Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Replication Study for the Association of 9 East Asian GWAS-Derived Loci with Susceptibility to Type 2 Diabetes in a Japanese Population

  • Kensuke Sakai ,

    Contributed equally to this work with: Kensuke Sakai, Minako Imamura

    Affiliations Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan, Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki, Japan

  • Minako Imamura ,

    Contributed equally to this work with: Kensuke Sakai, Minako Imamura

    Affiliation Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan

  • Yasushi Tanaka,

    Affiliation Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki, Japan

  • Minoru Iwata,

    Affiliation First Department of Internal Medicine, University of Toyama, Toyama, Japan

  • Hiroshi Hirose,

    Affiliation Health Center, Keio University School of Medicine, Tokyo, Japan

  • Kohei Kaku,

    Affiliation Division of Diabetes, Endocrinology and Metabolism, Department of Internal Medicine, Kawasaki medical school, Kurashiki, Japan

  • Hiroshi Maegawa,

    Affiliation Department of Medicine, Shiga University of Medical Science, Otsu, Japan

  • Hirotaka Watada,

    Affiliations Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan, Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan

  • Kazuyuki Tobe,

    Affiliation First Department of Internal Medicine, University of Toyama, Toyama, Japan

  • Atsunori Kashiwagi,

    Affiliation Department of Medicine, Shiga University of Medical Science, Otsu, Japan

  • Ryuzo Kawamori,

    Affiliation Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan

  • Shiro Maeda

    smaeda@src.riken.jp

    Affiliation Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan

Abstract

Aims

East Asian genome-wide association studies (GWAS) for type 2 diabetes identified 8 loci with genome-wide significance, and 2 loci with a borderline association. However, the associations of these loci except MAEA locus with type 2 diabetes have not been evaluated in independent East Asian cohorts. We performed a replication study to investigate the association of these susceptibility loci with type 2 diabetes in an independent Japanese population.

Methods

We genotyped 7,379 Japanese participants (5,315 type 2 diabetes and 2,064 controls) for each of the 9 single nucleotide polymorphisms (SNPs), rs7041847 in GLIS3, rs6017317 in FITM2R3HDMLHNF4A, rs6467136 near GCCIPAX4, rs831571 near PSMD6, rs9470794 in ZFAND3, rs3786897 in PEPD, rs1535500 in KCNK16, rs16955379 in CMIP, and rs17797882 near WWOX. Because the sample size in this study was not sufficient to replicate single SNP associations, we constructed a genetic risk score (GRS) by summing a number of risk alleles of the 9 SNPs, and examined the association of the GRS with type 2 diabetes using logistic regression analysis.

Results

With the exception of rs1535500 in KCNK16, all SNPs had the same direction of effect (odds ratio [OR]>1.0) as in the original reports. The GRS constructed from the 9 SNPs was significantly associated with type 2 diabetes in the Japanese population (p = 4.0 × 10-4, OR = 1.05, 95% confidence interval: 1.02–1.09). In quantitative trait analyses, rs16955379 in CMIP was nominally associated with a decreased homeostasis model assessment of β-cell function and with increased fasting plasma glucose, but neither the individual SNPs nor the GRS showed a significant association with the glycemic traits.

Conclusions

These results indicate that 9 loci that were identified in the East Asian GWAS meta-analysis have a significant effect on the susceptibility to type 2 diabetes in the Japanese population.

Introduction

Diabetes mellitus affects more than 300 million individuals worldwide, and its prevalence is progressively increasing, particularly in the East Asia, South Asia and Western Pacific regions [1]. Although the precise mechanisms underlying the development and progression of type 2 diabetes have not been elucidated, previous epidemiological studies have linked diverse factors, such as obesity, hypertension and dyslipidemia, with the risk of type 2 diabetes [2]. Additionally, genetic factors are thought to play an important role in the pathogenesis of type 2 diabetes [3]. Genetic studies of type 2 diabetes have made rapid progress through genome-wide association studies (GWAS) [4,5], and the number of susceptibility loci identified for type 2 diabetes has been increased through GWAS to nearly 70 [625]. In 2012, an East Asian GWAS meta-analysis identified 8 susceptibility loci for type 2 diabetes [23], namely rs6815464 in MAEA, rs7041847 in GLIS3, rs6017317 in FITM2R3HDMLHNF4A locus, rs6467136 near GCC1-PAX4 locus, rs831571 near PSMD6, rs9470794 in ZFAND3, rs3786897 in PEPD, and rs1535500 in KCNK16, along with 2 additional loci with borderline association (p < 10-6), namely rs16955379 in CMIP and rs17797882 near WWOX. Of these, the association of rs6815464 in MAEA with type 2 diabetes has been already evaluated and confirmed in an independent Japanese population [25], but the associations of the remaining 9 loci have not been evaluated in an independent East Asian cohort. In the East Asian GWAS meta-analysis reported by Cho et al. [23], over 50,000 individuals were analyzed, and sample sizes that are considered large by conventional standards (~ several thousands) may be underpowered to conduct individual single nucleotide polymorphism (SNP) analyses. It was also shown that a genetic risk score (GRS) constructed by summing the number of risk alleles for multiple loci had a much stronger effect on disease susceptibility than individual SNPs; thus, the GRS is considered to be a useful and tractable measure to evaluate the effect of multiple candidate loci of interest [2628]. In this study, to evaluate the contribution of these 9 loci identified in East Asian GWAS meta-analysis to conferring susceptibility to type 2 diabetes in an independent population, we performed a replication study for the association of these 9 susceptibility loci with type 2 diabetes in an independent Japanese population using the GRS that was constructed from the 9 loci.

Materials and Methods

Ethics Statements

All participants agreed to the protocol of this study and provided written informed consent before enrolling. DNA samples were obtained from the peripheral blood of each participant. The study protocol conformed to the provisions of the Declaration of Helsinki, and was approved by the ethics committees of RIKEN Yokohama Institute and each of the participating institutes, i.e., Shiga University of Medical Science, Kawasaki Medical School, St. Marianna University, Juntendo University, the University of Toyama, and Keio University.

Participants and DNA Preparation

We enrolled 5,315 type 2 diabetes patients who regularly visited the outpatient clinics of Shiga University of Medical Science, Kawasaki Medical School, St. Marianna University, Juntendo University, and the University of Toyama or who were registered in BioBank Japan [12]. Diabetes mellitus was diagnosed according to the World Health Organization (WHO) criteria [29], and type 2 diabetes was defined by a gradual adult onset. Patients who tested positive for antibodies to glutamic acid decarboxylase or who were diagnosed with mitochondrial disease or maturity-onset diabetes of the young were excluded from the present study. We also recruited 2,064 controls, who underwent annual health check-ups at Keio University, St. Marianna University, or Toyama University Hospital.

Single nucleotide polymorphism (SNP) Genotyping

We first selected the following 8 SNPs identified by GWAS in individuals of East Asian ancestry and were reported by Cho et al. in 2012 [23], rs6815464 in MAEA, rs7041847 in GLIS3, rs6017317 in FITM2R3HDMLHNF4A locus, rs6467136 near GCC1-PAX4 locus, rs831571 near PSMD6, rs9470794 in ZFAND3, rs3786897 in PEPD, and rs1535500 in KCNK16. Because the association of rs6815464 in MAEA with type 2 diabetes was already evaluated, and confirmed in the present Japanese population [25], we excluded this SNP from the present study. We also examined 2 additional SNP loci, that showed a borderline association (p < 10-6) in the East Asian GWAS meta-analysis, rs16955379 in CMIP and rs17797882 near WWOX.

Genotyping was performed using the multiplex-polymerase chain reaction (PCR) invader assay as previously described [30]. The success rates of the assay were > 95% for each SNP, and the concordance rates in the duplicate samples were ≥ 99.8%.

Statistical Analysis

We performed Hardy-Weinberg equilibrium (HWE) tests according to the method described by Nielsen et al [31]. The SNP genotypes were scored using an additive model (0, 1, and 2 for homozygous for the non-effect allele, heterozygous, and homozygous for the effect allele, respectively). We calculated a GRS by counting the number of risk alleles of the 9 SNPs in each individual. The association of the GRS or each SNP with type 2 diabetes was evaluated by a logistic regression analysis with or without adjustment for age, sex, and body mass index (BMI). The association of the GRS or each SNP with quantitative traits, including fasting plasma glucose (FPG), the homeostasis model assessment of β-cell function (HOMA-β), and the HOMA of insulin resistance (HOMA-IR) [32,33] was evaluated by multiple linear regression analysis. The values of these traits in the present Japanese population showed a skewed distribution; therefore, we used log-transformed values for the analyses.

Statistical analyses were performed using StatView software (SAS Institute, Cary, NC, USA). The level of significance was determined by the Bonferroni correction for multiple tests, and a p value of <0.0055 (0.05 divided by 9) was considered to be statistically significant.

Results

The clinical characteristics of the participants are shown in Table 1. The male ratio, age and BMI in the type 2 diabetes cases were significantly higher than those in the controls. The genotype distributions for all 9 SNPs did not deviate from HWE proportions, except rs7041847 in the type 2 diabetes cases (p = 0.015, Table 2). Since our samples were collected at 7 distinct sites for patients with type 2 diabetes and at 3 sites for the control individuals, we compared the genotype distributions of the 9 SNPs among the different collection sites. The results indicated that the genotype distributions of each SNP were not significantly different among the 7 collection sites for type 2 diabetes or among the 3 collection sites for the controls with 1 exception; the genotype distribution of rs16955379 was slightly different among the 7 type 2 diabetes collection sites (p = 0.0339, Table S1), although the difference was not statistically significant after correction for multiple testing error.

Sample size (case/control)Type 2 diabetesControlp
n5,3152,064
Sex (male:female)5,315/2,0643,249 : 2,066982 : 1,082< 0.0001b
Age (year)a5,278/2,05863.0 ± 11.549.8 ± 17.0< 0.0001c
BMI (kg/m2)a4,951/2,01824.2 ± 4.122.4 ± 3.1< 0.0001c
HbA1c (%)a4,730/1,3867.9 ± 2.15.4 ± 0.3< 0.0001c
FPG (mmol/L)a2,249/1,3208.4 ± 3.05.3 ± 0.6< 0.0001c
Diabetes duration (year)a3,474/-13.7 ± 9.7

Table 1. Clinical characteristics of participants.

CSV
Download CSV
SNPGeneAllele1/Allele2aAllele 11/12/22p for HWE test
Type 2 diabetesControlType 2 diabetesControl
rs7041847GLIS3A/G1,256/2,477/1,399464/982/5360.01510.7293
rs6017317FITM2-R3HDML-HNF4AG/T1,646/2,639/983613/1,038/3930.19080.2128
rs6467136GCC1-PAX4G/A3,144/1,807/2791,196/727/1180.35780.5884
rs831571PSMD6C/T2,224/2,372/594824/949/2540.31000.4505
rs9470794ZFAND3T/C3,338/1,711/2271,332/655/680.67920.2493
rs3786897PEPDA/G1,733/2,518/1,013606/1,048/3910.06890.0998
rs1535500KCNK16G/T2,104/2,400/747821/945/2830.13800.6738
rs16955379CMIPC/T2,944/2,012/3221,181/729/1370.37490.0912
rs17797882WWOXC/T3,349/1,709/1991,289/664/920.31590.5821

Table 2. Genotype distributions of 9 single nucleotide polymorphisms (SNPs) in case and control groups.

CSV
Download CSV

The results of the association study for each SNP with type 2 diabetes in the present Japanese population are shown in Table 3. All SNPs showed the same direction of effect (odds ratio [OR] >1.0) as in the original reports [23], except rs1535500 in KCNK16 (OR = 1.00 in the unadjusted data, OR = 0.99 in the data adjusted for age, sex and BMI). Two SNPs, rs831571 in PSMD6 and rs3786897 in PEPD were nominally associated with type 2 diabetes (rs831571: p = 0.0135, OR = 1.11, 95% confidence interval [CI] 1.02–1.22; rs3786897: p = 0.0407, OR = 1.09, 95% CI 1.004–1.18, adjusted for age, sex, and BMI); however, these associations were not significant after Bonferroni’s correction (p > 0.0055). The remaining 7 SNPs were not associated with type 2 diabetes in the present Japanese population (p≥0.05). Although we did not observe a significant association of 9 SNPs with type 2 diabetes by themselves, a GRS constructed from the 9 SNPs was significantly associated with type 2 diabetes in the present Japanese population (p = 0.002, per allele OR = 1.04, 95% CI 1.01–1.07, unadjusted data, p = 0.0004, per allele OR = 1.05, 95% CI 1.02–1.09, adjusted for age, sex and BMI).

SNPGeneRisk AlleleaRAFb (case/control)UnadjustedAdjustedc
p valueOR (95%CI)p valueOR (95%CI)
rs7041847GLIS3A0.486/0.4810.65501.01 (0.94–1.09)0.38891.03 (0.95–1.12)
rs6017317FITM2-R3HDML-HNF4AG0.562/0.5530.31411.03 (0.96–1.11)0.15541.06 (0.97–1.15)
rs6467136GCC1-PAX4G0.773/0.7640.20841.05 (0.97–1.15)0.13321.07 (0.97–1.19)
rs831571PSMD6C0.657/0.6400.06061.07 (0.99–1.16)0.01351.11 (1.02–1.22)
rs9470794ZFAND3C0.205/0.1920.08441.08 (0.98–1.18)0.15131.08 (0.97–1.20)
rs3786897PEPDA0.568/0.5520.08461.06 (0.99–1.14)0.04071.09 (1.004–1.18)
rs1535500KCNK16T0.370/0.3680.81751.00 (0.93–1.12)0.85990.99 (0.91–1.08)
rs16955379CMIPT0.251/0.2440.40671.03 (0.95–1.12)0.11991.08 (0.98–1.19)
rs17797882WWOXC0.799/0.7920.33551.04 (0.95–1.14)0.75211.01 (0.91–1.12)
GRSd0.00201.04 (1.01–1.07)0.00041.05 (1.02–1.09)

Table 3. Association of 9 SNPs with type 2 diabetes in the Japanese population.

The results of logistic regression analyses are shown
aThe risk allele reported in the previous report
bRisk allele frequency
cAdjusted for age, sex and log-transformed BMI
dThe genetic risk score (GRS) was calculated according to the number of risk alleles by counting the 9 East Asian genome-wide association study derived SNPs; Individuals who had complete genotype data for the 9 SNPs were used for the analysis (n = 6,819)
CSV
Download CSV

We next examined the associations between these SNPs and quantitative traits related to glucose metabolism, such as HOMA-IR, HOMA-β, and FPG, using the control individuals (Table 4). Rs16955379-T in CMIP had a nominal effect on decreasing HOMA-β (p = 0.0442, β= -0.055, se = 0.027) or on increasing FPG (p = 0.0271, β= 0.009, se = 0.004). However, neither the individual SNPs nor the GRS was significantly associated with these quantitative traits after Bonferroni’s correction in the present Japanese population.

SNPGeneRisk AlleleaHOMA-IRb (n=893)HOMA-βb (n=893)FPGb (n=1,325)
Effect (SE)p valueEffect (SE)p valueEffect (SE)p value
rs7041847GLIS3A-0.011 (0.023)0.6441-0.015 (0.024)0.54290.003 (0.004)0.3839
rs6017317FITM2-R3HDML-HNF4AG-0.015 (0.023)0.51230.038 (0.024)0.1184-0.006 (0.004)0.1052
rs6467136GCC1-PAX4G0.008 (0.027)0.76720.028 (0.028)0.3094-0.001 (0.004)0.7666
rs831571PSMD6C-0.006 (0.024)0.80660.029 (0.025)0.2384-0.006 (0.004)0.0963
rs9470794ZFAND3C-0.027 (0.029)0.3613-0.011 (0.031)0.7129-0.004 (0.005)0.4310
rs3786897PEPDA0.040 (0.023)0.08940.014 (0.025)0.57280.005 (0.004)0.1517
rs1535500KCNK16T-0.007 (0.023)0.7586-0.003 (0.024)0.90320.003 (0.004)0.3892
rs16955379CMIPT-0.028 (0.026)0.2870-0.055 (0.027)0.04420.009 (0.004)0.0271
rs17797882WWOXC0.0015 (0.028)0.59410.003 (0.030)0.90640.002 (0.004)0.5919
GRSc-0.004 (0.009)0.62710.006 (0.009)0.53980.0003 (0.001)0.8405

Table 4. Association of 9 SNPs with quantitative traits related to glucose metabolism in controls.

CSV
Download CSV

Discussion

In this study, we examined the association of 9 SNP loci derived from an East Asian GWAS meta-analysis with type 2 diabetes in an independent Japanese population, and found that a GRS that was constructed with the 9 SNPs was significantly associated with type 2 diabetes in the present Japanese population.

To date, nearly 70 susceptibility loci have been identified for type 2 diabetes [625], through GWAS, and a few plausible candidates were reported through candidate gene approaches [34,35]. Of these loci, several have been shown to be associated with type 2 diabetes in different ethnic groups, including the Japanese [28,3641]. In 2012, a GWAS meta-analysis that was conducted by the Asian Genetic Epidemiology Network Consortium for type 2 diabetes (AGEN-T2D) identified 8 loci that were associated with susceptibility to type 2 diabetes at genome-wide significance levels, and 2 additional loci with borderline association (p < 10-6) [23]. Although these loci are considered to be important suceptibility loci for type 2 diabetes in East Asian populations, there are some differences in genetic background among different East Asian populations [42]. The MAEA locus was previously shown to be associated with type 2 diabetes in the present Japanese sample [25], but the remaining 9 loci have not been examined in an independent East Asian cohort. Therefore, the associations of these loci with type 2 diabetes need to be evaluated in independent ethnic groups to know the contribution of each locus to susceptibility to the disease in individual ethnic groups.

In the present Japanese population, we did not observe a significant association of all the 9 SNPs with type 2 diabetes. The accuracy of the genotyping was well validated, and the genotype distribution for each SNP in the controls did not deviate from HWE proportions, indicating that techinical factors are not likely to be a principal cause of the discrepancy between the present study and the original study. Although we collected samples for case and control individuals from several different Japanese institutes, the genotype distributions for the individual SNPs were not significantly different among the different collection sites (Table S1); therefore, we considered that our finding was not affected by sample heterogeneity.

Because the control individuals in this study were younger than the type 2 diabetes patients, the control group might include several individuals who would develop the disease later, and the possibility of type 2 error may be increased, although the results were not affected by adjusting for age. We evaluated the association of the 9 SNPs with type 2 diabetes using the older control individuals (age ≥ 40, ≥ 50 or ≥ 60). The results indicated that the effect sizes of individual SNPs were almost the same in the analysis using older controls as those in the original findings (Table S2). Most SNPs showed the same direction of effect as those in the original reports, except rs1535500 in KCNK16 and the estimated power of the present study to replicate an original association for the individual SNPs is between 42% and 79% (Table S3); therefore, insufficient study power is likely to be a principal cause for the discrepancy between this Japanese study and the original East Asian GWAS meta-analysis. Since a GRS constructed by summing the number of risk alleles for multiple loci was shown to be useful and tractable measure to evaluate the effect of multiple candidate loci of interest [2628], we examined the combined effect of the 9 SNPs on conferring susceptibility to type 2 diabetes, and found that the GRS constructed using the 9 SNPs was significantly associated with type 2 diabetes; therefore, we suggest that the 9 SNPs are important susceptibility loci for type 2 diabetes also in the Japanese population, and the construction of a GRS is considered to be a useful and alternative approach to evaluate the effect of multiple candidate loci in a underpowered sample for single SNP analyses, like the present study. However, since the association did not attain a genome-wide significance level, the contribution of the 9 loci to susceptibility to Japanese type 2 diabetes needs to be evaluated further in larger Japanese cohorts.

The mechanisms of how these loci contribute to susceptibility to type 2 diabetes are still unknown. The analyses of quantitative traits related to glucose metabolism, such as HOMA-β, HOMA-IR and FPG, revealed that rs16955379 in CMIP had a nominal effect on decreased HOMA-β and on increased FPG, but neither individual SNPs nor the GRS were significantly associated with these quantitative traits, probably because of insufficient study power (Table S4). Further study is required to elucidate the precise mechanisms how these loci contribute to conferring susceptibility to type 2 diabetes.

In conclusion, we examined the association of 9 SNPs identified in an East Asian GWAS meta-analysis with type 2 diabetes in an independent Japanese population. Our results indicate that the 9 East Asian GWAS-derived loci have a significant effect on conferring susceptibility to type 2 diabetes in the Japanese population.

Supporting Information

Table S1.

Comparison of risk allele frequencies among individual areas for sample collection. acollection 1 (BioBank Japan1), collection 2 (Kawasaki Medical University), collection 3 (BioBank Japan2), collection 4 (Juntendo University), collection 5 (Case; Shiga University of Medical Science, Control; Keio University), collection 6 (St. Mariannna University School of Medicine), collection 7 (Toyama University). bChi square test.

https://doi.org/10.1371/journal.pone.0076317.s001

(DOCX)

Table S2.

Association study of 9 SNPs with type 2 diabetes using older control (age ≥ 40, n=1,424, age ≥ 50, n=1,057, age ≥ 60n=622) and all cases (n=5,315). Results of logistic regression analysis are shown. arisk allele reported in the previous reports. badjusted for age, sex and log-transformed BMI.

https://doi.org/10.1371/journal.pone.0076317.s002

(DOCX)

Table S3. Power estimation for each SNP locus in the present study.

aRisk allele for type 2 diabetes reported in the previous reports. bPower estimation was performed using CaTS power calculator, CaTS: http://www.sph.umich.edu/csg/abecasis/CaTS/). The prevalence of type 2 diabetes is assumed to be 10%, α = 0.05. cRequired sample size estimation was performed using the Quanto software package (Version 1.2.4, http://hydra.usc.edu/gxe/).

https://doi.org/10.1371/journal.pone.0076317.s003

(DOCX)

Table S4. Estimation of statistical power for the present study to detect associations of 9 SNPs with quantitative metabolic traits.

Power calculations were carried out using the Quanto software package (Version 1.2.4, http://hydra.usc.edu/gxe/). avalues are log-transformed for the analysis.

https://doi.org/10.1371/journal.pone.0076317.s004

(DOCX)

Acknowledgments

We thank the technical staff at the Laboratory for Endocrinology, Metabolism and Kidney Diseases, RIKEN Center for Integrative Medical Sciences, for their technical assistance.

Author Contributions

Conceived and designed the experiments: SM. Performed the experiments: KS M. Imamura SM. Analyzed the data: KS M. Imamura SM. Contributed reagents/materials/analysis tools: M.Iwata HM HW HH YT KT KK AK RK. Wrote the manuscript: KS SM.

References

  1. 1. International Diabetes Federation (2012) Diabetes Atlas. 5th edition update.. Brussels, Belgium: International Diabetes Federation. p. 2012. Accessed 27/03/2013.
  2. 2. Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI et al. (2009) Harmonizing the metabolic syndrome. Circulation 120: 1640–1645. doi:https://doi.org/10.1161/CIRCULATIONAHA.109.192644. PubMed: 19805654.
  3. 3. O’Rahilly S, Barroso I, Wareham NJ (2005) Genetic factors in type 2 diabetes: the end of the beginning? Science 307: 370–373. doi:https://doi.org/10.1126/science.1104346. PubMed: 15662000.
  4. 4. McCarthy MI (2010) Genomics, Type 2 Diabetes, and Obesity. N Engl J Med 363: 2339–2350. doi:https://doi.org/10.1056/NEJMra0906948. PubMed: 21142536.
  5. 5. Imamura M, Maeda S (2011) Genetics of type 2 diabetes: the GWAS era and future perspectives. Endocr J 58: 723–739. doi:https://doi.org/10.1507/endocrj.EJ11-0113. PubMed: 21778616.
  6. 6. Sladek R, Rocheleau G, Rung J, Dina C, Shen L et al. (2007) A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445: 881–885. doi:https://doi.org/10.1038/nature05616. PubMed: 17293876.
  7. 7. Zeggini E, Weedon MN, Lindgren CM, Frayling TM, Elliott KS et al. (2007) Replication of genome-wide association signal in U.K. samples reveals risk loci for type 2 diabetes. Science 316: 1336–1341. doi:https://doi.org/10.1126/science.1142364. PubMed: 17463249.
  8. 8. Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y et al. (2007) A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316: 1341–1345. doi:https://doi.org/10.1126/science.1142382. PubMed: 17463248.
  9. 9. Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI et al. (2007) Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316: 1331–1336. doi:https://doi.org/10.1126/science.1142358. PubMed: 17463246.
  10. 10. Steinthorsdottir V, Thorleifsson G, Reynisdottir I, Benediktsson R, Jonsdottir T et al. (2007) A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet 39: 770–775. doi:https://doi.org/10.1038/ng2043. PubMed: 17460697.
  11. 11. Zeggini E, Scott LJ, Saxena R, Voight BF, Marchini JL et al. (2008) Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet 40: 638-645. doi:https://doi.org/10.1038/ng.120. PubMed: 18372903.
  12. 12. Unoki H, Takahashi A, Kawaguchi T, Hara K, Horikoshi M et al. (2008) SNPs in KCNQ1 are associated with susceptibility to type 2 diabetes in East Asian and European populations. Nat Genet 40: 1098–1102. doi:https://doi.org/10.1038/ng.208. PubMed: 18711366.
  13. 13. Yasuda K, Miyake K, Horikawa Y, Hara K, Osawa H et al. (2008) Variants in KCNQ1 are associated with susceptibility to type 2 diabetes mellitus. Nat Genet 40: 1092–1097. doi:https://doi.org/10.1038/ng.207. PubMed: 18711367.
  14. 14. Rung J, Cauchi S, Albrechtsen A, Shen L, Rocheleau G et al. (2009) Genetic variant near IRS1 is associated with type 2 diabetes, insulin resistance and hyperinsulinemia. Nat Genet 41: 1110–1115. doi:https://doi.org/10.1038/ng.443. PubMed: 19734900.
  15. 15. Lyssenko V, Nagorny CL, Erdos MR, Wierup N, Jonsson A et al. (2009) Common variant in MTNR1B associated with increased risk of type 2 diabetes and impaired early insulin secretion. Nat Genet 41: 82–88. doi:https://doi.org/10.1038/ng.288. PubMed: 19060908.
  16. 16. Bouatia-Naji N, Bonnefond A, Cavalcanti-Proença C, Sparsø T, Holmkvist J et al. (2009) A variant near MTNR1B is associated with increased fasting plasma glucose levels and type 2 diabetes risk. Nat Genet 41: 89–94. doi:https://doi.org/10.1038/ng.277. PubMed: 19060909.
  17. 17. Prokopenko I, Langenberg C, Florez JC, Saxena R, Soranzo N et al. (2009) Variants in MTNR1B influence fasting glucose levels. Nat Genet 41: 77–81. doi:https://doi.org/10.1038/ng.290. PubMed: 19060907.
  18. 18. Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N et al. (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42: 105–116. doi:https://doi.org/10.1038/ng.520. PubMed: 20081858.
  19. 19. Voight BF, Scott LJ, Steinthorsdottir V, Morris AP, Dina C et al. (2010) Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat Genet 42: 579–589. doi:https://doi.org/10.1038/ng.609. PubMed: 20581827.
  20. 20. Yamauchi T, Hara K, Maeda S, Yasuda K, Takahashi A et al. (2010) A genome-wide association study in the Japanese population identifies susceptibility loci for type2 diabetes at UBE2E2 and C2CD4A-C2CD4B. Nat Genet 42: 864–868. doi:https://doi.org/10.1038/ng.660. PubMed: 20818381.
  21. 21. Shu XO, Long J, Cai Q, Qi L, Xiang YB et al. (2010) Identification of new genetic risk variants for type 2 diabetes. PLOS Genet 6: e1001127. PubMed: 20862305.
  22. 22. Kooner JS, Saleheen D, Sim X, Sehmi J, Zhang W et al. (2011) Genome-wide association study in individuals of South Asian ancestry identifies six new type 2 diabetes susceptibility loci. Nat Genet 43: 984–989. doi:https://doi.org/10.1038/ng.921. PubMed: 21874001.
  23. 23. Cho YS, Chen CH, Hu C, Long J, Zhang W et al. (2012) Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in Eaat Asians. Nat Genet 44: 67–72.
  24. 24. Morris AP, Voight BF, Teslovich TM, Ferreira T, Segrè AV et al. (2012) Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet 44: 981–990. doi:https://doi.org/10.1038/ng.2383. PubMed: 22885922.
  25. 25. Imamura M, Maeda S, Yamauchi T, Hara K, Yasuda K et al. (2012) A single-nucleotide polymorphism in ANK1 is associated with susceptibility to type 2 diabetes in Japanese populations. Hum Mol Genet 21: 3042–3049. doi:https://doi.org/10.1093/hmg/dds113. PubMed: 22456796.
  26. 26. Roseann EP, Hermine HM, Peter H, Alan RS, Douglas FL et al. (2011) Genetic risk sum score comprised of common polygenic variation is associated with body mass index. Hum Genet 129: 221–230. doi:https://doi.org/10.1007/s00439-010-0917-1. PubMed: 21104096.
  27. 27. Iwata M, Maeda S, Kamura Y, Takano A, Kato H et al. (2012) Genetic risk score constructed using 14 susceptibility alleles for type2 diabetes is associated with the early onset of diabetes and may predict the future requirement of insulin injections among Japanese individuals. Diabetes Care 35: 1763–1770. doi:https://doi.org/10.2337/dc11-2006. PubMed: 22688542.
  28. 28. Fukuda H, Imamura M, Tanaka Y, Iwata M, Hirose H et al. (2012) A single nucleotide polymorphism within DUSP9 is associated with susceptibility to type 2 diabetes in a Japanese population. PLOS ONE 7: e46263. doi:https://doi.org/10.1371/journal.pone.0046263. PubMed: 23029454.
  29. 29. Alberti KG, Zimmet PZ (1998) Definition, diagnosis and classification of diabetes mellitus and its complications.Part1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med 15: 539–553.
  30. 30. Maeda S, Tsukada S, Kanazawa A, Sekine A, Tsunoda T et al. (2005) Genetic variations in the gene encoding TFAP2B are associated with type 2 diabetes mellitus. J Hum Genet 50: 283–292. doi:https://doi.org/10.1007/s10038-005-0253-9. PubMed: 15940393.
  31. 31. Nielsen DM, Ehm MG, Weir BS (1998) Detecting marker-disease association by testing for Hardy-Weinberg disequilibrium at a marker locus. Am J Hum Genet 63: 1531–1540. doi:https://doi.org/10.1086/302114. PubMed: 9867708.
  32. 32. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF et al. (1985) Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetlogia 28: 412–419. doi:https://doi.org/10.1007/BF00280883. PubMed: 3899825.
  33. 33. Wallace TM, Levy JC, Matthews DR (2004) Use and abuse of HOMA modeling. Diabetes Care 27: 1487–1495. doi:https://doi.org/10.2337/diacare.27.6.1487. PubMed: 15161807.
  34. 34. Chiefari E, Tanyolaç S, Paonessa F, Pullinger CR, Capula C et al. (2011) Functional variants of the HMGA1 gene and type 2 diabetes mellitus. JAMA 305: 903–912. doi:https://doi.org/10.1001/jama.2011.207. PubMed: 21364139.
  35. 35. Liu L, Ding H, Wang HR, Xu YJ, Cui GL et al. (2012) Polymorphism of HMGA1 is associated with increased risk of type 2 diabetes among Chinese individuals. Diabetologia 55: 1685–1688. doi:https://doi.org/10.1007/s00125-012-2518-0. PubMed: 22411136.
  36. 36. Omori S, Tanaka Y, Takahashi A, Hirose H, Kashiwagi A et al. (2008) Association of CDKAL1 IGF2BP2, CDKN2A/B, HHEX, SLC30A8, and KCNJ11 with susceptibility to type 2 diabetes in a Japanese population. Diabetes 57: 791–795.
  37. 37. Horikoshi M, Hara K, Ito C, Nagai R, Froguel P et al. (2007) A genetic variation of the transcription factor 7-like 2 gene is associated with the risk of type 2 diabetes in the Japanese population. Diabetologia 50: 747–751. doi:https://doi.org/10.1007/s00125-006-0588-6. PubMed: 17245589.
  38. 38. Hayashi T, Iwamoto Y, Kaku K, Hirose H, Maeda S (2007) Replication study for the association of TCF7L2 with susceptibility to type 2 diabetes in a Japanese population. Diabetologia 50: 980–984. doi:https://doi.org/10.1007/s00125-007-0618-z. PubMed: 17340123.
  39. 39. Onuma H, Tabara Y, Kawamoto R, Shimizu I, Kawamura R et al. (2010) The GCKR rs780094 polymorphism is associated with susceptibility of type 2 diabetes, reduced fasting plasma glucose levels, increased triglycerides levels and lower HOMA-IR in Japanese population. J Hum Genet 55: 600–604. doi:https://doi.org/10.1038/jhg.2010.75. PubMed: 20574426.
  40. 40. Ohshige T, Iwata M, Omori S, Tanaka Y, Hirose H et al. (2011) Association of New Loci Identified in European Genome-wide Association Studies with Susceptibility to Type 2 Diabetes in the Japanese. PLOS ONE 6: e26911. doi:https://doi.org/10.1371/journal.pone.0026911. PubMed: 22046406.
  41. 41. Imamura M, Iwata M, Maegawa H, Watada H, Hirose H et al. (2011) Genetic variants at CDC123/CAMK1D and SPRY2 are associated with susceptibility to type 2 diabetes in Japanese population. Diabetologia 54: 3071–3077. doi:https://doi.org/10.1007/s00125-011-2293-3. PubMed: 21909839.
  42. 42. Imamura M, Iwata M, Maegawa H, Watada H, Hirose H et al. (2013) A replication study for the association of rs391300 in SRR and rs17584499 in PTPRD with susceptibility to type 2 diabetes in the Japanese. J Diabetes Investig 4: 168–173. doi:https://doi.org/10.1111/jdi.12017.