38 (95% CI: 1.17; 1.64, p = 1.5*10(-4)), for lymphocytes 1.26 (95% CI: 1.02; 1.56, p = 0.029), and for monocytes 0.93 (95% CI: 0.68; 1.28, p = 0.67) comparing top to bottom tertile. In cross-sectional studies, RR was 1.74 (95% CI: 1.49; 2.02, p = 7.7*10(-13)), while in cohort studies it was 1.48 (95% CI: 1.22; 1.79, p = 7.7*10(-5)). We assessed the impact of confounding in EPIC-Norfolk study and found that the age and sex adjusted HR of 2.19 (95% CI: 1.74; 2.75) was attenuated to 1.82 (95% CI: 1.45; 2.29) after further accounting for smoking, T2D family history, physical activity, education, BMI and waist circumference.A raised WBC is associated with higher risk of T2D. The presence of publication bias and failure to control for all potential confounders in all studies means the observed association is likely an overestimate.

    View details for DOI 10.1371/journal.pone.0013405

    View details for Web of Science ID 000283045300009

    View details for PubMedID 20976133

  • Hundreds of variants clustered in genomic loci and biological pathways affect human height NATURE Allen, H. L., Estrada, K., Lettre, G., Berndt, S. I., Weedon, M. N., Rivadeneira, F., Willer, C. J., Jackson, A. U., Vedantam, S., Raychaudhuri, S., Ferreira, T., Wood, A. R., Weyant, R. J., Segre, A. V., Speliotes, E. K., Wheeler, E., Soranzo, N., Park, J., Yang, J., Gudbjartsson, D., Heard-Costa, N. L., Randall, J. C., Qi, L., Smith, A. V., Maegi, R., Pastinen, T., Liang, L., Heid, I. M., Luan, J., Thorleifsson, G., Winkler, T. W., Goddard, M. E., Lo, K. S., Palmer, C., Workalemahu, T., Aulchenko, Y. S., Johansson, A., Zillikens, M. C., Feitosa, M. F., Esko, T., Johnson, T., Ketkar, S., Kraft, P., Mangino, M., Prokopenko, I., Absher, D., Albrecht, E., Ernst, F., Glazer, N. L., Hayward, C., Hottenga, J., Jacobs, K. B., Knowles, J. W., Kutalik, Z., Monda, K. L., Polasek, O., Preuss, M., Rayner, N. W., Robertson, N. R., Steinthorsdottir, V., Tyrer, J. P., Voight, B. F., Wiklund, F., Xu, J., Zhao, J. H., Nyholt, D. R., Pellikka, N., Perola, M., Perry, J. R., Surakka, I., Tammesoo, M., Altmaier, E. L., Amin, N., Aspelund, T., Bhangale, T., Boucher, G., Chasman, D. I., Chen, C., Coin, L., Cooper, M. N., Dixon, A. L., Gibson, Q., Grundberg, E., Hao, K., Junttila, M. J., Kaplan, L. M., Kettunen, J., Koenig, I. R., Kwan, T., Lawrence, R. W., Levinson, D. F., Lorentzon, M., McKnight, B., Morris, A. P., Mueller, M., Ngwa, J. S., Purcell, S., Rafelt, S., Salem, R. M., Salvi, E., Sanna, S., Shi, J., Sovio, U., Thompson, J. R., Turchin, M. C., Vandenput, L., Verlaan, D. J., Vitart, V., White, C. C., Ziegler, A., Almgren, P., Balmforth, A. J., Campbell, H., Citterio, L., de Grandi, A., Dominiczak, A., Duan, J., Elliott, P., Elosua, R., Eriksson, J. G., Freimer, N. B., Geus, E. J., Glorioso, N., Haiqing, S., Hartikainen, A., Havulinna, A. S., Hicks, A. A., Hui, J., Igl, W., Illig, T., Jula, A., Kajantie, E., Kilpelaeinen, T. O., Koiranen, M., Kolcic, I., Koskinen, S., Kovacs, P., Laitinen, J., Liu, J., Lokki, M., Marusic, A., Maschio, A., Meitinger, T., Mulas, A., Pare, G., Parker, A. N., Peden, J. F., Petersmann, A., Pichler, I., Pietilainen, K. H., Pouta, A., Riddertrale, M., Rotter, J. I., Sambrook, J. G., Sanders, A. R., Schmidt, C. O., Sinisalo, J., Smit, J. H., Stringham, H. M., Walters, G. B., Widen, E., Wild, S. H., Willemsen, G., Zagato, L., Zgaga, L., Zitting, P., Alavere, H., Farrall, M., McArdle, W. L., Nelis, M., Peters, M. J., Ripatti, S., vVan Meurs, J. B., Aben, K. K., Ardlie, K. G., Beckmann, J. S., Beilby, J. P., Bergman, R. N., Bergmann, S., Collins, F. S., Cusi, D., den Heijer, M., Eiriksdottir, G., Gejman, P. V., Hall, A. S., Hamsten, A., Huikuri, H. V., Iribarren, C., Kahonen, M., Kaprio, J., Kathiresan, S., Kiemeney, L., Kocher, T., Launer, L. J., Lehtimaki, T., Melander, O., Mosley, T. H., Musk, A. W., Nieminen, M. S., O'Donnell, C. J., Ohlsson, C., Oostra, B., Palmer, L. J., Raitakari, O., Ridker, P. M., Rioux, J. D., Rissanen, A., Rivolta, C., Schunkert, H., Shuldiner, A. R., Siscovick, D. S., Stumvoll, M., Toenjes, A., Tuomilehto, J., van Ommen, G., Viikari, J., Heath, A. C., Martin, N. G., Montgomery, G. W., Province, M. A., Kayser, M., Arnold, A. M., Atwood, L. D., Boerwinkle, E., Chanock, S. J., Deloukas, P., Gieger, C., Gronberg, H., Hall, P., Hattersley, A. T., Hengstenberg, C., Hoffman, W., Lathrop, G. M., Salomaa, V., Schreiber, S., Uda, M., Waterworth, D., Wright, A. F., Assimes, T. L., Barroso, I., Hofman, A., Mohlke, K. L., Boomsma, D. I., Caulfield, M. J., Cupples, L. A., Erdmann, J., Fox, C. S., Gudnason, V., Gyllensten, U., Harris, T. B., Hayes, R. B., Jarvelin, M., Mooser, V., Munroe, P. B., Ouwehand, W. H., Penninx, B. W., Pramstaller, P. P., Quertermous, T., Rudan, I., Samani, N. J., Spector, T. D., Voelzke, H., Watkins, H., Wilson, J. F., Groop, L. C., Haritunians, T., Hu, F. B., Kaplan, R. C., Metspalu, A., North, K. E., Schlessinger, D., Wareham, N. J., Hunter, D. J., O'Connell, J. R., Strachan, D. P., Schadt, H., Thorsteinsdottir, U., Peltonen, L., Uitterlinden, A. G., Visscher, P. M., Chatterjee, N., Loos, R. J., Boehnke, M., McCarthy, M. I., Ingelsson, E., Lindgren, C. M., Abecasis, G. R., Stefansson, K., Frayling, T. M., Hirschhorn, J. N. 2010; 467 (7317): 832-838

    Abstract

    Most common human traits and diseases have a polygenic pattern of inheritance: DNA sequence variants at many genetic loci influence the phenotype. Genome-wide association (GWA) studies have identified more than 600 variants associated with human traits, but these typically explain small fractions of phenotypic variation, raising questions about the use of further studies. Here, using 183,727 individuals, we show that hundreds of genetic variants, in at least 180 loci, influence adult height, a highly heritable and classic polygenic trait. The large number of loci reveals patterns with important implications for genetic studies of common human diseases and traits. First, the 180 loci are not random, but instead are enriched for genes that are connected in biological pathways (P = 0.016) and that underlie skeletal growth defects (P < 0.001). Second, the likely causal gene is often located near the most strongly associated variant: in 13 of 21 loci containing a known skeletal growth gene, that gene was closest to the associated variant. Third, at least 19 loci have multiple independently associated variants, suggesting that allelic heterogeneity is a frequent feature of polygenic traits, that comprehensive explorations of already-discovered loci should discover additional variants and that an appreciable fraction of associated loci may have been identified. Fourth, associated variants are enriched for likely functional effects on genes, being over-represented among variants that alter amino-acid structure of proteins and expression levels of nearby genes. Our data explain approximately 10% of the phenotypic variation in height, and we estimate that unidentified common variants of similar effect sizes would increase this figure to approximately 16% of phenotypic variation (approximately 20% of heritable variation). Although additional approaches are needed to dissect the genetic architecture of polygenic human traits fully, our findings indicate that GWA studies can identify large numbers of loci that implicate biologically relevant genes and pathways.

    View details for DOI 10.1038/nature09410

    View details for Web of Science ID 000282898700065

    View details for PubMedID 20881960

  • Serum Cathepsin S Is Associated with Serum C-Reactive Protein and Interleukin-6 Independently of Obesity in Elderly Men JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Jobs, E., Riserus, U., Ingelsson, E., Helmersson, J., Nerpin, E., Jobs, M., Sundstrom, J., Lind, L., Larsson, A., Basu, S., Arnlov, J. 2010; 95 (9): 4460-4464

    Abstract

    Cathepsin S has been suggested provide a mechanistic link between obesity and atherosclerosis, possibly mediated via adipose tissue-derived inflammation. Previous data have shown an association between circulating cathepsin S and inflammatory markers in the obese, but to date, community-based reports are lacking. Accordingly, we aimed to investigate the association between serum levels of cathepsin S and markers of cytokine-mediated inflammation in a community-based sample, with prespecified subgroup analyses in nonobese participants.Serum cathepsin S, C-reactive protein (CRP), and IL-6 were measured in a community-based cohort of elderly men (Uppsala Longitudinal Study of Adult Men; mean age 71 years, n = 991). CRP and IL-6 were also measured at a reexamination after 7 yr.After adjustment for age, body mass index, fasting plasma glucose, diabetes treatment, systolic blood pressure, diastolic blood pressure, hypertension treatment, serum cholesterol, serum high-density lipoprotein cholesterol, prior cardiovascular disease, smoking, and leisure time physical activity, higher cathepsin S was associated with higher CRP (regression coefficient for 1 sd increase, 0.13; 95% confidence interval 0.07-0.19; P < 0.001) and higher serum IL-6 (regression coefficient for 1 sd increase, 0.08; 95% confidence interval 0.01-0.14; P = 0.02). These associations remained similar in normal-weight participants (body mass index <25 kg/m(2), n = 375). In longitudinal analyses, higher cathepsin S at baseline was associated with higher serum CRP and IL-6 after 7 yr.These results provide additional evidence for the interplay between cathepsin S and inflammatory activity and suggest that this association is present also in normal-weight individuals in the community.

    View details for DOI 10.1210/jc.2010-0328

    View details for Web of Science ID 000281640300065

    View details for PubMedID 20610597

  • Biological, clinical and population relevance of 95 loci for blood lipids NATURE Teslovich, T. M., Musunuru, K., Smith, A. V., Edmondson, A. C., Stylianou, I. M., Koseki, M., Pirruccello, J. P., Ripatti, S., Chasman, D. I., Willer, C. J., Johansen, C. T., Fouchier, S. W., Isaacs, A., Peloso, G. M., Barbalic, M., Ricketts, S. L., Bis, J. C., Aulchenko, Y. S., Thorleifsson, G., Feitosa, M. F., Chambers, J., Orho-Melander, M., Melander, O., Johnson, T., Li, X., Guo, X., Li, M., Cho, Y. S., Go, M. J., Kim, Y. J., Lee, J., Park, T., Kim, K., Sim, X., Ong, R. T., Croteau-Chonka, D. C., Lange, L. A., Smith, J. D., Song, K., Zhao, J. H., Yuan, X., Luan, J., Lamina, C., Ziegler, A., Zhang, W., Zee, R. Y., Wright, A. F., Witteman, J. C., Wilson, J. F., Willemsen, G., Wichmann, H., Whitfield, J. B., Waterworth, D. M., Wareham, N. J., Waeber, G., Vollenweider, P., Voight, B. F., Vitart, V., Uitterlinden, A. G., Uda, M., Tuomilehto, J., Thompson, J. R., Tanaka, T., Surakka, I., Stringham, H. M., Spector, T. D., Soranzo, N., Smit, J. H., Sinisalo, J., Silander, K., Sijbrands, E. J., Scuteri, A., Scott, J., Schlessinger, D., Sanna, S., Salomaa, V., Saharinen, J., Sabatti, C., Ruokonen, A., Rudan, I., Rose, L. M., Roberts, R., Rieder, M., Psaty, B. M., Pramstaller, P. P., Pichler, I., Perola, M., Penninx, B. W., Pedersen, N. L., Pattaro, C., Parker, A. N., Pare, G., Oostra, B. A., O'Donnell, C. J., Nieminen, M. S., Nickerson, D. A., Montgomery, G. W., Meitinger, T., McPherson, R., McCarthy, M. I., McArdle, W., Masson, D., Martin, N. G., Marroni, F., Mangino, M., Magnusson, P. K., Lucas, G., Luben, R., Loos, R. J., Lokki, M., Lettre, G., Langenberg, C., Launer, L. J., Lakatta, E. G., Laaksonen, R., Kyvik, K. O., Kronenberg, F., Koenig, I. R., Khaw, K., Kaprio, J., Kaplan, L. M., Johansson, A., Jarvelin, M., Janssens, A. C., Ingelsson, E., Igi, W., Hovingh, G. K., Hottenga, J., Hofman, A., Hicks, A. A., Hengstenberg, C., Heid, I. M., Hayward, C., Havulinna, A. S., Hastie, N. D., Harris, T. B., Haritunians, T., Hall, A. S., Gyllensten, U., Guiducci, C., Groop, L. C., Gonzalez, E., Gieger, C., Freimer, N. B., Ferrucci, L., Erdmann, J., Elliott, P., Ejebe, K. G., Doering, A., Dominiczak, A. F., Demissie, S., Deloukas, P., de Geus, E. J., de Faire, U., Crawford, G., Collins, F. S., Chen, Y. I., Caulfield, M. J., Campbell, H., Burtt, N. P., Bonnycastle, L. L., Boomsma, D. I., Boekholdt, S. M., Bergman, R. N., Barroso, I., Bandinelli, S., Ballantyne, C. M., Assimes, T. L., Quertermous, T., Altshuler, D., Seielstad, M., Wong, T. Y., Tai, E., Feranil, A. B., Kuzawa, C. W., Adair, L. S., Taylor, H. A., Borecki, I. B., Gabriel, S. B., Wilson, J. G., Holm, H., Thorsteinsdottir, U., Gudnason, V., Krauss, R. M., Mohlke, K. L., Ordovas, J. M., Munroe, P. B., Kooner, J. S., Tall, A. R., Hegele, R. A., Kastelein, J. J., Schadt, E. E., Rotter, J. I., Boerwinkle, E., Strachan, D. P., Mooser, V., Stefansson, K., Reilly, M. P., Samani, N. J., Schunkert, H., Cupples, L. A., Sandhu, M. S., Ridker, P. M., Rader, D. J., van Duijn, C. M., Peltonen, L., Abecasis, G. R., Boehnke, M., Kathiresan, S. 2010; 466 (7307): 707-713

    Abstract

    Plasma concentrations of total cholesterol, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol and triglycerides are among the most important risk factors for coronary artery disease (CAD) and are targets for therapeutic intervention. We screened the genome for common variants associated with plasma lipids in >100,000 individuals of European ancestry. Here we report 95 significantly associated loci (P < 5 x 10(-8)), with 59 showing genome-wide significant association with lipid traits for the first time. The newly reported associations include single nucleotide polymorphisms (SNPs) near known lipid regulators (for example, CYP7A1, NPC1L1 and SCARB1) as well as in scores of loci not previously implicated in lipoprotein metabolism. The 95 loci contribute not only to normal variation in lipid traits but also to extreme lipid phenotypes and have an impact on lipid traits in three non-European populations (East Asians, South Asians and African Americans). Our results identify several novel loci associated with plasma lipids that are also associated with CAD. Finally, we validated three of the novel genes-GALNT2, PPP1R3B and TTC39B-with experiments in mouse models. Taken together, our findings provide the foundation to develop a broader biological understanding of lipoprotein metabolism and to identify new therapeutic opportunities for the prevention of CAD.

    View details for DOI 10.1038/nature09270

    View details for Web of Science ID 000280562500029

    View details for PubMedID 20686565

  • Absolute and Relative Risk of Cardiovascular Disease in Men With Prostate Cancer: Results From the Population-Based PCBaSe Sweden JOURNAL OF CLINICAL ONCOLOGY Van Hemelrijck, M., Garmo, H., Holmberg, L., Ingelsson, E., Bratt, O., Bill-Axelson, A., Lambe, M., Stattin, P., Adolfsson, J. 2010; 28 (21): 3448-3456

    Abstract

    Cardiovascular disease (CVD) is a potential adverse effect of endocrine treatment (ET) for prostate cancer (PC). We investigated absolute and relative CVD risk in 76,600 patients with PC undergoing ET, curative treatment, or surveillance.PCBaSe Sweden is based on the National Prostate Cancer Register, which covers more than 96% of PC cases. Standardized incidence ratios (SIRs) and standardized mortality ratios (SMRs) of ischemic heart disease (IHD), acute myocardial infarction (MI), arrhythmia, heart failure, and stroke were calculated to compare observed and expected (using total Swedish population) numbers of CVD, taking into account age, calendar time, and previous CVD.Between 1997 and 2007, 30,642 patients with PC received primary ET, 26,432 curative treatment, and 19,527 surveillance. SIRs for CVD were elevated in all men with the highest for those undergoing ET, independent of circulatory disease history (SIR MI for men without circulatory disease history: 1.40 [95% CI, 1.31 to 1.49], 1.15 [95% CI, 1.01 to 1.31], and 1.20 [95% CI, 1.11 to 1.30] for men undergoing ET, curative treatment, and surveillance, respectively). Absolute risk differences (ARD) showed that two (arrhythmia) to eight (IHD) extra cases of CVD would occur per 1,000 person-years. SMRs showed similar patterns, with ARD of zero (arrhythmia) to three (IHD) per 1,000 person-years.Increased relative risks of nonfatal and fatal CVD were found among all men with PC, especially those treated with ET. Because ET is currently the only effective treatment for metastatic disease and the ARDs were rather small, our findings indicate that CVD risk should be considered when prescribing ET but should not constitute a contraindication when the expected gain is tangible.

    View details for DOI 10.1200/JCO.2010.29.1567

    View details for Web of Science ID 000280003700009

    View details for PubMedID 20567006

  • Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies LANCET Sarwar, N., Gao, P., Seshasai, S. R., Gobin, R., Kaptoge, S., Di Angelantonio, E., Ingelsson, E., Lawlor, D. A., Selvin, E., Stampfer, M., Stehouwer, C. D., Lewington, S., Pennells, L., Thompson, A., Sattar, N., White, I. R., Ray, K. K., Danesh, J. 2010; 375 (9733): 2215-2222

    Abstract

    Uncertainties persist about the magnitude of associations of diabetes mellitus and fasting glucose concentration with risk of coronary heart disease and major stroke subtypes. We aimed to quantify these associations for a wide range of circumstances.We undertook a meta-analysis of individual records of diabetes, fasting blood glucose concentration, and other risk factors in people without initial vascular disease from studies in the Emerging Risk Factors Collaboration. We combined within-study regressions that were adjusted for age, sex, smoking, systolic blood pressure, and body-mass index to calculate hazard ratios (HRs) for vascular disease.Analyses included data for 698 782 people (52 765 non-fatal or fatal vascular outcomes; 8.49 million person-years at risk) from 102 prospective studies. Adjusted HRs with diabetes were: 2.00 (95% CI 1.83-2.19) for coronary heart disease; 2.27 (1.95-2.65) for ischaemic stroke; 1.56 (1.19-2.05) for haemorrhagic stroke; 1.84 (1.59-2.13) for unclassified stroke; and 1.73 (1.51-1.98) for the aggregate of other vascular deaths. HRs did not change appreciably after further adjustment for lipid, inflammatory, or renal markers. HRs for coronary heart disease were higher in women than in men, at 40-59 years than at 70 years and older, and with fatal than with non-fatal disease. At an adult population-wide prevalence of 10%, diabetes was estimated to account for 11% (10-12%) of vascular deaths. Fasting blood glucose concentration was non-linearly related to vascular risk, with no significant associations between 3.90 mmol/L and 5.59 mmol/L. Compared with fasting blood glucose concentrations of 3.90-5.59 mmol/L, HRs for coronary heart disease were: 1.07 (0.97-1.18) for lower than 3.90 mmol/L; 1.11 (1.04-1.18) for 5.60-6.09 mmol/L; and 1.17 (1.08-1.26) for 6.10-6.99 mmol/L. In people without a history of diabetes, information about fasting blood glucose concentration or impaired fasting glucose status did not significantly improve metrics of vascular disease prediction when added to information about several conventional risk factors.Diabetes confers about a two-fold excess risk for a wide range of vascular diseases, independently from other conventional risk factors. In people without diabetes, fasting blood glucose concentration is modestly and non-linearly associated with risk of vascular disease.British Heart Foundation, UK Medical Research Council, and Pfizer.

    View details for Web of Science ID 000279652900018

    View details for PubMedID 20609967

  • Associations of Circulating Adiponectin with Measures of Vascular Function and Morphology JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Gustafsson, S., Lind, L., Soderberg, S., Ingelsson, E. 2010; 95 (6): 2927-2934

    Abstract

    Some previous studies have reported an association between circulating adiponectin and selected measures of vascular function and morphology, but most of these studies have been performed in small samples of patients with preexisting disease.We aimed to evaluate associations between circulating adiponectin and comprehensive measures of vascular function and morphology in a large sample of individuals from the community.We conducted a cross-sectional investigation of 981 70-yr-old participants (50% women) of the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS).Measures of outcome included vascular function [common carotid artery (CCA) distensibility, flow-mediated dilation, endothelium-dependent and endothelium-independent vasodilation using invasive methods] and vascular morphology [intima-media (IM) thickness, plaque presence, gray scale median (GSM) in the IM and plaques].In age- and sex-adjusted models, adiponectin was positively associated with IM-GSM, plaque GSM, CCA distensibility, endothelium-dependent and endothelium-independent vasodilation. In multivariable models (with additional adjustment for body mass index; systolic blood pressure; antihypertensive, antidiabetic, and lipid-lowering medication; fasting blood glucose; total cholesterol; high-density lipoprotein cholesterol; creatinine; and smoking), adiponectin remained positively associated with IM-GSM [beta = 2.06; 95% confidence interval (CI), 0.54, 3.58], plaque GSM (beta = 3.11; 95% CI, 0.36, 5.86), and CCA distensibility (beta = 0.04; 95% CI, 0.00, 0.07).Serum levels of adiponectin were positively associated with IM-GSM and plaque GSM (indicating lower fat content in the IM and plaques) and CCA distensibility (indicating higher wall elasticity), independent of potential confounders. Our results imply that adiponectin is associated with less arterial pathology.

    View details for DOI 10.1210/jc.2009-2685

    View details for Web of Science ID 000278444000052

    View details for PubMedID 20375206

  • Triglyceride-mediated pathways and coronary disease: collaborative analysis of 101 studies LANCET Sarwar, N., Sandhu, M. S., Ricketts, S. L., Butterworth, A. S., Di Angelantonio, E., Boelcholdt, S. M., Ouwehand, W., Watkins, H., Samani, N. J., Saleheen, D., Lawlor, D., Reilly, M. P., Hingorani, A. D., Talmud, P. J., Danesh, J., Braund, P. S., Hall, A. S., Samani, N. J., Thompson, J., Marz, W., Ouwehand, W., Sivapalaratnam, S., Soranzo, N., Trip, M., Lawlor, D. A., Casas, J. P., Ebrahim, S., Arsenault, B. J., Boekholdt, S. M., Khaw, K. T., Ricketts, S. L., Sandhu, M. S., Wareham, N. J., Grallert, H., Illig, T., Humphries, S. E., Talmud, P. J., Rader, D. J., He, J., Reilly, M. P., Clarke, R., Hamsten, R., Hopewell, J. C., Watkins, K., Saleheen, D., Frossard, P., Deloukas, P., Danesh, J., Ye, S., Simpson, J. A., Onat, A., Komurcu-Bayrak, E., Martinelli, N., Olivieri, O., Girelli, D., Hingorani, A. D., Kivimaki, M., Kumari, M., Aouizerat, B. E., Baum, L., CAMPOS, H., Chaaba, R., Chen, B. S., Cho, E. Y., Evans, D., Hill, J., Hsu, L. A., Hubacek, J. A., Lai, C. Q., Lee, J. H., Klos, K., Liu, H., Masana, L., Melegh, B., Nabika, T., RIBALTA, J., Ruiz-Narvaez, E., Thomas, G. N., Tomlinson, B., Szalai, C., Vaverkova, H., Yamada, Y., Yang, Y., Tipping, R. W., Ford, C. E., Pressel, S. L., Ballantyne, C., Brautbar, A., Knuiman, M., Winchup, P. H., Wannamethee, S. G., Morris, R. W., Kiechl, S., Willeit, J., Santer, P., Mayr, A., WALD, N., Ebrahim, S., Lawlor, D. A., Yarnell, J. W., Gallacher, J., Casiglia, E., Tikhonoff, V., Cushman, M., Psaty, B. M., Tracy, R. P., Tybjaerg-Hansen, A., Nordestgaard, B. G., Benn, M., Frikke-Schmidt, R., Giampaoli, S., Palmieri, L., Panico, S., Vanuzzo, D., Pilotto, L., Gomez de la Camara, A., Gomez-Gerique, J. A., Simons, L., McCallum, J., Friedlander, Y., Fowkes, F. G., Lee, A. J., Taylor, J., Guralnik, J. M., Phillips, C. L., WALLACE, W. R., Guralnik, J. M., Blazer, D. G., Khaw, K., Brenner, H., Raum, E., Mueller, H., Rothenbacher, D., Jansson, J. H., Wennberg, P., Nissinen, A., Donfrancesco, C., Giampaoli, S., Salomaa, V., Harald, K., Pencina, M. J., Vartiainen, E., D'Agostino, R. B., Vasan, R. S., Pencina, M. J., Bladbjerg, E. M., Jorgensen, T., Moller, L., Jespersen, J., Dankner, R., Chetrit, A., Lubin, F., Bjoerkelund, C., Lissner, L., Bengtsson, C., Cremer, P., Nagel, D., Rodriguez, B., Dekker, J. M., Nijpels, G., Stehouwer, C. D., Sato, S., Iso, H., Kitamura, A., NODA, H., Salonen, J. T., Nyssoenen, K., Tuimainen, T., Voutilainen, S., Meade, T. W., Cooper, J. A., Kuller, L. H., Grandits, G., GILLUM, R., Mussolino, M., Rimm, E., Hankinson, S., Manson, J. A., Pai, J. K., Bauer, K. A., Sato, S., Kitamura, A., Naito, Y., Iso, H., Amouyel, P., Arveiler, D., Evans, A., Ferrieres, J., Schulte, H., Assmann, G., Packard, C. J., Sattar, N., Westendorp, R. G., Buckley, B. M., Cantin, B., Lamarche, B., Despres, J., Dagenais, G. R., Barrett-Connor, E., Wingard, D. L., Bettencourt, R., Gudnason, V., Aspelund, T., Sigurdsson, G., Thorsson, B., Trevisan, M., Tunstall-Pedoe, H., Tavendale, R., Lowe, G. D., Woodward, M., Howard, B. V., Zhang, Y., Best, L., Umans, J., Ben-Shlomo, Y., Davey-Smith, G., Onat, A., Njolstad, I., Mathiesen, E. B., Lochen, M. L., Wilsgaard, T., Ingelsson, E., Lind, I., Giedraitis, V., Michaeelsson, K., Brunner, E., Shipley, M., RIDKER, P., Buring, J., Shepherd, J., Cobbe, S. M., Ford, I., Robertson, M., Marin Ibanez, A., Feskens, E. J., Kromhout, D., Walker, M., Watson, S., Collins, R., Di Angelantonio, E., Kaptoge, S., Perry, P. L., Sarwar, N., Thompson, A., Thompson, S. G., Walker, M., Watson, S., White, I. R., Wood, A. M., Danesh, J. 2010; 375 (9726): 1634-1639

    Abstract

    Whether triglyceride-mediated pathways are causally relevant to coronary heart disease is uncertain. We studied a genetic variant that regulates triglyceride concentration to help judge likelihood of causality.We assessed the -1131T>C (rs662799) promoter polymorphism of the apolipoprotein A5 (APOA5) gene in relation to triglyceride concentration, several other risk factors, and risk of coronary heart disease. We compared disease risk for genetically-raised triglyceride concentration (20,842 patients with coronary heart disease, 35,206 controls) with that recorded for equivalent differences in circulating triglyceride concentration in prospective studies (302 430 participants with no history of cardiovascular disease; 12,785 incident cases of coronary heart disease during 2.79 million person-years at risk). We analysed -1131T>C in 1795 people without a history of cardiovascular disease who had information about lipoprotein concentration and diameter obtained by nuclear magnetic resonance spectroscopy.The minor allele frequency of -1131T>C was 8% (95% CI 7-9). -1131T>C was not significantly associated with several non-lipid risk factors or LDL cholesterol, and it was modestly associated with lower HDL cholesterol (mean difference per C allele 3.5% [95% CI 2.6-4.6]; 0.053 mmol/L [0.039-0.068]), lower apolipoprotein AI (1.3% [0.3-2.3]; 0.023 g/L [0.005-0.041]), and higher apolipoprotein B (3.2% [1.3-5.1]; 0.027 g/L [0.011-0.043]). By contrast, for every C allele inherited, mean triglyceride concentration was 16.0% (95% CI 12.9-18.7), or 0.25 mmol/L (0.20-0.29), higher (p=4.4x10(-24)). The odds ratio for coronary heart disease was 1.18 (95% CI 1.11-1.26; p=2.6x10(-7)) per C allele, which was concordant with the hazard ratio of 1.10 (95% CI 1.08-1.12) per 16% higher triglyceride concentration recorded in prospective studies. -1131T>C was significantly associated with higher VLDL particle concentration (mean difference per C allele 12.2 nmol/L [95% CI 7.7-16.7]; p=9.3x10(-8)) and smaller HDL particle size (0.14 nm [0.08-0.20]; p=7.0x10(-5)), factors that could mediate the effects of triglyceride.These data are consistent with a causal association between triglyceride-mediated pathways and coronary heart disease.British Heart Foundation, UK Medical Research Council, Novartis.

    View details for Web of Science ID 000277734700029

    View details for PubMedID 20452521

  • Risk of thromboembolic diseases in men with prostate cancer: results from the population-based PCBaSe Sweden LANCET ONCOLOGY Van Hemelrijck, M., Adolfsson, J., Garmo, H., Bill-Axelson, A., Bratt, O., Ingelsson, E., Lambe, M., Stattin, P., Holmberg, L. 2010; 11 (5): 450-458

    Abstract

    Cancer is associated with an increased risk of thromboembolic diseases, but data on the association between prostate cancer and thromboembolic diseases are scarce. We investigated the risk of thromboembolic disease in men with prostate cancer who were receiving endocrine treatment, curative treatment, or surveillance.We analysed data from PCBaSe Sweden, a database based on the National Prostate Cancer Register, which covers over 96% of prostate cancer cases in Sweden. Standardised incidence ratios (SIR) of deep-venous thrombosis (DVT), pulmonary embolism, and arterial embolism were calculated by comparing observed and expected (using the total Swedish male population) occurrences of thromboembolic disease, taking into account age, calendar-time, number of thromboembolic diseases, and time since previous thromboembolic disease.Between Jan 1, 1997, and Dec 31, 2007, 30 642 men received primary endocrine therapy, 26 432 curative treatment, and 19 526 surveillance. 1881 developed a thromboembolic disease. For men on endocrine therapy, risks for DVT (SIR 2.48, 95% CI 2.25-2.73) and pulmonary embolism (1.95, 1.81-2.15) were increased, although this was not the case for arterial embolism (1.00, 0.82-1.20). Similar patterns were seen for men who received curative treatment (DVT: 1.73, 1.47-2.01; pulmonary embolism: 2.03, 1.79-2.30; arterial embolism: 0.95, 0.69-1.27) and men who were on surveillance (DVT: 1.27, 1.08-1.47; pulmonary embolism: 1.57, 1.38-1.78; arterial embolism: 1.08, 0.87-1.33). Increased risks for thromboembolic disease were maintained when patients were stratified by age and tumour stage.All men with prostate cancer were at higher risk of thromboembolic diseases, with the highest risk for those on endocrine therapy. Our results indicate that prostate cancer itself, prostate cancer treatments, and selection mechanisms all contribute to increased risk of thromboembolic disease. Thromboembolic disease should be a concern when managing patients with prostate cancer.Swedish Research Council, Stockholm Cancer Society, and Cancer Research UK.

    View details for DOI 10.1016/S1470-2045(10)70038-3

    View details for Web of Science ID 000278478800020

    View details for PubMedID 20395174

  • Adiponectin and cardiac geometry and function in elderly: results from two community-based cohort studies EUROPEAN JOURNAL OF ENDOCRINOLOGY Gustafsson, S., Lind, L., Zethelius, B., Venge, P., Flyvbjerg, A., Soderberg, S., Ingelsson, E. 2010; 162 (3): 543-550

    Abstract

    Several smaller studies have indicated that adiponectin might be associated with left ventricular (LV) mass and function, but community-based studies with adequate sample size and adjustment for potential confounders are lacking. Our objective was to investigate such associations in two large community-based studies of elderly.Cross-sectional.We evaluated cross-sectional relations between serum adiponectin and echocardiographic measures of cardiac geometry and function (LV mass index, LV relative wall thickness, LV end-diastolic diameter, left atrial diameter, ejection fraction, LV isovolumic relaxation time, and E/A ratio) in 954 70-year-old participants (50% women) of the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS), and in 427 71-year-old men from the Uppsala Longitudinal Study of Adult Men (ULSAM).In models adjusted for age, sex, body mass index, systolic blood pressure, antihypertensive treatment, antidiabetic treatment, lipid-lowering medication, fasting blood glucose, total cholesterol, high-density lipoprotein cholesterol, creatinine, and smoking, adiponectin was inversely associated with ejection fraction in men (beta, -1.62; 95% confidence interval (CI), -2.50, -0.75 in PIVUS; beta, -1.35; 95% CI, -2.41, -0.29 in ULSAM), but not in women. After additional adjustment for N-terminal pro-brain natriuretic peptide (NT-proBNP), the association between adiponectin and ejection fraction was attenuated (beta, -0.98; 95% CI, -1.86, -0.10 in PIVUS; beta, -0.75; 95% CI, -1.84, 0.35 in ULSAM).Serum adiponectin concentrations were associated with ejection fraction in men, and these associations were partially attenuated by NT-proBNP. Our results imply that adiponectin may be associated with systolic function through pathways that involve natriuretic peptides.

    View details for DOI 10.1530/EJE-09-1006

    View details for Web of Science ID 000276612100012

    View details for PubMedID 20008076

  • Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge NATURE GENETICS Saxena, R., Hivert, M., Langenberg, C., Tanaka, T., Pankow, J. S., Vollenweider, P., Lyssenko, V., Bouatia-Naji, N., Dupuis, J., Jackson, A. U., Kao, W. H., Li, M., Glazer, N. L., Manning, A. K., Luan, J., Stringham, H. M., Prokopenko, I., Johnson, T., Grarup, N., Boesgaard, T. W., Lecoeur, C., Shrader, P., O'Connell, J., Ingelsson, E., Couper, D. J., Rice, K., Song, K., Andreasen, C. H., Dina, C., Koettgen, A., Le Bacquer, O., Pattou, F., Taneera, J., Steinthorsdottir, V., Rybin, D., Ardlie, K., Sampson, M., Qi, L., van Hoek, M., Weedon, M. N., Aulchenko, Y. S., Voight, B. F., Grallert, H., Balkau, B., Bergman, R. N., Bielinski, S. J., Bonnefond, A., Bonnycastle, L. L., Borch-Johnsen, K., Boettcher, Y., Brunner, E., Buchanan, T. A., Bumpstead, S. J., Cavalcanti-Proenca, C., Charpentier, G., Chen, Y. I., Chines, P. S., Collins, F. S., Cornelis, M., Crawford, G. J., Delplanque, J., Doney, A., Egan, J. M., Erdos, M. R., Firmann, M., Forouhi, N. G., Fox, C. S., Goodarzi, M. O., Graessler, J., Hingorani, A., Isomaa, B., Jorgensen, T., Kivimaki, M., Kovacs, P., Krohn, K., Kumari, M., Lauritzen, T., Levy-Marchal, C., Mayor, V., McAteer, J. B., Meyre, D., Mitchell, B. D., Mohlke, K. L., Morken, M. A., Narisu, N., Palmer, C. N., Pakyz, R., Pascoe, L., Payne, F., Pearson, D., Rathmann, W., Sandbaek, A., Sayer, A. A., Scott, L. J., Sharp, S. J., Sijbrands, E., Singleton, A., Siscovick, D. S., Smith, N. L., Sparso, T., Swift, A. J., Syddall, H., Thorleifsson, G., Toenjes, A., Tuomi, T., Tuomilehto, J., Valle, T. T., Waeber, G., Walley, A., Waterworth, D. M., Zeggini, E., Zhao, J. H., Illig, T., Wichmann, H. E., Wilson, J. F., Van Duijn, C., Hu, F. B., Morris, A. D., Frayling, T. M., Hattersley, A. T., Thorsteinsdottir, U., Stefansson, K., Nilsson, P., Syvanen, A., Shuldiner, A. R., Walker, M., Bornstein, S. R., Schwarz, P., Williams, G. H., Nathan, D. M., Kuusisto, J., Laakso, M., Cooper, C., Marmot, M., Ferrucci, L., Mooser, V., Stumvoll, M., Loos, R. J., Altshuler, D., Psaty, B. M., Rotter, J. I., Boerwinkle, E., Hansen, T., Pedersen, O., Florez, J. C., McCarthy, M. I., Boehnke, M., Barroso, I., Sladek, R., Froguel, P., Meigs, J. B., Groop, L., Wareham, N. J., Watanabe, R. M. 2010; 42 (2): 142-U75

    Abstract

    Glucose levels 2 h after an oral glucose challenge are a clinical measure of glucose tolerance used in the diagnosis of type 2 diabetes. We report a meta-analysis of nine genome-wide association studies (n = 15,234 nondiabetic individuals) and a follow-up of 29 independent loci (n = 6,958-30,620). We identify variants at the GIPR locus associated with 2-h glucose level (rs10423928, beta (s.e.m.) = 0.09 (0.01) mmol/l per A allele, P = 2.0 x 10(-15)). The GIPR A-allele carriers also showed decreased insulin secretion (n = 22,492; insulinogenic index, P = 1.0 x 10(-17); ratio of insulin to glucose area under the curve, P = 1.3 x 10(-16)) and diminished incretin effect (n = 804; P = 4.3 x 10(-4)). We also identified variants at ADCY5 (rs2877716, P = 4.2 x 10(-16)), VPS13C (rs17271305, P = 4.1 x 10(-8)), GCKR (rs1260326, P = 7.1 x 10(-11)) and TCF7L2 (rs7903146, P = 4.2 x 10(-10)) associated with 2-h glucose. Of the three newly implicated loci (GIPR, ADCY5 and VPS13C), only ADCY5 was found to be associated with type 2 diabetes in collaborating studies (n = 35,869 cases, 89,798 controls, OR = 1.12, 95% CI 1.09-1.15, P = 4.8 x 10(-18)).

    View details for DOI 10.1038/ng.521

    View details for Web of Science ID 000274084400011

    View details for PubMedID 20081857

  • Conjoint Effects of Serum Calcium and Phosphate on Risk of Total, Cardiovascular, and Noncardiovascular Mortality in the Community ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY Larsson, T. E., Olauson, H., Hagstrom, E., Ingelsson, E., Arnlov, J., Lind, L., Sundstrom, J. 2010; 30 (2): 333-U379

    Abstract

    Hyperphosphatemia is a cardiovascular risk factor in patients with chronic kidney disease. Relations of circulating calcium (Ca) and phosphorus (Pi) to long-term mortality risk in the community require further investigation.Associations of serum Ca and Pi to mortality were evaluated in a community-based cohort of 2176 men (mean age, 50.1 years). During follow-up (median, 29.8 years), 1009 men died, and 466 of these deaths resulted from cardiovascular causes. In Cox proportional hazards models, serum Pi and [CaxPi] were independent predictors of total mortality (hazard ratio per SD, 1.06; 95% CI, 1.01-1.12; P=0.03; 1.07; 95% CI, 1.01-1.12; P=0.01) and cardiovascular mortality (1.10; 95% CI, 1.02-1.18; P=0.01; 1.10; 95% CI, 1.03-1.19; P=0.008). Serum Ca was associated with risk of total mortality (1.08; 95% CI, 1.01-1.16; P=0.02) and noncardiovascular mortality (1.10; 95% CI, 1.01-1.21; P=0.04). Results were consistent after multivariate adjustments in subsamples of individuals with estimated glomerular filtration rate >90 mL/min and low-to-normal serum Ca and Pi.Circulating Ca and Pi levels are associated with risks of total, cardiovascular, and noncardiovascular mortality in the community, and their conjoint effects are additive. Additional studies are warranted to evaluate whether Ca and Pi are modifiable risk factors in the general population.

    View details for DOI 10.1161/ATVBAHA.109.196675

    View details for Web of Science ID 000273799900033

    View details for PubMedID 19948843

  • Parity and risk of later-life maternal cardiovascular disease AMERICAN HEART JOURNAL Parikh, N. I., Cnattingius, S., Dickman, P. W., Mittleman, M. A., Ludvigsson, J. F., Ingelsson, E. 2010; 159 (2)

    Abstract

    Prior studies relating parity with maternal cardiovascular disease (CVD) have been performed in relatively small study samples without accounting for pregnancy-related complications associated with CVD.We examined the associations between parity and maternal risk of later-life CVD in a population-based cohort study using data from the Swedish population registers. Women born from 1932 to 1955 were followed until the occurrence of CVD, death, emigration, or end of follow-up (December 31, 2005). Cox proportional hazards models were used to estimate associations between parity and risk of CVD accounting for birth year, yearly income, education level, country of birth, hypertension (pregestational hypertension or gestational hypertension, with or without proteinuria), diabetes (type 1, type 2, or gestational diabetes), preterm birth, small for gestational age, and stillbirth.During a median follow-up time of 9.5 years (range 0-23.5), there were 65,204 CVD events in the full sample of women. Among 1,332,062 women, parity was associated with CVD in a J-shaped fashion, with 2 births representing the nadir of risk (global P value < .0001). Upon accounting for pregnancy-related complications in a subset of women with at least 1 childbirth after 1973 (n = 590,725), the association of parity with CVD was similar. Compared with women with 2 childbirths, the multivariable-adjusted hazard ratios (95% CIs) for women with 1 and >/=5 births were 1.09 (1.03-1.15) and 1.47 (1.37-1.57), respectively.In conclusion, parity was associated with incident maternal CVD in a J-shaped fashion, even after accounting for socioeconomic factors and pregnancy-related complications.

    View details for DOI 10.1016/j.ahj.2009.11.017

    View details for Web of Science ID 000274287200008

    View details for PubMedID 20152219

  • New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk NATURE GENETICS Dupuis, J., Langenberg, C., Prokopenko, I., Saxena, R., Soranzo, N., Jackson, A. U., Wheeler, E., Glazer, N. L., Bouatia-Naji, N., Gloyn, A. L., Lindgren, C. M., Magi, R., Morris, A. P., Randall, J., Johnson, T., Elliott, P., Rybin, D., Thorleifsson, G., Steinthorsdottir, V., Henneman, P., Grallert, H., Dehghan, A., Hottenga, J. J., Franklin, C. S., Navarro, P., Song, K., Goel, A., Perry, J. R., Egan, J. M., Lajunen, T., Grarup, N., Sparso, T., Doney, A., Voight, B. F., Stringham, H. M., Li, M., Kanoni, S., Shrader, P., Cavalcanti-Proenca, C., Kumari, M., Qi, L., Timpson, N. J., Gieger, C., Zabena, C., Rocheleau, G., Ingelsson, E., An, P., O'Connell, J., Luan, J., Elliott, A., McCarroll, S. A., Payne, F., Roccasecca, R. M., Pattou, F., Sethupathy, P., Ardlie, K., Ariyurek, Y., Balkau, B., Barter, P., Beilby, J. P., Ben-Shlomo, Y., Benediktsson, R., Bennett, A. J., Bergmann, S., Bochud, M., Boerwinkle, E., Bonnefond, A., Bonnycastle, L. L., Borch-Johnsen, K., Boettcher, Y., Brunner, E., Bumpstead, S. J., Charpentier, G., Chen, Y. I., Chines, P., Clarke, R., Coin, L. J., Cooper, M. N., Cornelis, M., Crawford, G., Crisponi, L., Day, I. N., de Geus, E. J., Delplanque, J., Dina, C., Erdos, M. R., Fedson, A. C., Fischer-Rosinsky, A., Forouhi, N. G., Fox, C. S., Frants, R., Franzosi, M. G., Galan, P., Goodarzi, M. O., Graessler, J., Groves, C. J., Grundy, S., Gwilliam, R., Gyllensten, U., Hadjadj, S., Hallmans, G., Hammond, N., Han, X., Hartikainen, A., Hassanali, N., Hayward, C., Heath, S. C., Hercberg, S., Herder, C., Hicks, A. A., Hillman, D. R., Hingorani, A. D., Hofman, A., Hui, J., Hung, J., Isomaa, B., Johnson, P. R., Jorgensen, T., Jula, A., Kaakinen, M., Kaprio, J., Kesaniemi, Y. A., Kivimaki, M., Knight, B., Koskinen, S., Kovacs, P., Kyvik, K. O., Lathrop, G. M., Lawlor, D. A., Le Bacquer, O., Lecoeur, C., Li, Y., Lyssenko, V., Mahley, R., Mangino, M., Manning, A. K., Teresa Martinez-Larrad, M., McAteer, J. B., McCulloch, L. J., McPherson, R., Meisinger, C., Melzer, D., Meyre, D., Mitchell, B. D., Morken, M. A., Mukherjee, S., Naitza, S., Narisu, N., Neville, M. J., Oostra, B. A., Orru, M., Pakyz, R., Palmer, C. N., Paolisso, G., Pattaro, C., Pearson, D., Peden, J. F., Pedersen, N. L., Perola, M., Pfeiffer, A. F., Pichler, I., Polasek, O., Posthuma, D., Potter, S. C., Pouta, A., Province, M. A., Psaty, B. M., Rathmann, W., Rayner, N. W., Rice, K., Ripatti, S., Rivadeneira, F., Roden, M., Rolandsson, O., Sandbaek, A., Sandhu, M., Sanna, S., Sayer, A. A., Scheet, P., Scott, L. J., Seedorf, U., Sharp, S. J., Shields, B., Sigurosson, G., Sijbrands, E. J., Silveira, A., Simpson, L., Singleton, A., Smith, N. L., Sovio, U., Swift, A., Syddall, H., Syvanen, A., Tanaka, T., Thorand, B., Tichet, J., Tonjes, A., Tuomi, T., Uitterlinden, A. G., Van Dijk, K. W., van Hoek, M., Varma, D., Visvikis-Siest, S., Vitart, V., Vogelzangs, N., Waeber, G., Wagner, P. J., Walley, A., Walters, G. B., Ward, K. L., Watkins, H., Weedon, M. N., Wild, S. H., Willemsen, G., Witteman, J. C., Yarnell, J. W., Zeggini, E., Zelenika, D., Zethelius, B., Zhai, G., Zhao, J. H., Zillikens, M. C., Borecki, I. B., Loos, R. J., Meneton, P., Magnusson, P. K., Nathan, D. M., Williams, G. H., Hattersley, A. T., Silander, K., Salomaa, V., Smith, G. D., Bornstein, S. R., Schwarz, P., Spranger, J., Karpe, F., Shuldiner, A. R., Cooper, C., Dedoussis, G. V., Serrano-Rios, M., Morris, A. D., Lind, L., Palmer, L. J., Hu, F. B., Franks, P. W., Ebrahim, S., Marmot, M., Kao, W. H., Pankow, J. S., Sampson, M. J., Kuusisto, J., Laakso, M., Hansen, T., Pedersen, O., Pramstaller, P. P., Wichmann, H. E., Illig, T., Rudan, I., Wright, A. F., Stumvoll, M., Campbell, H., Wilson, J. F., Bergman, R. N., Buchanan, T. A., Collins, F. S., Mohlke, K. L., Tuomilehto, J., Valle, T. T., Altshuler, D., Rotter, J. I., Siscovick, D. S., Penninx, B. W., Boomsma, D. I., Deloukas, P., Spector, T. D., Frayling, T. M., Ferrucci, L., Kong, A., Thorsteinsdottir, U., Stefansson, K., van Duijn, C. M., Aulchenko, Y. S., Cao, A., Scuteri, A., Schlessinger, D., Uda, M., Ruokonen, A., Jarvelin, M., Waterworth, D. M., Vollenweider, P., Peltonen, L., Mooser, V., Abecasis, G. R., Wareham, N. J., Sladek, R., Froguel, P., Watanabe, R. M., Meigs, J. B., Groop, L., Boehnke, M., McCarthy, M. I., Florez, J. C., Barroso, I. 2010; 42 (2): 105-U32

    Abstract

    Levels of circulating glucose are tightly regulated. To identify new loci influencing glycemic traits, we performed meta-analyses of 21 genome-wide association studies informative for fasting glucose, fasting insulin and indices of beta-cell function (HOMA-B) and insulin resistance (HOMA-IR) in up to 46,186 nondiabetic participants. Follow-up of 25 loci in up to 76,558 additional subjects identified 16 loci associated with fasting glucose and HOMA-B and two loci associated with fasting insulin and HOMA-IR. These include nine loci newly associated with fasting glucose (in or near ADCY5, MADD, ADRA2A, CRY2, FADS1, GLIS3, SLC2A2, PROX1 and C2CD4B) and one influencing fasting insulin and HOMA-IR (near IGF1). We also demonstrated association of ADCY5, PROX1, GCK, GCKR and DGKB-TMEM195 with type 2 diabetes. Within these loci, likely biological candidate genes influence signal transduction, cell proliferation, development, glucose-sensing and circadian regulation. Our results demonstrate that genetic studies of glycemic traits can identify type 2 diabetes risk loci, as well as loci containing gene variants that are associated with a modest elevation in glucose levels but are not associated with overt diabetes.

    View details for DOI 10.1038/ng.520

    View details for Web of Science ID 000274084400005

    View details for PubMedID 20081858

  • Impact of Body Mass Index and the Metabolic Syndrome on the Risk of Cardiovascular Disease and Death in Middle-Aged Men CIRCULATION Arnlov, J., Ingelsson, E., Sundstrom, J., Lind, L. 2010; 121 (2): 230-U88

    Abstract

    The purpose of this study was to investigate associations between combinations of body mass index (BMI) categories and metabolic syndrome (MetS) and the risk of cardiovascular disease and death in middle-aged men.At age 50 years, cardiovascular risk factors were assessed in 1758 participants without diabetes in the community-based Uppsala Longitudinal Study of Adult Men (ULSAM). According to BMI-MetS status, they were categorized as normal weight (BMI <25 kg/m(2)) without MetS (National Cholesterol Education Program criteria; n=891), normal weight with MetS (n=64), overweight (BMI 25 to 30 kg/m(2)) without MetS (n=582), overweight with MetS (n=125), obese (BMI >30 kg/m(2)) without MetS (n=30), or obese with MetS (n=66). During follow-up (median 30 years), 788 participants died, and 681 developed cardiovascular disease (composite of cardiovascular death or hospitalization for myocardial infarction, stroke, or heart failure). In Cox proportional-hazards models that adjusted for age, smoking, and low-density lipoprotein cholesterol, an increased risk for cardiovascular disease was observed in normal-weight participants with MetS (hazard ratio 1.63, 95% confidence interval 1.11 to 2.37), overweight participants without MetS (hazard ratio 1.52, 95% confidence interval 1.28 to 1.80), overweight participants with MetS (hazard ratio 1.74, 95% confidence interval 1.32 to 2.30), obese participants without MetS (hazard ratio 1.95, 95% confidence interval 1.14 to 3.34), and obese participants with MetS (hazard ratio 2.55, 95% confidence interval 1.81 to 3.58) compared with normal-weight individuals without MetS. These BMI-MetS categories significantly predicted total mortality rate in a similar pattern.Middle-aged men with MetS had increased risk for cardiovascular events and total death regardless of BMI status during more than 30 years of follow-up. In contrast to previous reports, overweight and obese individuals without MetS also had an increased risk. The present data refute the notion that overweight and obesity without MetS are benign conditions.

    View details for DOI 10.1161/CIRCULATIONAHA.109.887521

    View details for Web of Science ID 000273693500009

    View details for PubMedID 20038741

  • C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis LANCET Kaptoge, S., Di Angelantonio, E., Lowe, G., Pepys, M. B., Thompson, S. G., Collins, R., Danesh, J., Tipping, R. W., Ford, C. E., Pressel, S. L., Walldius, G., Jungner, I., Folsom, A. R., Chambless, L., Ballantyne, C. M., Panagiotakos, D., Pitsavos, C., Chrysohoou, C., Stefanadis, C., Knuiman, M. W., Goldbourt, U., Benderly, M., Tanne, D., Whincup, P., Wannamethee, S. G., Morris, R. W., Kiechl, S., Willeit, J., Mayr, A., Schett, G., WALD, N., Ebrahim, S., Lawlor, D., Yarnell, J., Gallacher, J., Casiglia, E., Tikhonoff, V., Nietert, P. J., Sutherland, S. E., Bachman, D. L., KEIL, J. E., Cushman, M., Psaty, B. M., Tracy, R., Tybjaerg-Hansen, A., Nordestgaard, B. G., Zacho, J., Frikke-Schmidt, R., Giampaoli, S., Palmieri, L., Panico, S., Vanuzzo, D., Pilotto, L., de la Camara, A. G., Gerique, J. A., Simons, L., McCallum, J., Friedlander, Y., Fowkes, F. G., Lee, A., Taylor, J., Guralnik, J. M., Phillips, C. L., Wallace, R. B., Blazer, D. G., Khaw, K., Brenner, H., Raum, E., Mueller, H., Rothenbacher, D., Jansson, J., Wennberg, P., Nissinen, A., Donfrancesco, C., Salomaa, V., Harald, K., Jousilahti, P., Vartiainen, E., Woodward, M., D'Agostino, R. B., Wolf, P. A., Vasan, R. S., Benjamin, E. J., Bladbjerg, E., Jorgensen, T., Moller, L., Jespersen, J., Dankner, R., Chetrit, A., Lubin, F., Rosengren, A., WILHELMSEN, L., Lappas, G., Eriksson, H., Bjorkelund, C., Lissner, L., Bengtsson, C., Cremer, P., Nagel, D., Tilvis, R. S., Strandberg, T. E., Kiyohara, Y., Arima, H., DOI, Y., Ninomiya, T., Rodriguez, B., Dekker, J., Nijpels, G., Stehouwer, C. D., Rimm, E., Pai, J. K., Sato, S., Iso, H., Kitamura, A., NODA, H., Salonen, J. T., Nyyssonen, K., Tuomainen, T., Laukkanen, J. A., Deeg, D. J., Bremmer, M. A., Meade, T. W., Cooper, J. A., Hedblad, B., Berglund, G., Engstrom, G., Verschuren, W. M., Blokstra, A., Shea, S., Doring, A., Koenig, W., Meisinger, C., Bueno-de-Mesquita, H. B., Kuller, L. H., Grandits, G., Selmer, R., Tverdal, A., Nystad, W., Gillum, R. F., Mussolino, M., Hankinson, S., Manson, J. E., Knottenbelt, C., Bauer, K. A., Davidson, K., Kirkland, S., Shaffer, J., Korin, M. R., Naito, Y., Holme, I., Nakagawa, H., Miura, K., Ducimetiere, P., Jouven, X., Luc, G., Crespo, J., Garcia-Palmieri, M. R., Amouyel, P., Arveiler, D., Evans, A., Ferrieres, J., Schulte, H., Assmann, G., Packard, C. J., Sattar, N., Westendorp, R. G., Buckley, B. M., Cantin, B., Lamarche, B., Despres, J., Dagenais, G. R., Barrett-Connor, E., Wingard, D. L., Bettencourt, R. R., Gudnason, V., Aspelund, T., Sigurdsson, G., Thorsson, B., Trevisan, M., Witteman, J., Kardys, I., Breteler, M. M., Hofman, A., Tunstall-Pedoe, H., Tavendale, R., Lowe, G., Howard, B. V., Zhang, Y., Bes, L., Umans, J., Ben-Shlomo, Y., Davey-Smith, G., Onat, A., Njostad, I., Mathiesen, E. B., Lohen, M., Wilsgaard, T., Ingelsson, E., Basu, S., Cederholm, T., Byberg, L., Gaziano, J. M., Stampfer, M., Ridker, P. M., Ulmer, H., Diem, G., Concin, H., Tosetto, A., Rodeghiero, F., Wassertheil-Smoller, S., Marmot, M., Clarke, R., Collins, R., FLETCHER, A., Brunner, E., Shipley, M., Buring, J., Shepherd, J., Cobbe, S., Ford, I., Robertson, M., He, Y., Ibanez, A. M., Feskens, E. J., Walker, M., Watson, S., Erqou, S., Kaptoge, S., Lewington, S., Pennells, L., Perry, P. L., Ray, K. K., Sarwar, N., Alexander, M., Thompson, A., Thompson, S. G., White, I. R., Wood, A. M., Danesh, J. 2010; 375 (9709): 132-140

    Abstract

    Associations of C-reactive protein (CRP) concentration with risk of major diseases can best be assessed by long-term prospective follow-up of large numbers of people. We assessed the associations of CRP concentration with risk of vascular and non-vascular outcomes under different circumstances.We meta-analysed individual records of 160 309 people without a history of vascular disease (ie, 1.31 million person-years at risk, 27 769 fatal or non-fatal disease outcomes) from 54 long-term prospective studies. Within-study regression analyses were adjusted for within-person variation in risk factor levels.Log(e) CRP concentration was linearly associated with several conventional risk factors and inflammatory markers, and nearly log-linearly with the risk of ischaemic vascular disease and non-vascular mortality. Risk ratios (RRs) for coronary heart disease per 1-SD higher log(e) CRP concentration (three-fold higher) were 1.63 (95% CI 1.51-1.76) when initially adjusted for age and sex only, and 1.37 (1.27-1.48) when adjusted further for conventional risk factors; 1.44 (1.32-1.57) and 1.27 (1.15-1.40) for ischaemic stroke; 1.71 (1.53-1.91) and 1.55 (1.37-1.76) for vascular mortality; and 1.55 (1.41-1.69) and 1.54 (1.40-1.68) for non-vascular mortality. RRs were largely unchanged after exclusion of smokers or initial follow-up. After further adjustment for fibrinogen, the corresponding RRs were 1.23 (1.07-1.42) for coronary heart disease; 1.32 (1.18-1.49) for ischaemic stroke; 1.34 (1.18-1.52) for vascular mortality; and 1.34 (1.20-1.50) for non-vascular mortality.CRP concentration has continuous associations with the risk of coronary heart disease, ischaemic stroke, vascular mortality, and death from several cancers and lung disease that are each of broadly similar size. The relevance of CRP to such a range of disorders is unclear. Associations with ischaemic vascular disease depend considerably on conventional risk factors and other markers of inflammation.British Heart Foundation, UK Medical Research Council, BUPA Foundation, and GlaxoSmithKline.

    View details for DOI 10.1016/S0140-6736(09)61717-7

    View details for Web of Science ID 000273664500030

    View details for PubMedID 20031199

  • Circulating retinol-binding protein 4, cardiovascular risk factors and prevalent cardiovascular disease in elderly ATHEROSCLEROSIS Ingelsson, E., Sundstrom, J., Melhus, H., Michaelsson, K., Berne, C., Vasan, R. S., Riserus, U., Blomhoff, R., Lind, L., Arnlov, J. 2009; 206 (1): 239-244

    Abstract

    Our aim was to examine relations of serum retinol-binding protein 4 (RBP4) to cardiovascular risk factors, and prevalent metabolic syndrome (MetS) and cardiovascular disease (CVD) in a large community-based sample of elderly.We evaluated cross-sectional relations of serum RBP4 to cardiovascular risk factors including anthropometrical measures, blood pressure, lipid measures, fasting glucose and insulin, body fat distribution including truncal fat by dual-energy x-ray absorptiometry (DXA), homeostasis model assessment insulin resistance (HOMA-IR) and prevalent MetS in one thousand eight 70-year old participants (50% women) of the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS), and in five hundred seven 82-year old men from Uppsala Longitudinal Study of Adult Men (ULSAM). In ULSAM, we also examined associations with prevalent CVD.RBP4 concentrations were positively correlated with serum triglycerides (r=0.30; P<0.0001 in both samples), whereas correlations with body mass index (BMI), waist circumference, sagittal abdominal diameter, total and truncal fat mass, total cholesterol, fasting glucose and HOMA-IR were weak. In multivariable-adjusted models, RBP-4 was associated with MetS (odds ratio (OR), 1.16 and 1.33; 95% confidence interval (CI), 0.99-1.37 and 1.05-1.67 per 1-standard deviation (SD) increase in PIVUS and ULSAM, respectively), and prior cerebrovascular disease (OR, 1.37; 95% CI, 1.00-1.88 per 1-SD increase in ULSAM), but not with prior myocardial infarction.In elderly, RBP4 concentrations were associated with MetS and its components in both sexes, and prior cerebrovascular disease in men. These findings are consistent with the hypothesis that circulating RBP4 could be a marker of metabolic complications and possibly also atherosclerosis and overt CVD.

    View details for DOI 10.1016/j.atherosclerosis.2009.02.029

    View details for Web of Science ID 000270002000038

    View details for PubMedID 19339013

  • Genetic Variants Associated With Cardiac Structure and Function A Meta-analysis and Replication of Genome-wide Association Data JAMA-JOURNAL OF THE AMERICAN MEDICAL ASSOCIATION Vasan, R. S., Glazer, N. L., Felix, J. F., Lieb, W., Wild, P. S., Felix, S. B., Watzinger, N., Larson, M. G., Smith, N. L., Dehghan, A., Grosshennig, A., Schillert, A., Teumer, A., Schmidt, R., Kathiresan, S., Lumley, T., Aulchenko, Y. S., Koenig, I. R., Zeller, T., Homuth, G., Struchalin, M., Aragam, J., Bis, J. C., Rivadeneira, F., Erdmann, J., Schnabel, R. B., Doerr, M., Zweiker, R., Lind, L., Rodeheffer, R. J., Greiser, K. H., Levy, D., Haritunians, T., Deckers, J. W., Stritzke, J., Lackner, K. J., Voelker, U., Ingelsson, E., O'Donnell, C. J., Heckbert, S. R., Stricker, B. H., Kullo, I., Ziegler, A., Reffelmann, T., Redfield, M. M., Werdan, K., Mitchell, G. F., Rice, K., Arnett, D. K., Hofman, A., Gottdiener, J. S., Uitterlinden, A. G., Meitinger, T., Blettner, M., Friedrich, N., Wang, T. J., Psaty, B. M., van Duijn, C. M., Wichmann, H., Munzel, T., Kroemer, H. K., J Benjamin, E., Rotter, J. I., Witteman, J. C., Schunkert, H., Schmidt, H., Voelzke, H., Blankenberg, S. 2009; 302 (2): 168-178

    Abstract

    Echocardiographic measures of left ventricular (LV) structure and function are heritable phenotypes of cardiovascular disease.To identify common genetic variants associated with cardiac structure and function by conducting a meta-analysis of genome-wide association data in 5 population-based cohort studies (stage 1) with replication (stage 2) in 2 other community-based samples.Within each of 5 community-based cohorts comprising the EchoGen consortium (stage 1; n = 12 612 individuals of European ancestry; 55% women, aged 26-95 years; examinations between 1978-2008), we estimated the association between approximately 2.5 million single-nucleotide polymorphisms (SNPs; imputed to the HapMap CEU panel) and echocardiographic traits. In stage 2, SNPs significantly associated with traits in stage 1 were tested for association in 2 other cohorts (n = 4094 people of European ancestry). Using a prespecified P value threshold of 5 x 10(-7) to indicate genome-wide significance, we performed an inverse variance-weighted fixed-effects meta-analysis of genome-wide association data from each cohort.Echocardiographic traits: LV mass, internal dimensions, wall thickness, systolic dysfunction, aortic root, and left atrial size.In stage 1, 16 genetic loci were associated with 5 echocardiographic traits: 1 each with LV internal dimensions and systolic dysfunction, 3 each with LV mass and wall thickness, and 8 with aortic root size. In stage 2, 5 loci replicated (6q22 locus associated with LV diastolic dimensions, explaining <1% of trait variance; 5q23, 12p12, 12q14, and 17p13 associated with aortic root size, explaining 1%-3% of trait variance).We identified 5 genetic loci harboring common variants that were associated with variation in LV diastolic dimensions and aortic root size, but such findings explained a very small proportion of variance. Further studies are required to replicate these findings, identify the causal variants at or near these loci, characterize their functional significance, and determine whether they are related to overt cardiovascular disease.

    View details for Web of Science ID 000267696500026

    View details for PubMedID 19584346

  • Breastfeeding in Infancy and Adult Cardiovascular Disease Risk Factors AMERICAN JOURNAL OF MEDICINE Parikh, N. I., Hwang, S., Ingelsson, E., Benjamin, E. J., Fox, C. S., Vasan, R. S., Murabito, J. M. 2009; 122 (7): 656-U80

    Abstract

    Public health recommendations advocate breastfeeding in infancy as a means to reduce obesity in later life. Several prior studies relating breastfeeding to cardiovascular risk factors have been limited by lack of adjustment for maternal and participant confounding factors.We ascertained breastfeeding history via questionnaire from mothers enrolled in the Framingham Offspring Study. In their young to middle-aged adult children enrolled in the Framingham Third Generation, we examined the relations between maternal breastfeeding history (yes, no) and cardiovascular risk factors, including body mass index (BMI), high-density lipoprotein (HDL) cholesterol, total cholesterol, triglycerides, fasting blood glucose, and systolic and diastolic blood pressure levels. We applied generalized estimating equations to account for sibling correlations and adjusted for maternal and participant lifestyle, education, and cardiovascular risk factors.In Third Generation participants (n = 962, mean age = 41 years, 54% were women), 26% of their mothers reported breastfeeding. Compared with non-breastfed individuals, breastfed adult participants had lower multivariable-adjusted BMI (26.1 kg/m2 vs 26.9 kg/m2, P = .04) and higher HDL cholesterol levels (HDL 56.6 mg/dL vs 53.7 mg/dL, P = .01). On additional adjustment for BMI, the association between breastfeeding and HDL cholesterol was attenuated (P = .09). Breastfeeding was not associated with total cholesterol, triglycerides, fasting blood glucose, systolic blood pressure, or diastolic blood pressure.Breastfeeding in infancy is inversely associated with adult BMI and positively associated with HDL cholesterol. Associations between breastfeeding and BMI may mediate the association between breastfeeding and HDL cholesterol.

    View details for DOI 10.1016/j.amjmed.2008.11.034

    View details for Web of Science ID 000267341000013

    View details for PubMedID 19559168

  • Plasma Parathyroid Hormone and the Risk of Cardiovascular Mortality in the Community CIRCULATION Hagstrom, E., Hellman, P., Larsson, T. E., Ingelsson, E., Berglund, L., Sundstrom, J., Melhus, H., Held, C., Lind, L., Michaelsson, K., Arnlov, J. 2009; 119 (21): 2765-U34

    Abstract

    Diseases with elevated levels of parathyroid hormone (PTH) such as primary and secondary hyperparathyroidism are associated with increased incidence of cardiovascular disease and death. However, data on the prospective association between circulating PTH levels and cardiovascular mortality in the community are lacking.The Uppsala Longitudinal Study of Adult Men (ULSAM), a community-based cohort of elderly men (mean age, 71 years; n=958), was used to investigate the association between plasma PTH and cardiovascular mortality. During follow-up (median, 9.7 years), 117 participants died of cardiovascular causes. In Cox proportional-hazards models adjusted for established cardiovascular risk factors (age, systolic blood pressure, diabetes, smoking, body mass index, total cholesterol, high-density lipoprotein cholesterol, antihypertensive treatment, lipid-lowering treatment, and history of cardiovascular disease), higher plasma PTH was associated with higher risk for cardiovascular mortality (hazard ratio for 1-SD increase in PTH, 1.38; 95% confidence interval, 1.18 to 1.60; P<0.001). This association remained essentially unaltered in participants without previous cardiovascular disease and in participants with normal PTH (<6.8 pmol/L) with no other signs of a disturbed mineral metabolism (normal serum calcium, 2.2 to 2.6 mmol/L; normal glomerular filtration rate, >50 mL . min(-1) . 1.73 m(-2) and without vitamin D deficiency, plasma 25-OH vitamin D >37.5 nmol/L). Interestingly, elevated plasma PTH (>5.27 pmol/L) accounted for 20% (95% confidence interval, 10 to 26) of the population-attributable risk proportion for cardiovascular mortality.Plasma PTH levels predict cardiovascular mortality in the community, even in individuals with PTH within the normal range. Further studies are warranted to evaluate the clinical implications of measuring PTH in cardiovascular risk prediction and to elucidate whether PTH is a modifiable risk factor.

    View details for DOI 10.1161/CIRCULATIONAHA.108.808733

    View details for Web of Science ID 000266521400004

    View details for PubMedID 19451355

  • Cardiac troponin-I and risk of heart failure: a community-based cohort study EUROPEAN HEART JOURNAL Sundstrom, J., Ingelsson, E., Berglund, L., Zethelius, B., Lind, L., Venge, P., Arnlov, J. 2009; 30 (7): 773-781

    Abstract

    We examined if circulating levels of cardiac troponin-I (cTnI) predict subsequent heart failure in the community.Using Cox proportional hazards models, we examined the risk of a first hospitalization for heart failure during a maximum of 11.4 years in a community-based sample of 1089 70-year-old men without heart failure, valvular disease, or electrocardiographic left ventricular hypertrophy. Adjusting for smoking, systolic blood pressure, antihypertensive medication use, diabetes, body mass index, serum cholesterol, and myocardial infarction before baseline or during follow-up, 0.01 microg/L higher cTnI conferred a hazard ratio (HR) of 1.26 (95% confidence interval 1.15-1.38) for subsequent heart failure. Persons with cTnI > or =0.03 microg/L had an HR of 5.25 (2.00-13.77) compared with persons with cTnI <0.01 microg/L. Adjusting additionally for serum NTproBNP attenuated the estimates somewhat [HR 1.22 (1.11-1.34) per 0.01 microg/L of cTnI]. Excluding persons with myocardial infarction before baseline and censoring at time of myocardial infarction during follow-up, 0.01 microg/L higher cTnI was associated with a multivariable-adjusted HR of 1.31 (1.16-1.47) for heart failure.In a community-based sample, a direct measure of cardiomyocyte damage, cTnI, indicated a substantially increased risk of heart failure, accounting for other risk factors. Studies investigating the clinical utility of measuring cTnI in asymptomatic individuals are warranted.

    View details for DOI 10.1093/eurheartj/ehp047

    View details for Web of Science ID 000264889600011

    View details for PubMedID 19264749

  • Circulating Retinol-Binding Protein 4 and Subclinical Cardiovascular Disease in the Elderly DIABETES CARE Ingelsson, E., Lind, L. 2009; 32 (4): 733-735

    Abstract

    We evaluated associations of serum retinol-binding protein 4 (RBP4) with subclinical cardiovascular disease (CVD).Subclinical CVD was measured with echocardiography, carotid artery ultrasound, brachial artery ultrasound, and invasive forearm endothelial vasoreactivity in 1,008 70-year-old participants (50% women) of the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study.In analyses adjusted for multiple CVD risk factors, we observed inverse associations of RBP4 with carotid artery intima-media (beta -0.39, 95% CI -0.55 to -0.22) and plaque (beta -0.33, 95% CI -0.60 to -0.05) echogenicity (gray scale median).Circulating RBP4 concentrations were inversely associated with intima-media and plaque echogenicity in carotid arteries. These findings imply that RBP4 could be involved in the development of atherosclerosis.

    View details for DOI 10.2337/dc08-1656

    View details for Web of Science ID 000264819800040

    View details for PubMedID 19114616

  • Associations of Serum Adiponectin with Skeletal Muscle Morphology and Insulin Sensitivity JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Ingelsson, E., Arnlov, J., Zethelius, B., Vasan, R. S., Flyvbjerg, A., Frystyk, J., Berne, C., Hanni, A., Lind, L., Sundstrom, J. 2009; 94 (3): 953-957

    Abstract

    Skeletal muscle morphology and function are strongly associated with insulin sensitivity.The objective of the study was to test the hypothesis that circulating adiponectin is associated with skeletal muscle morphology and that adiponectin mediates the relation of muscle morphology to insulin sensitivity.This was a cross-sectional investigation of 461 men aged 71 yr, participants of the community-based Uppsala Longitudinal Study of Adult Men study.Measures included serum adiponectin, insulin sensitivity measured with euglycemic insulin clamp technique, and capillary density and muscle fiber composition determined from vastus lateralis muscle biopsies.In multivariable linear regression models (adjusting for age, physical activity, fasting glucose, and pharmacological treatment for diabetes), serum adiponectin levels rose with increasing capillary density (beta, 0.30 per 50 capillaries per square millimeter increase; P = 0.041) and higher proportion of type I muscle fibers (beta, 0.27 per 10% increase; P = 0.036) but declined with a higher proportion of type IIb fibers (beta, -0.39 per 10% increase; P = 0.014). Using bootstrap methods to examine the potential role of adiponectin in associations between muscle morphology and insulin sensitivity and the associations of capillary density (beta difference, 0.041; 95% confidence interval 0.001, 0.085) and proportion of type IIb muscle fibers (beta difference, -0.053; 95% confidence interval -0.107, -0.002) with insulin sensitivity were significantly attenuated when adiponectin was included in the models.Circulating adiponectin concentrations were higher with increasing skeletal muscle capillary density and in individuals with higher proportion of slow oxidative muscle fibers. Furthermore, our results indicate that adiponectin could be a partial mediator of the relations between skeletal muscle morphology and insulin sensitivity.

    View details for DOI 10.1210/jc.2008-1772

    View details for Web of Science ID 000264020700037

    View details for PubMedID 19106268

  • Contemporary Trends in Dyslipidemia in the Framingham Heart Study ARCHIVES OF INTERNAL MEDICINE Ingelsson, E., Massaro, J. M., Sutherland, P., Jacques, P. F., Levy, D., D'Agostino, R. B., Vasan, R. S., Robins, S. J. 2009; 169 (3): 279-286

    Abstract

    Recent cross-sectional population studies in the United States have shown an increase in obesity, a decrease in cholesterol values, but no changes in levels of high-density lipoprotein cholesterol (HDL-C) or triglycerides (TG).Plasma total cholesterol, HDL-C, and TG levels, measured by the same methods at the 3 most recently completed examinations of Framingham Offspring Study participants (1991-2001), were compared in 1666 participants without prevalent cardiovascular disease, lipid therapy, or hormone replacement therapy (56% were men; mean ages of participants at the first and last examinations, 53 and 60 years, respectively). Changes in age- and multivariate-adjusted mean lipid levels were related to changes in body mass index (BMI).Over the 3 examinations, comparing the findings of the earliest examination with those of the most recent examination, the mean HDL-C level was significantly increased (multivariate-adjusted means, 44.4 and 46.6 mg/dL in men; 56.9 and 60.1 mg/dL in women; P value for trend, P <.001 in both sexes), whereas levels of TG were decreased (144.5 and 134.1 mg/dL in men; 122.3 and 112.3 mg/dL in women; P value for trend, P = .004 in men and <.001 in women). Over the same time interval, BMI (calculated as weight in kilograms divided by height in meters squared) increased (27.8 and 28.5 in men; 27.0 and 27.6 in women; P value for trend, P < .001 in men and P = .001 in women). There was an inverse relationship between changes in BMI and magnitude of dyslipidemia (ie, individuals with the least increase in BMI had the most favorable changes in levels of HDL-C and TG).During a 10-year period of recent examinations in the Framingham Heart Study there was a decrease in dyslipidemia with an increase in HDL-C levels and a decrease in levels of TG despite an overall increase in BMI.

    View details for Web of Science ID 000263202600010

    View details for PubMedID 19204219

  • Relative importance and conjoint effects of obesity and physical inactivity for the development of insulin resistance EUROPEAN JOURNAL OF CARDIOVASCULAR PREVENTION & REHABILITATION Ingelsson, E., Arnlov, J., Sundstrom, J., Riserus, U., Michaelsson, K., Byberg, L. 2009; 16 (1): 28-33

    Abstract

    Obesity and physical inactivity are related to the development of insulin resistance, but their relative importance and conjoint effects are unclear.We related body mass index (BMI) and self-reported leisure-time physical activity (PA) at the age of 50 years to insulin sensitivity measured with euglycemic insulin clamp technique and the presence of metabolic syndrome (MetS) at a subsequent examination, 20 years later, in 862 men free from diabetes and MetS at baseline.In a multivariable model including BMI, PA, homeostasis model assessment insulin resistance index, erythrocyte sedimentation rate, and all components of MetS at baseline, both BMI (beta, -0.19 mg/kg bodyweight/min per 1 kg/m; P<0.0001) and PA (adjusted least square means, 5.1, 5.2, 5.4, and 6.2 mg/kg bodyweight/min in individuals with sedentary, moderate, regular, and athletic PA, respectively; P=0.0035) were significant predictors of insulin sensitivity at age 70. When categorizing individuals into four groups by BMI and PA at baseline, insulin sensitivity at the age of 70 years decreased significantly over the following categories: multivariable-adjusted least square means, 5.8 (low BMI/high PA); 5.6 (low BMI/low PA); 5.1 (high BMI/high PA); and 4.6 (high BMI/low PA) mg/kg bodyweight/min, respectively; P value of less than 0.0001.In our community-based sample of middle-aged men, BMI and PA were independent predictors of insulin resistance after 20 years of follow-up. Our results imply that obesity and physical inactivity may increase insulin resistance and metabolic risk by partly independent pathways, and emphasize the importance of strategies that address both obesity and physical inactivity to achieve increased public health.

    View details for DOI 10.1097/HJR.0b013e32831gbf8c

    View details for Web of Science ID 000264365000005

    View details for PubMedID 19237994

  • The PPARGC1A Gly482Ser polymorphism is associated with left ventricular diastolic dysfunction in men BMC CARDIOVASCULAR DISORDERS Ingelsson, E., Bennet, L., Ridderstrale, M., Soderstrom, M., Rastam, L., Lindblad, U. 2008; 8

    Abstract

    The Gly482Ser polymorphism in peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PPARGC1A) has been demonstrated to be associated with diabetes, obesity and hypertension, all of which are important risk factors for left ventricular diastolic dysfunction.The PPARGC1A Gly482Ser polymorphism was genotyped in a community-based cohort of 499 men and 533 women, who also underwent an echocardiographic examination to determine their left ventricular diastolic function. The association between the polymorphism and the presence of diastolic dysfunction was evaluated using logistic regression models.The Ser allele of the PPARGC1A Gly482Ser polymorphism was significantly associated with a lower risk of diastolic dysfunction in men, but not in women. In a model adjusting for potential confounders (age, body mass index, leisure time physical activity, hypertension and diabetes) the results were still significant and substantial (odds ratio 0.13, 95% confidence interval 0.03-0.54, p for trend = 0.004). The results were consistent in a series of models, and they imply a multiplicative, protective effect of the Ser allele, with lower risk of diastolic dysfunction for each copy of the allele.The Ser allele of the PPARGC1A Gly482Ser polymorphism was associated with decreased risk of diastolic left ventricular dysfunction in men, but not in women, in our large community-based sample. It was associated with a substantially decreased risk, even after adjustment for potential confounders. The clinical importance of the findings has to be established in further studies.

    View details for DOI 10.1186/1471-2261-8-37

    View details for Web of Science ID 000266881200001

    View details for PubMedID 19077249

  • Effects of trans10cis12CLA-induced insulin resistance on retinol-binding protein 4 concentrations in abdominally obese men. Diabetes research and clinical practice Ingelsson, E., Risérus, U. 2008; 82 (3): e23-4

    Abstract

    In this randomized, placebo-controlled, double-blind study of 57 abdominally obese middle-aged men, conjugated linoleic acid (CLA) did not induce changes in retinol-binding protein 4 concentrations (RBP4), despite marked induced insulin resistance. Further, there were no associations between CLA-induced insulin resistance and changes in RBP4.

    View details for DOI 10.1016/j.diabres.2008.09.034

    View details for PubMedID 18980783

  • Serum cystatin C and the risk of Alzheimer disease in elderly men NEUROLOGY Sundelof, J., Arnlov, J., Ingelsson, E., Sundstrom, J., Basu, S., Zethelius, B., Larsson, A., Irizarry, M. C., Giedraitis, V., Ronnemaa, E., Degerman-Gunnarsson, M., Hyman, B. T., Basun, H., Kilander, L., Lannfelt, L. 2008; 71 (14): 1072-1079

    Abstract

    Multiple lines of research suggest that increased cystatin C activity in the brain protects against the development of Alzheimer disease (AD).Serum cystatin C levels were analyzed at two examinations of the Uppsala Longitudinal Study of Adult Men, a longitudinal, community-based study of elderly men (age 70 years, n = 1,153 and age 77 years, n = 761, a subset of the age 70 examination). Cox regressions were used to examine associations between serum cystatin C and incident AD. AD cases were identified by cognitive screening and comprehensive medical chart review in all subjects.On follow-up (median 11.3 years), 82 subjects developed AD. At age 70 years, lower cystatin C was associated with higher risk of AD independently of age, APOE4 genotype, glomerular filtration rate, diabetes, hypertension, stroke, cholesterol, body mass index, smoking, education level, and plasma amyloid-beta protein 40 and 42 levels (hazard ratio [HR] for lowest [<1.12 micromol/L] vs highest [>1.30 micromol/L] tertile = 2.67, 95% CI 1.22-5.83, p < 0.02). The results were similar at age 77 years (43 participants developed AD during follow-up). Furthermore, a 0.1-mumol/L decrease of cystatin C between ages 70 and 77 years was associated with a 29% higher risk of incident AD (HR 1.29, 95% CI 1.03-1.63, p < 0.03).Low levels of serum cystatin C precede clinically manifest Alzheimer disease (AD) in elderly men free of dementia at baseline and may be a marker of future risk of AD. These findings strengthen the evidence for a role for cystatin C in the development of clinical AD.

    View details for Web of Science ID 000259649100007

    View details for PubMedID 18824671

  • Aortic root diameter and longitudinal blood pressure tracking HYPERTENSION Ingelsson, E., Pencina, M. J., Levy, D., Aragam, J., Mitchell, G. F., Benjamin, E. J., Vasan, R. S. 2008; 52 (3): 473-477

    Abstract

    Proximal aortic diameter, including aortic root (AoR) diameter, has been inversely related to pulse pressure in cross-sectional studies. So, investigators have hypothesized that a smaller AoR diameter may increase the risk of developing hypertension. Prospective studies are lacking to test this hypothesis. We measured AoR diameter in 3195 Framingham Study participants (mean age: 49 years; 57% women; 8460 person-examinations) free from hypertension and previous cardiovascular disease who underwent routine echocardiography. We related AoR to hypertension incidence and blood pressure (BP) progression (increment of >or=1 category, as defined by the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure). On follow-up (median: 4 years), 1267 individuals (15%; 661 women) developed hypertension, and 2978 participants experienced BP progression (35%; 1588 women). In logistic regression models adjusted for age, sex, and height, AoR was positively associated with hypertension incidence (odds ratio: 1.15; 95% CI: 1.08 to 1.23) and BP progression (odds ratio: 1.09; 95% CI: 1.04 to 1.14) on follow-up. However, adjustment for other factors known to influence BP tracking (baseline systolic and diastolic BP, smoking, diabetes, and weight) rendered these relations statistically nonsignificant (odds ratio: 1.03; 95% CI: 0.96 to 1.11 for hypertension incidence; odds ratio: 1.03; 95% CI: 0.97 to 1.08 for BP progression). In our large community-based sample of nonhypertensive individuals, AoR diameter was not associated with hypertension incidence or BP progression prospectively after adjustment for potential confounders. Our prospective study does not support the notion that a smaller AoR predisposes to hypertension.

    View details for DOI 10.1161/HYPERTENSIONAHA.108.114157

    View details for Web of Science ID 000258609500009

    View details for PubMedID 18663156

  • Circulating ghrelin, leptin, and soluble leptin receptor concentrations and cardiometabolic risk factors in a community-based sample JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Ingelsson, E., Larson, M. G., Yin, X., Wang, T. J., Meigs, J. B., Lipinska, I., Benjamin, E. J., Keaney, J. F., Vasan, R. S. 2008; 93 (8): 3149-3157

    Abstract

    The conjoint effects and relative importance of ghrelin, leptin, and soluble leptin receptor (sOB-R), adipokines involved in appetite control and energy expenditure in mediating cardiometabolic risk, is unknown.The objective of the study was to study the cross-sectional relations of these adipokines to cardiometabolic risk factors in a community-based sample.We measured circulating ghrelin, leptin, and sOB-R in 362 participants (mean age 45 yr; 54% women) of the Framingham Third Generation Cohort.Body mass index, waist circumference (WC), blood pressure, lipid measures, fasting glucose, smoking, and metabolic syndrome (MetS) were measured.Ghrelin and leptin concentrations were significantly higher in women (P < 0.0001). In multivariable models, ghrelin was inversely associated with age and systolic blood pressure, and leptin was positively related to body mass index and WC. sOB-R was positively associated with age, total cholesterol, and fasting glucose and inversely with WC and high-density lipoprotein cholesterol. Ghrelin and sOB-R concentrations were significantly lower with number of MetS components (P for trend = 0.022 and < 0.0001, respectively), whereas leptin concentrations were higher (P for trend = 0.0001). Relating all adipokines to MetS conjointly, higher ghrelin and leptin concentrations were associated with decreased and increased odds of MetS (odds ratio 0.55, P < 0.0001; odds ratio 4.44, P = 0.0002, per 1 sd increase of respective log adipokine).In our community-based sample, we observed a sexual dimorphism in circulating ghrelin and leptin concentrations. Ghrelin, leptin, and sOB-R were associated with number of MetS components cross-sectionally, consistent with the hypothesis that these adipokines may have a central role in cardiometabolic risk.

    View details for DOI 10.1210/jc.2008-0207

    View details for Web of Science ID 000258240500031

    View details for PubMedID 18492761

  • Endothelium-dependent vasodilation in conduit and resistance vessels in relation to the endothelial nitric oxide synthase gene JOURNAL OF HUMAN HYPERTENSION Ingelsson, E., Syvaenen, A., Lind, L. 2008; 22 (8): 569-578

    Abstract

    Single nucleotide polymorphisms (SNPs) in the endothelial nitric oxide synthase (NOS3) gene have been related to endothelium-dependent vasodilation in either conduit or resistance arteries with divergent results. In the Prospective Study of the Vasculature in Uppsala Seniors study, 959 participants aged 70 (51% men) were evaluated with brachial artery ultrasound to assess flow-mediated vasodilation (FMD; reflecting conduit arteries) and invasive forearm technique with intrabrachial infusion of acetylcholine (endothelium-dependent vasodilation (EDV); reflecting resistance arteries). The 23 SNPs analysed by minisequencing captured >90% of the common genetic variation in the NOS3 gene, using the HapMap population of European ancestry (CEU) as reference. One SNP (Glu298Asp) was related to FMD (nominal P=0.0018), but not to EDV (nominal P=0.76) after adjustment for sex, systolic blood pressure, diastolic blood pressure, pulse rate, antihypertensive treatment, total cholesterol, high-density cholesterol, lipid-lowering medication, fasting glucose, antidiabetic medication, body mass index, current smoking and prior diagnosis of cardiovascular disease. This relation was significant in both men and women in sex-specific analyses, and remained significant after adjusting for multiple testing (empirical P=0.029 from bootstrap resampling). None of the constructed haplotypes were related to vasodilation. The Glu298Asp SNP in the NOS3 gene was related to endothelium-dependent vasodilation in conduit, but not in resistance arteries. This SNP has previously been related to coronary heart disease, and our findings should stimulate to replication and exploration of the association of NOS3 variation with endothelial function in other settings.

    View details for DOI 10.1038/jhh.2008.37

    View details for Web of Science ID 000257692100008

    View details for PubMedID 18463668

  • Insulin sensitivity measured with euglycemic clamp is independently associated with glomerular filtration rate in a community-based cohort DIABETES CARE Nerpin, E., Riserus, U., Ingelsson, E., Sundstrom, J., Jobs, M., Larsson, A., Basu, S., Arnlov, J. 2008; 31 (8): 1550-1555

    Abstract

    To investigate the association between insulin sensitivity and glomerular filtration rate (GFR) in the community, with prespecified subgroup analyses in normoglycemic individuals with normal GFR.We investigated the cross-sectional association between insulin sensitivity (M/I, assessed using euglycemic clamp) and cystatin C-based GFR in a community-based cohort of elderly men (Uppsala Longitudinal Study of Adult Men [ULSAM], n = 1,070). We also investigated whether insulin sensitivity predicted the incidence of renal dysfunction at a follow-up examination after 7 years.Insulin sensitivity was directly related to GFR (multivariable-adjusted regression coefficient for 1-unit higher M/I 1.19 [95% CI 0.69-1.68]; P < 0.001) after adjusting for age, glucometabolic variables (fasting plasma glucose, fasting plasma insulin, and 2-h glucose after an oral glucose tolerance test), cardiovascular risk factors (hypertension, dyslipidemia, and smoking), and lifestyle factors (BMI, physical activity, and consumption of tea, coffee, and alcohol). The positive multivariable-adjusted association between insulin sensitivity and GFR also remained statistically significant in participants with normal fasting plasma glucose, normal glucose tolerance, and normal GFR (n = 443; P < 0.02). In longitudinal analyses, higher insulin sensitivity at baseline was associated with lower risk of impaired renal function (GFR <50 ml/min per 1.73 m(2)) during follow-up independently of glucometabolic variables (multivariable-adjusted odds ratio for 1-unit higher of M/I 0.58 [95% CI 0.40-0.84]; P < 0.004).Our data suggest that impaired insulin sensitivity may be involved in the development of renal dysfunction at an early stage, before the onset of diabetes or prediabetic glucose elevations. Further studies are needed in order to establish causality.

    View details for DOI 10.2337/dc08-0369

    View details for Web of Science ID 000258482000017

    View details for PubMedID 18509205

  • Altered blood pressure progression in the community and its relation to clinical events ARCHIVES OF INTERNAL MEDICINE Ingelsson, E., Gona, P., Larson, M. G., Lloyd-Jones, D. M., Kannel, W. B., Vasan, R. S., Levy, D. 2008; 168 (13): 1450-1457

    Abstract

    Long-term blood pressure (BP) progression and its importance as a predictor of clinical outcome have not been well characterized across different periods.We evaluated period trends for 3 BP variables (long-term slope and mean BP during a baseline period of 16 years, and last baseline value) in an earlier period (1953-1971; n = 1644, mean participant age, 61 years) and in a later period (1971-1990; n = 1040, mean participant age, 58 years) in participants in the Framingham Heart Study who initially did not have hypertension. In addition, we explored the relation of BP to cardiovascular disease incidence and all-cause mortality in the 2 periods, each with up to 16 years of follow-up.Long-term slope, mean, and last baseline BP measurements were significantly lower in the later period (P < .001). Rates of hypertension control (BP <140/90 mm Hg) were higher in the later vs the earlier period (32% vs 23%; P < .001). Multivariate hazard ratios for the relation of BP to outcomes were generally lower in the later period; this was statistically significant for the relation of last baseline BP to all-cause mortality (hazard ratio for 1-SD increase in systolic BP, 1.02 vs 1.25, P = .03; hazard ratio for diastolic BP, 1.00 vs 1.23, P = .04).We found evidence that BP levels in the community have changed over time, coinciding with improved rates of hypertension control and attenuation of BP-mortality relations. These findings are consistent with the hypothesis that hypertension treatment in the community has altered the natural history of BP progression and its relation to clinical outcome.

    View details for Web of Science ID 000257595100012

    View details for PubMedID 18625926

  • Polymorphisms in the estrogen receptor alpha gene and endothelial function in resistance and conduit arteries in the elderly ATHEROSCLEROSIS Ingelsson, E., Syvanen, A., Lind, L. 2008; 199 (1): 162-171

    Abstract

    Prior evidence suggests an important role for estrogen in the regulation of endothelium-dependent vasodilation, but the mechanisms modulating this are not known. Our aim was to examine the relations of single nucleotide polymorphisms (SNPs) in the estrogen receptor alpha gene (ESR1) to endothelium-dependent vasodilation.We evaluated 959 70-year-old participants (51% men) of the Prospective Study of the Vasculature in Uppsala Seniors (PIVUS) study, using invasive forearm technique with intra-brachial infusion of acetylcholine (EDV; reflecting vasodilation in resistance arteries), and brachial artery ultrasound to assess flow-mediated vasodilation (FMD; reflecting vasodilation in conduit arteries). We genotyped 25 common SNPs in the ESR1 gene, and related them to EDV and FMD using multivariable linear regression, adjusting for sex and other potential confounders, such as major cardiovascular risk factors and medications. Haplotypes were estimated using the PHASE software.We observed an association between rs1709183 in the ESR1 gene and EDV (nominal P=0.0012), with a lower EDV in carriers of the minor allele (C). This association remained significant after adjustment for multiple testing (empirical P=0.031, obtained using bootstrap re-sampling). Two ESR1 haplotypes in the block containing rs1709183 were associated with EDV (individual haplotype P=0.0015 and P=0.025); the directions of effect were consistent with individual SNP analyses. FMD was not associated with any of the ESR1 SNPs.In our community-based study of elderly, a polymorphism in the estrogen receptor alpha gene was associated with endothelium-dependent vasodilation in resistance, but not conduit arteries. Our findings should stimulate further exploration in other settings.

    View details for DOI 10.1016/j.atherosclerosis.2007.10.025

    View details for Web of Science ID 000257837100023

    View details for PubMedID 18062975

  • Inflammatory markers in relation to insulin resistance and the metabolic syndrome EUROPEAN JOURNAL OF CLINICAL INVESTIGATION Ingelsson, E., Hulthe, J., Lind, L. 2008; 38 (7): 502-509

    Abstract

    Inflammation has repeatedly been demonstrated to be associated with the metabolic syndrome (MetS) and insulin resistance, but the relative importance of different aspects of the inflammatory process is largely unexplored.We measured circulating interleukins (IL-1alpha, IL-1beta, IL-2, IL-4, IL-6, IL-8, IL-10); other cytokines (tumour necrosis factor-alpha, interferon gamma and monocyte chemotactic protein-1), cell adhesion molecules [vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1, E-selectin, P-selectin, l-selectin], and systemic inflammation markers [C-reactive protein (CRP) and leukocyte count] in 943 70 year old participants (50% women) of the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study. We related these biomarkers to MetS and the homeostasis model assessment insulin resistance index (HOMA-IR).In a multivariable model including all inflammatory markers conjointly together with sex, log VCAM-1 [odds ratio (OR), 1.45; 95% confidence interval (CI), 1.22-1.72 per 1 SD increase; P < 0.0001], log E-selectin (OR, 1.33; 95% CI, 1.12-1.57 per 1SD increase; P = 0.001), and log CRP (OR, 1.41; 95% CI, 1.20-1.66 per 1-SD increase; P < 0.0001) were independently associated with MetS. These biomarkers were also independently associated with HOMA-IR.Among 17 inflammatory biomarkers, most of them previously not examined in relation to MetS and insulin resistance, VCAM-1, E-selectin and CRP demonstrated the strongest associations with MetS and insulin resistance in our community based sample of the elderly. The relative importance of these biomarkers in predicting the development of MetS, insulin resistance and cardiovascular disease needs to be further examined in a longitudinal setting.

    View details for DOI 10.1111/j.1365-2362.2008.01962.x

    View details for Web of Science ID 000256686000007

    View details for PubMedID 18489581

  • Use of multiple biomarkers to improve the prediction of death from cardiovascular causes NEW ENGLAND JOURNAL OF MEDICINE Zethelius, B., Berglund, L., Sundstrom, J., Ingelsson, E., Basu, S., Larsson, A., Venge, P., Arnlov, J. 2008; 358 (20): 2107-2116

    Abstract

    The incremental usefulness of adding multiple biomarkers from different disease pathways for predicting the risk of death from cardiovascular causes has not, to our knowledge, been evaluated among the elderly.We used data from the Uppsala Longitudinal Study of Adult Men (ULSAM), a community-based cohort of elderly men, to investigate whether a combination of biomarkers that reflect myocardial cell damage, left ventricular dysfunction, renal failure, and inflammation (troponin I, N-terminal pro-brain natriuretic peptide, cystatin C, and C-reactive protein, respectively) improved the risk stratification of a person beyond an assessment that was based on the established risk factors for cardiovascular disease (age, systolic blood pressure, use or nonuse of antihypertensive treatment, total cholesterol, high-density lipoprotein cholesterol, use or nonuse of lipid-lowering treatment, presence or absence of diabetes, smoking status, and body-mass index).During follow-up (median, 10.0 years), 315 of the 1135 participants in our study (mean age, 71 years at baseline) died; 136 deaths were the result of cardiovascular disease. In Cox proportional-hazards models adjusted for established risk factors, all of the biomarkers significantly predicted the risk of death from cardiovascular causes. The C statistic increased significantly when the four biomarkers were incorporated into a model with established risk factors, both in the whole cohort (C statistic with biomarkers vs. without biomarkers, 0.766 vs. 0.664; P<0.001) and in the group of 661 participants who did not have cardiovascular disease at baseline (0.748 vs. 0.688, P=0.03). The improvement in risk assessment remained strong when it was estimated by other statistical measures of model discrimination, calibration, and global fit.Our data suggest that in elderly men with or without prevalent cardiovascular disease, the simultaneous addition of several biomarkers of cardiovascular and renal abnormalities substantially improves the risk stratification for death from cardiovascular causes beyond that of a model that is based only on established risk factors.

    View details for Web of Science ID 000255849300004

    View details for PubMedID 18480203

  • Plasma beta amyloid and the risk of Alzheimer disease and dementia in elderly men ARCHIVES OF NEUROLOGY Sundelof, J., Giedraitis, V., Irizarry, M. C., Sundstrom, J., Ingelsson, E., Ronnemaa, E., Arnlov, J., Gunnarsson, M. D., Hyman, B. T., Basun, H., Ingelsson, M., Lannfelt, L., Kilander, L. 2008; 65 (2): 256-263

    Abstract

    Beta amyloid (Abeta) protein accumulates in the brains of individuals with Alzheimer disease (AD) and is detectable in cerebrospinal fluid and plasma.To examine plasma levels of Abeta peptides Abeta(40) and Abeta(42) as predictors of incident AD and other types of dementia.Prospective, population-based cohort study.The Uppsala Longitudinal Study of Adult Men.Plasma Abeta(40) and Abeta(42) levels were analyzed as predictors of incident AD in 1045 men at age 70 years and 680 men at age 77 years using Cox proportional hazards analyses. Alzheimer disease and other types of dementia were diagnosed by standardized screening, clinical evaluation, and medical record review.Hazard ratios of AD (primary outcome) and vascular dementia or other dementia (secondary outcomes) according to baseline levels of plasma Abeta(40) and Abeta(42).From the age of 77 years at baseline, 46 individuals developed AD at follow-up (median, 5.3 years). A low plasma Abeta(40) level at age 77 years was associated with higher incidence of AD. The multivariate-adjusted hazard ratio was 4.87 (95% confidence interval, 1.63-14.6) for the lowest Abeta(40) tertile compared with the highest tertile. On follow-up from age 70 years at baseline (median, 11.2 years), 82 individuals developed AD. Plasma Abeta(40) and Abeta(42) levels measured at age 70 years were not significantly associated with incident AD.Low plasma Abeta(40) levels predicted incident AD in elderly men independently of potential confounders. Plasma Abeta(42) levels were not significantly associated with AD incidence. The clinical value of Abeta measurement in plasma remains to be established in future studies.

    View details for Web of Science ID 000253058800015

    View details for PubMedID 18268197

  • Vitamin D deficiency and risk of cardiovascular disease CIRCULATION Wang, T. J., Pencina, M. J., Booth, S. L., Jacques, P. F., Ingelsson, E., Lanier, K., Benjamin, E. J., D'Agostino, R. B., Wolf, M., Vasan, R. S. 2008; 117 (4): 503-511

    Abstract

    Vitamin D receptors have a broad tissue distribution that includes vascular smooth muscle, endothelium, and cardiomyocytes. A growing body of evidence suggests that vitamin D deficiency may adversely affect the cardiovascular system, but data from longitudinal studies are lacking.We studied 1739 Framingham Offspring Study participants (mean age 59 years; 55% women; all white) without prior cardiovascular disease. Vitamin D status was assessed by measuring 25-dihydroxyvitamin D (25-OH D) levels. Prespecified thresholds were used to characterize varying degrees of 25-OH D deficiency (< 15 ng/mL, < 10 ng/mL). Multivariable Cox regression models were adjusted for conventional risk factors. Overall, 28% of individuals had levels < 15 ng/mL, and 9% had levels < 10 ng/mL. During a mean follow-up of 5.4 years, 120 individuals developed a first cardiovascular event. Individuals with 25-OH D < 15 ng/mL had a multivariable-adjusted hazard ratio of 1.62 (95% confidence interval 1.11 to 2.36, P=0.01) for incident cardiovascular events compared with those with 25-OH D > or = 15 ng/mL. This effect was evident in participants with hypertension (hazard ratio 2.13, 95% confidence interval 1.30 to 3.48) but not in those without hypertension (hazard ratio 1.04, 95% confidence interval 0.55 to 1.96). There was a graded increase in cardiovascular risk across categories of 25-OH D, with multivariable-adjusted hazard ratios of 1.53 (95% confidence interval 1.00 to 2.36) for levels 10 to < 15 ng/mL and 1.80 (95% confidence interval 1.05 to 3.08) for levels < 10 ng/mL (P for linear trend=0.01). Further adjustment for C-reactive protein, physical activity, or vitamin use did not affect the findings.Vitamin D deficiency is associated with incident cardiovascular disease. Further clinical and experimental studies may be warranted to determine whether correction of vitamin D deficiency could contribute to the prevention of cardiovascular disease.

    View details for DOI 10.1161/CIRCULATIONAHA.107.706127

    View details for Web of Science ID 000252711900008

    View details for PubMedID 18180395

  • Multimarker approach to evaluate the incidence of the metabolic syndrome and longitudinal changes in metabolic risk factors - The framingham offspring study CIRCULATION Ingelsson, E., Pencina, M. J., Tofler, G. H., Benjamin, E. J., Lanier, K. J., Jacques, P. F., Fox, C. S., Meigs, J. B., Levy, D., Larson, M. G., Selhub, J., D'Agostino, R. B., Wang, T. J., Vasan, R. S. 2007; 116 (9): 984-992

    Abstract

    The metabolic syndrome (MetS) is associated with increased cardiovascular risk. We evaluated the relative contributions of circulating biomarkers representing distinct biological pathways to the incidence of MetS and to longitudinal changes of its constituent risk factors.We measured 8 circulating biomarkers reflecting inflammation (C-reactive protein), hemostasis (plasminogen activator inhibitor-1, fibrinogen), neurohormonal activity (aldosterone, renin, B-type natriuretic peptide, N-terminal proatrial natriuretic peptide), and endothelial dysfunction (homocysteine) in 2292 Framingham Offspring Study participants (mean age, 57 years; 56% women). We related the biomarker panel to incidence of MetS on follow-up initially and then related biomarkers associated with incident MetS to longitudinal change in its components. On follow-up (mean, 2.9 years), 282 participants (of 1473 participants without prevalent MetS at baseline) developed MetS. After adjustment for clinical risk factors, the biomarker panel was associated with incident MetS (P=0.008). On backward elimination, plasminogen activator inhibitor-1 and aldosterone remained associated with incident MetS (multivariable-adjusted odds ratio per 1-SD increment log marker, 1.31 [P=0.004] and 1.21 [P=0.015], respectively). In multivariable analyses evaluating longitudinal change in MetS components (analyzed as continuous variables), plasminogen activator inhibitor-1 was significantly and positively associated with changes in fasting glucose, systolic blood pressure, and triglycerides (all P<0.05). Serum aldosterone was associated positively with change in systolic blood pressure (P=0.023) and inversely with change in high-density lipoprotein cholesterol (P=0.001).Higher circulating plasminogen activator inhibitor-1 and aldosterone levels are associated with the development of MetS and with longitudinal change of its components, suggesting that these biomarkers and related pathways play a key role in mediating metabolic risk.

    View details for DOI 10.1161/CIRCULATIONAHA.107.708537

    View details for Web of Science ID 000249031100004

    View details for PubMedID 17698726

  • Clinical utility of different lipid measures for prediction of coronary heart disease in men and women JAMA-JOURNAL OF THE AMERICAN MEDICAL ASSOCIATION Ingelsson, E., Schaefer, E. J., Contois, J. H., McNamara, J. R., Sullivan, L., Keyes, M. J., Pencina, M. J., Schoonmaker, C., Wilson, P. W., D'Agostino, R. B., Vasan, R. S. 2007; 298 (7): 776-785

    Abstract

    Evidence is conflicting regarding the performance of apolipoproteins vs traditional lipids for predicting coronary heart disease (CHD) risk.To compare performance of different lipid measures for CHD prediction using discrimination and calibration characteristics and reclassification of risk categories; to assess incremental utility of apolipoproteins over traditional lipids for CHD prediction.Population-based, prospective cohort from, Framingham, Massachusetts. We evaluated serum total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), non-HDL-C, apolipoprotein (apo) A-I and apo B, and 3 lipid ratios (total cholesterol:HDL-C, LDL-C:HDL-C, and apo B:apo A-I) in 3322 middle-aged white participants who attended the fourth offspring examination cycle (1987-1991) and were without cardiovascular disease. Fifty-three percent of the participants were women.Incidence of first CHD event (recognized or unrecognized myocardial infarction, angina pectoris, coronary insufficiency, or coronary heart disease death).After a median follow-up of 15.0 years, 291 participants, 198 of whom were men, developed CHD. In multivariate models adjusting for nonlipid risk factors, the apo B:apo A-I ratio predicted CHD (hazard ratio [HR] per SD increment, 1.39; 95% confidence interval [CI], 1.23-1.58 in men and HR, 1.40; 95% CI, 1.16-1.67 in women), but risk ratios were similar for total cholesterol:HDL-C (HR, 1.39; 95% CI, 1.22-1.58 in men and HR, 1.39; 95% CI, 1.17-1.66 in women) and for LDL-C:HDL-C (HR, 1.35; 95% CI, 1.18-1.54 in men and HR, 1.36; 95% CI 1.14-1.63 in women). In both sexes, models using the apo B:apo A-I ratio demonstrated performance characteristics comparable with but not better than that for other lipid ratios. The apo B:apo A-I ratio did not predict CHD risk in a model containing all components of the Framingham risk score including total cholesterol:HDL-C (P = .12 in men; P = .58 in women).In this large, population-based cohort, the overall performance of apo B:apo A-I ratio for prediction of CHD was comparable with that of traditional lipid ratios but did not offer incremental utility over total cholesterol:HDL-C. These data do not support measurement of apo B or apo A-I in clinical practice when total cholesterol and HDL-C measurements are available.

    View details for Web of Science ID 000248728800022

    View details for PubMedID 17699011

  • Burden and prognostic importance of subclinical cardiovascular disease in overweight and obese individuals CIRCULATION Ingelsson, E., Sullivan, L. M., Fox, C. S., Murabito, J. M., Benjamin, E. J., Polak, J. F., Meigs, J. B., Keyes, M. J., O'Donnell, C. J., Wang, T. J., D'Agostino, R. B., Wolf, P. A., Vasan, R. S. 2007; 116 (4): 375-384

    Abstract

    The burden and prognostic importance of subclinical cardiovascular disease (CVD) in obesity has not been investigated systematically.We examined prevalence of subclinical disease in 1938 Framingham Study participants (mean age, 57 years; 59% women) by use of 5 tests (electrocardiography, echocardiography, carotid ultrasound, ankle-brachial pressure, and urinary albumin excretion) and stratified by body mass index (BMI) (normal, < 25; overweight, 25 to < 30.0; obese, > or = 30 kg/m2) and waist circumference (WC) (increased, > or = 88 cm for women or > or = 102 cm for men). We investigated risk of overt CVD associated with adiposity according to presence versus absence of subclinical disease on any of the 5 tests. Prevalence of subclinical disease was higher in overweight (40.0%; adjusted odds ratio, 1.68) and obese individuals (49.7%; odds ratio, 2.82) compared with individuals with normal BMI (29.3%) and in individuals with increased WC (44.9%; odds ratio, 1.67) compared with normal WC (31.9%). On follow-up (mean 7.2 years), 139 participants had developed overt CVD. Presence of subclinical disease was associated with > 2-fold risk of overt CVD in all BMI and WC strata, with no evidence of an interaction between BMI and subclinical disease. CVD risk was attenuated in participants with obesity or increased WC but without subclinical disease (adjusted hazard ratio for obesity, 1.57; 95% confidence interval, 0.74 to 3.33; adjusted hazard ratio for increased WC, 1.22; 95% confidence interval, 0.69 to 2.15), compared with individuals with normal BMI or WC and no subclinical disease, respectively.In our community-based sample, overweight and obesity were associated with high prevalence of subclinical disease, which partly contributed to the increased risk of overt CVD in these strata.

    View details for DOI 10.1161/CIRCULATIONAHA.107.688788

    View details for Web of Science ID 000248244300004

    View details for PubMedID 17620505

  • Alcohol intake, insulin resistance, and abdominal obesity in elderly men OBESITY Riserus, U., Ingelsson, E. 2007; 15 (7): 1766-1773

    Abstract

    Moderate and high alcohol intake have been associated with decreased and increased risk of type 2 diabetes, respectively. Insulin resistance, insulin secretion, and abdominal obesity are major predictors of diabetes, but the links with alcohol intake remain contradictory because of limited data.In a population-based cohort of 807 men (age, 70 years), we studied whether alcohol intake was related to insulin sensitivity, measured with the gold standard technique (euglycemic clamp), insulin secretion (early insulin response), or adiposity [BMI, waist circumference (WC), waist-to-hip ratio]. Alcohol intake was self-reported (questionnaire) and was assessed from a validated 7-day dietary record. The cross-sectional associations were evaluated using multivariable linear regression, adjusting for smoking, education level, physical activity, dietary total energy intake, hypertension, diabetes, triglycerides, and cholesterol.In multivariable models, self-estimated alcohol intake was not related to insulin sensitivity, early insulin response, or BMI, but was positively related to WC (beta-coefficient, 0.77; 95% confidence interval, 0.15 to 1.39; p=0.02) and waist-to-hip ratio (0.006 [0.002-0.009], p=0.003). The association with WC and waist-to-hip ratio was most pronounced in men in the lowest tertile of BMI. The results using dietary records were similar.Evaluated in a large sample in elderly men, neither insulin sensitivity measured by clamp technique nor insulin secretion was significantly associated with alcohol intake. However, high alcohol intake was associated with abdominal obesity, which might explain the higher diabetes risk previously observed in high alcohol consumers.

    View details for Web of Science ID 000248242900018

    View details for PubMedID 17636095

  • Polymorphisms in the SCD1 gene: Associations with body fat distribution and insulin sensitivity OBESITY Warensjo, E., Ingelsson, E., Lundmark, P., Lannfelt, L., Syvanen, A., Vessby, B., Riserus, U. 2007; 15 (7): 1732-1740

    Abstract

    Obesity and insulin resistance are major risk factors for metabolic diseases and are influenced by lifestyle and genetics. The lipogenic enzyme, stearoyl-coenzyme A-desaturase (SCD), is related to obesity. Further, SCD1-deficent mice are protected against obesity and insulin resistance. We hypothesized that genetic polymorphisms in the SCD1 gene would be associated with obesity, insulin sensitivity, and estimated SCD activity in humans.The study population was 1143 elderly Swedish men taking part of a population-based cohort study, the Uppsala Longitudinal Study of Adult Men. Associations between single nucleotide polymorphisms and obesity (waist circumference and BMI), insulin sensitivity (assessed by hyperinsulinemic euglycemic clamp), and estimated SCD activity (fatty acid ratios) were analyzed using linear regression analysis.Subjects homozygous for the rare alleles of rs10883463, rs7849, rs2167444, and rs508384 had decreased BMI and waist circumference and improved insulin sensitivity. The rare allele of rs7849 demonstrated the strongest effect on both insulin sensitivity [regression coefficient (beta)=1.19, p=0.007] and waist circumference (beta=-4.4, p=0.028), corresponding to 23% higher insulin sensitivity and 4 cm less waist circumference.This study indicates that genetic variations in the SCD1 gene are associated with body fat distribution and insulin sensitivity, results that accord well with animal data. These results need confirmation in other populations with a larger sample size.

    View details for Web of Science ID 000248242900014

    View details for PubMedID 17636091

  • Low-grade albuminuria and the incidence of heart failure in a community-based cohort of elderly men EUROPEAN HEART JOURNAL Ingelsson, E., Sundstrom, J., Lind, L., Riserus, U., Larsson, A., Basu, S., Arnlov, J. 2007; 28 (14): 1739-1745

    Abstract

    To investigate associations of urinary albumin excretion rate (UAER) and heart failure (HF) incidence in a community-based sample.In a prospective study of 70-year-old men free from HF at baseline (n = 1106), UAER (from timed overnight samples) was analysed with established risk factors for HF [acute MI before baseline, acute MI during follow-up (modelled as a time-dependent covariate), hypertension, diabetes, left ventricular hypertrophy, smoking, body mass index, and glomerular filtration rate] and more recently described risk factors [high-sensitive C-reactive protein and insulin sensitivity (clamp glucose disposal rate)] as predictors of HF incidence. Ninety-eight participants developed HF during a median follow-up of 9.0 years. In Cox proportional hazards models adjusted for established and novel risk factors for HF, a 1 SD increase in log UAER increased the risk of HF in individuals without anti-hypertensive treatment (hazard ratio 1.49; 95% CI 1.13-1.98; P = 0.005). Furthermore, UAER remained an independent predictor of HF, also in participants without diabetes at baseline or myocardial infarction at baseline or during follow-up. There were no significant associations between UAER and HF incidence in individuals with anti-hypertensive treatment.Our observations support the notion that low-grade albuminuria is a marker for subclinical cardiovascular damage that predisposes to future HF in the community.

    View details for DOI 10.1093/eurheartj/ehm130

    View details for Web of Science ID 000248731000017

    View details for PubMedID 17495987

  • Heritability, linkage, and genetic associations of exercise treadmill test responses CIRCULATION Ingelsson, E., Larson, M. G., Vasan, R. S., O'Donnell, C. J., Yin, X., Hirschhorn, J. N., Newton-Cheh, C., Drake, J. A., Musone, S. L., Heard-Costa, N. L., Benjamin, E. J., Levy, D., Atwood, L. D., Wang, T. J., Kathiresan, S. 2007; 115 (23): 2917-2924

    Abstract

    The blood pressure (BP) and heart rate responses to exercise treadmill testing predict incidence of cardiovascular disease, but the genetic determinants of hemodynamic and chronotropic responses to exercise are largely unknown.We assessed systolic BP, diastolic BP, and heart rate during the second stage of the Bruce protocol and at the third minute of recovery in 2982 Framingham Offspring participants (mean age 43 years; 53% women). With use of residuals from multivariable models adjusted for clinical correlates of exercise treadmill testing responses, we estimated the heritability (variance-components methods), genetic linkage (multipoint quantitative trait analyses), and association with 235 single-nucleotide polymorphisms in 14 candidate genes selected a priori from neurohormonal pathways for their potential role in exercise treadmill testing responses. Heritability estimates for heart rate during exercise and during recovery were 0.32 and 0.34, respectively. Heritability estimates for BP variables during exercise were 0.25 and 0.26 (systolic and diastolic BP) and during recovery, 0.16 and 0.13 (systolic and diastolic BP), respectively. Suggestive linkage was found for systolic BP during recovery from exercise (locus 1q43-44, log-of-the-odds score 2.59) and diastolic BP during recovery from exercise (locus 4p15.3, log-of-the-odds score 2.37). Among 235 single-nucleotide polymorphisms tested for association with exercise treadmill testing responses, the minimum nominal probability value was 0.003, which was nonsignificant after adjustment for multiple testing.Hemodynamic and chronotropic responses to exercise are heritable and demonstrate suggestive linkage to select loci. Genetic mapping with newer approaches such as genome-wide association may yield novel insights into the physiological responses to exercise.

    View details for DOI 10.1161/CIRCULATIONAHA.106.683821

    View details for Web of Science ID 000247153300004

    View details for PubMedID 17548724

  • Prevalence and prognostic impact of subclinical cardiovascular disease in individuals with the metabolic syndrome and diabetes DIABETES Ingelsson, E., Sullivan, L. M., Murabito, J. M., Fox, C. S., Benjamin, E. J., Polak, J. F., Meigs, J. B., Keyes, M. J., O'Donnell, C. J., Wang, T. J., D'Agostino, R. B., Wolf, P. A., Vasan, R. S. 2007; 56 (6): 1718-1726

    Abstract

    Data are limited regarding prevalence and prognostic significance of subclinical cardiovascular disease (CVD) in individuals with metabolic syndrome (MetS). We investigated prevalence of subclinical CVD in 1,945 Framingham Offspring Study participants (mean age 58 years; 59% women) using electrocardiography, echocardiography, carotid ultrasound, ankle-brachial blood pressure, and urinary albumin excretion. We prospectively evaluated the incidence of CVD associated with MetS and diabetes according to presence versus absence of subclinical disease. Cross-sectionally, 51% of 581 participants with MetS had subclinical disease in at least one test, a frequency higher than individuals without MetS (multivariable-adjusted odds ratio 2.06 [95% CI 1.67-2.55]; P < 0.0001). On follow-up (mean 7.2 years), 139 individuals developed overt CVD, including 59 with MetS (10.2%). Overall, MetS was associated with increased CVD risk (multivariable-adjusted hazards ratio [HR] 1.61 [95% CI 1.12-2.33]). Participants with MetS and subclinical disease experienced increased risk of overt CVD (2.67 [1.62-4.41] compared with those without MetS, diabetes, or subclinical disease), whereas the association of MetS with CVD risk was attenuated in absence of subclinical disease (HR 1.59 [95% CI 0.87-2.90]). A similar attenuation of CVD risk in absence of subclinical disease was observed also for diabetes. Subclinical disease was a significant predictor of overt CVD in participants without MetS or diabetes (1.93 [1.15-3.24]). In our community-based sample, individuals with MetS have a high prevalence of subclinical atherosclerosis that likely contributes to the increased risk of overt CVD associated with the condition.

    View details for DOI 10.2337/db07-0078

    View details for Web of Science ID 000246930000029

    View details for PubMedID 17369522

  • Clinical correlates of circulating visfatin levels in a community-based sample DIABETES CARE Ingelsson, E., Larson, M. G., Fox, C. S., Yin, X., Wang, T. J., Lipinska, I., Pou, K. M., Hoffmann, U., Benjamin, E. J., Keaney, J. F., Vasan, R. S. 2007; 30 (5): 1278-1280

    View details for Web of Science ID 000246291400043

    View details for PubMedID 17261750

  • Sleep disturbances independently predict heart failure in overweight middle-aged men EUROPEAN JOURNAL OF HEART FAILURE Ingelsson, E., Lind, L., Arnlov, J., Sundstrom, J. 2007; 9 (2): 184-190

    Abstract

    Sleep disturbances are associated with manifest heart failure (HF). However, the relationship between sleep disturbances and incident HF has been less studied.To investigate self-reported sleep disturbances as predictors of HF in a longitudinal, community-based cohort of 2314 middle-aged men.Data on self-reported sleep disturbances, as well as established risk factors for HF were collected and analyzed using Cox proportional hazards analyses. In multivariable Cox proportional hazards models adjusted for established risk factors for HF, the presence at baseline of sleep disturbances (hazard ratio [HR], 1.52; 95% confidence interval [CI], 1.16-1.99; p=0.002) was an independent risk factor for HF. There was evidence of effect modification between BMI and sleep disturbances. In multivariable-adjusted models, sleep disturbance (HR, 1.58; 95% CI, 1.13-2.21; p=0.008) was an independent risk factor for HF in overweight participants (BMI>25), but not in normal-weight participants (BMI< or =25). All results remained similar in a sub-sample excluding all participants suffering from a myocardial infarction during follow-up.Self-reported sleep disturbances imply an increased risk of subsequent HF in overweight middle-aged men, via mechanisms largely independent of established risk factors for HF, including an interim myocardial infarction.

    View details for DOI 10.1016/j.ejheart.2006.05.012

    View details for Web of Science ID 000245310700015

    View details for PubMedID 16884954

  • Metabolic syndrome and risk for heart failure in middle-aged men HEART Ingelsson, E., Arnlov, J., Lind, L., Sundstrom, J. 2006; 92 (10): 1409-1413

    Abstract

    To explore metabolic syndrome as a possible risk factor for development of heart failure (HF).Community-based cohort study.Uppsala, Sweden.2314 50-year-old men free from HF, myocardial infarction and valvular disease at baseline were enrolled between 1970 and 1974 and were followed up until the age of 70. A modified National Cholesterol Education Program definition of metabolic syndrome was used with body mass index in the place of waist circumference.First hospitalisation for HF.In multivariable Cox proportional hazards models adjusted for established risk factors for HF (hypertension, diabetes, ECG left ventricular hypertrophy, smoking and body mass index), the presence at baseline of metabolic syndrome (hazard ratio 1.66, 95% confidence interval (CI) 1.02 to 2.70) was a predictor of subsequent HF. This relation was even stronger after adjustment for the presence of an acute myocardial infarction during follow up in addition to the other established risk factors for HF (hazard ratio 1.80, 95% CI 1.11 to 2.91).Metabolic syndrome was a significant predictor of HF, independent of established risk factors for HF including an interim myocardial infarction, during two decades of follow up in a community-based sample of middle-aged men. This implies that metabolic syndrome provides important risk information beyond that of established risk factors for HF.

    View details for DOI 10.1136/hrt.2006.089011

    View details for Web of Science ID 000240508300012

    View details for PubMedID 16717067

  • Socioeconomic factors as predictors of incident heart failure JOURNAL OF CARDIAC FAILURE Ingelsson, E., Lind, L., Arnlov, J., Sundstrom, J. 2006; 12 (7): 540-545

    Abstract

    Studies of socioeconomic factors as predictors of heart failure (HF) are few and have given opposing results. Further, it is unknown if these factors predict incident HF independently of myocardial infarction and other established risk factors for HF.In a community-based cohort of 2314 middle-age men free from HF, valvular disease, and previous myocardial infarction at baseline, socioeconomic factors were examined as predictors for HF using Cox proportional hazards analyses. In multivariable Cox proportional hazards models adjusted for established risk factors for HF (hypertension, diabetes, electrocardiographic left ventricular hypertrophy, smoking, serum cholesterol, and interim myocardial infarction), low occupational classification (hazard ratio [HR] 1.55, 95% confidence interval [CI] 1.03-2.35 for low vs. high occupational classification), low education level (HR 1.98, 95% CI 1.07-3.68 for elementary school vs. college exam) and being unmarried (HR 1.44, 95% CI 0.99-2.10 for being unmarried vs. being married) increased the risk of HF.High occupational classification and high education level decreased, and being unmarried increased, the risk of subsequent HF in middle-age men, via mechanisms largely independent of established risk factors for HF, including an interim myocardial infarction. Further studies are needed to understand the mechanisms behind these associations.

    View details for DOI 10.1016/j.cardfail.2006.05.010

    View details for Web of Science ID 000240708700008

    View details for PubMedID 16952788

  • Risk associated with the metabolic syndrome versus the sum of its individual components DIABETES CARE Sundstrom, J., Vallhagen, E., Riserus, U., Byberg, L., Zethelius, B., Berne, C., Lind, L., Ingelsson, E. 2006; 29 (7): 1673-1674

    View details for DOI 10.2337/dc06-0664

    View details for Web of Science ID 000238854000038

    View details for PubMedID 16801601

  • Diurnal blood pressure pattern and risk of congestive heart failure JAMA-JOURNAL OF THE AMERICAN MEDICAL ASSOCIATION Ingelsson, E., Bjorklund-Bodegard, K., Lind, L., Arnlov, J., Sundstrom, J. 2006; 295 (24): 2859-2866

    Abstract

    High blood pressure is the most important risk factor for congestive heart failure (CHF) at a population level, but the relationship of an altered diurnal blood pressure pattern to risk of subsequent CHF is unknown.To explore 24-hour ambulatory blood pressure characteristics as predictors of CHF incidence and to investigate whether altered diurnal blood pressure patterns confer any additional risk information beyond that provided by conventional office blood pressure measurements.Prospective, community-based, observational cohort in Uppsala, Sweden, including 951 elderly men free of CHF, valvular disease, and left ventricular hypertrophy at baseline between 1990 and 1995, followed up until the end of 2002. Twenty-four-hour ambulatory blood pressure monitoring was performed at baseline, and the blood pressure variables were analyzed as predictors of subsequent CHF.First hospitalization for CHF.Seventy men developed heart failure during follow-up, with an incidence rate of 8.6 per 1000 person-years at risk. In multivariable Cox proportional hazards models adjusted for antihypertensive treatment and established risk factors for CHF (myocardial infarction, diabetes, smoking, body mass index, and serum cholesterol level), a 1-SD (9-mm Hg) increase in nighttime ambulatory diastolic blood pressure (hazard ratio [HR], 1.26; 95% confidence interval [CI], 1.02-1.55) and the presence of "nondipping" blood pressure (night-day ambulatory blood pressure ratio > or =1; HR, 2.29; 95% CI, 1.16-4.52) were associated with an increased risk of CHF. After adjusting for office-measured systolic and diastolic blood pressures, nondipping blood pressure remained a significant predictor of CHF (HR, 2.21; 95% CI, 1.12-4.36 vs normal night-day pattern). Nighttime ambulatory diastolic blood pressure and nondipping blood pressure were also significant predictors of CHF after exclusion of all participants who had an acute myocardial infarction before baseline or during follow-up.Nighttime blood pressure appears to convey additional risk information about CHF beyond office-measured blood pressure and other established risk factors for CHF. The clinical value of this association remains to be established in future studies.

    View details for Web of Science ID 000238577700023

    View details for PubMedID 16804152

  • Novel metabolic risk factors for heart failure JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY Ingelsson, E., Arnlov, J., Sundstrom, J., Zethelius, B., Vessby, B., Lind, L. 2005; 46 (11): 2054-2060

    Abstract

    Our objectives were to explore novel metabolic risk factors for development of heart failure (HF).In the past decade, considerable knowledge has been gained from limited samples regarding novel risk factors for HF, but the importance of these in the general population is largely unexplored.In a community-based prospective study of 2,321 middle-aged men free from HF and valvular disease at baseline, variables reflecting glucose and lipid metabolism and variables involved in oxidative processes were compared with established risk factors for HF (prior myocardial infarction, hypertension, diabetes, electrocardiographic left ventricular hypertrophy, smoking, obesity, and serum cholesterol) using Cox proportional hazards analyses.During a median follow-up time of 29 years, 259 subjects developed HF. In a multivariable Cox proportional hazards backward stepwise model, a 1-SD increase of fasting proinsulin (hazard ratio [HR] 1.38, 95% confidence interval [CI] 1.15 to 1.66) and apolipoprotein B/A-I-ratio (HR 1.27, 95% CI 1.09 to 1.48) increased the risk, whereas a 1-SD increase in serum beta-carotene (HR 0.79, 95% CI 0.66 to 0.94) decreased the risk of HF. These variables also remained significant when adjusting for acute myocardial infarction during follow-up.Novel variables reflecting insulin resistance and dyslipidemia, together with a low beta-carotene level, were found to predict HF independently of established risk factors. If confirmed, our observations could have large clinical implications, as they may offer new approaches in the prevention of HF.

    View details for DOI 10.1016/j.jacc.2005.07.059

    View details for Web of Science ID 000233755700016

    View details for PubMedID 16325042

  • The validity of a diagnosis of heart failure in a hospital discharge register EUROPEAN JOURNAL OF HEART FAILURE Ingelsson, E., Arnlov, J., Sundstrom, J., Lind, L. 2005; 7 (5): 787-791

    Abstract

    The accuracy of a diagnosis of heart failure (HF) in hospital discharge registers is largely unknown. We aimed to determine the validity of such a diagnosis in the Swedish hospital discharge register.In a population-based study of 2322 middle-aged men (the ULSAM study), 321 participants were diagnosed with HF according to the Swedish hospital discharge register, during a median follow-up time of 29 years. A review board examined the validity of the diagnosis according to the European Society of Cardiology definition of HF. Eighty-two percent of the possible cases were classified as having definite HF. An echocardiographic examination increased the validity to 88%. For patients treated at an internal medicine or cardiology clinic the validity was 86% and 91%, respectively. If HF was the primary diagnosis, the validity was 95%, irrespective of clinic type.The HF diagnosis in the Swedish hospital discharge register appears slightly less precise than for acute myocardial infarction and stroke. For population-based research, only those with a primary diagnosis of HF in the hospital discharge register should be regarded as definite HF cases, or alternatively the cases should be validated individually.

    View details for DOI 10.1016/j.ejheart.2004.12.007

    View details for Web of Science ID 000236408500013

    View details for PubMedID 15916919

  • Insulin resistance and risk of congestive heart failure JAMA-JOURNAL OF THE AMERICAN MEDICAL ASSOCIATION Ingelsson, E., Sundstrom, J., Arnlov, J., Zethelius, B., Lind, L. 2005; 294 (3): 334-341

    Abstract

    Diabetes and obesity are established risk factors for congestive heart failure (CHF) and are both associated with insulin resistance.To explore if insulin resistance may predict CHF and may provide the link between obesity and CHF.The Uppsala Longitudinal Study of Adult Men, a prospective, community-based, observational cohort in Uppsala, Sweden. We investigated 1187 elderly (>or=70 years) men free from CHF and valvular disease at baseline between 1990 and 1995, with follow-up until the end of 2002. Variables reflecting insulin sensitivity (including euglycemic insulin clamp glucose disposal rate) and obesity were analyzed together with established risk factors (prior myocardial infarction, hypertension, diabetes, electrocardiographic left ventricular hypertrophy, smoking, and serum cholesterol level) as predictors of subsequent incidence of CHF, using Cox proportional hazards analyses.First hospitalization for heart failure.One hundred four men developed CHF during a median follow-up of 8.9 (range, 0.01-11.4) years. In multivariable Cox proportional hazards models adjusted for established risk factors for CHF, increased risk of CHF was associated with a 1-SD increase in the 2-hour glucose value of an oral glucose tolerance test (hazard ratio [HR], 1.44; 95% confidence interval [CI], 1.08-1.93), fasting serum proinsulin level (HR, 1.29; 95% CI, 1.02-1.64), body mass index (HR, 1.35; 95% CI, 1.11-1.65), and waist circumference (HR, 1.36; 95% CI, 1.10-1.69), whereas a 1-SD increase in clamp glucose disposal rate decreased the risk (HR, 0.66; 95% CI, 0.51-0.86). When adding clamp glucose disposal rate to these models as a covariate, the obesity variables were no longer significant predictors of subsequent CHF.Insulin resistance predicted CHF incidence independently of established risk factors including diabetes in our large community-based sample of elderly men. The previously described association between obesity and subsequent CHF may be mediated largely by insulin resistance.

    View details for Web of Science ID 000230609100018

    View details for PubMedID 16030278

  • Inflammation, as measured by the erythrocyte sedimentation rate, is an independent predictor for the development of heart failure JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY Ingelsson, E., Arnlov, J., Sundstrom, J., Lind, L. 2005; 45 (11): 1802-1806

    Abstract

    Our objective was to explore inflammation, measured as erythrocyte sedimentation rate (ESR), as a predictor for the development of heart failure (HF).In recent years, evidence of the importance of inflammation in the pathophysiology of HF has emerged, and various inflammatory markers have been found to predict future HF. Erythrocyte sedimentation rate is an inexpensive and easily accessible marker of systemic inflammation, but to this date it is unknown whether ESR predicts subsequent HF.In a community-based prospective study of 2,314 middle-aged men free from HF, myocardial infarction, and valvular disease at baseline, ESR was analyzed in multivariable models together with established risk factors for HF (hypertension, diabetes, electrocardiographic left ventricular hypertrophy, smoking, obesity, and serum cholesterol) and hematocrit.A total of 282 men developed HF during a median follow-up time of 30 years. In Cox proportional hazards analyses, ESR was an independent predictor of HF (hazard ratio 1.46 for highest quartile vs. the lowest, 95% confidence interval 1.04 to 2.06). This observation remained significant when also adjusting for interim myocardial infarction during follow-up.Erythrocyte sedimentation rate was a significant predictor of HF, independent of established risk factors for HF, and interim myocardial infarction after three decades of follow-up in a population-based sample of middle-aged men. Our findings indicate that inflammation occurs early in the process leading to HF and that ESR could be used to evaluate this process.

    View details for DOI 10.1016/j.jacc.2005.002.066

    View details for Web of Science ID 000229593000012

    View details for PubMedID 15936609

  • Myocardial performance index, a Doppler-derived index of global left ventricular function, predicts congestive heart failure in elderly men EUROPEAN HEART JOURNAL Arnlov, J., Ingelsson, E., Riserus, U., Andren, B., Lind, L. 2004; 25 (24): 2220-2225

    Abstract

    There is limited data on echocardiographic and Doppler indices of cardiac function as predictors for congestive heart failure (CHF) in the general population. Myocardial performance index (MPI, also denoted TEI-Doppler index) reflects both left ventricular (LV) systolic and diastolic function.We compared eight different echocardiographic and Doppler indices of cardiac function as predictors of CHF using a population-based cohort of 552 seventy-year-old men without CHF and significant valve disease at baseline (median follow-up time 8.2 years). In a stepwise multivariable Cox proportional-hazard analysis including the different indices of cardiac function, high MPI (above the 90th percentile of MPI [> or =0.91]), abnormal LV wall motion score index and a pseudo-normalized/restrictive E/A-ratio pattern independently predicted future CHF morbidity. After adding traditional CHF risk factors (age, previous myocardial infarction, hypertension, diabetes mellitus, hyperlipidaemia, smoking, LV hypertrophy and body mass index) to the above model, only a high MPI remained a significant predictor (hazard ratio 4.72, 95% CI 1.75-12.76, p=0.002).MPI provides important prognostic information for the risk of future CHF, beyond other measurements of cardiac function and traditional heart failure risk factors in elderly men. MPI seems to be a clinically relevant indicator of cardiac function and may prove to be a valuable tool in assessing the risk of future CHF.

    View details for DOI 10.1016/j.ehj.2004.10.021

    View details for Web of Science ID 000225936600009

    View details for PubMedID 15589639