Skip to main content
Erschienen in: Diabetologia 12/2017

Open Access 13.09.2017 | Article

Transethnic insight into the genetics of glycaemic traits: fine-mapping results from the Population Architecture using Genomics and Epidemiology (PAGE) consortium

verfasst von: Stephanie A. Bien, James S. Pankow, Jeffrey Haessler, Yinchang N. Lu, Nathan Pankratz, Rebecca R. Rohde, Alfred Tamuno, Christopher S. Carlson, Fredrick R. Schumacher, Petra Bůžková, Martha L. Daviglus, Unhee Lim, Myriam Fornage, Lindsay Fernandez-Rhodes, Larissa Avilés-Santa, Steven Buyske, Myron D. Gross, Mariaelisa Graff, Carmen R. Isasi, Lewis H. Kuller, JoAnn E. Manson, Tara C. Matise, Ross L. Prentice, Lynne R. Wilkens, Sachiko Yoneyama, Ruth J. F. Loos, Lucia A. Hindorff, Loic Le Marchand, Kari E. North, Christopher A. Haiman, Ulrike Peters, Charles Kooperberg

Erschienen in: Diabetologia | Ausgabe 12/2017

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

Elevated levels of fasting glucose and fasting insulin in non-diabetic individuals are markers of dysregulation of glucose metabolism and are strong risk factors for type 2 diabetes. Genome-wide association studies have discovered over 50 SNPs associated with these traits. Most of these loci were discovered in European populations and have not been tested in a well-powered multi-ethnic study. We hypothesised that a large, ancestrally diverse, fine-mapping genetic study of glycaemic traits would identify novel and population-specific associations that were previously undetectable by European-centric studies.

Methods

A multiethnic study of up to 26,760 unrelated individuals without diabetes, of predominantly Hispanic/Latino and African ancestries, were genotyped using the Metabochip. Transethnic meta-analysis of racial/ethnic-specific linear regression analyses were performed for fasting glucose and fasting insulin. We attempted to replicate 39 fasting glucose and 17 fasting insulin loci. Genetic fine-mapping was performed through sequential conditional analyses in 15 regions that included both the initially reported SNP association(s) and denser coverage of SNP markers. In addition, Metabochip-wide analyses were performed to discover novel fasting glucose and fasting insulin loci. The most significant SNP associations were further examined using bioinformatic functional annotation.

Results

Previously reported SNP associations were significantly replicated (p ≤ 0.05) in 31/39 fasting glucose loci and 14/17 fasting insulin loci. Eleven glycaemic trait loci were refined to a smaller list of potentially causal variants through transethnic meta-analysis. Stepwise conditional analysis identified two loci with independent secondary signals (G6PC2-rs477224 and GCK-rs2908290), which had not previously been reported. Population-specific conditional analyses identified an independent signal in G6PC2 tagged by the rare variant rs77719485 in African ancestry. Further Metabochip-wide analysis uncovered one novel fasting insulin locus at SLC17A2-rs75862513.

Conclusions/interpretation

These findings suggest that while glycaemic trait loci often have generalisable effects across the studied populations, transethnic genetic studies help to prioritise likely functional SNPs, identify novel associations that may be population-specific and in turn have the potential to influence screening efforts or therapeutic discoveries.

Data availability

The summary statistics from each of the ancestry-specific and transethnic (combined ancestry) results can be found under the PAGE study on dbGaP here: https://​www.​ncbi.​nlm.​nih.​gov/​projects/​gap/​cgi-bin/​study.​cgi?​study_​id=​phs000356.​v1.​p1
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-017-4405-1) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s00125-017-4476-z.
Abkürzungen
AA
African ancestry
AFR
African ancestry (1000 Genomes Super Population Code)
AI/AN
American Indian/Alaskan Native
AMR
Admixed American ancestry (1000 Genomes Super Population Code)
ARIC
Atherosclerosis Risk in Communities
ASN
Asian and Pacific Islander
CARDIA
Coronary Artery Risk Development in Young Adults
CEU
Utah Residents (CEPH) with Northern and Western European Ancestry (HapMap Population Code)
EUR
European ancestry (1000 Genomes Super Population Code)
GWAS
Genome-wide association studies
HCHS/SOL
Hispanic Community Health Study/Study of Latinos
H/L
Hispanic/Latino
MAF
Minor allele frequency
MAGIC
Meta-Analyses of Glucose and Insulin-related traits
MEC
The Multiethnic Cohort
NHGRI
National Human Genome Research Institute
PAGE
Population Architecture using Genetic Epidemiology
SHARe
WHI SNP Health Association Resource
WHI
Women’s Health Initiative

Introduction

Type 2 diabetes is a growing epidemic that disproportionally burdens US minority populations [1]. Elevated levels of fasting glucose and fasting insulin in individuals without diabetes are markers of dysregulated glucose metabolism and are strong risk factors for type 2 diabetes [2]. Although twin and family studies provide heritability estimates of 10–50% for these traits [3, 4], family-based linkage studies have been largely unsuccessful in identifying specific contributing loci. Genome-wide association studies (GWAS) greatly accelerated the pace of discovery of genetic variants contributing to glycaemic traits. For example, the Meta-Analyses of Glucose and Insulin-related traits (MAGIC) consortium performed a large-scale investigation of glycaemic traits in individuals of European descent without diabetes and identified 24 fasting glucose loci and eight fasting insulin loci, three of which were associated with both traits [5, 6]. These findings have implicated genes and pathways known to be related to glucose metabolism (e.g. GCK/G6PC2 and glucose dephosphorylation), as well as novel pathways (e.g. MTNR1B and circadian rhythmicity). However, in some instances, the interpretation of GWAS findings has been challenging. For instance, many of the known loci are positioned in non-coding, putative regulatory regions of the genome, which in turn makes it difficult to identify the gene target(s). Additionally, the most significant variant is often not the causal variant but is a correlated variant in linkage disequilibrium with the functional variant(s).
While early GWAS efforts were focused on populations of European descent, initial attempts to generalise GWAS findings to more diverse populations have had limited success [79]. Importantly, these studies tended to be small and only included the initial most significant GWAS variant (index SNP). However, it is critical that transethnic investigation of GWAS loci include both the index variant and all correlated variants, given that patterns of linkage disequilibrium vary by ancestry and the functional SNP(s) are rarely known. On average, European populations have more highly correlated SNPs and extended haplotypes in comparison with populations of African ancestry (AA). Hispanic/Latino (H/L) populations, on the other hand, are more admixed with highly variable contributions of African, European and New World ancestry. Due in part to reduction in linkage disequilibrium with neighbouring SNPs, transethnic studies can utilise these differences across and within admixed populations to localise causal variants, and discover novel population-specific associations that were undetectable in genetically homogeneous studies. Thus, transethnic studies may provide insight into the underlying biology of complex traits, which may differ among groups.
The Metabochip was developed to fine-map GWAS loci for metabolic and cardiovascular traits, as well as replicate promising loci with suggestive, but not genome-wide, significant p values [10]. Among the 196,725 Metabochip variants selected for fine-mapping metabolic and cardiovascular-related loci, approximately 40,000 were selected for type 2 diabetes and related biomarkers. Among the 39 fasting glucose loci and 17 fasting insulin loci [5, 6] that were available for replication, 15 loci included not only the index SNP but also denser coverage of SNPs on the Metabochip that could be utilised for fine-mapping. Importantly, despite very large sample sizes, attempted Metabochip fine-mapping in a population of European descent generally did not yield stronger associations than the original GWAS index SNP and did not reduce the number of SNPs reaching similar levels of significance [11]. As such, this effort was unable to narrow in on functional candidate SNP(s).
This study examined the association of Metabochip SNPs with fasting glucose and fasting insulin in a multiethnic study of up to 26,760 participants: 14,953 H/L, 10,380 AA, 998 Asian and Pacific Islander (ASN) and 429 American Indian/Alaskan Native (AI/AN) populations from the Population Architecture using Genetic Epidemiology (PAGE) consortium. Specifically, we carried out the following procedures: (1) tested the association of index SNPs previously reported for 39 fasting glucose and 17 fasting insulin loci from studies of individuals of European descent; (2) used transethnic meta-analysis to refine known glycaemic trait loci in 15 loci which were densely covered with SNPs on the Metabochip; (3) investigated remaining metabolic and cardiovascular trait loci on the Metabochip for association with these glycaemic traits and (4) performed bioinformatic functional annotation of the most significant (lead) SNPs to further prioritise likely causal variants.

Methods

Ethics statement
This study was performed in accordance with the tenets of the Declaration of Helsinki and approved by the Institutional Review Boards of each participating study. All study participants provided written informed consent.
Study population and trait measurement
The PAGE consortium was funded by the National Human Genome Research Institute (NHGRI) to investigate the epidemiological architecture of well-replicated genetic variants associated with human diseases or traits [12]. This analysis includes self-reported H/L, AA, ASN and AI/AN individuals without diabetes, aged 18 years or over, from the Multiethnic Cohort Study (MEC), the Women’s Health Initiative (WHI), Atherosclerosis Risk in Communities (ARIC), Coronary Artery Risk Development in Young Adults (CARDIA), the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) and the Mount Sinai School of Medicine’s (MSSM) DNA biobank (BioMe). Further details about each cohort can be found in the electronic supplementary materials (ESM) Methods (study population and trait measurement section).
Fasting glucose and fasting insulin concentrations were measured using standard assays, at laboratories specific to each PAGE site (ESM Table 1). Individuals self-reporting that they had ever been diagnosed with diabetes or taken diabetes medications or who had fasting blood glucose levels ≥ 6.99 mmol/l (≥ 126 mg/dl) were excluded from analyses. Individuals with BMI < 16.5 kg/m2 or BMI > 70 kg/m2 were also excluded on the assumption that these extremes could be attributable to data coding errors or underlying illness or could reflect a familial syndrome. Prior to analyses, each study removed race/ethnicity outliers using ancestry informative principal components.
After exclusions, fasting glucose analyses consisted of 14,953 H/L, 10,380 AA, 998 ASN and 429 AI/AN individuals. Fasting insulin analyses involved fewer individuals: 12,895 H/L, 8361 AA, 998 ASN and 420 AI/AN. Fasting insulin was not available for BioMe. Race/ethnicity was self-reported. Descriptive characteristics of PAGE study participants by cohort can be found in ESM Table 2. While ASN and AI/AN were included for transethnic meta-analysis, population-specific analyses were underpowered due to small sample sizes. As such, ASN and AI/AN population-specific analyses were used as a comparison for consistency in the direction of effect.
Genotyping and quality control
Genotyping was performed using the Metabochip, the design of which has been described elsewhere [10]. In brief, the 200K Metabochip is designed to cost effectively analyse putative association signals identified through GWAS of many glucose- and insulin-related metabolic and cardiovascular traits and to fine-map established loci [10]. More than 122,000 SNPs were included to fine-map 257 GWAS loci for 23 traits [10]. Fine-mapping loci were defined as the GWAS index SNP and all correlated SNPs (r 2 ≥ 0.5) that were within 0.02 cM of the index and having a minor allele frequency (MAF) > 1% in at least one HapMap Phase I population. SNPs were excluded if the Illumina design score was < 0.5 or there were SNPs within 15 bp of the SNP of interest with MAF of > 2% among Europeans (CEU [HapMap Population Code for Utah residents (CEPH) with Northern and Western European ancestry]).
Metobochip genotyping was performed for MEC, ARIC, CARDIA, HCHS/SOL and WHI [13] individuals. Standard quality control filters were applied for samples and SNPs, including missing rate and Hardy–Weinberg equilibrium (p < 1 × 10−7). A portion of WHI individuals of AA had both Metabochip and the Affymetrix 6.0 genotype data available from the SNP Health Association Resource (SHARe); this was used to impute Metabochip SNPs in the remaining SHARe participants with only Affymetrix 6.0 GWAS [8] and only dosages with imputation R 2 > 0.3 were included in the analyses. In BioMe, genotypes from the Illumina HumanOmniExpress array were imputed to 1000 Genome Phase I haplotype panels (March 2012) [14]. Metabochip SNPs with ‘proper info’ score ≥ 0.4 were included in the analysis. Principal components were determined within each study using the Eigensoft software [15]. We excluded SNPs with a minor allele count less than 5 within each study by racial/ethnic population. The sample success rate and concordance rate for duplicate pairs across all studies was ≥ 95% and ≥ 99%, respectively. Further genotyping and analytical characteristics of the participating studies are further summarised in ESM Methods (genotyping and quality control section) and ESM Table 1.
Replication and fine-mapping approach
The overall study design for replication, fine-mapping and discovery of novel loci is summarised in Fig. 1. For replication of known loci, unconditional association analyses were performed for previously reported index SNPs listed in ESM Table 3. A nominal significance level (α = 0.05) was used to define replication of a locus. Next, unconditional association analyses were performed for all SNPs in a locus by race/ethnicity and by transethnic meta-analysis. A locus-specific p value threshold was defined as 0.05 divided by the number of SNPs passing quality control in each region (ranging from α = 1.4 × 10−5 to α = 4.1 × 10−4, Table 1). Locus-specific significance was used to conservatively adjust for multiple testing, while also acknowledging that genetic variation is known to influence glycaemic traits in these regions. Linkage disequilibrium was calculated for PAGE H/L, AA and Asian samples with 500 kb sliding windows using PLINK [16]. Metabochip linkage disequilibrium and frequency information in Europeans was provided by the 1000 Genomes Phase 3 population. These linkage disequilibrium patterns were used to evaluate locus refinement. Additionally, LocusZoom plots [17] were used to graphically display the fine-mapping results and linkage disequilibrium for these plots used 1000 Genomes Phase I Super Populations (European ancestry [EUR], admixed American ancestry [AMR], African ancestry [AFR]). After identifying the most significant lead SNP in each region, we searched for additional independent association signals by including the lead SNP in the conditional model and then testing each of the remaining SNPs in a region. These conditional analyses were repeated, adding in the lead SNP and conditional lead SNP(s), until no SNP in the model had a conditional p value less than the locus-specific significance. Sequential conditional analyses were performed for each race/ethnicity and transethnic meta-analysis. Further details on our approach to locus refinement are provided in ESM Methods (replication and fine-mapping of known glycaemic trait loci section).
Table 1
Characterisation of 15 fine-mapping genomic regions analysed for fasting glucose and fasting insulin
Chromosome
Locus
Base pair range (GRCh37/hg19)
No. of SNPs on Metabochip
No. of SNPsa
α
Trait
1q32.3
PROX1
214,124,818–214,167,508
153
129
3.9 × 10−4
Glucose
2p23.3
GCKR
27,389,634–27,951,658
1099
966
5.2 × 10−5
Both
2q31.1
G6PC2
169,752,640–169,814,655
240
211
2.4 × 10−4
Glucose
3q21.1
ADCY5
122,976,919–123,206,919
924
786
6.2 × 10−5
Glucose
3q26.2
SLC2A2
170,532,111–170,769,171
717
653
7.7 × 10−5
Glucose
7p21.2
DGKB
14,185,088–15,145,520
3894
3555
1.4 × 10−5
Glucose
7p13
GCK
44,222,003–44,266,077
148
122
4.1 × 10−4
Glucose
9p24.2
GLIS3
4,243,162–4,310,558
419
385
1.3 × 10−4
Glucose
10q25.2
ADRA2A/TCF7L2
112,967,738–113,053,039
462
424
1.2 × 10−4
Glucose
11p15.4
CRY2
45,706,162–46,162,829
1082
921
5.4 × 10−5
Glucose
11p11.2
MADD
46,921,641–48,091,303
2392
2037
2.5 × 10−5
Glucose
11q12.2
FADS2
61,505,583–61,751,624
726
643
7.8 × 10−5
Glucose
11q14.3
MTNR1B
92,667,047–92,725,321
214
180
2.8 × 10−4
Glucose
12q23.2
IGF1
103,851,897–104,450,976
1307
1059
4.7 × 10−5
Insulin
15q22.2
C2CD4A
62,099,182–62,520,109
1143
949
5.3 × 10−5
Glucose
α is the Bonferroni significance threshold (0.05/no. of SNPs passing quality control) used to define region-specific significance
aNo. of SNPs passing quality control in the transethnic meta-analysis
Discovery of novel loci
Metabochip-wide analyses were performed to identify novel associations with fasting glucose and fasting insulin. Statistical significance for the Metabochip-wide analysis was set at 0.05 divided by the number of Metabochip SNPs passing quality control (α = 2.7 × 10−7). Results were examined through qq plots and Manhattan plots for each model, highlighting known regions defined in ESM Table 4. Further details are provided in ESM Methods (strategy for selecting novel associations section).
Statistical analysis
First, in each study with unrelated individuals we performed race/ethnic-specific analyses for fasting glucose and natural log-transformed fasting insulin, excluding ancestry outliers and first-degree relatives. In HCHS/SOL, a weighted version of generalised estimation equations was used to account for unequal inclusion probabilities and complex family-based sampling designs [18]. Models adjusted for age, sex (except WHI), study site (as applicable), smoking status (current vs former/never), continuous BMI and ancestry principal components. Like previous studies [11], primary analyses adjusted for BMI because it is a major risk factor for type 2 diabetes and is correlated with glycaemic traits. For comparison, all models were also run without adjustment for BMI. Next, fixed-effect models with inverse-variance weighting were used to pool the study-specific SNP effect estimates and their standard errors by race/ethnicity as implemented in METAL [19]. Finally, summary statistics from METAL for H/L, AA, NA/AI and ASN were combined using inverse-variance weighted fixed effects meta-analysis in METAL. Q statistics and I 2 were used to evaluate heterogeneity across studies and race/ethnicity. Further details are provided in ESM Methods (statistical analysis section).
Functional annotation
Detailed information on the functional annotation methods and various datasets used is provided in ESM Methods (functional annotation section). In brief, it is expected that the lead SNPs are more likely to be functional or to be in stronger linkage disequilibrium with underlying functional variant(s). Therefore, lead SNPs and all correlated SNPs (r 2 > 0.2 in 1000 Genomes Phase 3 AFR/AMR populations) were annotated using publicly available functional datasets. Potential functional effects were assessed using PolyPhen2 [20] (http://​genetics.​bwh.​harvard.​edu/​pph2/​, accessed 24 August 2016) for non-synonymous variants, SPANR (http://​tools.​genes.​toronto.​edu/) [21] for variants near splice sites, TargetScan miRNA Regulatory Sites for 3′-UTR regions [22], ENCODE/NIH Roadmap data [2325] and GTEx (https://​www.​gtexportal.​org/​home/​) [26] to identify non-coding variants positioned in predicted regulatory elements.

Results

Demographics
We included a total of 26,760 participants (14,953 H/L, 10,380 AA, 998 ASN, and 429 AI/AN) in fasting glucose analyses. The sample sizes for fasting insulin analyses were slightly smaller, with a total of 22,674 participants (12,895 H/L, 8361 AA, 998 ASN and 420 AI/AN). The mean age across the five cohorts was 55 years for men and 59 years for women (range 18–93 years). Study-specific descriptive characteristics are shown in ESM Table 2. Particularly due to the inclusion of the WHI cohort, the proportion of women in the total study population was high, with the highest fraction observed in AA (82.6% for fasting glucose and 97.1% for fasting insulin). Glycaemic trait distributions were similar across studies and ethnicities, with average fasting glucose levels ranging from 4.7 ± 0.7 mmol/l to 5.5 ± 0.6 mmol/l and average fasting insulin levels ranging from 43.3 ± 23.6 pmol/l to 75.9 ± 38.8 pmol/l.
Generalisation of European glycaemic trait loci
We found that 31/39 (79.5%) fasting glucose loci and 14/17 (82.3%) fasting insulin loci had a p value smaller than 0.05. Index SNP associations were directionally consistent in our transethnic PAGE meta-analysis and only four SNPs had heterogeneity p values less than 0.05 (Table 2). The effect estimates (βs) of index SNPs in the transethnic meta-analysis were very similar to those published in Metabochip analysis of individuals of European descent (Pearson’s r 2 = 0.86, 95% CI 0.78, 0.91; p < 2.2 × 10−16; ESM Fig. 1). At three loci (WARS, GIPR and DPYSLS) we observed replication in only H/L and not the transethnic meta-analysis. Interestingly, while the sample sizes were much smaller for Asian individuals than for H/L and AA individuals, the transethnic meta-analysis of the PROX1 index (rs340874) was only nominally significant and directionally consistent in the Asian samples. In the remaining loci that did not replicate in transethnic meta-analysis or the race/ethnic-specific analyses, the effects were generally similar or at least in the same direction. Analyses without inclusion of BMI as a covariate were generally similar, with slightly lower significance at some loci. Full summary statistics for models with and without BMI covariate are reported in ESM Table 5 and ESM Table 6, respectively.
Table 2
Replication of European Metabochip index SNPs for 39 fasting glucose and 17 fasting insulin loci via transethnic meta-analysis
Locus/gene
Lead EUR
C/NC allele
Coded allele frequency
Effect β of coded allele (SE)
Analyses with p < 0.05
p value TE Meta (Het.)
   
EUR
H/L
AA
ASN
TE Meta
EUR
H/L
AA
ASN
TE Meta
  
Fasting glucose loci (NTE = 26,760, NEUR = 118,881)
 1q32.3
PROX1
rs340874
A/G
0.48
0.60
0.82
0.61
0.67
−0.015 (0.002)
−0.004 (0.006)
−0.009 (0.009)
0.076 (0.027)
−0.003 (0.005)
ASN
0.59 (0.02)
 2p23.3
GCKR
rs780094
A/G
0.39
0.35
0.19
0.52
0.30
−0.029 (0.002)
−0.033 (0.007)
−0.016 (0.010)
−0.051 (0.027)
−0.029 (0.005)
H/L, ASN, TE
2 × 10−8 (0.2)
 2q31.1
G6PC2
rs560887
A/G
0.30
0.17
0.07
0.03
0.14
−0.075 (0.003)
−0.086 (0.008)
−0.063 (0.014)
−0.065 (0.077)
−0.079 (0.007)
H/L, AA, TE
1 × 10−29 (0.48)
 3q21.1
ADCY5
rs11708067
A/G
0.79
0.75
0.84
0.96
0.78
0.024 (0.003)
0.021 (0.007)
0.052 (0.010)
−0.254 (0.171)
0.031 (0.006)
H/L, AA, TE
5 × 10−8 (0.02)
 3q26.2
SLC2A2
rs1280
A/G
0.86
0.84
0.65
0.97
0.73
0.031 (0.003)
0.052 (0.009)
−0.001 (0.007)
0.043 (0.082)
0.021 (0.006)
H/L, TE
1 × 10−4 (2 × 10−5)
 7p21.2
DGKB
rs2191349
A/C
0.53
0.48
0.57
0.69
0.51
0.032 (0.002)
0.023 (0.006)
0.005 (0.009)
0.003 (0.028)
0.017 (0.005)
H/L, TE
8 × 10−4 (0.42)
 7p13
GCK
rs730497
A/G
0.16
0.20
0.18
0.18
0.20
0.061 (0.003)
0.061 (0.008)
0.056 (0.009)
0.004 (0.034)
0.057 (0.006)
H/L, AA, TE
3 × 10−22 (0.37)
 8q24.11
SLC30A8
rs11558471
A/G
0.68
0.75
0.90
0.57
0.77
0.032 (0.002)
0.018 (0.007)
0.014 (0.012)
−0.004 (0.026)
0.017 (0.006)
H/L, TE
4 × 10−3 (0.22)
 9p24.2
GLIS3
rs10814916
A/C
0.49
0.43
0.33
0.54
0.40
−0.017 (0.002)
−0.016 (0.006)
−0.009 (0.008)
−0.066 (0.027)
−0.015 (0.005)
H/L, ASN, TE
1 × 10−3 (0.21)
 10q25.2
ADRA2A
rs11195502
A/G
0.09
0.13
0.34
0.07
0.25
−0.036 (0.004)
−0.014 (0.010)
−0.012 (0.008)
−0.022 (0.054)
−0.013 (0.006)
TE
0.04 (0.62)
 10q25.2
TCF7L2
rs4506565
A/T
0.70
0.71
0.56
0.93
0.64
−0.024 (0.002)
−0.030 (0.007)
−0.019 (0.007)
−0.137 (0.060)
−0.025 (0.005)
All
3 × 10−7 (0.19)
 11p11.2
CRY2
rs11605924
A/C
0.49
0.54
0.86
0.81
0.63
0.022 (0.002)
0.017 (0.006)
0.027 (0.011)
−0.066 (0.034)
0.018 (0.005)
All
1 × 10−3 (0.03)
 11p11.2
MADD
rs11039182
A/G
0.73
0.82
0.95
0.97
0.85
0.023 (0.003)
0.000 (0.009)
0.021 (0.016)
−0.002 (0.091)
0.004 (0.007)
None
0.55 (0.67)
 11q12.2
FADS2
rs174550
A/G
0.66
0.52
0.91
0.57
0.60
0.018 (0.002)
0.026 (0.007)
0.036 (0.013)
0.039 (0.027)
0.029 (0.006)
H/L, AA, TE
7 × 10−7 (0.9)
 11q14.3
MTNR1B
rs10830963
C/G
0.71
0.79
0.93
0.60
0.81
−0.078 (0.003)
−0.062 (0.008)
−0.090 (0.014)
−0.078 (0.026)
−0.068 (0.006)
All
7 × 10−27 (0.21)
 15q22.2
C2CD4A
rs4502156
A/G
0.55
0.40
0.26
0.52
0.35
0.023 (0.002)
0.017 (0.007)
0.006 (0.008)
0.008 (0.026)
0.012 (0.005)
H/L, TE
0.01 (0.77)
 9p21.3
CDKN2B
rs10811661
A/G
0.82
0.86
0.93
0.56
0.86
0.024 (0.003)
0.021 (0.009)
0.017 (0.014)
0.072 (0.026)
0.024 (0.007)
H/L, ASN, TE
0.02 (0.29)
 5q15
PCSK1
rs4869272
A/G
0.69
0.75
0.78
0.73
0.76
0.018 (0.002)
0.021 (0.007)
0.019 (0.008)
0.032 (0.029)
0.020 (0.005)
H/L, AA, TE
1 × 10−3 (0.97)
 13q12.2
PDX1
rs11619319
A/G
0.77
0.71
0.83
0.55
0.75
−0.020 (0.002)
−0.008 (0.007)
−0.017 (0.010)
−0.054 (0.026)
−0.012 (0.006)
AA, ASN, TE
0.05 (0.32)
 8p23.1
PPP1R3B
rs983309
A/C
0.12
0.21
0.28
0.02
0.24
0.026 (0.003)
0.023 (0.008)
0.017 (0.008)
0.004 (0.104)
0.020 (0.006)
H/L, AA, TE
2 × 10−3 (0.96)
 7p12.1
GRB10
rs6943153
A/G
0.34
0.45
0.68
0.28
0.54
0.015 (0.002)
0.019 (0.006)
−0.004 (0.008)
−0.010 (0.030)
0.009 (0.005)
H/L, TE
0.07 (0.11)
 11q13.4
ARAP1
rs11603334
A/G
0.17
0.08
0.05
0.05
0.07
−0.019 (0.003)
−0.030 (0.011)
−0.039 (0.016)
−0.086 (0.067)
−0.033 (0.009)
H/L, AA, TE
1 × 10−5 (0.69)
 20p11.21
FOXA2
rs6113722
A/G
0.04
0.05
0.16
0.18
0.13
−0.035 (0.005)
−0.042 (0.014)
−0.040 (0.010)
−0.090 (0.033)
−0.043 (0.008)
All
2 × 10−6 (0.55)
 9q31.3
IKBKAP
rs16913693
A/C
0.97
0.96
0.77
1
0.81
0.043 (0.007)
0.010 (0.017)
−0.012 (0.008)
0.334 (0.333)
−0.008 (0.008)
None
0.51 (0.48)
 9q34.3
DNLZ
rs3829109
A/G
0.29
0.33
0.17
0.13
0.28
−0.017 (0.003)
−0.021 (0.007)
−0.026 (0.010)
0.000 (0.040)
−0.022 (0.006)
H/L, AA, TE
5 × 10−4 (0.91)
 14q32.2
WARS
rs3783347
A/C
0.21
0.12
0.06
0.1
0.11
−0.017 (0.003)
−0.023 (0.010)
0.000 (0.014)
0.000 (0.044)
−0.014 (0.008)
H/L
0.08 (0.40)
 19q13.32
GIPR
rs2302593
C/G
0.5
0.51
0.28
0.39
0.42
0.014 (0.002)
−0.013 (0.006)
−0.002 (0.008)
0.019 (0.027)
−0.008 (0.005)
H/L
0.05 (0.55)
 6p22.3
CDKAL1
rs9368222
A/C
0.28
0.23
0.19
0.41
0.23
0.014 (0.002)
0.025 (0.007)
0.025 (0.009)
0.041 (0.026)
0.026 (0.006)
H/L, AA, TE
3 × 10−5 (0.94)
 12q24.33
P2RX2
rs10747083
A/G
0.66
0.69
0.85
0.83
0.74
0.013 (0.002)
0.010 (0.007)
0.012 (0.011)
−0.017 (0.034)
0.010 (0.006)
None
0.12 (0.88)
 20q12
TOP1
rs6072275
A/G
0.16
0.12
0.08
0.02
0.11
0.016 (0.003)
0.021 (0.010)
0.019 (0.013)
−0.075 (0.121)
0.021 (0.008)
H/L, TE
5 × 10−3 (0.53)
 3q27.2
IGF2BP2
rs7651090
A/G
0.69
0.7
0.46
0.7
0.59
−0.013 (0.002)
−0.011 (0.007)
−0.011 (0.007)
−0.023 (0.029)
−0.011 (0.005)
TE
0.07 (0.90)
 13q13.1
KL
rs576674
A/G
0.85
0.68
0.4
0.85
0.56
−0.017 (0.003)
−0.026 (0.007)
−0.014 (0.007)
0.054 (0.038)
−0.019 (0.005)
H/L, AA, TE
7 × 10−4 (0.08)
 3p21.31
AMT
rs11715915
A/G
0.32
0.21
0.24
0.08
0.22
−0.012 (0.002)
−0.007 (0.008)
0.003 (0.008)
0.053 (0.051)
−0.002 (0.006)
None
0.59 (0.56)
 6p24.3
RREB1
rs17762454
A/G
0.26
0.33
0.16
0.41
0.28
0.012 (0.002)
0.017 (0.007)
0.012 (0.010)
0.011 (0.027)
0.015 (0.005)
H/L, TE
0.02 (0.97)
 5q13.3
ZBED3
rs7708285
A/G
0.73
0.69
0.85
0.91
0.74
−0.011 (0.003)
−0.004 (0.007)
0.003 (0.010)
0.002 (0.060)
−0.003 (0.006)
None
0.4 (0.47)
 12q13.3
GLS2
rs2657879
A/G
0.82
0.81
0.93
NA
0.83
−0.012 (0.003)
−0.011 (0.008)
0.016 (0.015)
−0.005 (0.007)
None
0.43 (0.11)
 2p23.3
DPYSL5
rs1371614
A/G
0.25
0.38
0.35
0.16
0.36
0.020 (0.004)
0.021 (0.007)
−0.006 (0.007)
−0.021 (0.036)
0.009 (0.005)
H/L
0.03 (0.05)
 15q22.2
C2CD4B
rs12440695*
A/G
0.63
0.57
0.83
0.71
0.65
0.008 (0.003)
0.004 (0.007)
−0.002 (0.009)
−0.011 (0.028)
0.003 (0.005)
None
0.63 (0.58)
 11p11.2
OR4S1
rs1483121
A/G
0.14
0.09
0.03
0.03
0.08
−0.027 (0.005)
0.008 (0.011)
−0.022 (0.022)
−0.101 (0.220)
0.002 (0.010)
None
0.59 (0.62)
Fasting insulin loci (NTE = 22,674, NEUR = 99,029)
 1q41
LYPLAL1
rs4846565
A/G
0.33
0.41
0.09
0.34
0.32
−0.013 (0.002)
−0.023 (0.008)
−0.007 (0.013)
0.022 (0.028)
−0.017 (0.007)
H/L, TE
0.01 (0.34)
 2p23.3
GCKR
rs780094
A/G
0.39
0.35
0.19
0.52
0.30
−0.029 (0.002)
−0.031 (0.008)
−0.029 (0.010)
−0.011 (0.027)
−0.030 (0.006)
H/L, AA, TE
2 × 10−7 (0.41)
 2q24.3
GRB14
rs10195252
A/G
0.60
0.67
0.28
0.89
0.49
0.017 (0.002)
0.041 (0.008)
0.036 (0.008)
−0.044 (0.044)
0.037 (0.006)
H/L, AA, TE
1 × 10−10 (0.29)
 2q36.3
IRS1
rs2943645
A/G
0.63
0.74
0.63
0.90
0.68
0.019 (0.002)
0.018 (0.009)
0.012 (0.008)
0.062 (0.046)
0.016 (0.006)
H/L, TE
4 × 10−3 (0.54)
 3p25.2
PPARG
rs17036328
A/G
0.86
0.89
0.83
0.95
0.85
0.021 (0.003)
0.038 (0.012)
0.009 (0.010)
0.036 (0.068)
0.022 (0.007)
H/L, TE
2 × 10−3 (0.15)
 4q22.1
FAM13A
rs3822072
A/G
0.48
0.44
0.51
0.63
0.47
0.012 (0.002)
0.008 (0.008)
0.018 (0.010)
0.024 (0.028)
0.012 (0.006)
AA, TE
0.04 (0.82)
 4q24
TET2
rs974801
A/G
0.62
0.58
0.72
0.40
0.64
−0.014 (0.002)
−0.018 (0.008)
−0.009 (0.008)
−0.023 (0.027)
−0.015 (0.006)
H/L, TE
6 × 10−3 (0.31)
 4q32.1
PDGFC
rs6822892
A/G
0.68
0.59
0.27
0.70
0.45
0.014 (0.002)
0.012 (0.008)
0.003 (0.008)
0.009 (0.029)
0.009 (0.006)
None
0.12 (0.76)
 5q11.2
ARL15
rs4865796
A/G
0.67
0.79
0.75
0.81
0.77
0.015 (0.002)
0.016 (0.009)
0.024 (0.008)
0.006 (0.036)
0.020 (0.006)
AA, TE
9 × 10−4 (0.80)
 5q11.2
ANKRD55
rs459193
A/G
0.27
0.27
0.42
0.52
0.36
−0.015 (0.002)
−0.025 (0.009)
−0.022 (0.008)
−0.040 (0.026)
−0.022 (0.006)
All
4 × 10−5 (0.30)
 6p21.31
UHRF1BP1
rs6912327
A/G
0.80
0.69
0.35
NA
0.51
0.016 (0.003)
0.004 (0.008)
−0.004 (0.008)
0.001 (0.006)
None
0.83 (0.08)
 6q22.33
RSPO3
rs2745353
A/G
0.51
0.58
0.60
0.61
0.59
0.011 (0.002)
0.016 (0.008)
0.010 (0.008)
−0.039 (0.027)
0.011 (0.005)
H/L, TE
0.03 (0.25)
 7q11.23
HIP1
rs1167800
A/G
0.54
0.67
0.84
0.69
0.73
0.011 (0.002)
0.018 (0.008)
0.009 (0.010)
−0.004 (0.028)
0.011 (0.006)
H/L
0.08 (0.07)
 8p23.1
PPP1R3B
rs983309
A/C
0.13
0.21
0.28
0.02
0.25
0.022 (0.003)
0.026 (0.010)
0.024 (0.008)
−0.082 (0.103)
0.026 (0.006)
All
2 × 10−5 (0.02)
 10q25.2
TCF7L2
rs7903146
A/G
0.27
0.25
0.28
0.08
0.27
−0.013 (0.002)
−0.014 (0.009)
−0.022 (0.008)
0.023 (0.057)
−0.019 (0.006)
AA, TE
1 × 10−3 (0.51)
 12q23.2
IGF1
rs35767
A/G
0.18
0.24
0.44
0.33
0.36
−0.003 (0.003)
−0.014 (0.011)
0.006 (0.008)
−0.050 (0.032)
−0.004 (0.006)
None
0.43 (0.28)
 19q13.11
PEPD
rs731839
A/G
0.66
0.61
0.63
0.48
0.61
−0.015 (0.002)
−0.016 (0.008)
−0.003 (0.008)
−0.037 (0.026)
−0.012 (0.005)
H/L, TE
0.03 (0.23)
EUR, individuals of European descent from Scott et al. [11] genotyped on Metabochip. Models included continuous BMI covariate, *rs12440695 used as a linkage disequilibrium proxy (r 2 = 0.98) for the index SNP rs11071657, which did not pass quality control. β, allelic effect size for an additive genetic model corresponding to the coded (C) allele, is shown in units of mmol/l for fasting glucose and natural log-transformed pmol/l for fasting insulin. Full results for models with and without BMI covariate for fasting glucose and fasting insulin are shown in ESM Table 5 and ESM Table 6, respectively
p values are shown for the transethnic (TE) meta-analysis and heterogeneity (Het.) in effect across populations
Fine-mapping of European glycaemic trait loci
Among the 15 glycaemic trait loci for which fine-mapping was attempted on the Metabochip, ten fasting glucose loci and two fasting insulin loci had one or more SNPs that reached locus-specific significance (α = 0.05/number of SNPs in the locus) in the transethnic meta-analysis. The p values ranged from 1.0 × 10−29 at G6PC2-rs560887 to 1.5 × 10−4 at PROX1-rs10494973 (Table 3). Although AI/AN ancestries were included in the transethnic meta-analysis, the AI/AN results are not shown because the small sample size was underpowered for population-specific analysis. At four fasting glucose loci, the most significant lead SNP in PAGE transethnic meta-analysis was the same as the European index SNP from prior Metabochip evaluation (G6PC2, ADCY5, MTNR1B and FADS2). For six fasting glucose loci (PROX1, GCKR, SLC2A2, DGKB, GCK and GLIS3) and the one fasting insulin locus (GCKR), the lead SNP in PAGE transethnic meta-analysis was in moderate or weak linkage disequilibrium with the index SNP in 1000 Genomes Population EUR (r 2 > 0.2). At these six fasting glucose loci and one fasting insulin locus, the PAGE lead SNP and EUR index SNP were not independent of each other as only one of the two SNP associations maintained nominal significance in transethnic conditional meta-analysis where both lead and index variants were included in the model. This was further supported by investigation of potential fine-mapping through locus zoom plots.
Table 3
Most significant lead SNPs in ten fasting glucose and two fasting insulin fine-mapping loci identified in transethnic meta-analysis
Region
Lead PAGE SNP
Frequency of coded (C) allele
Effect β of coded allele (SE)
p value
r 2 with EUR index SNPc
No. of LD SNPse
  
C/N
TEa
EUR
H/L
AA
ASN
TE Meta
H/L
AA
ASN
TE Metab
Het.
EUR SNPd
EUR
H/L
AA
ASN
EUR
TE (% red.)f
Fasting glucose loci
 1q32.3
PROX1
rs10494973
C/G
0.03
0.48
0.03
0.01
0.01
0.060 (0.016)
0.050 (0.018)**
0.100 (0.036)**
−0.274 (0.384)
2 × 10−4
0.44
rs340874
<0.10
<0.10
<0.10
<0.10
4
1 (75)
 2p23.3
GCKR
rs1260326
A/G
0.29
0.41
0.34
0.15
0.52
−0.032 (0.005)
−0.036 (0.007)***
−0.020 (0.010)*
−0.051 (0.026)*
2 × 10−9
0.44
rs780094
0.92
0.91
0.42
0.93
274
90 (67)
 2q31.1
G6PC2
rs560887
A/G
0.14
0.31
0.17
0.07
0.03
−0.079 (0.007)
−0.086 (0.008)***
−0.063 (0.014)***
−0.065 (0.077)
1 × 10−29
0.48
Same
1
1
1
1
118
9 (92)
 3q21.1
ADCY5
rs11708067
A/G
0.78
0.82
0.75
0.84
0.97
0.031 (0.006)
0.021 (0.007)**
0.052 (0.010)***
−0.254 (0.171)
5 × 10−8
0.02
Same
1
1
1
1
72
18 (75)
 3q26.2
SLC2A2
rs1604038
A/G
0.44
0.29
0.34
0.58
0.23
−0.026 (0.005)
−0.031 (0.007)***
−0.023 (0.007)**
0.037 (0.032)
1 × 10−7
0.2
rs1280
0.38
0.45
0.34
0.09
318
162 (49)
 7p21.2
DGKB
rs62448618
A/T
0.34
0.50
0.38
0.27
0.50
0.022 (0.005)
0.030 (0.007)***
0.014 (0.008)
−0.001 (0.026)
1 × 10−5
0.33
rs2191349
0.81
0.61
0.03
0.39
133
12 (91)
 7p13
GCK
rs2908286
A/G
0.19
0.18
0.2
0.18
0.20
0.060 (0.006)
0.064 (0.008)***
0.061 (0.009)***
0.002 (0.032)
9 × 10−25
0.27
rs730497
0.99
0.9
0.52
0.91
25
18 (28)
 9p24.2
GLIS3
rs10974438
A/C
0.76
0.62
0.71
0.86
0.63
−0.023 (0.006)
−0.019 (0.007)**
−0.021 (0.010)*
−0.080 (0.028)**
6 × 10−5
0.16
rs10814916
0.53
0.27
0.08
0.69
54
7 (87)
 11q12.2
FADS2
rs174547
A/G
0.60
0.66
0.52
0.91
0.55
0.029 (0.006)
0.026 (0.007)***
0.038 (0.013)**
0.039 (0.027)
4 × 10−7
0.86
Same
1
1
1
1
147
44 (70)
 11q14.3
MTNR1B
rs10830963
C/G
0.81
0.78
0.79
0.93
0.59
−0.068 (0.006)
−0.062 (0.008)***
−0.090 (0.014)***
−0.078 (0.026)**
7 × 10−27
0.21
Same
1
1
1
1
94
1 (99)
Fasting insulin loci
 2p23.3
GCKR
rs1260326
A/G
0.29
0.41
0.35
0.16
0.52
−0.035 (0.006)
−0.034 (0.008)***
−0.034 (0.010)***
−0.010 (0.027)
1 × 10−8
0.20
rs780094
0.92
0.91
0.42
0.93
274
90 (67)
 12q23.2
IGF1
rs10860845
A/C
0.6
0.83
0.48
0.74
0.65
−0.023 (0.006)
−0.025 (0.008)***
−0.023 (0.008)**
0.002 (0.028)
3 × 10−5
0.76
rs860598
<0.10
<0.10
<0.10
<0.10
322
64 (80)
β: effect size from an additive multivariate model including BMI and corresponding to the coded (C) allele, is shown in units of mmol/l for fasting glucose and natural log-transformed pmol/l for fasting insulin
aMAF averaged across ethnicities H/L, AI/AN and ASN from the transethnic (TE) meta-analysis for coded allele
b p value from the transethnic meta-analysis
cLinkage disequilibrium calculated from 1000 genomes Phase 3 super populations (EUR, AFR, AMR, and ASN)
dEuropean SNP index defined as most significant SNP from the Scott et al. [11] Metabochip analysis
eNo. of SNPs in linkage disequilibrium using r 2 > 0.2 calculated from 1000 genomes Phase 3 super populations with transethnic equal to the intersect of SNPs in EUR, AFR, AMR and ASN
fPercentage reduction in the number of SNPs
*p < 0.05, **p < 0.01 and ***p < 0.001 for race/ethnic-specific analyses
Significant at region-specific Bonferroni-corrected transethnic meta-analysis p values (ranging from α = 1.41 × 10−5 to α = 4.1 × 10−4)
EUR, Europeans, LD, linkage disequilibrium, TE, transethnic
For each of the 11 glycaemic trait loci with potential transethnic fine-mapping (fasting glucose loci–PROX1, G6PC2, ADCY5, MTNR1B, FADS2, GCKR, SLC2A2, DGKB, GCK and GLIS3; fasting insulin locus–GCKR), we found that the number of SNPs in linkage disequilibrium with the most significant marker in the transethnic results (r 2 ≥ 0.2 in the 1KG super populations AFR and AMR) were less than the number of SNPs tagged by the EUR marker (r 2 ≥ 0.2 in EUR). Visual inspection of locus zoom plots indicated that transethnic meta-analysis refined each of these loci by reducing the number of highly correlated SNPs reaching the same level of significance and/or narrowing the genomic region containing putative causal SNPs (ESM Fig. 2). On average, the number of variants in high linkage disequilibrium was reduced by 72.5% with the number of linkage disequilibrium SNPs ranging from one at MTNR1B to 162 at SLC2A2 in the PAGE transethnic meta-analysis results. Refinement was most evident at the SLC2A2 locus (Fig. 2). Bioinformatic functional follow-up was performed for each of the eleven glycaemic trait loci with one or more variants passing the region-specific significance threshold in our transethnic meta-analysis. We observed an overlap of promoter and enhancer sequences at each locus and identified potential target genes. These data not only provided further support for the fine-mapping results but also revealed additional insights into the aetiology of glycaemic traits. UCSC Genome Browser images of each locus are provided in ESM Fig. 3. The results of our in silico functional annotations are summarised in ESM Table 7.
Secondary associations at known glycaemic trait loci
To identify additional independent association signals at significant loci, conditional analyses were performed. Results of these analyses and population-specific associations are shown in Table 4. For transethnic conditional meta-analyses, ten fasting glucose loci and two fasting insulin loci were analysed. Independent secondary associations were identified at two fasting glucose loci (G6PC2-rs477224 and GCK-rs2908286). The second round of conditional analyses did not identify significant tertiary signals. Bioinformatic follow-up of rs477224 suggested that the variant is positioned within a pancreatic islet enhancer. The rs2908290 variant was in weak linkage disequilibrium (AMR r 2 = 0.26, AFR r 2 = 0.23) with a variant, rs2971677, predicted to alter splicing efficiency of GCK.
Table 4
Independent secondary signals at known fasting glucose and fasting insulin loci
Locus
Secondary SNPa
Frequency of coded (C) allele for secondary SNP
Effect of coded (C) allele for secondary SNP
p valueb
Primary SNPc
LD r 2d
Cond. p value (second./primary)e
  
C/N
TE
AA
H/L
AI/AN
ASN
TE
AA
H/L
AI/AN
ASN
    
Transethnic meta-analysis fasting glucose
   G6PC2
rs477224
A/G
0.575
0.486
0.645
0.659
0.820
−0.036 (0.005)
−0.034 (0.007)***
−0.042 (0.007)***
0.035 (0.042)
−0.006 (0.035)
3 × 10−14
rs560887
<0.1
2 × 10−5/5 × 10−26
   GCK
rs2908290
A/G
0.450
0.534
0.388
0.367
0.427
0.040 (0.005)
0.043 (0.007)***
0.038 (0.006)***
−0.009 (0.041)
0.058 (0.027)*
10 × 10−18
rs2908286
<0.1
2 × 10−8/6 × 10−16
Population-specific AA fasting glucose
   G6PC2
rs77719485
A/C
0.976
0.973
0.996
0.995
0.138 (0.020)
0.143 (0.022)***
0.115 (0.054)
−0.046 (0.283)
6x10−11
rs560887
<0.1
2 × 10−6/5 × 10−7
Sequential conditional analysis was performed on ten fasting glucose and two fasting insulin loci
In the AA fasting glucose analysis, rs77719485 was the most significant SNP in the locus and rs560887 was the second most significant. AA effects for rs560887 are shown in Table 3
aLead SNP from conditional analysis reaching locus-specific significance
b p value from the secondary SNP not adjusted for the primary SNP
cLead SNP from primary (unconditional) analysis
dLD r2 between primary and secondary SNP
e p values from conditional analysis
*p < 0.05 and ***p < 0.001 for race/ethnic-specific analyses
LD, linkage disequilibrium
To identify population-specific loci, we conducted separate conditional analyses for significant loci in the primary H/L (GCKR-rs1260326, G6PC2-rs560887, SLC2A2-rs1280, DGKB-rs1005256, GCK-rs1799884, FADS3-rs12577276, MTNR1B-rs10830963, C2CD4A-rs7167881), AA (G6PC2-rs77719485, GCK-rs2908286, CRY2-rs117493014, MADD-rs77082299, ADCY5-rs11708067, MTNR1B-rs10830963) and Asian populations (GLIS3-rs4395942). A population-specific variant was detected in the AA analysis of the G6PC2 locus. The lead fasting glucose SNP, rs77719485, is less frequent in AA population (MAF 2.4%) and rare or monomorphic in the other populations (MAF 0.4% in H/L). Like the transethnic lead SNP, rs560887, bioinformatic follow-up suggested that rs77719485 may affect splicing efficiency for exon 4 for G6PC2.
Association testing outside of glycaemic trait fine-mapping regions to identify potential novel variants
In secondary analyses, we conducted a Metabochip-wide scan to identify potential novel or pleiotropic variants, given that the chip included variants with suggestive signals in established loci for many known metabolic traits. Models were run with and without BMI as a covariate (ESM Table 8, ESM Figs 4,5). Using the Bonferroni significance threshold (0.05/182,055 = 2.7 × 10−7), we identified one novel association for fasting insulin (rs75862513, p = 4.3 × 10−8, Fig. 3) at the SLC17A2 locus previously associated with height and blood pressure [27, 28]. After BMI adjustment (ESM Fig. 5), the association was attenuated suggesting that the effects may be mediated by BMI.

Discussion

In this large multiethnic study population of close to 30,000 participants, we used transethnic fine-mapping to narrow the list of putative causal variants for eleven glycaemic trait loci. On average, we observed a 72% reduction in the number of candidate SNPs, before bioinformatic follow-up. We further demonstrated that many of the genetic variants associated with glycaemic traits likely exert their effects through regulatory mechanisms (splicing or enhancer activity), and provide detailed annotations for subsequent laboratory follow-up. These regulatory annotations provide putative targets for laboratory follow-up (e.g. genome editing) and important insights into strong targets for future therapeutic interventions. For example, this study found that most of the implicated enhancer elements were binding sites for the transcription factor FOXA2 in pancreatic islets, and previous studies have suggested that differential expression of FOXA2 is a genetic determinant of fasting glucose levels, as well as type 2 diabetes risk [29, 30]. Like the previous European Metabochip analysis, we found that rs6113722, which is positioned within a lncRNA adjacent to FOXA2, was associated (p = 3.2 × 10−8) with fasting glucose. As such, expression levels of FOXA2 could be a particularly important regulator of glucose homeostasis and a putative target for genome editing. Although the clinical application of genome editing is in its infancy, in vivo studies have already demonstrated the utility of the CRISPR/Cas9 technique. For example, to mimic observations of the naturally occurring loss-of-function mutation in the gene encoding LDL receptor antagonist PCSK9, a previous study in mice used CRISPR/Cas9 vectors to decrease PCSK9 protein levels, which resulted in increased hepatic LDL receptor levels, and a subsequent decrease in blood cholesterol levels [31]. Identification of key targets, such as FOXA2, and potential regulatory elements of these targets for laboratory follow-up is a critical first step in the translation of GWAS findings.
Analysis of known glycaemic trait loci in this diverse population study suggests the genetic determinants of glycaemic trait levels are likely to be similar across populations. In comparison with previous glycaemic trait studies conducted in diverse populations [7, 32], the replication of effects across populations is more extensive, likely due to the size of this study population. Although most of the loci in the European study were generalisable across populations, this study exemplifies the notion that analysis in diverse populations can refine known loci as well as help in the discovery of novel, sometimes population-specific, associations. For instance, in addition to the well-established splice variant rs560887 that has been robustly associated with fasting glucose, transethnic meta-analysis of the G6PC2 locus identified an additional signal that may implicate regulatory functionality in glycaemia-related tissues. At this same locus, an AA-specific variant, rs77719485, was found to be strongly associated with fasting glucose and, like rs560887 [33], is predicted to affect splicing efficiency. By expanding our analysis to the entire Metabochip, we discovered strong associations with SLC17A2, that were not previously reported by the Metabochip analysis carried out by Scott et al [11] in Europeans. rs75862513 is a relatively rare variant that appears to be monomorphic in Europeans and was most frequent in the Asian (MAF = 0.04-A) and H/L (MAF = 0.001-A) populations in this study. If replicated in an independent dataset, this finding may represent a new locus not previously detected in European- or AA-specific analyses. These examples illustrate the power of transethnic analysis for locus refinement and novel discovery.
Strengths of this study include the large study size, high-density genotyping and representation of multiple diverse populations. In light of the heavy burden of hyperglycaemia in H/L and AA populations, this study begins to address the major gap in knowledge related to the genetic architecture of glycaemic traits in understudied American minority populations. The large study population, combined with new annotation resources, allowed transethnic fine-mapping and prediction of regulatory elements. However, there were several limitations that should be noted. Although this study included populations from four major racial/ethnic groups, the greatest proportions of participants were H/L and AA. As such, this study was limited in its ability to detect associations with more prominent effects in Asian populations [34, 35]. We also acknowledge that fine-mapping approaches only serve as an initial step in determining the underlying causal variant(s) driving association signals by prioritising likely causal candidates for more onerous laboratory follow-up. To further meet this objective, functional elements and variants were identified using bioinformatics databases. However, given that the functional evidence detected by these datasets is incomplete, future functional studies are critical in determining the underlying causal variants. That being said, the combination of fine-mapping with bioinformatics data is particularly useful for reducing both the physical genomic regions of interest and prioritising candidates for molecular characterisation. Furthermore, the in silico approaches help to provide richer inferences regarding the biological mechanisms modulating fasting glucose and insulin levels. As such, fine-mapping is an essential step in functional interpretation of GWAS signals because laboratory follow-up of all possible variants in GWAS loci is prohibitively expensive and time-intensive.
This transethnic study comprehensively fine-mapped known common variants associated with concentrations of fasting glucose and insulin. Genomic regions harbouring known risk variants were refined, novel functional candidates were proposed, new independent signals in previously fasting glucose-implicated genes were identified and one novel locus was discovered. Thus, these results suggest that transethnic meta-analysis can help in transforming GWAS results into new biological insight.

Acknowledgements

The PAGE programme is supported by Genetic Epidemiology of Causal Variants Across the Life Course (CALiCo), MEC, WHI and the Coordinating Center. The contents of this paper are solely the responsibility of the authors and do not necessarily represent the official views of the NIH. The complete list of PAGE members can be found at www.​pagestudy.​org, accessed 29 April 2016. The data and materials included in this report result from a collaboration between the following studies: (1) The MEC characterisation of epidemiological architecture; (2) The Mount Sinai BioMe Biobank; (3) ‘Epidemiology of putative genetic variants: The Women’s Health Initiative’ study. The authors thank the WHI investigators and staff for their dedication and the study participants for making the program possible. Full listing of WHI investigators can be found at www.​whi.​org/​researchers/​Documents%20​%20​Write%20​a%20​Paper/​WHI%20​Investigator%20​Short%20​List.​pdf, accessed 2 June 2016; (4) The CALiCo programme and the ARIC, CARDIA and HCHS/SOL studies contributed to this manuscript. The authors thank the staff and participants of the ARIC study for their important contributions.
Assistance with phenotype harmonisation, SNP selection and annotation, data cleaning, data management, integration and dissemination and general study coordination was provided by the PAGE Coordinating Center. The authors gratefully acknowledge B. Voight for sharing the Metabochip SNP linkage disequilibrium and MAF statistics estimated in the Malmö Diet and Cancer Study. The PAGE consortium thanks the staff and participants of all PAGE studies for their important contributions.

Funding

The PAGE programme is funded by the NHGRI, supported by U01HG004803 (CALiCo), U01HG004802 (MEC), U01HG004790 (WHI) and U01HG004801 (Coordinating Center), and their respective NHGRI ARRA supplements.
The MEC characterisation of epidemiological architecture is funded through the NHGRI PAGE programme (U01HG004802 and its NHGRI ARRA supplement). The MEC study is funded by the National Cancer Institute (R37CA54281, R01 CA63, P01CA33619, U01CA136792 and U01CA98758).
Funding support for the ‘Epidemiology of putative genetic variants: The Women’s Health Initiative’ study is provided through the NHGRI PAGE programme (U01HG004790 and its NHGRI ARRA supplement). The WHI programme is funded by the National Heart, Lung, and Blood Institute (NHLBI), the National Institutes of Health (NIH) and the US Department of Health and Human Services through contracts N01WH22110, 24152, 32100-2, 32105-6, 32108-9, 32111-13, 32115, 32118-32119, 32122, 42107-26, 42129-32 and 44221.
Funding support for the CALiCo programme was provided by the NHGRI PAGE programme (U01HG004803 and its NHGRI ARRA supplement). The following studies are funded as follows: The ARIC study is carried out as a collaborative study supported by NHLBI contracts (HHSN268201100005C, HHSN268201100006C, HHSN268201100007C, HHSN268201100008C, HSN268201100009C, HHSN268201100010C, HHSN268201100011C and HHSN268201100012C), R01HL087641, R01HL59367 and R01HL086694; National Human Genome Research Institute contract U01HG004402; and NIH contract HHSN268200625226C. Infrastructure was partly supported by grant no. UL1RR025005, a component of the NIH and NIH Roadmap for Medical Research. The CARDIA study is supported by contracts HHSN268201300025C, HHSN268201300026C, HHSN268201300027C, HHSN268201300028C, HHSN268201300029C and HHSN268200900041C from the NHLBI, the Intramural Research Program of the National Institute on Aging (NIA) and an intra-agency agreement between NIA and NHLBI (AG0005).
The HCHS/SOL was carried out as a collaborative study supported by contracts from the NHLBI to the University of North Carolina (N01-HC65233), University of Miami (N01-HC65234), Albert Einstein College of Medicine (N01-HC65235), Northwestern University (N01-HC65236) and San Diego State University (N01-HC65237). Additional support was provided by 1R01DK101855-01 and 13GRNT16490017. The following Institutes/Centres/Offices contribute to the HCHS/SOL through a transfer of funds to the NHLBI: National Center on Minority Health and Health Disparities, the National Institute of Deafness and Other Communications Disorders, the National Institute of Dental and Craniofacial Research, the National Institute of Diabetes and Digestive and Kidney Diseases, the National Institute of Neurological Disorders and Stroke and the Office of Dietary Supplements.
The Mount Sinai BioMe Biobank is supported by The Andrea and Charles Bronfman Philanthropies.
Funding support for the PAGE Coordinating Center is provided through the NHGRI PAGE programme (U01HG004801-01 and its NHGRI ARRA supplement). The National Institute of Mental Health also contributes to the support for the Coordinating Center.

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

Each of the co-authors made substantial contributions in each of the three following areas: (1) conception and design, acquisition of data or analysis and interpretation of data; (2) drafting the article or revising it critically for important intellectual content and (3) final approval of the version to be published. CK and SAB are responsible for the integrity of the work as a whole.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Literatur
1.
Zurück zum Zitat Guariguata L, Whiting DR, Hambleton I et al (2014) Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract 103:137–149CrossRefPubMed Guariguata L, Whiting DR, Hambleton I et al (2014) Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract 103:137–149CrossRefPubMed
2.
Zurück zum Zitat Yki-Järvinen H (1994) Pathogenesis of non-insulin-dependent diabetes mellitus. Lancet 343:91–95CrossRefPubMed Yki-Järvinen H (1994) Pathogenesis of non-insulin-dependent diabetes mellitus. Lancet 343:91–95CrossRefPubMed
3.
Zurück zum Zitat Almgren P, Lehtovirta M, Isomaa B et al (2011) Heritability and familiality of type 2 diabetes and related quantitative traits in the Botnia study. Diabetologia 54:2811–2819CrossRefPubMed Almgren P, Lehtovirta M, Isomaa B et al (2011) Heritability and familiality of type 2 diabetes and related quantitative traits in the Botnia study. Diabetologia 54:2811–2819CrossRefPubMed
4.
Zurück zum Zitat Schousboe K, Visscher PM, Henriksen JE et al (2003) Twin study of genetic and environmental influences on glucose tolerance and indices of insulin sensitivity and secretion. Diabetologia 46:1276–1283CrossRefPubMed Schousboe K, Visscher PM, Henriksen JE et al (2003) Twin study of genetic and environmental influences on glucose tolerance and indices of insulin sensitivity and secretion. Diabetologia 46:1276–1283CrossRefPubMed
5.
Zurück zum Zitat Dupuis J, Langenberg C, Prokopenko I et al (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42:105–116CrossRefPubMedPubMedCentral Dupuis J, Langenberg C, Prokopenko I et al (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42:105–116CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Manning AK, Hivert M-F, Scott RA et al (2012) A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat Genet 44:659–669CrossRefPubMedPubMedCentral Manning AK, Hivert M-F, Scott RA et al (2012) A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat Genet 44:659–669CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Fesinmeyer MD, Meigs JB, North KE et al (2013) Genetic variants associated with fasting glucose and insulin concentrations in an ethnically diverse population: results from the population architecture using genomics and epidemiology (PAGE) study. BMC Med Genet 14:98CrossRefPubMedPubMedCentral Fesinmeyer MD, Meigs JB, North KE et al (2013) Genetic variants associated with fasting glucose and insulin concentrations in an ethnically diverse population: results from the population architecture using genomics and epidemiology (PAGE) study. BMC Med Genet 14:98CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Liu EY, Buyske S, Aragaki AK et al (2012) Genotype imputation of Metabochip SNPs using a study-specific reference panel of ~4,000 haplotypes in African Americans from the Women’s Health Initiative. Genet Epidemiol 36:107–117CrossRefPubMedPubMedCentral Liu EY, Buyske S, Aragaki AK et al (2012) Genotype imputation of Metabochip SNPs using a study-specific reference panel of ~4,000 haplotypes in African Americans from the Women’s Health Initiative. Genet Epidemiol 36:107–117CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Rasmussen-Torvik LJ, Guo X, Bowden DW et al (2012) Fasting glucose GWAS candidate region analysis across ethnic groups in the Multiethnic Study of Atherosclerosis (MESA). Genet Epidemiol 36:384–391CrossRefPubMedPubMedCentral Rasmussen-Torvik LJ, Guo X, Bowden DW et al (2012) Fasting glucose GWAS candidate region analysis across ethnic groups in the Multiethnic Study of Atherosclerosis (MESA). Genet Epidemiol 36:384–391CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Voight BF, Kang HM, Ding J et al (2012) The metabochip, a custom genotyping array for genetic studies of metabolic, cardiovascular, and anthropometric traits. PLoS Genet 8:e1002793CrossRefPubMedPubMedCentral Voight BF, Kang HM, Ding J et al (2012) The metabochip, a custom genotyping array for genetic studies of metabolic, cardiovascular, and anthropometric traits. PLoS Genet 8:e1002793CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Scott RA, Lagou V, Welch RP et al (2012) Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nat Genet 44:991–1005CrossRefPubMedPubMedCentral Scott RA, Lagou V, Welch RP et al (2012) Large-scale association analyses identify new loci influencing glycemic traits and provide insight into the underlying biological pathways. Nat Genet 44:991–1005CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Matise TC, Ambite JL, Buyske S et al (2011) The Next PAGE in understanding complex traits: design for the analysis of Population Architecture Using Genetics and Epidemiology (PAGE) Study. Am J Epidemiol 174:849–859CrossRefPubMedPubMedCentral Matise TC, Ambite JL, Buyske S et al (2011) The Next PAGE in understanding complex traits: design for the analysis of Population Architecture Using Genetics and Epidemiology (PAGE) Study. Am J Epidemiol 174:849–859CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Buyske S, Wu Y, Carty CL et al (2012) Evaluation of the metabochip genotyping array in African Americans and implications for fine mapping of GWAS-identified loci: the PAGE study. PLoS One 7:e35651CrossRefPubMedPubMedCentral Buyske S, Wu Y, Carty CL et al (2012) Evaluation of the metabochip genotyping array in African Americans and implications for fine mapping of GWAS-identified loci: the PAGE study. PLoS One 7:e35651CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Genomes Project Consortium, Abecasis GR, Auton A et al (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491:56–65CrossRef Genomes Project Consortium, Abecasis GR, Auton A et al (2012) An integrated map of genetic variation from 1,092 human genomes. Nature 491:56–65CrossRef
15.
Zurück zum Zitat Peterson LE (2003) Partitioning large-sample microarray-based gene expression profiles using principal components analysis. Comput Methods Prog Biomed 70:107–119CrossRef Peterson LE (2003) Partitioning large-sample microarray-based gene expression profiles using principal components analysis. Comput Methods Prog Biomed 70:107–119CrossRef
16.
Zurück zum Zitat Purcell S, Neale B, Todd-Brown K et al (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81:559–575CrossRefPubMedPubMedCentral Purcell S, Neale B, Todd-Brown K et al (2007) PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81:559–575CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Pruim RJ, Welch RP, Sanna S et al (2010) LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26:2336–2337CrossRefPubMedPubMedCentral Pruim RJ, Welch RP, Sanna S et al (2010) LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26:2336–2337CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Lin D-Y, Tao R, Kalsbeek WD et al (2014) Genetic association analysis under complex survey sampling: the Hispanic Community Health Study/Study of Latinos. Am J Hum Genet 95:675–688CrossRefPubMedPubMedCentral Lin D-Y, Tao R, Kalsbeek WD et al (2014) Genetic association analysis under complex survey sampling: the Hispanic Community Health Study/Study of Latinos. Am J Hum Genet 95:675–688CrossRefPubMedPubMedCentral
19.
20.
21.
Zurück zum Zitat Xiong HY, Alipanahi B, Lee LJ et al (2015) RNA splicing. The human splicing code reveals new insights into the genetic determinants of disease. Science 347:1254806CrossRefPubMed Xiong HY, Alipanahi B, Lee LJ et al (2015) RNA splicing. The human splicing code reveals new insights into the genetic determinants of disease. Science 347:1254806CrossRefPubMed
23.
Zurück zum Zitat Roadmap Epigenomics Consortium, Kundaje A, Meuleman W et al (2015) Integrative analysis of 111 reference human epigenomes. Nature 518:317–330CrossRefPubMedCentral Roadmap Epigenomics Consortium, Kundaje A, Meuleman W et al (2015) Integrative analysis of 111 reference human epigenomes. Nature 518:317–330CrossRefPubMedCentral
24.
Zurück zum Zitat ENCODE Project Consortium (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74CrossRef ENCODE Project Consortium (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74CrossRef
25.
Zurück zum Zitat Raney BJ, Dreszer TR, Barber GP et al (2014) Track data hubs enable visualization of user-defined genome-wide annotations on the UCSC Genome Browser. Bioinformatics 30:1003–1005CrossRefPubMed Raney BJ, Dreszer TR, Barber GP et al (2014) Track data hubs enable visualization of user-defined genome-wide annotations on the UCSC Genome Browser. Bioinformatics 30:1003–1005CrossRefPubMed
26.
Zurück zum Zitat GTEx Consortium (2013) The genotype-tissue expression (GTEx) project. Nat Genet 45:580–585CrossRef GTEx Consortium (2013) The genotype-tissue expression (GTEx) project. Nat Genet 45:580–585CrossRef
27.
Zurück zum Zitat He M, Xu M, Zhang B et al (2015) Meta-analysis of genome-wide association studies of adult height in east Asians identifies 17 novel loci. Hum Mol Genet 24:1791–1800CrossRefPubMed He M, Xu M, Zhang B et al (2015) Meta-analysis of genome-wide association studies of adult height in east Asians identifies 17 novel loci. Hum Mol Genet 24:1791–1800CrossRefPubMed
28.
Zurück zum Zitat Simino J, Shi G, Bis JC et al (2014) Gene-age interactions in blood pressure regulation: a large-scale investigation with the CHARGE, Global BPgen, and ICBP Consortia. Am J Hum Genet 95:24–38CrossRefPubMedPubMedCentral Simino J, Shi G, Bis JC et al (2014) Gene-age interactions in blood pressure regulation: a large-scale investigation with the CHARGE, Global BPgen, and ICBP Consortia. Am J Hum Genet 95:24–38CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Pasquali L, Gaulton KJ, Rodríguez-Seguí SA et al (2014) Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat Genet 46:136–143CrossRefPubMedPubMedCentral Pasquali L, Gaulton KJ, Rodríguez-Seguí SA et al (2014) Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat Genet 46:136–143CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Ng MCY, Saxena R, Li J et al (2013) Transferability and fine mapping of type 2 diabetes loci in African Americans: the Candidate Gene Association Resource Plus Study. Diabetes 62:965–976CrossRefPubMedPubMedCentral Ng MCY, Saxena R, Li J et al (2013) Transferability and fine mapping of type 2 diabetes loci in African Americans: the Candidate Gene Association Resource Plus Study. Diabetes 62:965–976CrossRefPubMedPubMedCentral
31.
32.
Zurück zum Zitat Liu C-T, Ng MCY, Rybin D et al (2012) Transferability and fine-mapping of glucose and insulin quantitative trait loci across populations: CARe, the Candidate Gene Association Resource. Diabetologia 55:2970–2984CrossRefPubMedPubMedCentral Liu C-T, Ng MCY, Rybin D et al (2012) Transferability and fine-mapping of glucose and insulin quantitative trait loci across populations: CARe, the Candidate Gene Association Resource. Diabetologia 55:2970–2984CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Baerenwald DA, Bonnefond A, Bouatia-Naji N et al (2013) Multiple functional polymorphisms in the G6PC2 gene contribute to the association with higher fasting plasma glucose levels. Diabetologia 56:1306–1316CrossRefPubMedPubMedCentral Baerenwald DA, Bonnefond A, Bouatia-Naji N et al (2013) Multiple functional polymorphisms in the G6PC2 gene contribute to the association with higher fasting plasma glucose levels. Diabetologia 56:1306–1316CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Wang H, Liu L, Zhao J et al (2013) Large scale meta-analyses of fasting plasma glucose raising variants in GCK, GCKR, MTNR1B and G6PC2 and their impacts on type 2 diabetes mellitus risk. PLoS One 8:e67665CrossRefPubMedPubMedCentral Wang H, Liu L, Zhao J et al (2013) Large scale meta-analyses of fasting plasma glucose raising variants in GCK, GCKR, MTNR1B and G6PC2 and their impacts on type 2 diabetes mellitus risk. PLoS One 8:e67665CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Sakai K, Imamura M, Tanaka Y et al (2013) Replication study for the association of 9 East Asian GWAS-derived loci with susceptibility to type 2 diabetes in a Japanese population. PLoS One 8:e76317CrossRefPubMedPubMedCentral Sakai K, Imamura M, Tanaka Y et al (2013) Replication study for the association of 9 East Asian GWAS-derived loci with susceptibility to type 2 diabetes in a Japanese population. PLoS One 8:e76317CrossRefPubMedPubMedCentral
Metadaten
Titel
Transethnic insight into the genetics of glycaemic traits: fine-mapping results from the Population Architecture using Genomics and Epidemiology (PAGE) consortium
verfasst von
Stephanie A. Bien
James S. Pankow
Jeffrey Haessler
Yinchang N. Lu
Nathan Pankratz
Rebecca R. Rohde
Alfred Tamuno
Christopher S. Carlson
Fredrick R. Schumacher
Petra Bůžková
Martha L. Daviglus
Unhee Lim
Myriam Fornage
Lindsay Fernandez-Rhodes
Larissa Avilés-Santa
Steven Buyske
Myron D. Gross
Mariaelisa Graff
Carmen R. Isasi
Lewis H. Kuller
JoAnn E. Manson
Tara C. Matise
Ross L. Prentice
Lynne R. Wilkens
Sachiko Yoneyama
Ruth J. F. Loos
Lucia A. Hindorff
Loic Le Marchand
Kari E. North
Christopher A. Haiman
Ulrike Peters
Charles Kooperberg
Publikationsdatum
13.09.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 12/2017
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-017-4405-1

Weitere Artikel der Ausgabe 12/2017

Diabetologia 12/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.