scispace - formally typeset
Search or ask a question

Showing papers by "Michael Boehnke published in 2017"


Journal ArticleDOI
Robert A. Scott1, Laura J. Scott2, Reedik Mägi3, Letizia Marullo4  +213 moreInstitutions (66)
01 Nov 2017-Diabetes
TL;DR: This article conducted a meta-analysis of genome-wide association data from 26,676 T2D case and 132,532 control subjects of European ancestry after imputation using the 1000 Genomes multiethnic reference panel.
Abstract: To characterize type 2 diabetes (T2D)-associated variation across the allele frequency spectrum, we conducted a meta-analysis of genome-wide association data from 26,676 T2D case and 132,532 control subjects of European ancestry after imputation using the 1000 Genomes multiethnic reference panel Promising association signals were followed up in additional data sets (of 14,545 or 7,397 T2D case and 38,994 or 71,604 control subjects) We identified 13 novel T2D-associated loci (P < 5 × 10-8), including variants near the GLP2R, GIP, and HLA-DQA1 genes Our analysis brought the total number of independent T2D associations to 128 distinct signals at 113 loci Despite substantially increased sample size and more complete coverage of low-frequency variation, all novel associations were driven by common single nucleotide variants Credible sets of potentially causal variants were generally larger than those based on imputation with earlier reference panels, consistent with resolution of causal signals to common risk haplotypes Stratification of T2D-associated loci based on T2D-related quantitative trait associations revealed tissue-specific enrichment of regulatory annotations in pancreatic islet enhancers for loci influencing insulin secretion and in adipocytes, monocytes, and hepatocytes for insulin action-associated loci These findings highlight the predominant role played by common variants of modest effect and the diversity of biological mechanisms influencing T2D pathophysiology

601 citations


Journal ArticleDOI
Dajiang J. Liu1, Gina M. Peloso2, Gina M. Peloso3, Haojie Yu4  +285 moreInstitutions (91)
TL;DR: It is found that beta-thalassemia trait carriers displayed lower TC and were protected from coronary artery disease (CAD), and only some mechanisms of lowering LDL-C appeared to increase risk for type 2 diabetes (T2D); and TG-lowering alleles involved in hepatic production of TG-rich lipoproteins tracked with higher liver fat, higher risk for T2D, and lower risk for CAD.
Abstract: We screened variants on an exome-focused genotyping array in >300,000 participants (replication in >280,000 participants) and identified 444 independent variants in 250 loci significantly associated with total cholesterol (TC), high-density-lipoprotein cholesterol (HDL-C), low-density-lipoprotein cholesterol (LDL-C), and/or triglycerides (TG). At two loci (JAK2 and A1CF), experimental analysis in mice showed lipid changes consistent with the human data. We also found that: (i) beta-thalassemia trait carriers displayed lower TC and were protected from coronary artery disease (CAD); (ii) excluding the CETP locus, there was not a predictable relationship between plasma HDL-C and risk for age-related macular degeneration; (iii) only some mechanisms of lowering LDL-C appeared to increase risk for type 2 diabetes (T2D); and (iv) TG-lowering alleles involved in hepatic production of TG-rich lipoproteins (TM6SF2 and PNPLA3) tracked with higher liver fat, higher risk for T2D, and lower risk for CAD, whereas TG-lowering alleles involved in peripheral lipolysis (LPL and ANGPTL4) had no effect on liver fat but decreased risks for both T2D and CAD.

465 citations


01 Jan 2017
TL;DR: The results demonstrate that sufficiently large sample sizes can uncover rare and low-frequency variants of moderate-to-large effect associated with polygenic human phenotypes, and that these variants implicate relevant genes and pathways.
Abstract: Height is a highly heritable, classic polygenic trait with approximately 700 common associated variants identified through genome-wide association studies so far. Here, we report 83 height-associated coding variants with lower minor-allele frequencies (in the range of 0.1–4.8%) and effects of up to 2 centimetres per allele (such as those in IHH, STC2, AR and CRISPLD2), greater than ten times the average effect of common variants. In functional follow-up studies, rare height-increasing alleles of STC2 (giving an increase of 1–2 centimetres per allele) compromised proteolytic inhibition of PAPP-A and increased cleavage of IGFBP-4 in vitro, resulting in higher bioavailability of insulin-like growth factors. These 83 height-associated variants overlap genes that are mutated in monogenic growth disorders and highlight new biological candidates (such as ADAMTS3, IL11RA and NOX4) and pathways (such as proteoglycan and glycosaminoglycan synthesis) involved in growth. Our results demonstrate that sufficiently large sample sizes can uncover rare and low-frequency variants of moderate-to-large effect associated with polygenic human phenotypes, and that these variants implicate relevant genes and pathways.

407 citations


Journal ArticleDOI
Eleanor Wheeler1, Aaron Leong2, Ching-Ti Liu3, Marie-France Hivert2  +255 moreInstitutions (89)
TL;DR: This multiancestry study recommends investigation of the possible benefits of screening for the G6PD genotype along with using HbA1c to diagnose T2D in populations of African ancestry or groups where G 6PD deficiency is common, and investigates the effect of genetic risk-scores comprised of erythrocytic or glycemic variants on incident diabetes prediction and on prevalent diabetes screening performance.
Abstract: Background: Glycated hemoglobin (HbA1c) is used to diagnose type 2 diabetes (T2D) and assess glycemic control in patients with diabetes. Previous genome-wide association studies (GWAS) have identif ...

304 citations


Journal ArticleDOI
Mariaelisa Graff1, Robert A. Scott2, Anne E. Justice1, Kristin L. Young1  +346 moreInstitutions (101)
TL;DR: In additional genome-wide meta-analyses adjusting for PA and interaction with PA, 11 novel adiposity loci are identified, suggesting that accounting for PA or other environmental factors that contribute to variation in adiposity may facilitate gene discovery.
Abstract: Physical activity (PA) may modify the genetic effects that give rise to increased risk of obesity. To identify adiposity loci whose effects are modified by PA, we performed genome-wide interaction meta-analyses of BMI and BMI-adjusted waist circumference and waist-hip ratio from up to 200,452 adults of European (n = 180,423) or other ancestry (n = 20,029). We standardized PA by categorizing it into a dichotomous variable where, on average, 23% of participants were categorized as inactive and 77% as physically active. While we replicate the interaction with PA for the strongest known obesity-risk locus in the FTO gene, of which the effect is attenuated by ~30% in physically active individuals compared to inactive individuals, we do not identify additional loci that are sensitive to PA. In additional genome-wide meta-analyses adjusting for PA and interaction with PA, we identify 11 novel adiposity loci, suggesting that accounting for PA or other environmental factors that contribute to variation in adiposity may facilitate gene discovery.

275 citations



01 Jan 2017
TL;DR: In this article, the effect of genetic risk-scores comprised of erythrocytic or glycemic variants on incident diabetes prediction and on prevalent diabetes screening performance was investigated.
Abstract: Background Glycated hemoglobin (HbA1c) is used to diagnose type 2 diabetes (T2D) and assess glycemic control in patients with diabetes. Previous genome-wide association studies (GWAS) have identified 18 HbA1c-associated genetic variants. These variants proved to be classifiable by their likely biological action as erythrocytic (also associated with erythrocyte traits) or glycemic (associated with other glucose-related traits). In this study, we tested the hypotheses that, in a very large scale GWAS, we would identify more genetic variants associated with HbA1c and that HbA1c variants implicated in erythrocytic biology would affect the diagnostic accuracy of HbA1c. We therefore expanded the number of HbA1c-associated loci and tested the effect of genetic risk-scores comprised of erythrocytic or glycemic variants on incident diabetes prediction and on prevalent diabetes screening performance. Throughout this multiancestry study, we kept a focus on interancestry differences in HbA1c genetics performance that might influence race-ancestry differences in health outcomes. Methods & findings Using genome-wide association meta-analyses in up to 159,940 individuals from 82 cohorts of European, African, East Asian, and South Asian ancestry, we identified 60 common genetic variants associated with HbA1c. We classified variants as implicated in glycemic, erythrocytic, or unclassified biology and tested whether additive genetic scores of erythrocytic variants (GS-E) or glycemic variants (GS-G) were associated with higher T2D incidence in multiethnic longitudinal cohorts (N = 33,241). Nineteen glycemic and 22 erythrocytic variants were associated with HbA1c at genome-wide significance. GS-G was associated with higher T2D risk (incidence OR = 1.05, 95% CI 1.04–1.06, per HbA1c-raising allele, p = 3 × 10−29); whereas GS-E was not (OR = 1.00, 95% CI 0.99–1.01, p = 0.60). In Europeans and Asians, erythrocytic variants in aggregate had only modest effects on the diagnostic accuracy of HbA1c. Yet, in African Americans, the X-linked G6PD G202A variant (T-allele frequency 11%) was associated with an absolute decrease in HbA1c of 0.81%-units (95% CI 0.66–0.96) per allele in hemizygous men, and 0.68%-units (95% CI 0.38–0.97) in homozygous women. The G6PD variant may cause approximately 2% (N = 0.65 million, 95% CI 0.55–0.74) of African American adults with T2D to remain undiagnosed when screened with HbA1c. Limitations include the smaller sample sizes for non-European ancestries and the inability to classify approximately one-third of the variants. Further studies in large multiethnic cohorts with HbA1c, glycemic, and erythrocytic traits are required to better determine the biological action of the unclassified variants. Conclusions As G6PD deficiency can be clinically silent until illness strikes, we recommend investigation of the possible benefits of screening for the G6PD genotype along with using HbA1c to diagnose T2D in populations of African ancestry or groups where G6PD deficiency is common. Screening with direct glucose measurements, or genetically-informed HbA1c diagnostic thresholds in people with G6PD deficiency, may be required to avoid missed or delayed diagnoses.

220 citations


Journal ArticleDOI
TL;DR: The largest integrated analysis to date of high-resolution, high-throughput human islet molecular profiling data is presented to characterize the genome, epigenome, and transcriptome and find that T2D genetic variants are enriched in regions of the genome where transcription Regulatory Factor X (RFX) is predicted to bind in an islet-specific manner.
Abstract: Genome-wide association studies (GWAS) have identified >100 independent SNPs that modulate the risk of type 2 diabetes (T2D) and related traits. However, the pathogenic mechanisms of most of these SNPs remain elusive. Here, we examined genomic, epigenomic, and transcriptomic profiles in human pancreatic islets to understand the links between genetic variation, chromatin landscape, and gene expression in the context of T2D. We first integrated genome and transcriptome variation across 112 islet samples to produce dense cis-expression quantitative trait loci (cis-eQTL) maps. Additional integration with chromatin-state maps for islets and other diverse tissue types revealed that cis-eQTLs for islet-specific genes are specifically and significantly enriched in islet stretch enhancers. High-resolution chromatin accessibility profiling using assay for transposase-accessible chromatin sequencing (ATAC-seq) in two islet samples enabled us to identify specific transcription factor (TF) footprints embedded in active regulatory elements, which are highly enriched for islet cis-eQTL. Aggregate allelic bias signatures in TF footprints enabled us de novo to reconstruct TF binding affinities genetically, which support the high-quality nature of the TF footprint predictions. Interestingly, we found that T2D GWAS loci were strikingly and specifically enriched in islet Regulatory Factor X (RFX) footprints. Remarkably, within and across independent loci, T2D risk alleles that overlap with RFX footprints uniformly disrupt the RFX motifs at high-information content positions. Together, these results suggest that common regulatory variations have shaped islet TF footprints and the transcriptome and that a confluent RFX regulatory grammar plays a significant role in the genetic component of T2D predisposition.

188 citations


Journal ArticleDOI
Anne E. Justice1, Thomas W. Winkler2, Mary F. Feitosa3, Misa Graff1  +367 moreInstitutions (97)
TL;DR: The results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution, and highlight the importance of accounting for environment in genetic analyses.
Abstract: Few genome-wide association studies (GWAS) account for environmental exposures, like smoking, potentially impacting the overall trait variance when investigating the genetic contribution to obesity-related traits. Here, we use GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI. We identify 23 novel genetic loci, and 9 loci with convincing evidence of gene-smoking interaction (GxSMK) on obesity-related traits. We show consistent direction of effect for all identified loci and significance for 18 novel and for 5 interaction loci in an independent study sample. These loci highlight novel biological functions, including response to oxidative stress, addictive behaviour, and regulatory functions emphasizing the importance of accounting for environment in genetic analyses. Our results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution.

159 citations


01 Jan 2017
TL;DR: Results from genetic risk score models raise the possibility of a precision medicine approach through early lifestyle intervention to offset the impact of blood pressure–raising genetic variants on future cardiovascular disease risk.
Abstract: Elevated blood pressure is the leading heritable risk factor for cardiovascular disease worldwide. We report genetic association of blood pressure (systolic, diastolic, pulse pressure) among UK Biobank participants of European ancestry with independent replication in other cohorts, and robust validation of 107 independent loci. We also identify new independent variants at 11 previously reported blood pressure loci. In combination with results from a range of in silico functional analyses and wet bench experiments, our findings highlight new biological pathways for blood pressure regulation enriched for genes expressed in vascular tissues and identify potential therapeutic targets for hypertension. Results from genetic risk score models raise the possibility of a precision medicine approach through early lifestyle intervention to offset the impact of blood pressure–raising genetic variants on future cardiovascular disease risk.

150 citations


Journal ArticleDOI
TL;DR: The generation of a phenotype and genotype resource in the METSIM study allows us to proceed toward a “systems genetics” approach, which includes statistical methods to quantitate and integrate intermediate phenotypes, such as transcript, protein, or metabolite levels, to provide a global view of the molecular architecture of complex traits.

Journal ArticleDOI
TL;DR: Trans-eQTL signals confirmed and extended the previously reported KLF14-mediated network to 55 target genes, validated the CIITA regulation of class II MHC genes, and identified ZNF800 as a candidate master regulator.
Abstract: Subcutaneous adipose tissue stores excess lipids and maintains energy balance. We performed expression quantitative trait locus (eQTL) analyses by using abdominal subcutaneous adipose tissue of 770 extensively phenotyped participants of the METSIM study. We identified cis-eQTLs for 12,400 genes at a 1% false-discovery rate. Among an approximately 680 known genome-wide association study (GWAS) loci for cardio-metabolic traits, we identified 140 coincident cis-eQTLs at 109 GWAS loci, including 93 eQTLs not previously described. At 49 of these 140 eQTLs, gene expression was nominally associated (p 5 Mb away. These trans-eQTL signals confirmed and extended the previously reported KLF14-mediated network to 55 target genes, validated the CIITA regulation of class II MHC genes, and identified ZNF800 as a candidate master regulator. Finally, we observed similar expression-clinical trait correlations of genes associated with GWAS loci in both humans and a panel of genetically diverse mice. These results provide candidate genes for further investigation of their potential roles in adipose biology and in regulating cardio-metabolic traits.

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

Journal ArticleDOI
TL;DR: Preliminary Whole Genome Sequencing for Psychiatric Disorders Consortium data will integrate data for 18,000 individuals with psychiatric disorders, beginning with autism spectrum disorder, schizophrenia, bipolar disorder, and major depressive disorder, along with over 150,000 controls.
Abstract: As technology advances, whole genome sequencing (WGS) is likely to supersede other genotyping technologies. The rate of this change depends on its relative cost and utility. Variants identified uniquely through WGS may reveal novel biological pathways underlying complex disorders and provide high-resolution insight into when, where, and in which cell type these pathways are affected. Alternatively, cheaper and less computationally intensive approaches may yield equivalent insights. Understanding the role of rare variants in the noncoding gene-regulating genome through pilot WGS projects will be critical to determining which of these two extremes best represents reality. With large cohorts, well-defined risk loci, and a compelling need to understand the underlying biology, psychiatric disorders have a role to play in this preliminary WGS assessment. The Whole Genome Sequencing for Psychiatric Disorders Consortium will integrate data for 18,000 individuals with psychiatric disorders, beginning with autism spectrum disorder, schizophrenia, bipolar disorder, and major depressive disorder, along with over 150,000 controls.

Journal ArticleDOI
Ioanna Tachmazidou1, Daniel Suveges1, Josine L. Min2, Graham R. S. Ritchie3, Graham R. S. Ritchie1, Julia Steinberg1, Klaudia Walter1, Valentina Iotchkova4, Valentina Iotchkova1, Jeremy Schwartzentruber1, Jie Huang, Yasin Memari1, Shane A. McCarthy1, Andrew A Crawford, Cristina Bombieri5, Massimiliano Cocca6, Aliki-Eleni Farmaki7, Tom R. Gaunt2, Pekka Jousilahti8, Marjolein N. Kooijman9, Benjamin Lehne10, Giovanni Malerba5, Satu Männistö8, Angela Matchan1, Carolina Medina-Gomez9, Sarah Metrustry11, Abhishek Nag11, Ioanna Ntalla12, Lavinia Paternoster2, Nigel W. Rayner1, Nigel W. Rayner13, Nigel W. Rayner14, Cinzia Sala15, William R. Scott10, William R. Scott16, Hashem A. Shihab2, Lorraine Southam1, Lorraine Southam13, Beate St Pourcain2, Michela Traglia15, Katerina Trajanoska9, Gialuigi Zaza, Weihua Zhang16, Weihua Zhang10, María Soler Artigas17, Narinder Bansal18, Marianne Benn19, Marianne Benn20, Zhongsheng Chen21, Petr Danecek20, Petr Danecek19, Wei-Yu Lin18, Adam E. Locke21, Adam E. Locke22, Jian'an Luan18, Alisa K. Manning23, Alisa K. Manning24, Antonella Mulas25, Carlo Sidore, Anne Tybjærg-Hansen19, Anne Tybjærg-Hansen20, Anette Varbo20, Anette Varbo19, Magdalena Zoledziewska, Chris Finan26, Konstantinos Hatzikotoulas1, Audrey E. Hendricks1, Audrey E. Hendricks27, John P. Kemp2, Alireza Moayyeri11, Alireza Moayyeri26, Kalliope Panoutsopoulou1, Michal Szpak1, Scott Wilson28, Scott Wilson29, Scott Wilson11, Michael Boehnke21, Francesco Cucca25, Emanuele Di Angelantonio18, Emanuele Di Angelantonio30, Claudia Langenberg18, Cecilia M. Lindgren13, Cecilia M. Lindgren14, Mark I. McCarthy14, Mark I. McCarthy13, Mark I. McCarthy31, Andrew P. Morris32, Andrew P. Morris13, Andrew P. Morris33, Børge G. Nordestgaard20, Børge G. Nordestgaard19, Robert A. Scott18, Martin D. Tobin30, Martin D. Tobin17, Nicholas J. Wareham18, Paul Burton2, John C. Chambers10, John C. Chambers34, John C. Chambers16, George Davey Smith2, George Dedoussis7, Janine F. Felix9, Oscar H. Franco9, Giovanni Gambaro35, Paolo Gasparini6, Christopher J Hammond11, Albert Hofman9, Vincent W. V. Jaddoe9, Marcus E. Kleber36, Jaspal S. Kooner34, Jaspal S. Kooner8, Jaspal S. Kooner16, Markus Perola32, Markus Perola37, Markus Perola8, Caroline L Relton2, Susan M. Ring2, Fernando Rivadeneira9, Veikko Salomaa8, Tim D. Spector11, Oliver Stegle4, Daniela Toniolo15, André G. Uitterlinden9, Inês Barroso18, Inês Barroso1, Celia M. T. Greenwood38, Celia M. T. Greenwood39, John R. B. Perry11, John R. B. Perry18, Brian R. Walker3, Adam S. Butterworth30, Adam S. Butterworth18, Yali Xue1, Richard Durbin1, Kerrin S. Small11, Nicole Soranzo2, Nicholas J. Timpson2, Eleftheria Zeggini1 
TL;DR: This work applied a hybrid whole-genome sequencing (WGS) and deep imputation approach to examine the broader allelic architecture of 12 anthropometric traits associated with height, body mass, and fat distribution in up to 267,616 individuals to report 106 genome-wide significant signals that have not been previously identified.
Abstract: Deep sequence-based imputation can enhance the discovery power of genome-wide association studies by assessing previously unexplored variation across the common- and low-frequency spectra We applied a hybrid whole-genome sequencing (WGS) and deep imputation approach to examine the broader allelic architecture of 12 anthropometric traits associated with height, body mass, and fat distribution in up to 267,616 individuals We report 106 genome-wide significant signals that have not been previously identified, including 9 low-frequency variants pointing to functional candidates Of the 106 signals, 6 are in genomic regions that have not been implicated with related traits before, 28 are independent signals at previously reported regions, and 72 represent previously reported signals for a different anthropometric trait 71% of signals reside within genes and fine mapping resolves 23 signals to one or two likely causal variants We confirm genetic overlap between human monogenic and polygenic anthropometric traits and find signal enrichment in cis expression QTLs in relevant tissues Our results highlight the potential of WGS strategies to enhance biologically relevant discoveries across the frequency spectrum

Journal ArticleDOI
Louise V. Wain1, Ahmad Vaez2, Rick Jansen2, Roby Joehanes3  +267 moreInstitutions (74)
TL;DR: 48 genes with evidence for involvement in blood pressure regulation that are significant in multiple resources are identified and these robustly implicated genes may provide new leads for therapeutic innovation.
Abstract: Elevated blood pressure is a major risk factor for cardiovascular disease and has a substantial genetic contribution. Genetic variation influencing blood pressure has the potential to identify new pharmacological targets for the treatment of hypertension. To discover additional novel blood pressure loci, we used 1000 Genomes Project-based imputation in 150 134 European ancestry individuals and sought significant evidence for independent replication in a further 228 245 individuals. We report 6 new signals of association in or near HSPB7, TNXB, LRP12, LOC283335, SEPT9, and AKT2, and provide new replication evidence for a further 2 signals in EBF2 and NFKBIA. Combining large whole-blood gene expression resources totaling 12 607 individuals, we investigated all novel and previously reported signals and identified 48 genes with evidence for involvement in blood pressure regulation that are significant in multiple resources. Three novel kidney-specific signals were also detected. These robustly implicated genes may provide new leads for therapeutic innovation.

01 Jan 2017
TL;DR: In this paper, the authors used GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI.
Abstract: Few genome-wide association studies (GWAS) account for environmental exposures, like smoking, potentially impacting the overall trait variance when investigating the genetic contribution to obesity-related traits. Here, we use GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI. We identify 23 novel genetic loci, and 9 loci with convincing evidence of gene-smoking interaction (GxSMK) on obesity-related traits. We show consistent direction of effect for all identified loci and significance for 18 novel and for 5 interaction loci in an independent study sample. These loci highlight novel biological functions, including response to oxidative stress, addictive behaviour, and regulatory functions emphasizing the importance of accounting for environment in genetic analyses. Our results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution.

Journal ArticleDOI
01 Aug 2017-Diabetes
TL;DR: The largest genome-wide association study of first-phase insulin secretion, as measured by intravenous glucose tolerance tests, using up to 5,567 individuals without diabetes from 10 studies is performed, providing further insight into the mechanisms by which common genetic variation influences type 2 diabetes risk and glycemic traits.
Abstract: Understanding the physiological mechanisms by which common variants predispose to type 2 diabetes requires large studies with detailed measures of insulin secretion and sensitivity. Here we performed the largest genome-wide association study of first-phase insulin secretion, as measured by intravenous glucose tolerance tests, using up to 5,567 individuals without diabetes from 10 studies. We aimed to refine the mechanisms of 178 known associations between common variants and glycemic traits and identify new loci. Thirty type 2 diabetes or fasting glucose–raising alleles were associated with a measure of first-phase insulin secretion at P HNF1A , IGF2BP2 , KCNQ1 , HNF1B , VPS13C/C2CD4A , FAF1 , PTPRD , AP3S2 , KCNK16 , MAEA , LPP, WFS1 , and TMPRSS6 loci. The fasting glucose–raising allele near PDX1 , a known key insulin transcription factor, was strongly associated with lower first-phase insulin secretion but has no evidence for an effect on type 2 diabetes risk. The diabetes risk allele at TCF7L2 was associated with a stronger effect on peak insulin response than on C-peptide–based insulin secretion rate, suggesting a possible additional role in hepatic insulin clearance or insulin processing. In summary, our study provides further insight into the mechanisms by which common genetic variation influences type 2 diabetes risk and glycemic traits.

Posted ContentDOI
31 May 2017-bioRxiv
TL;DR: Accurate identification of validated targets is dependent on correct specification of the contribution of coding and non-coding mediated mechanisms at associated loci, and convincing support is demonstrated that coding variants are causal for the association at 16 of the 37 signals.
Abstract: Identification of coding variant associations for complex diseases offers a direct route to biological insight, but is dependent on appropriate inference concerning the causal impact of those variants on disease risk. We aggregated exome-array and exome sequencing data for 81,412 type 2 diabetes (T2D) cases and 370,832 controls of diverse ancestry, identifying 40 distinct coding variant association signals (at 38 loci) reaching significance (p -7 ). Of these, 16 represent novel associations mapping outside known genome-wide association study (GWAS) signals. We make two important observations. First, despite a threefold increase in sample size over previous efforts, only five of the 40 signals are driven by variants with minor allele frequency 1.36. Second, we used GWAS data from 50,160 T2D cases and 465,272 controls to fine-map associated coding variants in their regional context, with and without additional weighting, to account for the global enrichment of complex trait association signals in coding exons. We demonstrate convincing support (posterior probability >80% under the ‘annotation-weighted’ model) that coding variants are causal for the association at 16 of the 40 signals (including novel signals involving POC5 p.His36Arg, ANKH p.Arg187Gln, WSCD2 p.Thr113Ile, PLCB3 p.Ser778Leu, and PNPLA3 p.Ile148Met). However, one third of coding variant association signals represent ‘false leads’ at which naive analysis would have led to an erroneous inference regarding the effector transcript mediating the signal. Accurate identification of validated targets is dependent on correct specification of the contribution of coding and non-coding mediated mechanisms at associated loci.

01 Jan 2017
TL;DR: Kiel et al. as discussed by the authors performed a meta-analysis of genome-wide association studies for whole body lean body mass and found five novel genetic loci to be significantly associated with lean body weight.
Abstract: Lean body mass, consisting mostly of skeletal muscle, is important for healthy aging. We performed a genome-wide association study for whole body (20 cohorts of European ancestry with n = 38,292) and appendicular (arms and legs) lean body mass (n = 28,330) measured using dual energy X-ray absorptiometry or bioelectrical impedance analysis, adjusted for sex, age, height, and fat mass. Twenty-one single-nucleotide polymorphisms were significantly associated with lean body mass either genome wide (p < 5 × 10−8) or suggestively genome wide (p < 2.3 × 10−6). Replication in 63,475 (47,227 of European ancestry) individuals from 33 cohorts for whole body lean body mass and in 45,090 (42,360 of European ancestry) subjects from 25 cohorts for appendicular lean body mass was successful for five single-nucleotide polymorphisms in/near HSD17B11, VCAN, ADAMTSL3, IRS1, and FTO for total lean body mass and for three single-nucleotide polymorphisms in/near VCAN, ADAMTSL3, and IRS1 for appendicular lean body mass. Our findings provide new insight into the genetics of lean body mass.Lean body mass is a highly heritable trait and is associated with various health conditions. Here, Kiel and colleagues perform a meta-analysis of genome-wide association studies for whole body lean body mass and find five novel genetic loci to be significantly associated.

03 Aug 2017
TL;DR: This paper performed the largest genome-wide association study of first-phase insulin secretion, as measured by intravenous glucose tolerance tests, using up to 5,567 individuals without diabetes from 10 studies.
Abstract: Understanding the physiological mechanisms by which common variants predispose to type 2 diabetes requires large studies with detailed measures of insulin secretion and sensitivity. Here we performed the largest genome-wide association study of first-phase insulin secretion, as measured by intravenous glucose tolerance tests, using up to 5,567 individuals without diabetes from 10 studies. We aimed to refine the mechanisms of 178 known associations between common variants and glycemic traits and identify new loci. Thirty type 2 diabetes or fasting glucose–raising alleles were associated with a measure of first-phase insulin secretion at P HNF1A , IGF2BP2 , KCNQ1 , HNF1B , VPS13C/C2CD4A , FAF1 , PTPRD , AP3S2 , KCNK16 , MAEA , LPP, WFS1 , and TMPRSS6 loci. The fasting glucose–raising allele near PDX1 , a known key insulin transcription factor, was strongly associated with lower first-phase insulin secretion but has no evidence for an effect on type 2 diabetes risk. The diabetes risk allele at TCF7L2 was associated with a stronger effect on peak insulin response than on C-peptide–based insulin secretion rate, suggesting a possible additional role in hepatic insulin clearance or insulin processing. In summary, our study provides further insight into the mechanisms by which common genetic variation influences type 2 diabetes risk and glycemic traits.

Journal ArticleDOI
01 Sep 2017-Diabetes
TL;DR: It is demonstrated that type 2 diabetes risk alleles are associated with decreased ADCY5 expression in human islets and candidate variants for regulatory function are examined, and data suggest that rs11708067-A risk allele contributes to type 1 diabetes by disrupting an islet enhancer, which results in reduced ADCy5 expression and impaired insulin secretion.
Abstract: Molecular mechanisms remain unknown for most type 2 diabetes genome-wide association study identified loci. Variants associated with type 2 diabetes and fasting glucose levels reside in introns of ADCY5, a gene that encodes adenylate cyclase 5. Adenylate cyclase 5 catalyzes the production of cyclic AMP, which is a second messenger molecule involved in cell signaling and pancreatic β-cell insulin secretion. We demonstrated that type 2 diabetes risk alleles are associated with decreased ADCY5 expression in human islets and examined candidate variants for regulatory function. rs11708067 overlaps a predicted enhancer region in pancreatic islets. The type 2 diabetes risk rs11708067-A allele showed fewer H3K27ac ChIP-seq reads in human islets, lower transcriptional activity in reporter assays in rodent β-cells (rat 832/13 and mouse MIN6), and increased nuclear protein binding compared with the rs11708067-G allele. Homozygous deletion of the orthologous enhancer region in 832/13 cells resulted in a 64% reduction in expression level of Adcy5, but not adjacent gene Sec22a, and a 39% reduction in insulin secretion. Together, these data suggest that rs11708067-A risk allele contributes to type 2 diabetes by disrupting an islet enhancer, which results in reduced ADCY5 expression and impaired insulin secretion.

Journal ArticleDOI
Josep M. Mercader1, Josep M. Mercader2, Josep M. Mercader3, Rachel G. Liao1, Avery Davis Bell1, Avery Davis Bell2, Zachary Dymek1, Karol Estrada2, Karol Estrada1, Taru Tukiainen2, Taru Tukiainen1, Alicia Huerta-Chagoya4, Hortensia Moreno-Macías5, Hortensia Moreno-Macías6, Kathleen A. Jablonski7, Robert L. Hanson8, Geoffrey A. Walford2, Geoffrey A. Walford1, Ignasi Moran9, Ling Chen1, Ling Chen2, Vineeta Agarwala1, María Luisa Ordóñez-Sánchez4, Rosario Rodríguez-Guillén4, Maribel Rodriguez-Torres4, Yayoi Segura-Kato4, Humberto García-Ortiz, Federico Centeno-Cruz, Francisco Barajas-Olmos, Lizz Caulkins1, Sobha Puppala10, Pierre Fontanillas1, Amy L. Williams11, Sílvia Bonàs-Guarch3, Christopher Hartl1, Stephan Ripke2, Stephan Ripke1, Stephan Ripke12, Katherine Tooley2, Katherine Tooley1, Jacqueline M. Lane1, Jacqueline M. Lane2, Carlos Zerrweck, Angélica Martínez-Hernández, Emilio J. Cordova, Elvia Mendoza-Caamal, Cecilia Contreras-Cubas, María Elena González-Villalpando, Ivette Cruz-Bautista, Liliana Muñoz-Hernandez, Donaji V. Gómez-Velasco, Ulises Alvirde, Brian E. Henderson13, Lynne R. Wilkens14, Loic Le Marchand14, Olimpia Arellano-Campos, Laura Riba, Maegan Harden1, Broad Genomics Platform1, Stacey Gabriel1, Hanna E. Abboud15, Maria L. Cortes1, Cristina Revilla-Monsalve16, Sergio Islas-Andrade16, Xavier Soberón, Joanne E. Curran17, Christopher P. Jenkinson17, Ralph A. DeFronzo15, Donna M. Lehman15, Craig L. Hanis18, Graeme I. Bell19, Michael Boehnke20, John Blangero17, Ravindranath Duggirala17, Richa Saxena2, Richa Saxena1, Daniel G. MacArthur2, Daniel G. MacArthur1, Jorge Ferrer9, Jorge Ferrer21, Steven A. McCarroll1, Steven A. McCarroll2, David Torrents3, David Torrents22, William C. Knowler8, Leslie J. Baier8, Noël P. Burtt1, Clicerio González-Villalpando, Christopher A. Haiman14, Carlos A. Aguilar-Salinas, Teresa Tusié-Luna6, Jason Flannick1, Jason Flannick2, Suzanne B.R. Jacobs2, Suzanne B.R. Jacobs1, Lorena Orozco, David Altshuler, Jose C. Florez2, Jose C. Florez1 
01 Nov 2017-Diabetes
TL;DR: It is suggested that reducing IGF2 isoform 2 expression in relevant tissues has potential as a new therapeutic strategy for T2D, even beyond the Latin American population, with no major adverse effects on health or reproduction.
Abstract: Type 2 diabetes (T2D) affects more than 415 million people worldwide, and its costs to the health care system continue to rise. To identify common or rare genetic variation with potential therapeutic implications for T2D, we analyzed and replicated genome-wide protein coding variation in a total of 8,227 individuals with T2D and 12,966 individuals without T2D of Latino descent. We identified a novel genetic variant in the IGF2 gene associated with ∼20% reduced risk for T2D. This variant, which has an allele frequency of 17% in the Mexican population but is rare in Europe, prevents splicing between IGF2 exons 1 and 2. We show in vitro and in human liver and adipose tissue that the variant is associated with a specific, allele-dosage–dependent reduction in the expression of IGF2 isoform 2. In individuals who do not carry the protective allele, expression of IGF2 isoform 2 in adipose is positively correlated with both incidence of T2D and increased plasma glycated hemoglobin in individuals without T2D, providing support that the protective effects are mediated by reductions in IGF2 isoform 2. Broad phenotypic examination of carriers of the protective variant revealed no association with other disease states or impaired reproductive health. These findings suggest that reducing IGF2 isoform 2 expression in relevant tissues has potential as a new therapeutic strategy for T2D, even beyond the Latin American population, with no major adverse effects on health or reproduction.

Journal ArticleDOI
01 Jul 2017-Diabetes
TL;DR: The allelic spectrum for coding variants in AKT2 associated with disorders of glucose homeostasis is extended and bidirectional effects of variants within the pleckstrin homology domain ofAKT2 are demonstrated.
Abstract: To identify novel coding association signals and facilitate characterization of mechanisms influencing glycemic traits and type 2 diabetes risk, we analyzed 109,215 variants derived from exome array genotyping together with an additional 390,225 variants from exome sequence in up to 39,339 normoglycemic individuals from five ancestry groups. We identified a novel association between the coding variant (p.Pro50Thr) in AKT2 and fasting plasma insulin (FI), a gene in which rare fully penetrant mutations are causal for monogenic glycemic disorders. The low-frequency allele is associated with a 12% increase in FI levels. This variant is present at 1.1% frequency in Finns but virtually absent in individuals from other ancestries. Carriers of the FI-increasing allele had increased 2-h insulin values, decreased insulin sensitivity, and increased risk of type 2 diabetes (odds ratio 1.05). In cellular studies, the AKT2-Thr50 protein exhibited a partial loss of function. We extend the allelic spectrum for coding variants in AKT2 associated with disorders of glucose homeostasis and demonstrate bidirectional effects of variants within the pleckstrin homology domain of AKT2.

Journal ArticleDOI
TL;DR: Gene-based associations (P<10−10) support a role for coding variants in LIPC and LIPG with lipoprotein subclass traits and novel association signals provide further insight into the molecular basis of dyslipidemia and the etiology of metabolic disorders.
Abstract: Lipid and lipoprotein subclasses are associated with metabolic and cardiovascular diseases, yet the genetic contributions to variability in subclass traits are not fully understood. We conducted single-variant and gene-based association tests between 15.1M variants from genome-wide and exome array and imputed genotypes and 72 lipid and lipoprotein traits in 8,372 Finns. After accounting for 885 variants at 157 previously identified lipid loci, we identified five novel signals near established loci at HIF3A, ADAMTS3, PLTP, LCAT, and LIPG. Four of the signals were identified with a low-frequency (0.005

Journal ArticleDOI
TL;DR: A glutamate-to-lysine variant (rs58542926-T) in transmembrane 6 superfamily member 2 (TM6SF2) is associated with increased fatty liver disease and diabetes in conjunction with decreased cardiovascular disease risk as discussed by the authors.

Posted ContentDOI
14 Feb 2017-bioRxiv
TL;DR: This work uses ∼36 million singleton variants from 3,560 whole-genome sequences to infer fine-scale patterns of mutation rate heterogeneity and provides the most refined portrait to date of the factors contributing to genome-wide variability of the human germline mutation rate.
Abstract: Precise estimates of the single-nucleotide mutation rate and its variability are essential to the study of human genome evolution and genetic diseases. Here we use ~36 million singleton variants observed in 3,716 whole-genome sequences to characterize the heterogeneity of germline mutation rates across the genome. Adjacent-nucleotide context is the strongest predictor of mutability, with mutation rates varying by >650-fold depending on the identity of three bases upstream or downstream of the mutated site. Histone modifications, replication timing, recombination rate, and other local genomic features further modify mutability; magnitude and direction of this modification varies with the sequence context. Compared to estimates based on common variants used in previous approaches, singleton-based estimates provide a more accurate prediction of the mutation patterns seen in an independent dataset of ~46,000 de novo mutations; and incorporating the effects of genomic features further improves the prediction. The effects of sequence contexts, genomic features, and their interactions reported here capture the most refined portrait to date of the germline mutation patterns in humans.

Journal ArticleDOI
Aldi T. Kraja1, James P. Cook2, Helen R. Warren3, Praveen Surendran4  +174 moreInstitutions (66)
TL;DR: This analysis highlights several candidate genes with variation that alter protein function or gene expression for potential follow-up in BP and identifies 4 novel loci associated with BP regulation, and 1 independent variant at an established BP locus.
Abstract: Background - Genome-wide association studies have recently identified >400 loci that harbor DNA sequence variants that influence blood pressure (BP). Our earlier studies identified and validated 56 single nucleotide variants (SNVs) associated with BP from meta-analyses of exome chip genotype data. An additional 100 variants yielded suggestive evidence of association. Methods and Results - Here, we augment the sample with 140 886 European individuals from the UK Biobank, in whom 77 of the 100 suggestive SNVs were available for association analysis with systolic BP or diastolic BP or pulse pressure. We performed 2 meta-analyses, one in individuals of European, South Asian, African, and Hispanic descent (pan-ancestry, ≈475 000), and the other in the subset of individuals of European descent (≈423 000). Twenty-one SNVs were genome-wide significant (P<5×10-8) for BP, of which 4 are new BP loci: rs9678851 (missense, SLC4A1AP), rs7437940 (AFAP1), rs13303 (missense, STAB1), and rs1055144 (7p15.2). In addition, we identified a potentially independent novel BP-associated SNV, rs3416322 (missense, SYNPO2L) at a known locus, uncorrelated with the previously reported SNVs. Two SNVs are associated with expression levels of nearby genes, and SNVs at 3 loci are associated with other traits. One SNV with a minor allele frequency <0.01, (rs3025380 at DBH) was genome-wide significant. Conclusions - We report 4 novel loci associated with BP regulation, and 1 independent variant at an established BP locus. This analysis highlights several candidate genes with variation that alter protein function or gene expression for potential follow-up.

Posted ContentDOI
07 Jul 2017-bioRxiv
TL;DR: The WGSPD consortium will integrate data for 18,000 individuals with psychiatric disorders, beginning with autism spectrum disorder, schizophrenia, bipolar disorder, and major depressive disorder, along with over 150,000 controls, to assess the role of rare variants in the noncoding gene-regulating genome.
Abstract: As technology advances, whole genome sequencing (WGS) is likely to supersede other genotyping technologies. The rate of this change depends on its relative cost and utility. Variants identified uniquely through WGS may reveal novel biological pathways underlying complex disorders and provide high-resolution insight into when, where, and in which cell type these pathways are affected. Alternatively, cheaper and less computationally intensive approaches may yield equivalent insights. Understanding the role of rare variants in the noncoding gene-regulating genome, through pilot WGS projects, will be critical to determine which of these two extremes best represents reality. With large cohorts, well-defined risk loci, and a compelling need to understand the underlying biology, psychiatric disorders have a role to play in this preliminary WGS assessment. The WGSPD consortium will integrate data for 18,000 individuals with psychiatric disorders, beginning with autism spectrum disorder, schizophrenia, bipolar disorder, and major depressive disorder, along with over 150,000 controls.

Journal ArticleDOI
14 Nov 2017-Diabetes
TL;DR: Data demonstrate that the p.P50T substitution of AKT2 influences insulin-mediated GU in multiple insulin-sensitive tissues and may explain, at least in part, the increased risk of type 2 diabetes in p.
Abstract: Rare fully penetrant mutations in AKT2 are an established cause of monogenic disorders of glucose metabolism. Recently, a novel partial loss-of-function AKT2 coding variant (p.Pro50Thr) was identified that is nearly specific to Finns (frequency 1.1%), with the low-frequency allele associated with an increase in fasting plasma insulin level and risk of type 2 diabetes. The effects of the p.Pro50Thr AKT2 variant (p.P50T/AKT2) on insulin-stimulated glucose uptake (GU) in the whole body and in different tissues have not previously been investigated. We identified carriers (N = 20) and matched noncarriers (N = 25) for this allele in the population-based Metabolic Syndrome in Men (METSIM)study and invited these individuals back for positron emission tomography study with [18F]-fluorodeoxyglucose during euglycemic hyperinsulinemia. When we compared p.P50T/AKT2 carriers to noncarriers, we found a 39.4% reduction in whole-body GU (P = 0.006) and a 55.6% increase in the rate of endogenous glucose production (P = 0.038). We found significant reductions in GU in multiple tissues-skeletal muscle (36.4%), liver (16.1%), brown adipose (29.7%), and bone marrow (32.9%)-and increases of 16.8-19.1% in seven tested brain regions. These data demonstrate that the p.P50T substitution of AKT2 influences insulin-mediated GU in multiple insulin-sensitive tissues and may explain, at least in part, the increased risk of type 2 diabetes in p.P50T/AKT2 carriers.