1251
|
Zaitlen N, Pasaniuc B, Patterson N, Pollack S, Voight B, Groop L, Altshuler D, Henderson BE, Kolonel LN, Le Marchand L, Waters K, Haiman CA, Stranger BE, Dermitzakis ET, Kraft P, Price AL. Analysis of case-control association studies with known risk variants. ACTA ACUST UNITED AC 2012; 28:1729-37. [PMID: 22556366 DOI: 10.1093/bioinformatics/bts259] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MOTIVATION The question of how to best use information from known associated variants when conducting disease association studies has yet to be answered. Some studies compute a marginal P-value for each Several Nucleotide Polymorphisms independently, ignoring previously discovered variants. Other studies include known variants as covariates in logistic regression, but a weakness of this standard conditioning strategy is that it does not account for disease prevalence and non-random ascertainment, which can induce a correlation structure between candidate variants and known associated variants even if the variants lie on different chromosomes. Here, we propose a new conditioning approach, which is based in part on the classical technique of liability threshold modeling. Roughly, this method estimates model parameters for each known variant while accounting for the published disease prevalence from the epidemiological literature. RESULTS We show via simulation and application to empirical datasets that our approach outperforms both the no conditioning strategy and the standard conditioning strategy, with a properly controlled false-positive rate. Furthermore, in multiple data sets involving diseases of low prevalence, standard conditioning produces a severe drop in test statistics whereas our approach generally performs as well or better than no conditioning. Our approach may substantially improve disease gene discovery for diseases with many known risk variants. AVAILABILITY LTSOFT software is available online http://www.hsph.harvard.edu/faculty/alkes-price/software/.
Collapse
Affiliation(s)
- Noah Zaitlen
- Department of Epidemiology, Department of Biostatistics, Harvard School of Public Health, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1252
|
Pausch H, Wang X, Jung S, Krogmeier D, Edel C, Emmerling R, Götz KU, Fries R. Identification of QTL for UV-protective eye area pigmentation in cattle by progeny phenotyping and genome-wide association analysis. PLoS One 2012; 7:e36346. [PMID: 22567150 PMCID: PMC3342244 DOI: 10.1371/journal.pone.0036346] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 04/01/2012] [Indexed: 02/07/2023] Open
Abstract
Pigmentation patterns allow for the differentiation of cattle breeds. A dominantly inherited white head is characteristic for animals of the Fleckvieh (FV) breed. However, a minority of the FV animals exhibits peculiar pigmentation surrounding the eyes (ambilateral circumocular pigmentation, ACOP). In areas where animals are exposed to increased solar ultraviolet radiation, ACOP is associated with a reduced susceptibility to bovine ocular squamous cell carcinoma (BOSCC, eye cancer). Eye cancer is the most prevalent malignant tumour affecting cattle. Selection for animals with ACOP rapidly reduces the incidence of BOSCC. To identify quantitative trait loci (QTL) underlying ACOP, we performed a genome-wide association study using 658,385 single nucleotide polymorphisms (SNPs). The study population consisted of 3579 bulls of the FV breed with a total of 320,186 progeny with phenotypes for ACOP. The proportion of progeny with ACOP was used as a quantitative trait with high heritability (h2 = 0.79). A variance component based approach to account for population stratification uncovered twelve QTL regions on seven chromosomes. The identified QTL point to MCM6, PAX3, ERBB3, KITLG, LEF1, DKK2, KIT, CRIM1, ATRN, GSDMC, MITF and NBEAL2 as underlying genes for eye area pigmentation in cattle. The twelve QTL regions explain 44.96% of the phenotypic variance of the proportion of daughters with ACOP. The chromosomes harbouring significantly associated SNPs account for 54.13% of the phenotypic variance, while another 19.51% of the phenotypic variance is attributable to chromosomes without identified QTL. Thus, the missing heritability amounts to 7% only. Our results support a polygenic inheritance pattern of ACOP in cattle and provide the basis for efficient genomic selection of animals that are less susceptible to serious eye diseases.
Collapse
Affiliation(s)
- Hubert Pausch
- Lehrstuhl fuer Tierzucht, Technische Universitaet Muenchen, Freising, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
1253
|
|
1254
|
Evidence-based psychiatric genetics, AKA the false dichotomy between common and rare variant hypotheses. Mol Psychiatry 2012; 17:474-85. [PMID: 21670730 DOI: 10.1038/mp.2011.65] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this article, we review some of the data that contribute to our understanding of the genetic architecture of psychiatric disorders. These include results from evolutionary modelling (hence no data), the observed recurrence risk to relatives and data from molecular markers. We briefly discuss the common-disease common-variant hypothesis, the success (or otherwise) of genome-wide association studies, the evidence for polygenic variance and the likely success of exome and whole-genome sequencing studies. We conclude that the perceived dichotomy between 'common' and 'rare' variants is not only false, but unhelpful in making progress towards increasing our understanding of the genetic basis of psychiatric disorders. Strong evidence has been accumulated that is consistent with the contribution of many genes to risk of disease, across a wide range of allele frequencies and with a substantial proportion of genetic variation in the population in linkage disequilibrium with single-nucleotide polymorphisms (SNPs) on commercial genotyping arrays. At the same time, most causal variants that segregate in the population are likely to be rare and in total these variants also explain a significant proportion of genetic variation. It is the combination of allele frequency, effect size and functional characteristics that will determine the success of new experimental paradigms such as whole exome/genome sequencing to detect such loci. Empirical results suggest that roughly half the genetic variance is tagged by SNPs on commercial genome-wide chips, but that individual causal variants have a small effect size, on average. We conclude that larger experimental sample sizes are essential to further our understanding of the biology underlying psychiatric disorders.
Collapse
|
1255
|
Hill WG. Quantitative genetics in the genomics era. Curr Genomics 2012; 13:196-206. [PMID: 23115521 PMCID: PMC3382274 DOI: 10.2174/138920212800543110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/22/2011] [Accepted: 10/01/2011] [Indexed: 01/02/2023] Open
Abstract
The genetic analysis of quantitative or complex traits has been based mainly on statistical quantities such as genetic variances and heritability. These analyses continue to be developed, for example in studies of natural populations. Genomic methods are having an impact on progress and prospects. Actual relationships of individuals can be estimated enabling novel quantitative analyses. Increasing precision of linkage mapping is feasible with dense marker panels and designed stocks allowing multiple generations of recombination, and large SNP panels enable the use of genome wide association analysis utilising historical recombination. Whilst such analyses are identifying many loci for disease genes and traits such as height, typically each individually contributes a small amount of the variation. Only by fitting all SNPs without regard to significance can a high proportion be accounted for, so a classical polygenic model with near infinitesimally small effects remains a useful one. Theory indicates that a high proportion of variants will have low minor allele frequency, making detection difficult. Genomic selection, based on simultaneously fitting very dense markers and incorporating these with phenotypic data in breeding value prediction is revolutionising breeding programmes in agriculture and has a major potential role in human disease prediction.
Collapse
Affiliation(s)
- William G Hill
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, West Mains Road, Edinburgh, EH9 3JT, UK
| |
Collapse
|
1256
|
Service SK, Verweij KJH, Lahti J, Congdon E, Ekelund J, Hintsanen M, Räikkönen K, Lehtimäki T, Kähönen M, Widen E, Taanila A, Veijola J, Heath AC, Madden PAF, Montgomery GW, Sabatti C, Järvelin MR, Palotie A, Raitakari O, Viikari J, Martin NG, Eriksson JG, Keltikangas-Järvinen L, Wray NR, Freimer NB. A genome-wide meta-analysis of association studies of Cloninger's Temperament Scales. Transl Psychiatry 2012; 2:e116. [PMID: 22832960 PMCID: PMC3365256 DOI: 10.1038/tp.2012.37] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Temperament has a strongly heritable component, yet multiple independent genome-wide studies have failed to identify significant genetic associations. We have assembled the largest sample to date of persons with genome-wide genotype data, who have been assessed with Cloninger's Temperament and Character Inventory. Sum scores for novelty seeking, harm avoidance, reward dependence and persistence have been measured in over 11,000 persons collected in four different cohorts. Our study had >80% power to identify genome-wide significant loci (P<1.25 × 10(-8), with correction for testing four scales) accounting for ≥0.4% of the phenotypic variance in temperament scales. Using meta-analysis techniques, gene-based tests and pathway analysis we have tested over 1.2 million single-nucleotide polymorphisms (SNPs) for association to each of the four temperament dimensions. We did not discover any SNPs, genes, or pathways to be significantly related to the four temperament dimensions, after correcting for multiple testing. Less than 1% of the variability in any temperament dimension appears to be accounted for by a risk score derived from the SNPs showing strongest association to the temperament dimensions. Elucidation of genetic loci significantly influencing temperament and personality will require potentially very large samples, and/or a more refined phenotype. Item response theory methodology may be a way to incorporate data from cohorts assessed with multiple personality instruments, and might be a method by which a large sample of a more refined phenotype could be acquired.
Collapse
Affiliation(s)
- S K Service
- Center for Neurobehavioral Genetics, University of California, Los Angeles, CA, USA
| | - K J H Verweij
- Genetic Epidemiology, Molecular Epidemiology and Psychiatric Genetics Laboratories, Queensland Institute of Medical Research, Brisbane, QLD, Australia,School of Psychology, University of Queensland, Brisbane, QLD, Australia
| | - J Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - E Congdon
- Center for Neurobehavioral Genetics, University of California, Los Angeles, CA, USA
| | - J Ekelund
- Department of Psychiatry, University of Helsinki and Finland National Public Health Institute, Helsinki, Finland,Finland Vaasa Hospital District, Vaasa, Finland
| | - M Hintsanen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland,Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - K Räikkönen
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - T Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland,University of Tampere School of Medicine, Tampere, Finland
| | - M Kähönen
- University of Tampere School of Medicine, Tampere, Finland,Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - E Widen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - A Taanila
- Institute of Health Sciences, Public Health and General Practice, University of Oulu, Oulu, Finland
| | - J Veijola
- Department of Psychiatry, Institute of Clinical Medicine, University of Oulu, Oulu, Finland
| | - A C Heath
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - P A F Madden
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - G W Montgomery
- Genetic Epidemiology, Molecular Epidemiology and Psychiatric Genetics Laboratories, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - C Sabatti
- Department of Health and Research Policy, Stanford University, Stanford, CA, USA,Department of Statistics, Stanford University, Stanford, CA, USA
| | - M-R Järvelin
- Department of Epidemiology and Biostatistics, School of Public Health, MRC-HPA Centre for Environment and Health, Imperial College London, London, UK,Institute of Health Sciences, University of Oulu, Oulu, Finland,Biocenter Oulu, University of Oulu, Oulu, Finland,Department of Lifecourse and Services, National Institute of Health and Welfare, Oulu Finland
| | - A Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK,Department of Medical Genetics, University of Helsinki, Helsinki, Finland,University Central Hospital, Helsinki, Finland
| | - O Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - J Viikari
- Department of Medicine, Turku University Hospital, Turku, Finland,University of Turku, Turku, Finland
| | - N G Martin
- Genetic Epidemiology, Molecular Epidemiology and Psychiatric Genetics Laboratories, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - J G Eriksson
- Finland Vaasa Hospital District, Vaasa, Finland,National Institute for Health and Welfare, Helsinki, Finland,Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland,Helsinki University Central Hospital, Unit of General Practice, Helsinki, Finland,Folkhalsan Research Centre, Helsinki, Finland
| | | | - N R Wray
- Genetic Epidemiology, Molecular Epidemiology and Psychiatric Genetics Laboratories, Queensland Institute of Medical Research, Brisbane, QLD, Australia
| | - N B Freimer
- Center for Neurobehavioral Genetics, University of California, Los Angeles, CA, USA,The Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA,Department of Psychiatry, University of California, Los Angeles, Los Angeles, CA, USA,Center for Neurobehavioral Genetics, University of California, Gonda Center Room 3506, 695 Charles E Young Dr South, Box 951761, Los Angeles, CA 90095, USA. E-mail:
| |
Collapse
|
1257
|
Chan Y, Jones F, McConnell E, Bryk J, Bünger L, Tautz D. Parallel Selection Mapping Using Artificially Selected Mice Reveals Body Weight Control Loci. Curr Biol 2012; 22:794-800. [DOI: 10.1016/j.cub.2012.03.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 02/17/2012] [Accepted: 03/05/2012] [Indexed: 12/21/2022]
|
1258
|
Robinette SL, Holmes E, Nicholson JK, Dumas ME. Genetic determinants of metabolism in health and disease: from biochemical genetics to genome-wide associations. Genome Med 2012; 4:30. [PMID: 22546284 PMCID: PMC3446258 DOI: 10.1186/gm329] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Increasingly sophisticated measurement technologies have allowed the fields of metabolomics and genomics to identify, in parallel, risk factors of disease; predict drug metabolism; and study metabolic and genetic diversity in large human populations. Yet the complementarity of these fields and the utility of studying genes and metabolites together is belied by the frequent separate, parallel applications of genomic and metabolomic analysis. Early attempts at identifying co-variation and interaction between genetic variants and downstream metabolic changes, including metabolic profiling of human Mendelian diseases and quantitative trait locus mapping of individual metabolite concentrations, have recently been extended by new experimental designs that search for a large number of gene-metabolite associations. These approaches, including metabolomic quantitiative trait locus mapping and metabolomic genome-wide association studies, involve the concurrent collection of both genomic and metabolomic data and a subsequent search for statistical associations between genetic polymorphisms and metabolite concentrations across a broad range of genes and metabolites. These new data-fusion techniques will have important consequences in functional genomics, microbial metagenomics and disease modeling, the early results and implications of which are reviewed.
Collapse
Affiliation(s)
- Steven L Robinette
- Biomolecular Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK.
| | | | | | | |
Collapse
|
1259
|
Abstract
Evolution has long provided a foundation for population genetics, but some major advances in evolutionary biology from the twentieth century that provide foundations for evolutionary medicine are only now being applied in molecular medicine. They include the need for both proximate and evolutionary explanations, kin selection, evolutionary models for cooperation, competition between alleles, co-evolution, and new strategies for tracing phylogenies and identifying signals of selection. Recent advances in genomics are transforming evolutionary biology in ways that create even more opportunities for progress at its interfaces with genetics, medicine, and public health. This article reviews 15 evolutionary principles and their applications in molecular medicine in hopes that readers will use them and related principles to speed the development of evolutionary molecular medicine.
Collapse
|
1260
|
Jarvis JP, Scheinfeldt LB, Soi S, Lambert C, Omberg L, Ferwerda B, Froment A, Bodo JM, Beggs W, Hoffman G, Mezey J, Tishkoff SA. Patterns of ancestry, signatures of natural selection, and genetic association with stature in Western African pygmies. PLoS Genet 2012; 8:e1002641. [PMID: 22570615 PMCID: PMC3343053 DOI: 10.1371/journal.pgen.1002641] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 02/21/2012] [Indexed: 11/18/2022] Open
Abstract
African Pygmy groups show a distinctive pattern of phenotypic variation, including short stature, which is thought to reflect past adaptation to a tropical environment. Here, we analyze Illumina 1M SNP array data in three Western Pygmy populations from Cameroon and three neighboring Bantu-speaking agricultural populations with whom they have admixed. We infer genome-wide ancestry, scan for signals of positive selection, and perform targeted genetic association with measured height variation. We identify multiple regions throughout the genome that may have played a role in adaptive evolution, many of which contain loci with roles in growth hormone, insulin, and insulin-like growth factor signaling pathways, as well as immunity and neuroendocrine signaling involved in reproduction and metabolism. The most striking results are found on chromosome 3, which harbors a cluster of selection and association signals between approximately 45 and 60 Mb. This region also includes the positional candidate genes DOCK3, which is known to be associated with height variation in Europeans, and CISH, a negative regulator of cytokine signaling known to inhibit growth hormone-stimulated STAT5 signaling. Finally, pathway analysis for genes near the strongest signals of association with height indicates enrichment for loci involved in insulin and insulin-like growth factor signaling. Africa is thought to be the location of origin of modern humans within the past 200,000 years and the source of our dispersion across the globe within the past 100,000 years. Africa is also a region of extreme environmental, cultural, linguistic, and phenotypic diversity, and human populations living there show the highest levels of genetic diversity in the world. Yet little is known about the genetic basis of the observed phenotypic variation in Africa or how local adaptation and demography have influenced these patterns in the recent past. Here, we analyze a set of admixing Bantu-speaking agricultural and Western Pygmy hunter-gatherer populations that show extreme differences in stature; Pygmies are ∼17 cm shorter on average than their Bantu neighbors and among the shortest populations globally. Our multifaceted approach identified several genomic regions that may have been targets of natural selection and so may harbor variants underlying the unique anatomy and physiology of Western African Pygmies. One region of chromosome three, in particular, harbors strong signals of natural selection, population differentiation, and association with height. This region also contains a significant association with height in Europeans as well as a candidate gene known to regulate growth hormone signaling.
Collapse
Affiliation(s)
- Joseph P. Jarvis
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Laura B. Scheinfeldt
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sameer Soi
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Charla Lambert
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Larsson Omberg
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Bart Ferwerda
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | | | - Jean-Marie Bodo
- Ministere de la Recherche Scientifique et de l'Innovation, Yaounde, Cameroon
| | - William Beggs
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gabriel Hoffman
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Jason Mezey
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Sarah A. Tishkoff
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
1261
|
Yashin AI, Wu D, Arbeev KG, Ukraintseva SV. Polygenic effects of common single-nucleotide polymorphisms on life span: when association meets causality. Rejuvenation Res 2012; 15:381-94. [PMID: 22533364 DOI: 10.1089/rej.2011.1257] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Recently we have shown that the human life span is influenced jointly by many common single-nucleotide polymorphisms (SNPs), each with a small individual effect. Here we investigate further the polygenic influence on life span and discuss its possible biological mechanisms. First we identified six sets of prolongevity SNP alleles in the Framingham Heart Study 550K SNPs data, using six different statistical procedures (normal linear, Cox, and logistic regressions; generalized estimation equation; mixed model; gene frequency method). We then estimated joint effects of these SNPs on human survival. We found that alleles in each set show significant additive influence on life span. Twenty-seven SNPs comprised the overlapping set of SNPs that influenced life span, regardless of the statistical procedure. The majority of these SNPs (74%) were within genes, compared to 40% of SNPs in the original 550K set. We then performed a review of current literature on functions of genes closest to these 27 SNPs. The review showed that the respective genes are largely involved in aging, cancer, and brain disorders. We concluded that polygenic effects can explain a substantial portion of genetic influence on life span. Composition of the set of prolongevity alleles depends on the statistical procedure used for the allele selection. At the same time, there is a core set of longevity alleles that are selected with all statistical procedures. Functional relevance of respective genes to aging and major diseases supports causal relationships between the identified SNPs and life span. The fact that genes found in our and other genetic association studies of aging/longevity have similar functions indicates high chances of true positive associations for corresponding genetic variants.
Collapse
Affiliation(s)
- Anatoliy I Yashin
- Center for Population Health and Aging, Duke University, Durham, NC 27708-0408, USA.
| | | | | | | |
Collapse
|
1262
|
Sampson JN, Jacobs K, Wang Z, Yeager M, Chanock S, Chatterjee N. A two-platform design for next generation genome-wide association studies. Genet Epidemiol 2012; 36:400-8. [PMID: 22508365 DOI: 10.1002/gepi.21634] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 02/16/2012] [Accepted: 03/01/2012] [Indexed: 12/13/2022]
Abstract
Genome-wide association studies (GWAS) have been successful in their search for common genetic variants associated with complex traits and diseases. With new advances in array technologies together with available genetic reference sets, the next generation of GWAS will extend the search for associations with uncommon SNPs (1% ≤ MAF ≤ 10%). Two possible approaches are genotyping all participants, a prohibitively expensive option for large GWAS, or using a combination of genotyping and imputation. Here, we consider a two platform method that genotypes all participants on a standard genotyping array, designed to identify common variants, and then supplements that data by genotyping only a small proportion of the participants on a platform that has higher coverage for uncommon SNPs. This subset of the study population is then included as part of the imputation reference set. To demonstrate the use of this two-platform design, we evaluate its potential efficiency using a newly available dataset containing 756 individuals genotyped on both the Illumina Human OmniExpress and Omni2.5 Quad. Although genotyping all individuals on the denser array would be ideal, we find that genotyping only 100 individuals on this array, in combination with imputation, leads to only a modest loss of power for detecting associations. However, the loss of power due to imputation can be more substantial if the relative risks for rare variants are significantly larger than those previously observed for common variants.
Collapse
Affiliation(s)
- Joshua N Sampson
- Biostatistics Branch, DCEG, National Cancer Institute, Rockville, Maryland 20852, USA.
| | | | | | | | | | | |
Collapse
|
1263
|
Taal HR, Pourcain BS, Thiering E, Das S, Mook-Kanamori DO, Warrington NM, Kaakinen M, Kreiner-Møller E, Bradfield JP, Freathy RM, Geller F, Guxens M, Cousminer DL, Kerkhof M, Timpson NJ, Ikram MA, Beilin LJ, Bønnelykke K, Buxton JL, Charoen P, Chawes BLK, Eriksson J, Evans DM, Hofman A, Kemp JP, Kim CE, Klopp N, Lahti J, Lye SJ, McMahon G, Mentch FD, Müller M, O'Reilly PF, Prokopenko I, Rivadeneira F, Steegers EAP, Sunyer J, Tiesler C, Yaghootkar H, Breteler MMB, Debette S, Fornage M, Gudnason V, Launer LJ, van der Lugt A, Mosley TH, Seshadri S, Smith AV, Vernooij MW, Blakemore AI, Chiavacci RM, Feenstra B, Fernandez-Benet J, Grant SFA, Hartikainen AL, van der Heijden AJ, Iñiguez C, Lathrop M, McArdle WL, Mølgaard A, Newnham JP, Palmer LJ, Palotie A, Pouta A, Ring SM, Sovio U, Standl M, Uitterlinden AG, Wichmann HE, Vissing NH, DeCarli C, van Duijn CM, McCarthy MI, Koppelman GH, Estivill X, Hattersley AT, Melbye M, Bisgaard H, Pennell CE, Widen E, Hakonarson H, Smith GD, Heinrich J, Jarvelin MR, Jaddoe VWV. Common variants at 12q15 and 12q24 are associated with infant head circumference. Nat Genet 2012; 44:532-538. [PMID: 22504419 PMCID: PMC3773913 DOI: 10.1038/ng.2238] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 03/07/2012] [Indexed: 12/20/2022]
Abstract
To identify genetic variants associated with head circumference in infancy, we performed a meta-analysis of seven genome-wide association studies (GWAS) (N = 10,768 individuals of European ancestry enrolled in pregnancy and/or birth cohorts) and followed up three lead signals in six replication studies (combined N = 19,089). rs7980687 on chromosome 12q24 (P = 8.1 × 10(-9)) and rs1042725 on chromosome 12q15 (P = 2.8 × 10(-10)) were robustly associated with head circumference in infancy. Although these loci have previously been associated with adult height, their effects on infant head circumference were largely independent of height (P = 3.8 × 10(-7) for rs7980687 and P = 1.3 × 10(-7) for rs1042725 after adjustment for infant height). A third signal, rs11655470 on chromosome 17q21, showed suggestive evidence of association with head circumference (P = 3.9 × 10(-6)). SNPs correlated to the 17q21 signal have shown genome-wide association with adult intracranial volume, Parkinson's disease and other neurodegenerative diseases, indicating that a common genetic variant in this region might link early brain growth with neurological disease in later life.
Collapse
Affiliation(s)
- H Rob Taal
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Beate St Pourcain
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Elisabeth Thiering
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Shikta Das
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Dennis O Mook-Kanamori
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
- Weill Cornell Medical College - Qatar, Doha, Qatar
| | - Nicole M Warrington
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
- Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Canada
| | - Marika Kaakinen
- Institute of Health Sciences, University of Oulu, Finland
- Biocenter Oulu, University of Oulu, Finland
| | - Eskil Kreiner-Møller
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan P Bradfield
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Rachel M Freathy
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, UK
| | - Frank Geller
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Mònica Guxens
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Catalonia, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Catalonia, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
| | - Diana L Cousminer
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Marjan Kerkhof
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nicholas J Timpson
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lawrence J Beilin
- School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Jessica L Buxton
- Department of Genomics of Common Disease, School of Public Health, Imperial College London
| | - Pimphen Charoen
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Bo Lund Krogsgaard Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Johan Eriksson
- National Institute for Health and Welfare, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Centre, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - David M Evans
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John P Kemp
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Cecilia E Kim
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Norman Klopp
- Research Unit for Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Jari Lahti
- Institute of Behavioural Sciences, University of Helsinki, Helsinki, Finland
| | - Stephen J Lye
- Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Canada
| | - George McMahon
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Frank D Mentch
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Martina Müller
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Paul F O'Reilly
- Department of Epidemiology and Biostatistics, Imperial College London, W2 1PG London, UK
| | - Inga Prokopenko
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eric A P Steegers
- Department of Obstetrics & Gynecology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Jordi Sunyer
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Catalonia, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Catalonia, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
- Pompeu Fabra University (UPF), Barcelona, Catalonia, Spain
| | - Carla Tiesler
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Dr Von Hauner Children's Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, UK
| | | | - Stephanie Debette
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Myriam Fornage
- Institute of Molecular Medicine, Human Genetics Center and Division of Epidemiology, School of Public Health, University of Texas, Houston Health Sciences Center, Houston, TX, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogus, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Lenore J Launer
- Laboratory of Epidemiology, Demography and Biometry, National Institute on Aging, National Institute of Health, Bethesda, MD, USA
| | - Aad van der Lugt
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thomas H Mosley
- Department of Medicine (Geriatrics), University of Mississippi Medical Center, Jackson, MS, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Albert V Smith
- Icelandic Heart Association, Kopavogus, Iceland
- University of Iceland, Reykjavik, Iceland
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Radiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Alexandra If Blakemore
- Department of Genomics of Common Disease, School of Public Health, Imperial College London
| | - Rosetta M Chiavacci
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Julio Fernandez-Benet
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, United Kingdom
| | - Struan F A Grant
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia PA 19104, USA
| | - Anna-Liisa Hartikainen
- Institute of Clinical Medicine/Obstetrics and Gynecology, University of Oulu, Oulu, Finland
| | | | - Carmen Iñiguez
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
- Division of Environment and Health, Center for Public Health Research-CSISP, Valencia, Spain
| | - Mark Lathrop
- Centre National de Génotypage, Evry, France
- Foundation Jean Dausset, CEPH, Paris, France
| | - Wendy L McArdle
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Anne Mølgaard
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - John P Newnham
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Lyle J Palmer
- Samuel Lunenfeld Research Institute, University of Toronto, Toronto, Canada
- Genetic Epidemiology and Biostatistics Platform, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Annneli Pouta
- National Institute for Health and Welfare, Oulu, Finland, Biocenter Oulu, University of Oulu, Finland
| | - Susan M Ring
- School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Ulla Sovio
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H-Erich Wichmann
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nadja Hawwa Vissing
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA, USA
| | | | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xavier Estivill
- CIBER Epidemiologia y Salud Pública (CIBERESP), Barcelona, Catalonia, Spain
- Pompeu Fabra University (UPF), Barcelona, Catalonia, Spain
- Genes and Disease Program, Center for Genomic Regulation (CRG-UPF), Barcelona, Catalonia, Spain
| | - Andrew T Hattersley
- Peninsula NIHR Clinical Research Facility, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, UK
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, University of Copenhagen, Copenhagen, Denmark
| | - Craig E Pennell
- School of Women's and Infants' Health, The University of Western Australia, Perth, Australia
| | - Elisabeth Widen
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, University of Pennsylvania, Philadelphia PA 19104, USA
| | - George Davey Smith
- MRC Centre for Causal Analyses in Translational Epidemiology, School of Social and Community Medicine, University of Bristol, Bristol, UK
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Marjo-Riitta Jarvelin
- Institute of Health Sciences, University of Oulu, Finland
- National Institute for Health and Welfare, Oulu, Finland, Biocenter Oulu, University of Oulu, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, MRC Health Protection Agency (HPA) Centre for Environment and Health, Imperial College London
| | - Vincent W V Jaddoe
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
1264
|
Stein JL, Medland SE, Vasquez AA, Hibar DP, Senstad RE, Winkler AM, Toro R, Appel K, Bartecek R, Bergmann Ø, Bernard M, Brown AA, Cannon DM, Chakravarty MM, Christoforou A, Domin M, Grimm O, Hollinshead M, Holmes AJ, Homuth G, Hottenga JJ, Langan C, Lopez LM, Hansell NK, Hwang KS, Kim S, Laje G, Lee PH, Liu X, Loth E, Lourdusamy A, Mattingsdal M, Mohnke S, Maniega SM, Nho K, Nugent AC, O'Brien C, Papmeyer M, Pütz B, Ramasamy A, Rasmussen J, Rijpkema M, Risacher SL, Roddey JC, Rose EJ, Ryten M, Shen L, Sprooten E, Strengman E, Teumer A, Trabzuni D, Turner J, van Eijk K, van Erp TGM, van Tol MJ, Wittfeld K, Wolf C, Woudstra S, Aleman A, Alhusaini S, Almasy L, Binder EB, Brohawn DG, Cantor RM, Carless MA, Corvin A, Czisch M, Curran JE, Davies G, de Almeida MAA, Delanty N, Depondt C, Duggirala R, Dyer TD, Erk S, Fagerness J, Fox PT, Freimer NB, Gill M, Göring HHH, Hagler DJ, Hoehn D, Holsboer F, Hoogman M, Hosten N, Jahanshad N, Johnson MP, Kasperaviciute D, Kent JW, Kochunov P, Lancaster JL, Lawrie SM, Liewald DC, Mandl R, Matarin M, Mattheisen M, Meisenzahl E, Melle I, Moses EK, Mühleisen TW, et alStein JL, Medland SE, Vasquez AA, Hibar DP, Senstad RE, Winkler AM, Toro R, Appel K, Bartecek R, Bergmann Ø, Bernard M, Brown AA, Cannon DM, Chakravarty MM, Christoforou A, Domin M, Grimm O, Hollinshead M, Holmes AJ, Homuth G, Hottenga JJ, Langan C, Lopez LM, Hansell NK, Hwang KS, Kim S, Laje G, Lee PH, Liu X, Loth E, Lourdusamy A, Mattingsdal M, Mohnke S, Maniega SM, Nho K, Nugent AC, O'Brien C, Papmeyer M, Pütz B, Ramasamy A, Rasmussen J, Rijpkema M, Risacher SL, Roddey JC, Rose EJ, Ryten M, Shen L, Sprooten E, Strengman E, Teumer A, Trabzuni D, Turner J, van Eijk K, van Erp TGM, van Tol MJ, Wittfeld K, Wolf C, Woudstra S, Aleman A, Alhusaini S, Almasy L, Binder EB, Brohawn DG, Cantor RM, Carless MA, Corvin A, Czisch M, Curran JE, Davies G, de Almeida MAA, Delanty N, Depondt C, Duggirala R, Dyer TD, Erk S, Fagerness J, Fox PT, Freimer NB, Gill M, Göring HHH, Hagler DJ, Hoehn D, Holsboer F, Hoogman M, Hosten N, Jahanshad N, Johnson MP, Kasperaviciute D, Kent JW, Kochunov P, Lancaster JL, Lawrie SM, Liewald DC, Mandl R, Matarin M, Mattheisen M, Meisenzahl E, Melle I, Moses EK, Mühleisen TW, Nauck M, Nöthen MM, Olvera RL, Pandolfo M, Pike GB, Puls R, Reinvang I, Rentería ME, Rietschel M, Roffman JL, Royle NA, Rujescu D, Savitz J, Schnack HG, Schnell K, Seiferth N, Smith C, Steen VM, Valdés Hernández MC, Van den Heuvel M, van der Wee NJ, Van Haren NEM, Veltman JA, Völzke H, Walker R, Westlye LT, Whelan CD, Agartz I, Boomsma DI, Cavalleri GL, Dale AM, Djurovic S, Drevets WC, Hagoort P, Hall J, Heinz A, Jack CR, Foroud TM, Le Hellard S, Macciardi F, Montgomery GW, Poline JB, Porteous DJ, Sisodiya SM, Starr JM, Sussmann J, Toga AW, Veltman DJ, Walter H, Weiner MW, Bis JC, Ikram MA, Smith AV, Gudnason V, Tzourio C, Vernooij MW, Launer LJ, DeCarli C, Seshadri S, Andreassen OA, Apostolova LG, Bastin ME, Blangero J, Brunner HG, Buckner RL, Cichon S, Coppola G, de Zubicaray GI, Deary IJ, Donohoe G, de Geus EJC, Espeseth T, Fernández G, Glahn DC, Grabe HJ, Hardy J, Hulshoff Pol HE, Jenkinson M, Kahn RS, McDonald C, McIntosh AM, McMahon FJ, McMahon KL, Meyer-Lindenberg A, Morris DW, Müller-Myhsok B, Nichols TE, Ophoff RA, Paus T, Pausova Z, Penninx BW, Potkin SG, Sämann PG, Saykin AJ, Schumann G, Smoller JW, Wardlaw JM, Weale ME, Martin NG, Franke B, Wright MJ, Thompson PM. Identification of common variants associated with human hippocampal and intracranial volumes. Nat Genet 2012; 44:552-61. [PMID: 22504417 PMCID: PMC3635491 DOI: 10.1038/ng.2250] [Show More Authors] [Citation(s) in RCA: 479] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 03/19/2012] [Indexed: 02/06/2023]
Abstract
Identifying genetic variants influencing human brain structures may reveal new biological mechanisms underlying cognition and neuropsychiatric illness. The volume of the hippocampus is a biomarker of incipient Alzheimer's disease and is reduced in schizophrenia, major depression and mesial temporal lobe epilepsy. Whereas many brain imaging phenotypes are highly heritable, identifying and replicating genetic influences has been difficult, as small effects and the high costs of magnetic resonance imaging (MRI) have led to underpowered studies. Here we report genome-wide association meta-analyses and replication for mean bilateral hippocampal, total brain and intracranial volumes from a large multinational consortium. The intergenic variant rs7294919 was associated with hippocampal volume (12q24.22; N = 21,151; P = 6.70 × 10(-16)) and the expression levels of the positional candidate gene TESC in brain tissue. Additionally, rs10784502, located within HMGA2, was associated with intracranial volume (12q14.3; N = 15,782; P = 1.12 × 10(-12)). We also identified a suggestive association with total brain volume at rs10494373 within DDR2 (1q23.3; N = 6,500; P = 5.81 × 10(-7)).
Collapse
Affiliation(s)
- Jason L Stein
- Laboratory of Neuro Imaging, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1265
|
Common variants at 6q22 and 17q21 are associated with intracranial volume. Nat Genet 2012; 44:539-44. [PMID: 22504418 DOI: 10.1038/ng.2245] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 03/10/2012] [Indexed: 12/21/2022]
Abstract
During aging, intracranial volume remains unchanged and represents maximally attained brain size, while various interacting biological phenomena lead to brain volume loss. Consequently, intracranial volume and brain volume in late life reflect different genetic influences. Our genome-wide association study (GWAS) in 8,175 community-dwelling elderly persons did not reveal any associations at genome-wide significance (P < 5 × 10(-8)) for brain volume. In contrast, intracranial volume was significantly associated with two loci: rs4273712 (P = 3.4 × 10(-11)), a known height-associated locus on chromosome 6q22, and rs9915547 (P = 1.5 × 10(-12)), localized to the inversion on chromosome 17q21. We replicated the associations of these loci with intracranial volume in a separate sample of 1,752 elderly persons (P = 1.1 × 10(-3) for 6q22 and 1.2 × 10(-3) for 17q21). Furthermore, we also found suggestive associations of the 17q21 locus with head circumference in 10,768 children (mean age of 14.5 months). Our data identify two loci associated with head size, with the inversion at 17q21 also likely to be involved in attaining maximal brain size.
Collapse
|
1266
|
Estrada K, Styrkarsdottir U, Evangelou E, Hsu YH, Duncan EL, Ntzani EE, Oei L, Albagha OME, Amin N, Kemp JP, Koller DL, Li G, Liu CT, Minster RL, Moayyeri A, Vandenput L, Willner D, Xiao SM, Yerges-Armstrong LM, Zheng HF, Alonso N, Eriksson J, Kammerer CM, Kaptoge SK, Leo PJ, Thorleifsson G, Wilson SG, Wilson JF, Aalto V, Alen M, Aragaki AK, Aspelund T, Center JR, Dailiana Z, Duggan DJ, Garcia M, Garcia-Giralt N, Giroux S, Hallmans G, Hocking LJ, Husted LB, Jameson KA, Khusainova R, Kim GS, Kooperberg C, Koromila T, Kruk M, Laaksonen M, Lacroix AZ, Lee SH, Leung PC, Lewis JR, Masi L, Mencej-Bedrac S, Nguyen TV, Nogues X, Patel MS, Prezelj J, Rose LM, Scollen S, Siggeirsdottir K, Smith AV, Svensson O, Trompet S, Trummer O, van Schoor NM, Woo J, Zhu K, Balcells S, Brandi ML, Buckley BM, Cheng S, Christiansen C, Cooper C, Dedoussis G, Ford I, Frost M, Goltzman D, González-Macías J, Kähönen M, Karlsson M, Khusnutdinova E, Koh JM, Kollia P, Langdahl BL, Leslie WD, Lips P, Ljunggren Ö, Lorenc RS, Marc J, Mellström D, Obermayer-Pietsch B, Olmos JM, Pettersson-Kymmer U, Reid DM, Riancho JA, Ridker PM, Rousseau F, Slagboom PE, Tang NLS, et alEstrada K, Styrkarsdottir U, Evangelou E, Hsu YH, Duncan EL, Ntzani EE, Oei L, Albagha OME, Amin N, Kemp JP, Koller DL, Li G, Liu CT, Minster RL, Moayyeri A, Vandenput L, Willner D, Xiao SM, Yerges-Armstrong LM, Zheng HF, Alonso N, Eriksson J, Kammerer CM, Kaptoge SK, Leo PJ, Thorleifsson G, Wilson SG, Wilson JF, Aalto V, Alen M, Aragaki AK, Aspelund T, Center JR, Dailiana Z, Duggan DJ, Garcia M, Garcia-Giralt N, Giroux S, Hallmans G, Hocking LJ, Husted LB, Jameson KA, Khusainova R, Kim GS, Kooperberg C, Koromila T, Kruk M, Laaksonen M, Lacroix AZ, Lee SH, Leung PC, Lewis JR, Masi L, Mencej-Bedrac S, Nguyen TV, Nogues X, Patel MS, Prezelj J, Rose LM, Scollen S, Siggeirsdottir K, Smith AV, Svensson O, Trompet S, Trummer O, van Schoor NM, Woo J, Zhu K, Balcells S, Brandi ML, Buckley BM, Cheng S, Christiansen C, Cooper C, Dedoussis G, Ford I, Frost M, Goltzman D, González-Macías J, Kähönen M, Karlsson M, Khusnutdinova E, Koh JM, Kollia P, Langdahl BL, Leslie WD, Lips P, Ljunggren Ö, Lorenc RS, Marc J, Mellström D, Obermayer-Pietsch B, Olmos JM, Pettersson-Kymmer U, Reid DM, Riancho JA, Ridker PM, Rousseau F, Slagboom PE, Tang NLS, Urreizti R, Van Hul W, Viikari J, Zarrabeitia MT, Aulchenko YS, Castano-Betancourt M, Grundberg E, Herrera L, Ingvarsson T, Johannsdottir H, Kwan T, Li R, Luben R, Medina-Gómez C, Palsson ST, Reppe S, Rotter JI, Sigurdsson G, van Meurs JBJ, Verlaan D, Williams FMK, Wood AR, Zhou Y, Gautvik KM, Pastinen T, Raychaudhuri S, Cauley JA, Chasman DI, Clark GR, Cummings SR, Danoy P, Dennison EM, Eastell R, Eisman JA, Gudnason V, Hofman A, Jackson RD, Jones G, Jukema JW, Khaw KT, Lehtimäki T, Liu Y, Lorentzon M, McCloskey E, Mitchell BD, Nandakumar K, Nicholson GC, Oostra BA, Peacock M, Pols HAP, Prince RL, Raitakari O, Reid IR, Robbins J, Sambrook PN, Sham PC, Shuldiner AR, Tylavsky FA, van Duijn CM, Wareham NJ, Cupples LA, Econs MJ, Evans DM, Harris TB, Kung AWC, Psaty BM, Reeve J, Spector TD, Streeten EA, Zillikens MC, Thorsteinsdottir U, Ohlsson C, Karasik D, Richards JB, Brown MA, Stefansson K, Uitterlinden AG, Ralston SH, Ioannidis JPA, Kiel DP, Rivadeneira F. Genome-wide meta-analysis identifies 56 bone mineral density loci and reveals 14 loci associated with risk of fracture. Nat Genet 2012; 44:491-501. [PMID: 22504420 PMCID: PMC3338864 DOI: 10.1038/ng.2249] [Show More Authors] [Citation(s) in RCA: 918] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 03/16/2012] [Indexed: 12/15/2022]
Abstract
Bone mineral density (BMD) is the most widely used predictor of fracture risk. We performed the largest meta-analysis to date on lumbar spine and femoral neck BMD, including 17 genome-wide association studies and 32,961 individuals of European and east Asian ancestry. We tested the top BMD-associated markers for replication in 50,933 independent subjects and for association with risk of low-trauma fracture in 31,016 individuals with a history of fracture (cases) and 102,444 controls. We identified 56 loci (32 new) associated with BMD at genome-wide significance (P < 5 × 10(-8)). Several of these factors cluster within the RANK-RANKL-OPG, mesenchymal stem cell differentiation, endochondral ossification and Wnt signaling pathways. However, we also discovered loci that were localized to genes not known to have a role in bone biology. Fourteen BMD-associated loci were also associated with fracture risk (P < 5 × 10(-4), Bonferroni corrected), of which six reached P < 5 × 10(-8), including at 18p11.21 (FAM210A), 7q21.3 (SLC25A13), 11q13.2 (LRP5), 4q22.1 (MEPE), 2p16.2 (SPTBN1) and 10q21.1 (DKK1). These findings shed light on the genetic architecture and pathophysiological mechanisms underlying BMD variation and fracture susceptibility.
Collapse
Affiliation(s)
- Karol Estrada
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | | | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece
| | - Yi-Hsiang Hsu
- Institute for Aging Research, Hebrew SeniorLife, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
| | - Emma L Duncan
- Human Genetics Group, University of Queensland Diamantina Institute, Brisbane, Australia
- Department of Endocrinology, Royal Brisbane and Women’s Hospital, Brisbane, Australia
| | - Evangelia E Ntzani
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece
| | - Ling Oei
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - Omar M E Albagha
- Rheumatic Diseases Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John P Kemp
- Medical Research Council (MRC) Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, UK
| | - Daniel L Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
| | - Guo Li
- Cardiovascular Health Research Unit, University of Washington, Seattle, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, USA
| | - Ryan L Minster
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alireza Moayyeri
- of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dana Willner
- Human Genetics Group, University of Queensland Diamantina Institute, Brisbane, Australia
- Australian Centre for Ecogenomics, University of Queensland, Brisbane, Australia
| | - Su-Mei Xiao
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Laura M Yerges-Armstrong
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hou-Feng Zheng
- Department of Human Genetics, Lady Davis Institute, McGill University, Montreal, Canada
| | - Nerea Alonso
- Rheumatic Diseases Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Candace M Kammerer
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen K Kaptoge
- of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Paul J Leo
- Human Genetics Group, University of Queensland Diamantina Institute, Brisbane, Australia
| | | | - Scott G Wilson
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - James F Wilson
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, UK
| | - Ville Aalto
- Department of Clinical Physiology, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Markku Alen
- Department of Medical Rehabilitation, Oulu University Hospital and Institute of Health Sciences, Oulu, Finland
| | - Aaron K Aragaki
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Thor Aspelund
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jacqueline R Center
- Osteoporosis and Bone Biology Program, Garvan Institute of Medical Research, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
- Department of Endocrinology, St Vincents Hospital, Sydney, Australia
| | - Zoe Dailiana
- Department of Orthopaedic Surgery, Medical School University of Thessalia, Larissa, Greece
| | | | - Melissa Garcia
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, MD, USA
| | - Natàlia Garcia-Giralt
- Department of Internal Medicine, Hospital del Mar, Instituto Municipal de Investigación Médica (IMIM), Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Universitat Autònoma de Barcelona (UAB), Barcelone, Spain
| | - Sylvie Giroux
- Unité de recherche en génétique humaine et moléculaire, Centre de recherche du Centre hospitalier universitaire de Québec - Hôpital St-François-d’Assise (CHUQ/HSFA), Québec City, Canada
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå Unviersity, Umeå, Sweden
| | - Lynne J Hocking
- Musculoskeletal Research Programme, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK
| | - Lise Bjerre Husted
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus C, Denmark
| | - Karen A Jameson
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Rita Khusainova
- Ufa Scientific Centre of Russian Academy of Sciences, Institute of Biochemistry and Genetics, Ufa, Russia
- Biological Department, Bashkir State University, Ufa, Russia
| | - Ghi Su Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Theodora Koromila
- Department of Genetics and Biotechnology, Faculty of Biology, University of Athens, Athens, Greece
| | - Marcin Kruk
- Department of Biochemistry and Experimental Medicine, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Marika Laaksonen
- Department of Food and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Andrea Z Lacroix
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ping C Leung
- Jockey Club Centre for Osteoporosis Care and Control, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joshua R Lewis
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - Laura Masi
- Department of Internal Medicine, University of Florence, Florence, Italy
| | - Simona Mencej-Bedrac
- Department of Clinical Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Tuan V Nguyen
- Osteoporosis and Bone Biology Program, Garvan Institute of Medical Research, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
| | - Xavier Nogues
- Department of Internal Medicine, Hospital del Mar, Instituto Municipal de Investigación Médica (IMIM), Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Universitat Autònoma de Barcelona (UAB), Barcelone, Spain
| | - Millan S Patel
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Janez Prezelj
- Department of Endocrinology, University Medical Center, Ljubljana, Slovenia
| | - Lynda M Rose
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, USA
| | - Serena Scollen
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Olle Svensson
- Department of Surgical and Perioperative Sciences, Umeå Unviersity, Umeå, Sweden
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Olivia Trummer
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University Graz, Graz, Austria
| | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, Extramuraal Geneeskundig Onderzoek (EMGO) Institute for Health and Care Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Jean Woo
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kun Zhu
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - Susana Balcells
- Department of Genetics, University of Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelone, Spain
| | - Maria Luisa Brandi
- Department of Internal Medicine, University of Florence, Florence, Italy
| | - Brendan M Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Sulin Cheng
- Department of Health Sciences, University of Jyväskylä, Jyväskylä, Finland
- Department of Orthopaedics and Traumatology, Kuopio University Hospital, Kuopio, Finland
| | | | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, United Kingdom
| | - Morten Frost
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Clinical Institute, University of Southern Denmark, Odense, Denmark
| | - David Goltzman
- Department of Medicine, McGill University, Montreal, Canada
| | - Jesús González-Macías
- Department of Medicine, University of Cantabria, Santander, Spain
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla and Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, University of Tampere School of Medicine, Tampere, Finland
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences and Department of Orthopaedics, Lund University, Malmö, Sweden
| | - Elza Khusnutdinova
- Ufa Scientific Centre of Russian Academy of Sciences, Institute of Biochemistry and Genetics, Ufa, Russia
- Biological Department, Bashkir State University, Ufa, Russia
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Panagoula Kollia
- Department of Genetics and Biotechnology, Faculty of Biology, University of Athens, Athens, Greece
| | - Bente Lomholt Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus C, Denmark
| | - William D Leslie
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Paul Lips
- Department of Endocrinology, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
- Extramuraal Geneeskundig Onderzoek (EMGO) Institute for Health and Care Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Östen Ljunggren
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Roman S Lorenc
- Department of Biochemistry and Experimental Medicine, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Janja Marc
- Department of Clinical Biochemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University Graz, Graz, Austria
| | - José M Olmos
- Department of Medicine, University of Cantabria, Santander, Spain
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla and Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | | | - David M Reid
- Musculoskeletal Research Programme, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK
| | - José A Riancho
- Department of Medicine, University of Cantabria, Santander, Spain
- Department of Internal Medicine, Hospital Universitario Marqués de Valdecilla and Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV), Santander, Spain
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, USA
- Harvard Medical School, Boston, USA
| | - François Rousseau
- Unité de recherche en génétique humaine et moléculaire, Centre de recherche du Centre hospitalier universitaire de Québec - Hôpital St-François-d’Assise (CHUQ/HSFA), Québec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec City, Canada
- The APOGEE-Net/CanGèneTest Network on Genetic Health Services and Policy, Université Laval, Québec City, Canada
| | - P Eline Slagboom
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nelson LS Tang
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Roser Urreizti
- Department of Genetics, University of Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelone, Spain
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Jorma Viikari
- Department of Medicine, Turku University Hospital, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
| | | | - Yurii S Aulchenko
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martha Castano-Betancourt
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - Elin Grundberg
- Department of Human Genetics, McGill University, Montreal, Canada
- McGill University and Genome Québec Innovation Centre, Montreal, Canada
- Wellcome Trust Sanger Institute, Hinxton, UK
| | - Lizbeth Herrera
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thorvaldur Ingvarsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Orthopedic Surgery, Akureyri Hospital, Akureyri, Iceland
- Institution of Health Science, University Of Akureyri, Akureyri, Iceland
| | | | - Tony Kwan
- Department of Human Genetics, McGill University, Montreal, Canada
- McGill University and Genome Québec Innovation Centre, Montreal, Canada
| | - Rui Li
- Department of Epidemiology and Biostatistics, Lady Davis Institute, McGill University, Montreal, Canada
| | - Robert Luben
- of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Carolina Medina-Gómez
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Sjur Reppe
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jerome I Rotter
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Gunnar Sigurdsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Endocrinology and Metabolism, University Hospital, Reykjavik, Iceland
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - Dominique Verlaan
- Department of Human Genetics, McGill University, Montreal, Canada
- McGill University and Genome Québec Innovation Centre, Montreal, Canada
| | - Frances MK Williams
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Andrew R Wood
- Genetics of Complex Traits, Peninsula College of Medicine and Dentistry, University of Exeter, Exeter, England
| | - Yanhua Zhou
- Department of Biostatistics, Boston University School of Public Health, Boston, USA
| | - Kaare M Gautvik
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Department of Clinical Biochemistry, Lovisenberg Deacon Hospital, Oslo, Norway
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Tomi Pastinen
- Department of Human Genetics, McGill University, Montreal, Canada
- McGill University and Genome Québec Innovation Centre, Montreal, Canada
- Department of Medical Genetics, McGill University Health Centre, Montreal, Canada
| | - Soumya Raychaudhuri
- Division of Genetics and Rheumatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, United States
- Program in Medical and Population Genetics, Broad Institute, Cambridge, United States
| | - Jane A Cauley
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, USA
- Harvard Medical School, Boston, USA
| | - Graeme R Clark
- Human Genetics Group, University of Queensland Diamantina Institute, Brisbane, Australia
| | | | - Patrick Danoy
- Human Genetics Group, University of Queensland Diamantina Institute, Brisbane, Australia
| | - Elaine M Dennison
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Richard Eastell
- National Institute for Health and Research (NIHR) Musculoskeletal Biomedical Research Unit, University of Sheffield, Sheffield, UK
| | - John A Eisman
- Osteoporosis and Bone Biology Program, Garvan Institute of Medical Research, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
- Department of Endocrinology, St Vincents Hospital, Sydney, Australia
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - Rebecca D Jackson
- Department of Internal Medicine, The Ohio State University, Columbus, USA
- Center for Clinical and Translational Science, The Ohio State University, Columbus, USA
| | - Graeme Jones
- Menzies Research Institute, University of Tasmania, Hobart, Australia
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Kay-Tee Khaw
- of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Tampere University Hospital, Tampere, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere, Finland
| | - Yongmei Liu
- Center for Human Genomics, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eugene McCloskey
- National Institute for Health and Research (NIHR) Musculoskeletal Biomedical Research Unit, University of Sheffield, Sheffield, UK
- Academic Unit of Bone Metabolism, Metabolic Bone Centre, University of Sheffield, Sheffield, UK
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kannabiran Nandakumar
- Institute for Aging Research, Hebrew SeniorLife, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
| | | | - Ben A Oostra
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Munro Peacock
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Huibert A P Pols
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Richard L Prince
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, Australia
| | - Olli Raitakari
- Department of Clinical Physiology, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Ian R Reid
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - John Robbins
- Department of Medicine, University of Davis, Sacramento, CA, USA
| | - Philip N Sambrook
- Kolling Institute, Royal North Shore Hospital, University of Sydney, Sydney, Australia
| | - Pak Chung Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong, China
- Centre for Reproduction, Development and Growth, The University of Hong Kong, Hong Kong, China
| | - Alan R Shuldiner
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Geriatric Research and Education Clinical Center (GRECC), Veterans Administration Medical Center, Baltimore, MD, USA
| | - Frances A Tylavsky
- Department of Preventive Medicine, University of Tennessee College of Medicine, Memphis, TN, USA
| | | | - Nick J Wareham
- MRC Epidemiology Unit Box 285, Medical Research Council, Cambridge, UK
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, USA
- Framingham Heart Study, Framingham, USA
| | - Michael J Econs
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - David M Evans
- Medical Research Council (MRC) Centre for Causal Analyses in Translational Epidemiology, University of Bristol, Bristol, UK
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, MD, USA
| | - Annie Wai Chee Kung
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Bruce M Psaty
- Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, USA
| | - Jonathan Reeve
- Medicine box 157, University of Cambridge, Cambridge, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
| | - Elizabeth A Streeten
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Geriatric Research and Education Clinical Center (GRECC), Veterans Administration Medical Center, Baltimore, MD, USA
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
| | - J Brent Richards
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, UK
- Departments of Medicine, Human Genetics, Epidemiology and Biostatistics, Lady Davis Institute, McGill University, Montreal, Canada
| | - Matthew A Brown
- Human Genetics Group, University of Queensland Diamantina Institute, Brisbane, Australia
| | - Kari Stefansson
- deCODE Genetics, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| | - Stuart H Ralston
- Rheumatic Diseases Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - John P A Ioannidis
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece
- Stanford Prevention Research Center, Stanford University, Stanford, USA
| | - Douglas P Kiel
- Institute for Aging Research, Hebrew SeniorLife, Boston, USA
- Department of Medicine, Harvard Medical School, Boston, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), Leiden, The Netherlands
| |
Collapse
|
1267
|
Cappola TP, Dorn GW. Clinical considerations of heritable factors in common heart failure. ACTA ACUST UNITED AC 2012; 4:701-9. [PMID: 22187448 DOI: 10.1161/circgenetics.110.959379] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Thomas P Cappola
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
1268
|
Wang Y, Gjuvsland AB, Vik JO, Smith NP, Hunter PJ, Omholt SW. Parameters in dynamic models of complex traits are containers of missing heritability. PLoS Comput Biol 2012; 8:e1002459. [PMID: 22496634 PMCID: PMC3320574 DOI: 10.1371/journal.pcbi.1002459] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 02/19/2012] [Indexed: 12/31/2022] Open
Abstract
Polymorphisms identified in genome-wide association studies of human traits rarely explain more than a small proportion of the heritable variation, and improving this situation within the current paradigm appears daunting. Given a well-validated dynamic model of a complex physiological trait, a substantial part of the underlying genetic variation must manifest as variation in model parameters. These parameters are themselves phenotypic traits. By linking whole-cell phenotypic variation to genetic variation in a computational model of a single heart cell, incorporating genotype-to-parameter maps, we show that genome-wide association studies on parameters reveal much more genetic variation than when using higher-level cellular phenotypes. The results suggest that letting such studies be guided by computational physiology may facilitate a causal understanding of the genotype-to-phenotype map of complex traits, with strong implications for the development of phenomics technology. Despite an ever-increasing number of genome locations reported to be associated with complex human diseases or quantitative traits, only a small proportion of phenotypic variations in a typical quantitative trait can be explained by the discovered variants. We argue that this problem can partly be resolved by combining the statistical methods of quantitative genetics with computational biology. We demonstrate this for the in silico genotype-to-phenotype map of a model heart cell in conjunction with publically accessible genomic data. We show that genome wide association studies (GWAS) on model parameters identify more causal variants and can build better prediction models for the higher-level phenotypes than by performing GWAS on the higher-level phenotypes themselves. Since model parameters are in principle measurable physiological phenotypes, our findings suggest that development of future phenotyping technologies could be guided by mathematical models of the biological systems being targeted.
Collapse
Affiliation(s)
- Yunpeng Wang
- Centre for Integrative Genetics, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Arne B. Gjuvsland
- Centre for Integrative Genetics, Department of Mathematical Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Jon Olav Vik
- Centre for Integrative Genetics, Department of Mathematical Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Nicolas P. Smith
- Department of Biomedical Engineering, St Thomas' Hospital, King's College London, London, United Kingdom
| | - Peter J. Hunter
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Stig W. Omholt
- Centre for Integrative Genetics, Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, Ås, Norway
- * E-mail:
| |
Collapse
|
1269
|
|
1270
|
Zhang M, Liang L, Morar N, Dixon AL, Lathrop GM, Ding J, Moffatt MF, Cookson WOC, Kraft P, Qureshi AA, Han J. Integrating pathway analysis and genetics of gene expression for genome-wide association study of basal cell carcinoma. Hum Genet 2012; 131:615-23. [PMID: 22006220 PMCID: PMC3303995 DOI: 10.1007/s00439-011-1107-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 10/08/2011] [Indexed: 10/16/2022]
Abstract
Genome-wide association studies (GWASs) have primarily focused on marginal effects for individual markers and have incorporated external functional information only after identifying robust statistical associations. We applied a new approach combining the genetics of gene expression and functional classification of genes to the GWAS of basal cell carcinoma (BCC) to identify potential biological pathways associated with BCC. We first identified 322,324 expression-associated single-nucleotide polymorphisms (eSNPs) from two existing GWASs of global gene expression in lymphoblastoid cell lines (n = 955), and evaluated the association of these functionally annotated SNPs with BCC among 2,045 BCC cases and 6,013 controls in Caucasians. We then grouped them into 99 KEGG pathways for pathway analysis and identified two pathways associated with BCC with p value <0.05 and false discovery rate (FDR) <0.5: the autoimmune thyroid disease pathway (mainly HLA class I and II antigens, p < 0.001, FDR = 0.24) and Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway (p = 0.02, FDR = 0.49). Seventy-nine (25.7%) out of 307 significant eSNPs in the JAK-STAT pathway were associated with BCC risk (p < 0.05) in an independent replication set of 278 BCC cases and 1,262 controls. In addition, the association of JAK-STAT signaling pathway was marginally validated using 16,691 eSNPs identified from 110 normal skin samples (p = 0.08). Based on the evidence of biological functions of the JAK-STAT pathway on oncogenesis, it is plausible that this pathway is involved in BCC pathogenesis.
Collapse
Affiliation(s)
- Mingfeng Zhang
- Clinical Research Program, Department of Dermatology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Nilesh Morar
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Anna L Dixon
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Jun Ding
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Peter Kraft
- Departments of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Abrar A. Qureshi
- Clinical Research Program, Department of Dermatology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jiali Han
- Clinical Research Program, Department of Dermatology, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
- Departments of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, MA, USA
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
1271
|
Abstract
Fourteen years ago, the first article on molecular genetics was published in this journal: Child Development, Molecular Genetics, andWhat to Do With Genes Once They Are Found (R. Plomin & M. Rutter, 1998). The goal of the article was to outline what developmentalists can do with genes once they are found. These new directions for developmental research are still relevant today. The problem lies with the phrase “once they are found”: It has been much more difficult than expected to identify genes responsible for the heritability of complex traits and common disorders, the so-called missing heritability problem. The present article considers reasons for the missing heritability problem and possible solutions.
Collapse
|
1272
|
Heritability and genetic correlations explained by common SNPs for metabolic syndrome traits. PLoS Genet 2012; 8:e1002637. [PMID: 22479213 PMCID: PMC3315484 DOI: 10.1371/journal.pgen.1002637] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 02/21/2012] [Indexed: 01/05/2023] Open
Abstract
We used a bivariate (multivariate) linear mixed-effects model to estimate the narrow-sense heritability (h2) and heritability explained by the common SNPs (hg2) for several metabolic syndrome (MetS) traits and the genetic correlation between pairs of traits for the Atherosclerosis Risk in Communities (ARIC) genome-wide association study (GWAS) population. MetS traits included body-mass index (BMI), waist-to-hip ratio (WHR), systolic blood pressure (SBP), fasting glucose (GLU), fasting insulin (INS), fasting trigylcerides (TG), and fasting high-density lipoprotein (HDL). We found the percentage of h2 accounted for by common SNPs to be 58% of h2 for height, 41% for BMI, 46% for WHR, 30% for GLU, 39% for INS, 34% for TG, 25% for HDL, and 80% for SBP. We confirmed prior reports for height and BMI using the ARIC population and independently in the Framingham Heart Study (FHS) population. We demonstrated that the multivariate model supported large genetic correlations between BMI and WHR and between TG and HDL. We also showed that the genetic correlations between the MetS traits are directly proportional to the phenotypic correlations. The narrow-sense heritability of a trait such as body-mass index is a measure of the variability of the trait between people that is accounted for by their additive genetic differences. Knowledge of these genetic differences provides insight into biological mechanisms and hence treatments for diseases. Genome-wide association studies (GWAS) survey a large set of genetic markers common to the population. They have identified several single markers that are associated with traits and diseases. However, these markers do not seem to account for all of the known narrow-sense heritability. Here we used a recently developed model to quantify the genetic information contained in GWAS for single traits and shared between traits. We specifically investigated metabolic syndrome traits that are associated with type 2 diabetes and heart disease, and we found that for the majority of these traits much of the previously unaccounted for heritability is contained within common markers surveyed in GWAS. We also computed the genetic correlation between traits, which is a measure of the genetic components shared by traits. We found that the genetic correlation between these traits could be predicted from their phenotypic correlation.
Collapse
|
1273
|
Bayesian inference analyses of the polygenic architecture of rheumatoid arthritis. Nat Genet 2012; 44:483-9. [PMID: 22446960 DOI: 10.1038/ng.2232] [Citation(s) in RCA: 304] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 03/01/2012] [Indexed: 12/15/2022]
Abstract
The genetic architectures of common, complex diseases are largely uncharacterized. We modeled the genetic architecture underlying genome-wide association study (GWAS) data for rheumatoid arthritis and developed a new method using polygenic risk-score analyses to infer the total liability-scale variance explained by associated GWAS SNPs. Using this method, we estimated that, together, thousands of SNPs from rheumatoid arthritis GWAS explain an additional 20% of disease risk (excluding known associated loci). We further tested this method on datasets for three additional diseases and obtained comparable estimates for celiac disease (43% excluding the major histocompatibility complex), myocardial infarction and coronary artery disease (48%) and type 2 diabetes (49%). Our results are consistent with simulated genetic models in which hundreds of associated loci harbor common causal variants and a smaller number of loci harbor multiple rare causal variants. These analyses suggest that GWAS will continue to be highly productive for the discovery of additional susceptibility loci for common diseases.
Collapse
|
1274
|
Yang J, Ferreira T, Morris AP, Medland SE, Madden PAF, Heath AC, Martin NG, Montgomery GW, Weedon MN, Loos RJ, Frayling TM, McCarthy MI, Hirschhorn JN, Goddard ME, Visscher PM. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat Genet 2012; 44:369-75, S1-3. [PMID: 22426310 PMCID: PMC3593158 DOI: 10.1038/ng.2213] [Citation(s) in RCA: 1127] [Impact Index Per Article: 86.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 02/06/2012] [Indexed: 12/14/2022]
Abstract
We present an approximate conditional and joint association analysis that can use summary-level statistics from a meta-analysis of genome-wide association studies (GWAS) and estimated linkage disequilibrium (LD) from a reference sample with individual-level genotype data. Using this method, we analyzed meta-analysis summary data from the GIANT Consortium for height and body mass index (BMI), with the LD structure estimated from genotype data in two independent cohorts. We identified 36 loci with multiple associated variants for height (38 leading and 49 additional SNPs, 87 in total) via a genome-wide SNP selection procedure. The 49 new SNPs explain approximately 1.3% of variance, nearly doubling the heritability explained at the 36 loci. We did not find any locus showing multiple associated SNPs for BMI. The method we present is computationally fast and is also applicable to case-control data, which we demonstrate in an example from meta-analysis of type 2 diabetes by the DIAGRAM Consortium.
Collapse
Affiliation(s)
- Jian Yang
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1275
|
A genome-wide search for genetic influences and biological pathways related to the brain's white matter integrity. Neurobiol Aging 2012; 33:1847.e1-14. [PMID: 22425255 DOI: 10.1016/j.neurobiolaging.2012.02.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 01/31/2012] [Accepted: 02/04/2012] [Indexed: 01/04/2023]
Abstract
A genome-wide search for genetic variants influencing the brain's white matter integrity in old age was conducted in the Lothian Birth Cohort 1936 (LBC1936). At ∼73 years of age, members of the LBC1936 underwent diffusion MRI, from which 12 white matter tracts were segmented using quantitative tractography, and tract-averaged water diffusion parameters were determined (n = 668). A global measure of white matter tract integrity, g(FA), derived from principal components analysis of tract-averaged fractional anisotropy measurements, accounted for 38.6% of the individual differences across the 12 white matter tracts. A genome-wide search was performed with g(FA) on 535 individuals with 542,050 single nucleotide polymorphisms (SNPs). No single SNP association was genome-wide significant (all p > 5 × 10(-8)). There was genome-wide suggestive evidence for two SNPs, one in ADAMTS18 (p = 1.65 × 10(-6)), which is related to tumor suppression and hemostasis, and another in LOC388630 (p = 5.08 × 10(-6)), which is of unknown function. Although no gene passed correction for multiple comparisons in single gene-based testing, biological pathways analysis suggested evidence for an over-representation of neuronal transmission and cell adhesion pathways relating to g(FA).
Collapse
|
1276
|
Fondon JW, Martin A, Richards S, Gibbs RA, Mittelman D. Analysis of microsatellite variation in Drosophila melanogaster with population-scale genome sequencing. PLoS One 2012; 7:e33036. [PMID: 22427938 PMCID: PMC3299726 DOI: 10.1371/journal.pone.0033036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 02/07/2012] [Indexed: 11/29/2022] Open
Abstract
Genome sequencing technologies promise to revolutionize our understanding of genetics, evolution, and disease by making it feasible to survey a broad spectrum of sequence variation on a population scale. However, this potential can only be realized to the extent that methods for extracting and interpreting distinct forms of variation can be established. The error profiles and read length limitations of early versions of next-generation sequencing technologies rendered them ineffective for some sequence variant types, particularly microsatellites and other tandem repeats, and fostered the general misconception that such variants are inherently inaccessible to these platforms. At the same time, tandem repeats have emerged as important sources of functional variation. Tandem repeats are often located in and around genes, and frequent mutations in their lengths exert quantitative effects on gene function and phenotype, rapidly degrading linkage disequilibrium between markers and traits. Sensitive identification of these variants in large-scale next-gen sequencing efforts will enable more comprehensive association studies capable of revealing previously invisible associations. We present a population-scale analysis of microsatellite repeats using whole-genome data from 158 inbred isolates from the Drosophila Genetics Reference Panel, a collection of over 200 extensively phenotypically characterized isolates from a single natural population, to uncover processes underlying repeat mutation and to enable associations with behavioral, morphological, and life-history traits. Analysis of repeat variation from next-generation sequence data will also enhance studies of genome stability and neurodegenerative diseases.
Collapse
Affiliation(s)
- John W. Fondon
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Andy Martin
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Stephen Richards
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - David Mittelman
- Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|
1277
|
Mendizabal I, Marigorta UM, Lao O, Comas D. Adaptive evolution of loci covarying with the human African Pygmy phenotype. Hum Genet 2012; 131:1305-17. [PMID: 22407027 PMCID: PMC3397127 DOI: 10.1007/s00439-012-1157-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 02/24/2012] [Indexed: 01/18/2023]
Abstract
African Pygmies are hunter-gatherer populations from the equatorial rainforest that present the lowest height averages among humans. The biological basis and the putative adaptive role of the short stature of Pygmy populations has been one of the most intriguing topics for human biologists in the last century, which still remains elusive. Worldwide convergent evolution of the Pygmy size suggests the presence of strong selective pressures on the phenotype. We developed a novel approach to survey the genetic architecture of phenotypes and applied it to study the genomic covariation between allele frequencies and height measurements among Pygmy and non-Pygmy populations. Among the regions that were most associated with the phenotype, we identified a significant excess of genes with pivotal roles in bone homeostasis, such as PPPT3B and the height associated SUPT3H-RUNX2. We hypothesize that skeletal remodeling could be a key biological process underlying the Pygmy phenotype. In addition, we showed that these regions have most likely evolved under positive selection. These results constitute the first genetic hint of adaptive evolution in the African Pygmy phenotype, which is consistent with the independent emergence of the Pygmy height in other continents with similar environments.
Collapse
Affiliation(s)
- Isabel Mendizabal
- Departament de Ciències de la Salut i de la Vida, Institut de Biologia Evolutiva (CSIC-UPF), Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | | | | | | |
Collapse
|
1278
|
Jorde R, Svartberg J, Joakimsen RM, Grimnes G. Associations between Polymorphisms Related to Calcium Metabolism and Human Height: The Tromsø Study. Ann Hum Genet 2012; 76:200-10. [DOI: 10.1111/j.1469-1809.2012.00703.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1279
|
Ratnayake M, Reynard LN, Raine EVA, Santibanez-Koref M, Loughlin J. Allelic expression analysis of the osteoarthritis susceptibility locus that maps to MICAL3. BMC MEDICAL GENETICS 2012; 13:12. [PMID: 22385522 PMCID: PMC3366887 DOI: 10.1186/1471-2350-13-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/02/2012] [Indexed: 11/17/2022]
Abstract
Background A genome-wide association scan with subsequent replication study that involved over 67,000 individuals of European ancestry has produced evidence of association of single nucleotide polymorphism rs2277831 to primary osteoarthritis (OA) with a P-value of 2.9 × 10-5. rs2277831, an A/G transition, is located in an intron of MICAL3. This gene is located on chromosome 22q11.21 and the association signal encompasses two additional genes, BCL2L13 and BID. It is becoming increasingly apparent that many common complex traits are mediated by cis-acting regulatory polymorphisms that influence, in a tissue-specific manner, gene expression or transcript stability. Methods We used total and allelic expression analysis to assess whether the OA association to rs2277831 is mediated by an influence on MICAL3, BCL2L13 or BID expression. Using RNA extracted from joint tissues of 60 patients who had undergone elective joint replacement surgery, we assessed whether rs2277831 correlated with allelic expression of either of the three genes by: 1) measuring the expression of each gene by quantitative PCR and then stratifying the data by genotype at rs2277831 and 2) accurately discriminating and quantifying the mRNA synthesised from the alleles of OA patients using allelic-quantitative PCR. Results We found no evidence for a correlation between gene expression and genotype at rs2277831, with P-values of 0.09 for BCL2L13, 0.07 for BID and 0.33 for MICAL3. In the allelic expression analysis we observed several examples of significant (p < 0.05) allelic imbalances, with an allelic expression ratio of 2.82 observed in BCL2L13 (P = 0.004), 2.09 at BID (P = 0.001) and the most extreme case being at MICAL3, with an allelic expression ratio of 5.47 (P = 0.001). However, there was no correlation observed between the pattern of allelic expression and the genotype at rs2277831. Conclusions In the tissues that we have studied, our data do not support our hypothesis that the association between rs2277831 and OA is due to the effect this SNP has on MICAL3, BCL2L13 or BID gene expression. Instead, our data point towards other functional effects accounting for the OA associated signal.
Collapse
Affiliation(s)
- Madhushika Ratnayake
- Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle, UK
| | | | | | | | | |
Collapse
|
1280
|
de Moor MHM, Costa PT, Terracciano A, Krueger RF, de Geus EJC, Toshiko T, Penninx BWJH, Esko T, Madden PAF, Derringer J, Amin N, Willemsen G, Hottenga JJ, Distel MA, Uda M, Sanna S, Spinhoven P, Hartman CA, Sullivan P, Realo A, Allik J, Heath AC, Pergadia ML, Agrawal A, Lin P, Grucza R, Nutile T, Ciullo M, Rujescu D, Giegling I, Konte B, Widen E, Cousminer DL, Eriksson JG, Palotie A, Peltonen L, Luciano M, Tenesa A, Davies G, Lopez LM, Hansell NK, Medland SE, Ferrucci L, Schlessinger D, Montgomery GW, Wright MJ, Aulchenko YS, Janssens ACJW, Oostra BA, Metspalu A, Abecasis GR, Deary IJ, Räikkönen K, Bierut LJ, Martin NG, van Duijn CM, Boomsma DI. Meta-analysis of genome-wide association studies for personality. Mol Psychiatry 2012; 17:337-49. [PMID: 21173776 PMCID: PMC3785122 DOI: 10.1038/mp.2010.128] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Revised: 11/14/2010] [Accepted: 11/16/2010] [Indexed: 01/22/2023]
Abstract
Personality can be thought of as a set of characteristics that influence people's thoughts, feelings and behavior across a variety of settings. Variation in personality is predictive of many outcomes in life, including mental health. Here we report on a meta-analysis of genome-wide association (GWA) data for personality in 10 discovery samples (17,375 adults) and five in silico replication samples (3294 adults). All participants were of European ancestry. Personality scores for Neuroticism, Extraversion, Openness to Experience, Agreeableness and Conscientiousness were based on the NEO Five-Factor Inventory. Genotype data of ≈ 2.4M single-nucleotide polymorphisms (SNPs; directly typed and imputed using HapMap data) were available. In the discovery samples, classical association analyses were performed under an additive model followed by meta-analysis using the weighted inverse variance method. Results showed genome-wide significance for Openness to Experience near the RASA1 gene on 5q14.3 (rs1477268 and rs2032794, P=2.8 × 10(-8) and 3.1 × 10(-8)) and for Conscientiousness in the brain-expressed KATNAL2 gene on 18q21.1 (rs2576037, P=4.9 × 10(-8)). We further conducted a gene-based test that confirmed the association of KATNAL2 to Conscientiousness. In silico replication did not, however, show significant associations of the top SNPs with Openness and Conscientiousness, although the direction of effect of the KATNAL2 SNP on Conscientiousness was consistent in all replication samples. Larger scale GWA studies and alternative approaches are required for confirmation of KATNAL2 as a novel gene affecting Conscientiousness.
Collapse
Affiliation(s)
- M H M de Moor
- Department of Biological Psychology, VU University Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1281
|
Abstract
Intrauterine growth restriction (IUGR) is prevalent worldwide and affects children and adults in multiple ways. These include predisposition to type 2 diabetes mellitus, the metabolic syndrome, cardiovascular disease, persistent reduction in stature, and possibly changes in the pattern of puberty. A review of recent literature confirms that the metabolic effects of being born small for gestational age are evident in the very young, persist with age, and are amplified by adiposity. Furthermore, the pattern of growth in the first few years of life has a significant bearing on a person's later health, with those that show increasing weight gain being at the greatest risk for future metabolic dysfunction. Treatment with exogenous human GH is used to improve height in children who remain short after being small for gestational age at birth, but the response of individuals remains variable and difficult to predict. The mechanisms involved in the metabolic programming of IUGR children are just beginning to be explored. It appears that IUGR leads to widespread changes in DNA methylation and that specific "epigenetic signatures" for IUGR are likely to be found in various fetal tissues. The challenge is to link such alterations with modifications in gene expression and ultimately the metabolic abnormalities of adulthood, and it represents one of the frontiers for research in the field.
Collapse
Affiliation(s)
- Steven D Chernausek
- Department of Pediatrics, University of Oklahoma Health Sciences Center, 1200 North Phillips Avenue, Suite 4500, Oklahoma City, Oklahoma 73104-4600, USA.
| |
Collapse
|
1282
|
Abstract
PURPOSE OF REVIEW Numerous genome-wide association studies (GWAS) of schizophrenia have been published in the past 6 years, with a number of key reports published in the last year. The studies have evolved in scale from small individual samples to large collaborative endeavors. This review aims to critically assess whether the results have improved as the sample size and scale of genetic association studies have grown. RECENT FINDINGS Genomic genotyping and increasing sample sizes for schizophrenia association studies have led to parallel increases in the number of risk genes discovered with high statistical confidence. Nearly 20 genes or loci have surpassed the genome-wide significance threshold (P = 5 × 10) in a single study, and several have been replicated in more than one GWAS. SUMMARY Identifying the genetic underpinnings of complex diseases offers insight into the etiological mechanisms leading to manifestation of the disease. New and more effective treatments for schizophrenia are desperately needed, and the ability to target the relevant biological processes grows with our understanding of the genes involved. As the size of GWAS samples has increased, more genes have been identified with high confidence that have begun to provide insight into the etiological and pathophysiological foundations of this disorder.
Collapse
Affiliation(s)
- Sarah E Bergen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetics Research, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
1283
|
Min JL, Nicholson G, Halgrimsdottir I, Almstrup K, Petri A, Barrett A, Travers M, Rayner NW, Mägi R, Pettersson FH, Broxholme J, Neville MJ, Wills QF, Cheeseman J, Allen M, Holmes CC, Spector TD, Fleckner J, McCarthy MI, Karpe F, Lindgren CM, Zondervan KT. Coexpression network analysis in abdominal and gluteal adipose tissue reveals regulatory genetic loci for metabolic syndrome and related phenotypes. PLoS Genet 2012; 8:e1002505. [PMID: 22383892 PMCID: PMC3285582 DOI: 10.1371/journal.pgen.1002505] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 12/11/2011] [Indexed: 01/11/2023] Open
Abstract
Metabolic Syndrome (MetS) is highly prevalent and has considerable public health impact, but its underlying genetic factors remain elusive. To identify gene networks involved in MetS, we conducted whole-genome expression and genotype profiling on abdominal (ABD) and gluteal (GLU) adipose tissue, and whole blood (WB), from 29 MetS cases and 44 controls. Co-expression network analysis for each tissue independently identified nine, six, and zero MetS–associated modules of coexpressed genes in ABD, GLU, and WB, respectively. Of 8,992 probesets expressed in ABD or GLU, 685 (7.6%) were expressed in ABD and 51 (0.6%) in GLU only. Differential eigengene network analysis of 8,256 shared probesets detected 22 shared modules with high preservation across adipose depots (DABD-GLU = 0.89), seven of which were associated with MetS (FDR P<0.01). The strongest associated module, significantly enriched for immune response–related processes, contained 94/620 (15%) genes with inter-depot differences. In an independent cohort of 145/141 twins with ABD and WB longitudinal expression data, median variability in ABD due to familiality was greater for MetS–associated versus un-associated modules (ABD: 0.48 versus 0.18, P = 0.08; GLU: 0.54 versus 0.20, P = 7.8×10−4). Cis-eQTL analysis of probesets associated with MetS (FDR P<0.01) and/or inter-depot differences (FDR P<0.01) provided evidence for 32 eQTLs. Corresponding eSNPs were tested for association with MetS–related phenotypes in two GWAS of >100,000 individuals; rs10282458, affecting expression of RARRES2 (encoding chemerin), was associated with body mass index (BMI) (P = 6.0×10−4); and rs2395185, affecting inter-depot differences of HLA-DRB1 expression, was associated with high-density lipoprotein (P = 8.7×10−4) and BMI–adjusted waist-to-hip ratio (P = 2.4×10−4). Since many genes and their interactions influence complex traits such as MetS, integrated analysis of genotypes and coexpression networks across multiple tissues relevant to clinical traits is an efficient strategy to identify novel associations. Metabolic Syndrome (MetS) is a highly prevalent disorder with considerable public health concern, but its underlying genetic factors remain elusive. Given that most cellular components exert their functions through interactions with other cellular components, even the largest of genome-wide association (GWA) studies may often not detect their effects, nor necessarily provide insight into the complex molecular mechanisms of the disease. Rather than focusing on individual genes, the analysis of coexpression networks can be used for finding clusters (modules) of correlated expression levels across samples. In this study, we used a gene network–based approach for integrating clinical MetS, genotypic, and gene expression data from abdominal and gluteal adipose tissue and whole blood. We identified modules of genes related to MetS significantly enriched for immune response and oxidative phosphorylation pathways. We tested SNPs for association with MetS–associated expression (eSNPs), and tested prioritised eSNPs for association with MetS–related phenotypes in two large-scale GWA datasets. We identified two loci, neither of which had reached genome-wide significance levels in GWAs, associated with expression levels of RARRES2 and HLA-DRB1 and with MetS–related phenotypes, demonstrating that the integrated analysis of genotype and expression data from relevant multiple tissues can identify novel associations with complex traits such as MetS.
Collapse
Affiliation(s)
- Josine L. Min
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- * E-mail: (JLM); (KTZ)
| | - George Nicholson
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | | | - Kristian Almstrup
- Department of Molecular Genetics, Novo Nordisk A/S, Maaloev, Denmark
| | - Andreas Petri
- Department of Molecular Genetics, Novo Nordisk A/S, Maaloev, Denmark
| | - Amy Barrett
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
| | - Mary Travers
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
| | - Nigel W. Rayner
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
| | - Reedik Mägi
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Fredrik H. Pettersson
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - John Broxholme
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Matt J. Neville
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, ORH Trust, Churchill Hospital, Oxford, United Kingdom
| | - Quin F. Wills
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Jane Cheeseman
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
| | | | | | - Maxine Allen
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
| | - Chris C. Holmes
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Tim D. Spector
- Twin Research Unit, King's College London, London, United Kingdom
| | - Jan Fleckner
- Department of Molecular Genetics, Novo Nordisk A/S, Maaloev, Denmark
| | - Mark I. McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, ORH Trust, Churchill Hospital, Oxford, United Kingdom
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, ORH Trust, Churchill Hospital, Oxford, United Kingdom
| | - Cecilia M. Lindgren
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Krina T. Zondervan
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- * E-mail: (JLM); (KTZ)
| |
Collapse
|
1284
|
A survey of genomic studies supports association of circadian clock genes with bipolar disorder spectrum illnesses and lithium response. PLoS One 2012; 7:e32091. [PMID: 22384149 PMCID: PMC3285204 DOI: 10.1371/journal.pone.0032091] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 01/23/2012] [Indexed: 11/19/2022] Open
Abstract
Circadian rhythm abnormalities in bipolar disorder (BD) have led to a search for genetic abnormalities in circadian “clock genes” associated with BD. However, no significant clock gene findings have emerged from genome-wide association studies (GWAS). At least three factors could account for this discrepancy: complex traits are polygenic, the organization of the clock is more complex than previously recognized, and/or genetic risk for BD may be shared across multiple illnesses. To investigate these issues, we considered the clock gene network at three levels: essential “core” clock genes, upstream circadian clock modulators, and downstream clock controlled genes. Using relaxed thresholds for GWAS statistical significance, we determined the rates of clock vs. control genetic associations with BD, and four additional illnesses that share clinical features and/or genetic risk with BD (major depression, schizophrenia, attention deficit/hyperactivity). Then we compared the results to a set of lithium-responsive genes. Associations with BD-spectrum illnesses and lithium-responsiveness were both enriched among core clock genes but not among upstream clock modulators. Associations with BD-spectrum illnesses and lithium-responsiveness were also enriched among pervasively rhythmic clock-controlled genes but not among genes that were less pervasively rhythmic or non-rhythmic. Our analysis reveals previously unrecognized associations between clock genes and BD-spectrum illnesses, partly reconciling previously discordant results from past GWAS and candidate gene studies.
Collapse
|
1285
|
Lee SH, DeCandia TR, Ripke S, Yang J, Sullivan PF, Goddard ME, Keller MC, Visscher PM, Wray NR. Estimating the proportion of variation in susceptibility to schizophrenia captured by common SNPs. Nat Genet 2012; 44:247-50. [PMID: 22344220 PMCID: PMC3327879 DOI: 10.1038/ng.1108] [Citation(s) in RCA: 434] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 11/17/2011] [Indexed: 12/15/2022]
Abstract
Schizophrenia is a complex disorder caused by both genetic and environmental factors. Using 9,087 cases, 12,171 controls and 915,354 imputed SNPs from the Psychiatric GWA Consortium for schizophrenia (PGC-SCZ) we estimate that 23% (s.e. 1%) of variation in liability to schizophrenia is captured by SNPs. We show that an important proportion of this variation must be due to common causal variants, that the variance explained by each chromosome is linearly related to its length (r = 0.89, p = 2.6 × 10−8), that the genetic basis of schizophrenia is the same in males and females, and that a disproportionate proportion of variation is attributable to a set of 2725 genes expressed in the central nervous system (CNS) (p = 7.6 ×10−8). These results are consistent with a polygenic genetic architecture and imply more individual SNP associations will be detected for this disease as sample size increases.
Collapse
Affiliation(s)
- S Hong Lee
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1286
|
Wells JC. Sexual dimorphism in body composition across human populations: Associations with climate and proxies for short- and long-term energy supply. Am J Hum Biol 2012; 24:411-9. [DOI: 10.1002/ajhb.22223] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 11/03/2011] [Accepted: 12/03/2011] [Indexed: 11/06/2022] Open
|
1287
|
Association of common genetic variants in GPCPD1 with scaling of visual cortical surface area in humans. Proc Natl Acad Sci U S A 2012; 109:3985-90. [PMID: 22343285 DOI: 10.1073/pnas.1105829109] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Visual cortical surface area varies two- to threefold between human individuals, is highly heritable, and has been correlated with visual acuity and visual perception. However, it is still largely unknown what specific genetic and environmental factors contribute to normal variation in the area of visual cortex. To identify SNPs associated with the proportional surface area of visual cortex, we performed a genome-wide association study followed by replication in two independent cohorts. We identified one SNP (rs6116869) that replicated in both cohorts and had genome-wide significant association (P(combined) = 3.2 × 10(-8)). Furthermore, a metaanalysis of imputed SNPs in this genomic region identified a more significantly associated SNP (rs238295; P = 6.5 × 10(-9)) that was in strong linkage disequilibrium with rs6116869. These SNPs are located within 4 kb of the 5' UTR of GPCPD1, glycerophosphocholine phosphodiesterase GDE1 homolog (Saccharomyces cerevisiae), which in humans, is more highly expressed in occipital cortex compared with the remainder of cortex than 99.9% of genes genome-wide. Based on these findings, we conclude that this common genetic variation contributes to the proportional area of human visual cortex. We suggest that identifying genes that contribute to normal cortical architecture provides a first step to understanding genetic mechanisms that underlie visual perception.
Collapse
|
1288
|
Tassopoulou-Fishell M, Deeley K, Harvey EM, Sciote J, Vieira AR. Genetic variation in myosin 1H contributes to mandibular prognathism. Am J Orthod Dentofacial Orthop 2012; 141:51-9. [PMID: 22196185 DOI: 10.1016/j.ajodo.2011.06.033] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 06/01/2011] [Accepted: 06/01/2011] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Several candidate loci have been suggested as influencing mandibular prognathism (1p22.1, 1p22.2, 1p36, 3q26.2, 5p13-p12, 6q25, 11q22.2-q22.3, 12q23, 12q13.13, and 19p13.2). The goal of this study was to replicate these results in a well-characterized homogeneous sample set. METHODS Thirty-three single nucleotide polymorphisms spanning all candidate regions were studied in 44 prognathic and 35 Class I subjects from the University of Pittsburgh School of Dental Medicine Dental Registry and DNA Repository. The 44 subjects with mandibular prognathism had an average age of 18.4 years; 31 were female and 13 male; and 24 were white, 15 African American, 2 Hispanic, and 3 Asian. The 36 Class I subjects had an average age of 17.6 years; 27 were female and 9 male; and 27 were white, 6 African American, 1 Hispanic, and 2 Asian. Skeletal mandibular prognathism diagnosis included cephalometric values indicative of Class III such as an ANB smaller than 2°, a negative Wits appraisal, and a positive A-B plane. Additional mandibular prognathism criteria included negative overjet and visually prognathic (concave) profile as determined by the subject's clinical evaluation. Orthognathic subjects without jaw deformations were used as the comparison group. The mandibular prognathic and orthognathic subjects were matched by race, sex, and age. Genetic markers were tested by polymerase chain reaction with TaqMan chemistry. Chi-square and Fisher exact tests were used to determine overrepresentation of marker allele with an alpha of 0.05. RESULTS An association was unveiled between a marker in MYO1H (rs10850110) and the mandibular prognathism phenotype (P = 0.03). MYO1H is a Class I myosin that is in a different protein group than the myosin isoforms of muscle sarcomeres, which are the basis of skeletal muscle fiber typing. Class I myosins are necessary for cell motility, phagocytosis, and vesicle transport. CONCLUSIONS More strict clinical definitions might increase homogeneity and aid the studies of genetic susceptibility to malocclusions. We provide evidence that MYO1H can contribute to mandibular prognathism.
Collapse
Affiliation(s)
- Maria Tassopoulou-Fishell
- Department of Orthodontics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
1289
|
Permutation-based approaches do not adequately allow for linkage disequilibrium in gene-wide multi-locus association analysis. Eur J Hum Genet 2012; 20:890-6. [PMID: 22317971 DOI: 10.1038/ejhg.2012.8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Additional information about risk genes or risk pathways for diseases can be extracted from genome-wide association studies through analyses of groups of markers. The most commonly employed approaches involve combining individual marker data by adding the test statistics, or summing the logarithms of their P-values, and then using permutation testing to derive empirical P-values that allow for the statistical dependence of single-marker tests arising from linkage disequilibrium (LD). In the present study, we use simulated data to show that these approaches fail to reflect the structure of the sampling error, and the effect of this is to give undue weight to correlated markers. We show that the results obtained are internally inconsistent in the presence of strong LD, and are externally inconsistent with the results derived from multi-locus analysis. We also show that the results obtained from regression and multivariate Hotelling T(2) (H-T2) testing, but not those obtained from permutations, are consistent with the theoretically expected distributions, and that the H-T2 test has greater power to detect gene-wide associations in real datasets. Finally, we show that while the results from permutation testing can be made to approximate those from regression and multivariate Hotelling T(2) testing through aggressive LD pruning of markers, this comes at the cost of loss of information. We conclude that when conducting multi-locus analyses of sets of single-nucleotide polymorphisms, regression or multivariate Hotelling T(2) testing, which give equivalent results, are preferable to the other more commonly applied approaches.
Collapse
|
1290
|
Affiliation(s)
- Patrick F Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
1291
|
Degner JF, Pai AA, Pique-Regi R, Veyrieras JB, Gaffney DJ, Pickrell JK, De Leon S, Michelini K, Lewellen N, Crawford GE, Stephens M, Gilad Y, Pritchard JK. DNase I sensitivity QTLs are a major determinant of human expression variation. Nature 2012; 482:390-4. [PMID: 22307276 PMCID: PMC3501342 DOI: 10.1038/nature10808] [Citation(s) in RCA: 480] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 12/15/2011] [Indexed: 01/25/2023]
Abstract
The mapping of expression quantitative trait loci (eQTLs) has emerged as an important tool for linking genetic variation to changes in gene regulation1-5. However, it remains difficult to identify the causal variants underlying eQTLs and little is known about the regulatory mechanisms by which they act. To address this gap, we used DNaseI sequencing to measure chromatin accessibility in 70 Yoruba lymphoblastoid cell lines (LCLs), for which genome-wide genotypes and estimates of gene expression levels are also available6-8. We obtained a total of 2.7 billion uniquely mapped DNase-seq reads, which allowed us to produce genome-wide maps of chromatin accessibility for each individual. We identified 9,595 locations at which DNase-seq read depth correlates significantly with genotype at a nearby SNP or indel (FDR=10%). We call such variants “DNaseI sensitivity Quantitative Trait Loci” (dsQTLs). We found that dsQTLs are strongly enriched within inferred transcription factor binding sites and are frequently associated with allele-specific changes in transcription factor binding. A substantial fraction (16%) of dsQTLs are also associated with variation in the expression levels of nearby genes, (namely, these loci are also classified as eQTLs). Conversely, we estimate that as many as 55% of eQTL SNPs are also dsQTLs. Our observations indicate that dsQTLs are highly abundant in the human genome, and are likely to be important contributors to phenotypic variation.
Collapse
Affiliation(s)
- Jacob F Degner
- Department of Human Genetics, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1292
|
Ladouceur M, Dastani Z, Aulchenko YS, Greenwood CMT, Richards JB. The empirical power of rare variant association methods: results from sanger sequencing in 1,998 individuals. PLoS Genet 2012; 8:e1002496. [PMID: 22319458 PMCID: PMC3271058 DOI: 10.1371/journal.pgen.1002496] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 12/08/2011] [Indexed: 01/09/2023] Open
Abstract
The role of rare genetic variation in the etiology of complex disease remains unclear. However, the development of next-generation sequencing technologies offers the experimental opportunity to address this question. Several novel statistical methodologies have been recently proposed to assess the contribution of rare variation to complex disease etiology. Nevertheless, no empirical estimates comparing their relative power are available. We therefore assessed the parameters that influence their statistical power in 1,998 individuals Sanger-sequenced at seven genes by modeling different distributions of effect, proportions of causal variants, and direction of the associations (deleterious, protective, or both) in simulated continuous trait and case/control phenotypes. Our results demonstrate that the power of recently proposed statistical methods depend strongly on the underlying hypotheses concerning the relationship of phenotypes with each of these three factors. No method demonstrates consistently acceptable power despite this large sample size, and the performance of each method depends upon the underlying assumption of the relationship between rare variants and complex traits. Sensitivity analyses are therefore recommended to compare the stability of the results arising from different methods, and promising results should be replicated using the same method in an independent sample. These findings provide guidance in the analysis and interpretation of the role of rare base-pair variation in the etiology of complex traits and diseases.
Collapse
Affiliation(s)
- Martin Ladouceur
- Department of Human Genetics, McGill University, Montreal, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Zari Dastani
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Yurii S. Aulchenko
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
- Institute of Cytology and Genetics SD RAS, Novosibirsk, Russia
| | - Celia M. T. Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
| | - J. Brent Richards
- Department of Human Genetics, McGill University, Montreal, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, Canada
- Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| |
Collapse
|
1293
|
Prickett TCR, Bothwell JC, Yandle TG, Richards AM, Espiner EA. Pharmacodynamic responses of plasma and tissue C-type natriuretic peptide to GH: correlation with linear growth in GH-deficient rats. J Endocrinol 2012; 212:217-25. [PMID: 22087017 DOI: 10.1530/joe-11-0387] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Studies from genetic modification and spontaneous mutations show that C-type natriuretic peptide (CNP) signalling plays an essential part in postnatal endochondral growth, but measurement of CNP proteins and changes in their abundance in tissues and plasma during normal growth has not been reported. Using rodent pups with GH deficiency, we now describe the pharmacodynamic response of CNP and rat amino-terminal proCNP (NTproCNP) in plasma and tissues, and relate these to changes in linear growth (nose-tail length, tibial length and tibial growth plate width) during the course of 1 week of GH or saline (control) administration. Compared with saline, significant increases in plasma and tissue CNP forms were observed after 24 h in GH-treated pups and before any detectable change in linear growth. Whereas CNP abundance was increased in most tissues (muscle, heart and liver) by GH, enrichment was the greatest in extracts from growth plates and kidney. Plasma and tissue concentrations in GH-treated pups were sustained or further increased at 1 week when strong positive associations were found between plasma NTproCNP and linear growth or tissue concentrations. High content of NTproCNP in kidney tissue strongly correlated with plasma concentrations, which is consistent with previous data showing renal extraction of the peptide. In showing a prompt and significant increase in CNP in tissues driving normal endochondral growth, these findings provide further rationale for CNP agonists in the treatment of growth disorders resistant to current therapies and support the use of CNP concentrations as biomarkers of linear growth.
Collapse
Affiliation(s)
- T C R Prickett
- Department of Medicine, University of Otago, Christchurch, PO Box 4345, Christchurch 8140, New Zealand.
| | | | | | | | | |
Collapse
|
1294
|
Peters U, Hutter CM, Hsu L, Schumacher FR, Conti DV, Carlson CS, Edlund CK, Haile RW, Gallinger S, Zanke BW, Lemire M, Rangrej J, Vijayaraghavan R, Chan AT, Hazra A, Hunter DJ, Ma J, Fuchs CS, Giovannucci EL, Kraft P, Liu Y, Chen L, Jiao S, Makar KW, Taverna D, Gruber SB, Rennert G, Moreno V, Ulrich CM, Woods MO, Green RC, Parfrey PS, Prentice RL, Kooperberg C, Jackson RD, LaCroix AZ, Caan BJ, Hayes RB, Berndt SI, Chanock SJ, Schoen RE, Chang-Claude J, Hoffmeister M, Brenner H, Frank B, Bézieau S, Küry S, Slattery ML, Hopper JL, Jenkins MA, Le Marchand L, Lindor NM, Newcomb PA, Seminara D, Hudson TJ, Duggan DJ, Potter JD, Casey G. Meta-analysis of new genome-wide association studies of colorectal cancer risk. Hum Genet 2012; 131:217-34. [PMID: 21761138 PMCID: PMC3257356 DOI: 10.1007/s00439-011-1055-0] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 06/23/2011] [Indexed: 12/28/2022]
Abstract
Colorectal cancer is the second leading cause of cancer death in developed countries. Genome-wide association studies (GWAS) have successfully identified novel susceptibility loci for colorectal cancer. To follow up on these findings, and try to identify novel colorectal cancer susceptibility loci, we present results for GWAS of colorectal cancer (2,906 cases, 3,416 controls) that have not previously published main associations. Specifically, we calculated odds ratios and 95% confidence intervals using log-additive models for each study. In order to improve our power to detect novel colorectal cancer susceptibility loci, we performed a meta-analysis combining the results across studies. We selected the most statistically significant single nucleotide polymorphisms (SNPs) for replication using ten independent studies (8,161 cases and 9,101 controls). We again used a meta-analysis to summarize results for the replication studies alone, and for a combined analysis of GWAS and replication studies. We measured ten SNPs previously identified in colorectal cancer susceptibility loci and found eight to be associated with colorectal cancer (p value range 0.02 to 1.8 × 10(-8)). When we excluded studies that have previously published on these SNPs, five SNPs remained significant at p < 0.05 in the combined analysis. No novel susceptibility loci were significant in the replication study after adjustment for multiple testing, and none reached genome-wide significance from a combined analysis of GWAS and replication. We observed marginally significant evidence for a second independent SNP in the BMP2 region at chromosomal location 20p12 (rs4813802; replication p value 0.03; combined p value 7.3 × 10(-5)). In a region on 5p33.15, which includes the coding regions of the TERT-CLPTM1L genes and has been identified in GWAS to be associated with susceptibility to at least seven other cancers, we observed a marginally significant association with rs2853668 (replication p value 0.03; combined p value 1.9 × 10(-4)). Our study suggests a complex nature of the contribution of common genetic variants to risk for colorectal cancer.
Collapse
Affiliation(s)
- Ulrike Peters
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, USA
| | - Carolyn M. Hutter
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Li Hsu
- Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Fredrick R. Schumacher
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - David V. Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | | | | | - Robert W. Haile
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Steven Gallinger
- Department of Surgery, University Health Network, Toronto General Hospital, Toronto, Canada
| | - Brent W. Zanke
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | | | | | | | - Andrew T. Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- Channing Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
| | - Aditi Hazra
- Channing Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, USA
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, USA
| | - Jing Ma
- Channing Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
| | - Charles S. Fuchs
- Channing Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, USA
| | - Edward L. Giovannucci
- Channing Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, USA
- Departments of Epidemiology and Nutrition, Harvard School of Public Health, Boston, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston, USA
| | - Yan Liu
- Quantitative Services, Baylor Health Care System, Dallas, USA
| | - Lin Chen
- Department of Health Studies, University of Chicago, Chicago, USA
| | - Shuo Jiao
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Karen W. Makar
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Darin Taverna
- Translational Genomics Research Institute, Phoenix, USA
| | - Stephen B. Gruber
- Department of Internal Medicine, University of Michigan, Ann Arbor, USA
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, CarmelMedical Center and Technion Faculty of Medicine, Haifa, Israel
| | - Victor Moreno
- Biostatistics and Bioinformatics Unit, Catalan Institute of Oncology-IDIBELL, Barcelona, Spain
| | - Cornelia M. Ulrich
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, USA
- Division of Preventive Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Michael O. Woods
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Canada
| | - Roger C. Green
- Discipline of Genetics, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Canada
| | - Patrick S. Parfrey
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Canada
| | - Ross L. Prentice
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Rebecca D. Jackson
- Division of Endocrinology, Diabetes and Metabolism, Ohio State University, Columbus, USA
| | - Andrea Z. LaCroix
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Bette J. Caan
- Division of Research, Kaiser Permanente Medical Care Program, Oakland, USA
| | - Richard B. Hayes
- Division of Epidemiology, Department of Environmental Medicine, New YorkUniversity School of Medicine, New York City, USA
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, USA
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, USA
| | - Robert E. Schoen
- Department of Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Bernd Frank
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Stéphane Bézieau
- Centre Hospitalier Universitaire (CHU) de Nantes, Pôle de Biologie, Service de Génétique Médicale, Nantes, France
| | - Sébastien Küry
- Centre Hospitalier Universitaire (CHU) de Nantes, Pôle de Biologie, Service de Génétique Médicale, Nantes, France
| | - Martha L. Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, USA
| | - John L. Hopper
- Centre for Molecular, Environmental, Genetic, and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - Mark A. Jenkins
- Centre for Molecular, Environmental, Genetic, and Analytical Epidemiology, University of Melbourne, Melbourne, Australia
| | - Loic Le Marchand
- Epidemiology Program, Cancer Research Center of Hawai’i, University of Hawai’i at Manoa, Honolulu, USA
| | | | - Polly A. Newcomb
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Daniela Seminara
- Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, USA
| | - Thomas J. Hudson
- Ontario Institute for Cancer Research, Toronto, Canada
- Departments of Medical Biophysics and Molecular Genetics, University of Toronto, Toronto, Canada
| | | | - John D. Potter
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Graham Casey
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| |
Collapse
|
1295
|
Liu J, Hoppman N, O'Connell JR, Wang H, Streeten EA, McLenithan JC, Mitchell BD, Shuldiner AR. A functional haplotype in EIF2AK3, an ER stress sensor, is associated with lower bone mineral density. J Bone Miner Res 2012; 27:331-41. [PMID: 22028037 PMCID: PMC3319695 DOI: 10.1002/jbmr.549] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
EIF2AK3 is a type I transmembrane protein that functions as an endoplasmic reticulum (ER) stress sensor to regulate global protein synthesis. Rare mutations in EIF2AK3 cause Wolcott-Rallison syndrome (OMIM 226980), an autosomal recessive disorder characterized by diabetes, epiphyseal dysplasia, osteoporosis, and growth retardation. To investigate the role of common genetic variation in EIF2AK3 as a determinant of bone mineral density (BMD) and osteoporosis, we sequenced all exons and flanking regions, then genotyped six potentially functional single nucleotide polymorphisms (SNPs) in this gene in 997 Amish subjects for association analysis, and attempted replication in 887 Mexican Americans. We found that the minor allele of a nonsynonymous SNP rs13045 had borderline associations with decreased forearm BMD in both discovery and replication cohorts (unadjusted p = 0.036 and β = -0.007 for the Amish; unadjusted p = 0.031 and β = -0.008 for Mexican Americans). A meta-analysis indicated this association achieved statistical significance in the combined sample (unadjusted p = 0.003; Bonferroni corrected p = 0.009). Rs13045 and three other potentially functional SNPs, a promoter SNP (rs6547787) and two nonsynonymous SNPs (rs867529 and rs1805165), formed two haplotypes: a low-BMD associated haplotype, denoted haplotype B [minor allele frequency (MAF) = 0.311] and a common haplotype A (MAF = 0.676). There were no differences in mRNA expression in lymphoblastoid cell lines between the two haplotypes. However, after treating lymphoblastoid cell lines with thapsigargin to induce ER stress, cell lines with haplotype B showed increased sensitivity to ER stress (p = 0.014) compared with cell lines with haplotype A. Taken together, our results suggest that common nonsynonymous sequence variants in EIF2AK3 have a modest effect on ER stress response and may contribute to the risk for low BMD through this mechanism.
Collapse
Affiliation(s)
- Jie Liu
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicole Hoppman
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Hong Wang
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Elizabeth A Streeten
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - John C McLenithan
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Braxton D Mitchell
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alan R Shuldiner
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Geriatric Research Education and Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| |
Collapse
|
1296
|
Gaffney DJ, Veyrieras JB, Degner JF, Pique-Regi R, Pai AA, Crawford GE, Stephens M, Gilad Y, Pritchard JK. Dissecting the regulatory architecture of gene expression QTLs. Genome Biol 2012; 13:R7. [PMID: 22293038 PMCID: PMC3334587 DOI: 10.1186/gb-2012-13-1-r7] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/26/2012] [Accepted: 01/31/2012] [Indexed: 01/08/2023] Open
Abstract
Background Expression quantitative trait loci (eQTLs) are likely to play an important role in the genetics of complex traits; however, their functional basis remains poorly understood. Using the HapMap lymphoblastoid cell lines, we combine 1000 Genomes genotypes and an extensive catalogue of human functional elements to investigate the biological mechanisms that eQTLs perturb. Results We use a Bayesian hierarchical model to estimate the enrichment of eQTLs in a wide variety of regulatory annotations. We find that approximately 40% of eQTLs occur in open chromatin, and that they are particularly enriched in transcription factor binding sites, suggesting that many directly impact protein-DNA interactions. Analysis of core promoter regions shows that eQTLs also frequently disrupt some known core promoter motifs but, surprisingly, are not enriched in other well-known motifs such as the TATA box. We also show that information from regulatory annotations alone, when weighted by the hierarchical model, can provide a meaningful ranking of the SNPs that are most likely to drive gene expression variation. Conclusions Our study demonstrates how regulatory annotation and the association signal derived from eQTL-mapping can be combined into a single framework. We used this approach to further our understanding of the biology that drives human gene expression variation, and of the putatively causal SNPs that underlie it.
Collapse
Affiliation(s)
- Daniel J Gaffney
- Department of Human Genetics, University of Chicago, 920 E58th Street, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
1297
|
Nagele P. Perioperative genomics. Best Pract Res Clin Anaesthesiol 2012; 25:549-55. [PMID: 22099920 DOI: 10.1016/j.bpa.2011.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 09/15/2011] [Indexed: 11/28/2022]
Abstract
Since the completion of the Human Genome Project 10 years ago, the world has witnessed an incredible progress in human genetics and genomics.(1) This progress was largely driven by the availability of better, faster and cheaper sequencing technology.(2) While it took more than 10 years and more than 1 billion dollars to complete the Human Genome Project,(3-5) an individual in the year 2011 can have his whole genome sequenced within a week for less than $30,000. With cheaper and faster sequencing came a wealth of novel discoveries which makes it timely to review how these newly found insights into the human genome are relevant for perioperative medicine. This article summarises the basics of genetic inheritance, the human genome and modern sequencing methods, as well as genetic variation and how this knowledge may be applied to patient care and research in the perioperative setting.
Collapse
Affiliation(s)
- Peter Nagele
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
1298
|
Zaina S, Lund G. Integrating genomic and epigenomic information: a promising strategy for identifying functional DNA variants of human disease. Clin Genet 2012; 81:334-40. [DOI: 10.1111/j.1399-0004.2011.01840.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
1299
|
Abstract
Hematological traits are essential biomedical indicators that are widely used in clinical practice. The elucidation of the etiology that determines an individual's hematological traits would have a substantial impact. Hematological traits are known to be heritable, and it has been suggested that genetic factors contribute significantly to the inter-individual variance of these traits. Here, we review our current knowledge regarding the genetic architecture of hematological traits in humans, most of which has been obtained through recent developments in genome-wide association studies (GWAS). In addition to current knowledge, which is based on the hematological traits of the three major blood-cell lineages (white blood cells; WBC, red blood cells; RBC, and platelets; PLT), we propose future approaches that would be useful as a next step in the post-GWAS era.
Collapse
|
1300
|
Rohland N, Reich D. Cost-effective, high-throughput DNA sequencing libraries for multiplexed target capture. Genome Res 2012; 22:939-46. [PMID: 22267522 PMCID: PMC3337438 DOI: 10.1101/gr.128124.111] [Citation(s) in RCA: 679] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Improvements in technology have reduced the cost of DNA sequencing to the point that the limiting factor for many experiments is the time and reagent cost of sample preparation. We present an approach in which 192 sequencing libraries can be produced in a single day of technician time at a cost of about $15 per sample. These libraries are effective not only for low-pass whole-genome sequencing, but also for simultaneously enriching them in pools of approximately 100 individually barcoded samples for a subset of the genome without substantial loss in efficiency of target capture. We illustrate the power and effectiveness of this approach on about 2000 samples from a prostate cancer study.
Collapse
Affiliation(s)
- Nadin Rohland
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | | |
Collapse
|