#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Genome-Wide Association Study Identifies Susceptibility Variants for Type 2 Diabetes in Han Chinese


To investigate the underlying mechanisms of T2D pathogenesis, we looked for diabetes susceptibility genes that increase the risk of type 2 diabetes (T2D) in a Han Chinese population. A two-stage genome-wide association (GWA) study was conducted, in which 995 patients and 894 controls were genotyped using the Illumina HumanHap550-Duo BeadChip for the first genome scan stage. This was further replicated in 1,803 patients and 1,473 controls in stage 2. We found two loci not previously associated with diabetes susceptibility in and around the genes protein tyrosine phosphatase receptor type D (PTPRD) (P = 8.54×10−10; odds ratio [OR] = 1.57; 95% confidence interval [CI] = 1.36–1.82), and serine racemase (SRR) (P = 3.06×10−9; OR = 1.28; 95% CI = 1.18–1.39). We also confirmed that variants in KCNQ1 were associated with T2D risk, with the strongest signal at rs2237895 (P = 9.65×10−10; OR = 1.29, 95% CI = 1.19–1.40). By identifying two novel genetic susceptibility loci in a Han Chinese population and confirming the involvement of KCNQ1, which was previously reported to be associated with T2D in Japanese and European descent populations, our results may lead to a better understanding of differences in the molecular pathogenesis of T2D among various populations.


Published in the journal: . PLoS Genet 6(2): e32767. doi:10.1371/journal.pgen.1000847
Category: Research Article
doi: https://doi.org/10.1371/journal.pgen.1000847

Summary

To investigate the underlying mechanisms of T2D pathogenesis, we looked for diabetes susceptibility genes that increase the risk of type 2 diabetes (T2D) in a Han Chinese population. A two-stage genome-wide association (GWA) study was conducted, in which 995 patients and 894 controls were genotyped using the Illumina HumanHap550-Duo BeadChip for the first genome scan stage. This was further replicated in 1,803 patients and 1,473 controls in stage 2. We found two loci not previously associated with diabetes susceptibility in and around the genes protein tyrosine phosphatase receptor type D (PTPRD) (P = 8.54×10−10; odds ratio [OR] = 1.57; 95% confidence interval [CI] = 1.36–1.82), and serine racemase (SRR) (P = 3.06×10−9; OR = 1.28; 95% CI = 1.18–1.39). We also confirmed that variants in KCNQ1 were associated with T2D risk, with the strongest signal at rs2237895 (P = 9.65×10−10; OR = 1.29, 95% CI = 1.19–1.40). By identifying two novel genetic susceptibility loci in a Han Chinese population and confirming the involvement of KCNQ1, which was previously reported to be associated with T2D in Japanese and European descent populations, our results may lead to a better understanding of differences in the molecular pathogenesis of T2D among various populations.

Introduction

Type 2 diabetes (T2D) affects at least 6% of the world's population; the worldwide prevalence is expected to double by 2025 [1]. T2D is a complex disorder that is characterized by hyperglycemia, which results from impaired pancreatic β cell function, decreased insulin action at target tissues, and increased glucose output by the liver [2]. Both genetic and environmental factors contribute to the pathogenesis of T2D. The disease is considered to be a polygenic disorder in which each genetic variant confers a partial and additive effect. Only 5%–10% of T2D cases are due to single gene defects; these include maturity-onset diabetes of the young (MODY), insulin resistance syndromes, mitochondrial diabetes, and neonatal diabetes [3][5]. Inherited variations have been identified from studies of monogenic diabetes, and have provided insights into β cell physiology, insulin release, and the action of insulin on target cells [6].

Much effort has been devoted to finding common T2D genes, including genome-wide linkage, candidate-gene, and genome-wide association studies (GWAS). Whole-genome linkage scans have identified chromosomal regions linked to T2D; however, with the exception of regions 1q [7][13] and 20q, which have been repeatedly mapped, linkage results vary from study to study [14][19]. Candidate-gene studies have provided strong evidence that common variants in the peroxisome proliferator-activated receptor-r (PPARG) [20], potassium inwardly-rectifying channel J11 (KCNJ11) [21][23], transcription factor 2 isoform b (TCF2) [24],[25], and Wolfram syndrome 1 (WFS1) [26] genes are associated with T2D. These genes all have strong biological links to diabetes, and rare, severe mutations cause monogenic diabetes. GWAS have accelerated the identification of T2D susceptibility genes, expanding the list from three in 2006 to over 20 genes in 2009. There are now at least 19 loci containing genes that increase risk of T2D, including PPARG [27], KCNJ11 [27], KCNQ1 [28],[29], CDKAL1 [27],[29][33], CDKN2A-2B [27],[32],[33], CDC123-CAMK1D [34], MTNR1B [35][37], TCF7L2 [31],[38],[39], TCF2 (HNF1B), HHEX-KIF11-IDE [27],[32],[33],[38], JAZF1 [34], IGF2BP2 [27],[29],[32], SLC30A8 [27],[32],[33],[38], THADA [34], ADAMTS9 [34], WFS1 [26], FTO [27],[31], NOTCH2 [34], and TSPAN8 [34]. Variants in these genes have been identified almost exclusively in populations of European descent, except for KCNQ1; individually, these variants confer a modest risk (odds ratio [OR] = 1.1–1.25) of developing T2D. KCNQ1 was identified as a T2D susceptibility gene in three GWA scans in Japanese individuals, highlighting the need to extend large-scale association efforts to different populations, such as Asian populations [28],[29],[40]. The association of other previously reported loci (CDKAL1, CDKN2A-2B, IGF2BP2, TCF7L2, SLC30A8, HHEX, and KCNJ11) with T2D were also replicated in the Japanese population [29],[40],[41].

To date, a GWA scan for T2D has not been conducted in the Han Chinese population, although the association of some known loci have been confirmed, including KCNQ1 and CDKAL1, CDKN2A-2B, MTNR1B, TCF7L2, HNF1β, and KCNJ11 [42][47]. Therefore, we conducted a two-stage GWA scan for T2D in a Han Chinese population residing in Taiwan. There were a total of 2,798 cases and 2,367 normal controls (995 cases and 894 controls in stage 1, 1,803 cases and 1,473 controls in stage 2). Our accomplished objective was to identify new diabetes susceptibility loci that were associated with increased risk of T2D in a Han Chinese population.

Results

Association analysis

We conducted a two-stage GWAS to identify genetic variants for T2D in the Han-Chinese residing in Taiwan. In the first stage, an exploratory genome-wide scan, we genotyped 995 T2D cases and 894 population controls using the Illumina Hap550duov3 chip (Figure 1 and Table S1). For each sample genotyped in this study, the average call rate was 99.92±0.12%. After applying stringent quality control criteria, high-quality genotypes for 516,737 SNPs (92.24%) were obtained, with an average call rate of 99.92±0.24% (Table S2). The results of principal component analysis in stage 1 revealed no evidence for population stratification between T2D cases and controls (P = 0.111, Fst statistics between populations <0.001) (Text S1; Figure S1). Multidimensional scaling analysis using PLINK [48] produced similar results (Text S1; Figure S2). Furthermore, genomic control (GC) with a variance inflation factor λ = 1.078 (trend test) did not substantially change the results of this GWAS (Table S3).

Graphical summary of T2D GWAS in a Han Chinese population.
Fig. 1. Graphical summary of T2D GWAS in a Han Chinese population.
T2D association was determined for SNPs on the Illumina HumanHap550K-Duo chip. The y-axis represents the −log10 P value and the x-axis represents each of the 516,212 SNPs used in the primary scan of 995 T2D cases and 894 controls.

We selected eight SNPs in seven regions: rs9985652 and rs2044844 on 4p13, rs7192960 on 16q23.1, rs7361808 on 20p13, rs1751960 on 10q11.23, rs4845624 on 1q21.3, rs391300 on 17p13.3, and rs648538 on 13q12.3. These SNPs had association P values of <10−5 at stage 1 with any of the genotype, allele, trend, dominant, and recessive models for subsequent cross-platform validation using Sequenom (Table 1; Table S3). For SNPs with weaker associations (P value between 10−4 and 10−5), we searched for novel susceptibility candidates for T2D as implicated by (1) gene function identified by a bioinformatics approach and (2) an animal model showing defects in glucose homeostasis caused by genes within the same subfamily. Therefore, we selected SNP rs17584499 (P = 2.4×10−5 under best model) for further investigation. rs17584499 lies within protein tyrosine phosphatase receptor type D (PTPRD). We hypothesized that PTPRD might play a role in the regulation of insulin signaling, because its subfamily members leukocyte common antigen-related (LAR) and protein tyrosine phosphatase sigma (PTPRS) exhibit defects in glucose homeostasis and insulin sensitivity in knockout and/or transgenic mice [49][51].

Tab. 1. Association results for Type 2 diabetes in Han Chinese.
Association results for Type 2 diabetes in Han Chinese.
aSNPs are arranged in order of decreasing P value under best statistical model in Stage 1.

We also evaluated the most significant SNP (rs231361) within KCNQ1, which was previously reported to be a diabetes susceptibility gene in a Japanese population, as well as in populations of Korean, Chinese, and European ancestry [28],[29]. Together, these ten SNPs—the 8 SNPs with association p<10−5, rs17584499, and rs231361—were cross-platform validated and yielded consistent results using both Illumina and Sequenom. The concordance rate for stage 1 samples typed on the Illumina and Sequenom platforms was 99.1%±0.84% (Table S4).

We took these ten SNPs and an additional 29 neighboring SNPs within the linkage disequilibrium (LD) block forward to replicate in 3,803 additional samples (stage 2; 1,803 cases and 1,473 controls). The average call rate for each sample was 96.13%±4.66%. After applying stringent quality control criteria, high-quality genotypes for 35 SNPs (89.7%) were obtained, with an average call rate of 98.96%±0.24% (Table S2). Of the ten SNPs selected in stage 1, only three SNPs still showed a strong association in the stage 2 analysis: rs17584499 in PTPRD at 9p24.1-p23, rs231359 in KCNQ1 at 11p15.5, and rs391300 in serine racemase (SRR) at 17p13.3 (Table 1). We were unable to replicate the association between T2D and the remaining seven SNPs in ATP8A1/GRXCR1, MAF/WWOX, SIRPA, LYZL1/SVIL, RORC/TMEM5, and KATNAL1 in the stage 2 analysis (Table 1). Joint analysis of stage 1 and stage 2 data revealed consistent results with stage 2. The most significant associations were found for rs391300, rs17584499, and rs231359 (Table 1; Figure 2). These associations remained significant after calculating P values using 108 permutations of the disease state labels. Joint association analysis was performed with all of the 2,798 T2D cases and 2,367 controls; this could achieve a power of 0.85 to detect a disease allele with a frequency of 0.15 and an OR of 1.5, assuming a disease prevalence of 0.06, at a significant level of 0.05 (Table S5).

Regional plots of three significant associations.
Fig. 2. Regional plots of three significant associations.
For each of the (A) PTPRD, (B) SRR, and (C) KCNQ1 regions, the −log10 P values for the trend test from the primary scan were plotted as a function of genomic position (NCBI Build 36). The SNPs with the strongest signal and neighboring genotyped SNPs in the joint analysis are denoted by blue diamonds. Green diamonds in the KCNQ1 region (C) represent reported T2D–associated SNPs genotyped in all samples of joint analysis. Estimated recombination rates (right y-axis) based on the Chinese HapMap population was plotted to reflect the local LD structure around the significant SNPs. Gene annotations were taken from NCBI.

Identification of two novel T2D loci and confirmation of KCNQ1 association

Two previously unknown loci were detected in our joint analysis of GWAS data. The strongest new association signal was found for rs17584499 in intron 10 of PTPRD (P = 8.54×10−10 [trend test]; allelic OR = 1.57, 95% confidence interval [CI] = 1.36–1.82) (Table 1; Figure 2). The second strongest signal was found with rs391300 (P = 3.06×10−9 [trend test]; OR = 1.28, 95% CI = 1.18–1.39). The nearby SNP rs4523957 also demonstrated a significant association (P = 1.44×10−8; OR = 1.27, 95% CI = 1.17–1.38). SNPs rs391300 and rs4523957 were in tight LD with one another (r2 = 0.942 in HapMap HCB), and were located within the serine racemase gene (SRR).

SNP rs231361, located in intron 11 of KCNQ1, had a less significant association with T2D, and was selected in stage 1 (P = 1.49×10−4 [trend test]; OR = 1.39, 95% CI = 1.17–1.64) (Table 1). We further genotyped eight additional SNPs within the same LD block from the HapMap Asian group data: rs231359 yielded a P value of 4.56×10−4 with a trend test (OR = 1.36, 95% CI = 1.14–1.61) (Figure 2). rs231361 and rs231359 were in strong LD with one another (r2 = 1 in HapMap HCB), and were located approximately 164 kb upstream of SNP rs2237897, which was previously reported to be significantly associated with T2D in a Japanese population [28],[29]. We took rs231361, rs231359, and neighboring SNPs within the LD block forward to replicate in stage 2. Joint analysis of stage 1 and stage 2 data revealed that rs231359 had an even stronger association with T2D than did rs231361 (rs231359: P = 3.43×10−8, OR = 1.33, 95% CI = 1.2–1.48; rs231361: P = 2.89×10−7, OR = 1.3, 95% CI = 1.18–1.44).

Additional SNPs that were reported to be significantly associated with T2D in a Japanese population were further genotyped [28],[29]. The average call rate for each sample was 99.12%±7.21%. After applying stringent quality control criteria, we obtained high-quality genotypes with an average call rate of 99.16%±0.18% (Table S2). SNP rs2237895 showed the strongest association with T2D of all the genotyped SNPs in KCNQ1 (P = 9.65×10−10; OR = 1.29, 95% CI = 1.19–1.40) (Figure 2 and Figure S3; Table S6). Conditioning on the rs2237895, the statistical significance of rs231361 (or rs231359) disappeared. It seems the same underlying biological effect between the 2 SNPs (Table S7).

Subsequently, we sequenced all of the exons, intron–exon boundaries, and up to 1.2 kb of the promoter region of the KCNQ1 gene in 50 individuals with T2D, and identified 42 polymorphic variations, including one nonsynonymous P448R polymorphism and two novel SNPs with minor allele frequency >0.03. We then genotyped the two novel SNPs and one nonsynonymous polymorphism; however, none of these SNPs showed an association with T2D (Table S6).

Discussion

Our GWAS for T2D in a Han Chinese population found two previously unreported susceptibility genes. All of the significant variants detected in our study showed modest effects, with an OR between 1.21 and 1.57. Two loci with less-significant associations in our primary scan (stage 1), PTPRD and KCNQ1, were selected for further replication; both showed compelling evidence of association in joint analysis. The susceptibility loci we identified in this study need to be further replicated in additional populations. Of the 18 loci previously reported to be associated with T2D (with the exception of KCNQ1), none of the P values for any of the SNPs within or near the genes reached 10−5 using allele, genotype, trend, dominant, or recessive models (Table S8; Figure S4). Three SNPs within CDKAL1, JAZF1, and HNF1B had the lowest P values, ranging from 5×10−4 to 10−5, among the 18 known loci (Table S8). No significant associations were found within these regions in our Han Chinese population.

The strongest new signal was observed for rs17584499 in PTPRD. The overall Fst among 11 HapMap groups for rs17584499 was estimated to be 0.068 [52], which indicated a significant difference in allele frequencies among the populations (P<0.0001, chi-square test ) (Table S9). PTPRD is widely expressed in tissues, including skeletal muscle and pancreas, and is expressed highest in the brain. PTPRD-deficient mice exhibit impaired learning and memory, early growth retardation, neonatal mortality, and posture and motor defects [53]. Multiple mRNA isoforms are expressed by alternative splicing and/or alternative transcription start sites in a developmental and tissue-specific manner [54],[55]. PTPRD belongs to the receptor type IIA (R2A) subfamily of protein tyrosine phosphatases (PTPs). The R2A PTP subfamily comprises LAR, PTPRS, and PTPRD. The R2A family has been implicated in neural development, cancer, and diabetes [56]. Although the complex phenotype including neurological defects seen in knockout mice could obscure the roles of these genes in glucose homeostasis, LAR- and PTPRS-deficient mice were demonstrated to have altered glucose homeostasis and insulin sensitivity [49][51]. Transgenic mice overexpressing LAR in skeletal muscle show whole-body insulin resistance [57]. Because R2A subfamily members are structurally very similar [54], PTPRD could play a role in T2D pathogenesis and should be further characterized.

The second new association locus was found for rs391300 and rs4523957 in the biologically plausible candidate gene SRR. SRR encodes a serine racemase that synthesizes D-serine from L-serine [58],[59]. D-serine is a physiological co-agonist of the N-methyl D-aspartate (NMDA) class of glutamate receptors, the major excitatory neurotransmitter receptors mediating synaptic neurotransmission in the brain [60],[61]. NMDA receptor activation requires binding of glutamate and D-serine, which plays a neuromodulatory role in NMDA receptor transmission, synaptic plasticity, cell migration, and neurotoxicity [62]. D-serine and SRR are also present in the pancreas [63]. Glutamate signaling functions in peripheral tissues, including the pancreas, and positively modulates secretion of both glucagon and insulin in pancreatic islets [64][66]. The nearby SNP rs216193 also showed significant association (P = 2.49×10−6); this SNP resides 3.8 kb upstream from SRR, within Smg-6 homolog, nonsense mediated mRNA decay factor (C. elegans) (SMG6). rs216193 was in tight LD with rs391300 (r2 = 0.942 in HapMap HCB). Based on their biological functions and the association results, neither SMG6 nor any of the nearby genes TSR1, SGSM2, MNT, and METT10D were compelling candidates for association withT2D. However, SRR was significantly associated with T2D; thus, we suggest that dysregulation of D-serine could alter glutamate signaling and affect insulin or glucagon secretion in T2D pathogenesis.

rs7192960 also had a suggestive association with T2D (P = 1.32×10−5; OR = 1.21, 95% CI = 1.11–1.33). This SNP which lies approximately 211 kb downstream of v-maf musculoaponeurotic fibrosarcoma oncogene homolog (avian) (MAF) and 170 kb downstream of WW domain containing oxidoreductase (WWOX). WWOX is a tumor suppressor gene that spans the second most common human fragile site FRA16D [67],[68], and is disrupted in many tumors, including pancreatic carcinoma [67], [69][73]. MAF encodes the transcription factor c-Maf, a member of the Maf family of basic-Zip (bZip) transcription factors. c-Maf is involved in development and differentiation of the lens [74],[75], kidney [76], immune system [77], adipose tissue [78], and pancreas [79]. It is expressed in α cells of the pancreatic islets [80], and is a strong transactivator of the glucagon promoter that regulates glucagon gene expression [80],[81]. c-Maf is also associated with early-onset and morbid adult obesity [82].

Our GWAS revealed that KCNQ1, which was previously reported to be associated with T2D in several populations, was also associated with T2D in a Han Chinese population residing in Taiwan. KCNQ1 encodes the pore-forming α subunit of a voltage-gated K+ channel (KvLQT1), which is involved in repolarization of the action potential in cardiac muscle [83],[84]. Mutations in KCNQ1 cause long QT syndrome [85],[86] and familial atrial fibrillation [87]. KCNQ1 is widely expressed, including in the heart, brain, kidney, liver, intestine, and pancreas [88][90]. It is also expressed in pancreatic islets, and blockade of the KvLQT1 channel stimulates insulin secretion in insulin-secreting INS-1 cells [91]. KCNQ1 knockout mice have cardiac dysfunctions [88],[92] and enhanced systemic insulin sensitivity [93]. In our study, variants in the coding region did not show an association with T2D. The functional variant(s) could be located in the regulatory element of KCNQ1, rather than in the coding region. We did not find an association between either CDKAL1 or IGF2BP2 and T2D, in contrast with the results described in a previous study [29], nor did we find T2D associated with various other genes identified in populations of European descent.

In conclusion, we identified two previously unknown loci that are associated with T2D in a Han Chinese population, and confirmed the reported association of KCNQ1 with T2D. The novel T2D risk loci may involve genes that are implicated in insulin sensitivity and control of glucagon and insulin secretion: PTPRD may participate in the regulation of insulin action on its target cells, while SRR variants may alter glutamate signaling in the pancreas, thus regulating insulin and/or glucagon secretion. Our study suggests that in different patient populations, different genes may confer risks for diabetes, which may lead to a better understanding of the molecular pathogenesis of T2D.

Materials and Methods

Ethical statement

The study was approved by the institutional review board and the ethics committee of each institution. Written informed consent was obtained from each participant in accordance with institutional requirements and the Declaration of Helsinki Principles.

Subject participants

A total of 2,798 unrelated individuals with T2D, age >20 years, were recruited from China Medical University Hospital (CMUH), Taichung, Taiwan; Chia-Yi Christian Hospital (CYCH), Chia-Yi, Taiwan; and National Taiwan University Hospital (NTU), Taipei, Taiwan. All of the T2D cases were diagnosed according to medical records and fasting plasma glucose levels using American Diabetic Association Criteria. Subjects with type 1 diabetes, gestational diabetes, and maturity-onset diabetes of the young (MODY) were excluded from this study. For the two-stage GWAS, we genotyped 995 T2D cases and 894 controls in the first exploratory genome-wide scan (stage 1). In the replication stage (stage 2), we genotyped selected SNPs in additional samples from 1,803 T2D cases and 1,473 controls. The controls were randomly selected from the Taiwan Han Chinese Cell and Genome Bank [94]. The criteria for controls in the association study were (1) no past diagnostic history of T2D, (2) HbA1C ranging from 3.4 to 6, and (3) BMI<32. The two control groups were comparable with respect to BMI, gender, age at study, and level of HbA1C. All of the participating T2D cases and controls were of Han Chinese origin, which is the origin of 98% of the Taiwan population. Details of demographic data are shown in Table S10.

Genotyping

Genomic DNA was extracted from peripheral blood using the Puregene DNA isolation kit (Gentra Systems, Minneapolis, MN, USA). In stage 1, whole genome genotyping using the Illumina HumanHap550-Duo BeadChip was performed by deCODE Genetics (Reykjavík, Iceland). Genotype calling was performed using the standard procedure implemented in BeadStudio (Illumina, Inc., San Diego, CA, USA), with the default parameters suggested by the platform manufacturer. Quality control of genotype data was performed by examining several summary statistics. First, the ratio of loci with heterozygous calls on the X chromosome was calculated to double-check the subject's gender. Total successful call rate and the minor allele frequency of cases and controls were also calculated for each SNP. SNPs were excluded if they: (1) were nonpolymorphic in both cases and controls, (2) had a total call rate <95% in the cases and controls combined, (3) had a minor allele frequency <5% and a total call rate <99% in the cases and controls combined, and (4) had significant distortion from Hardy–Weinberg equilibrium in the controls (P<10−7). Genotyping validation was performed using the Sequenom iPLEX assay (Sequenom MassARRAY system; Sequenom, San Diego, CA, USA). In the replication stage (stage 2), SNPs showing significant or suggestive associations with T2D and their neighboring SNPs within the same LD block were genotyped using the Sequenom iPLEX assay. The neighboring SNPs in the same LD were selected from the HapMap Asian (CHB + JPT) group data for fine mapping the significant signal.

Statistical analysis

T2D association analysis was carried out to compare allele frequency and genotype distribution between cases and controls using five single-point methods for each SNP: genotype, allele, trend (Cochran–Armitage test), dominant, and recessive models. The most significant test statistic obtained from the five models was chosen. SNPs with P values less than a = 2×10−8, a cut-off for the multiple comparison adjusted by Bonferroni correction, were considered to be significantly associated with the traits. The joint analysis was conducted by combining the data from the stage 1 and 2 samples. We also applied Fisher's method to combine P values for joint analysis. The permutation test was carried out genome-wide for 106 permutations, in which the phenotypes of subjects were randomly rearranged. For better estimation of empirical P values, the top SNPs were reexamined using 108 permutations. Each permutation proceeded as follows: (1) the case and control labels were shuffled and redistributed to subjects, and (2) the test statistics of the corresponding association test was calculated based on the shuffled labels. The empirical P value was defined as the number of permutations that were at least as extreme as the original divided by the total number of permutations. Detection of possible population stratification that might influence association analysis was carried out using principle component analysis, multidimensional scaling analysis, and genomic control (Text S1). Quantile–quantile (Q–Q) plots were then used to examine P value distributions (Figure 3 and Figure S5).

Q–Q plot for the trend test.
Fig. 3. Q–Q plot for the trend test.
Q–Q plots are shown for the trend test based on the 516,212 quality SNPs of the initial analysis of 995 cases and 894 controls. The red lines represent the upper and lower boundaries of the 95% confidence bands.

Supporting Information

Attachment 1

Attachment 2

Attachment 3

Attachment 4

Attachment 5

Attachment 6

Attachment 7

Attachment 8

Attachment 9

Attachment 10

Attachment 11

Attachment 12

Attachment 13

Attachment 14

Attachment 15

Attachment 16


Zdroje

1. ZimmetP

AlbertiKG

ShawJ

2001 Global and societal implications of the diabetes epidemic. Nature 414 782 787

2. StumvollM

GoldsteinBJ

van HaeftenTW

2005 Type 2 diabetes: principles of pathogenesis and therapy. Lancet 365 1333 1346

3. LedermannHM

1995 Maturity-onset diabetes of the young (MODY) at least ten times more common in Europe than previously assumed? Diabetologia 38 1482

4. MaassenJA

LMTH

Van EssenE

HeineRJ

NijpelsG

2004 Mitochondrial diabetes: molecular mechanisms and clinical presentation. Diabetes 53 Suppl 1 S103 109

5. FajansSS

BellGI

PolonskyKS

2001 Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N Engl J Med 345 971 980

6. MooreAF

FlorezJC

2008 Genetic susceptibility to type 2 diabetes and implications for antidiabetic therapy. Annu Rev Med 59 95 111

7. LangefeldCD

WagenknechtLE

RotterJI

WilliamsAH

HokansonJE

2004 Linkage of the metabolic syndrome to 1q23-q31 in Hispanic families: the Insulin Resistance Atherosclerosis Study Family Study. Diabetes 53 1170 1174

8. HansonRL

EhmMG

PettittDJ

ProchazkaM

ThompsonDB

1998 An autosomal genomic scan for loci linked to type II diabetes mellitus and body-mass index in Pima Indians. Am J Hum Genet 63 1130 1138

9. ElbeinSC

HoffmanMD

TengK

LeppertMF

HasstedtSJ

1999 A genome-wide search for type 2 diabetes susceptibility genes in Utah Caucasians. Diabetes 48 1175 1182

10. VionnetN

HaniEH

DupontS

GallinaS

FranckeS

2000 Genomewide search for type 2 diabetes-susceptibility genes in French whites: evidence for a novel susceptibility locus for early-onset diabetes on chromosome 3q27-qter and independent replication of a type 2-diabetes locus on chromosome 1q21-q24. Am J Hum Genet 67 1470 1480

11. AnP

HongY

WeisnagelSJ

RiceT

RankinenT

2003 Genomic scan of glucose and insulin metabolism phenotypes: the HERITAGE Family Study. Metabolism 52 246 253

12. WiltshireS

HattersleyAT

HitmanGA

WalkerM

LevyJC

2001 A genomewide scan for loci predisposing to type 2 diabetes in a U.K. population (the Diabetes UK Warren 2 Repository): analysis of 573 pedigrees provides independent replication of a susceptibility locus on chromosome 1q. Am J Hum Genet 69 553 569

13. HsuehWC

St JeanPL

MitchellBD

PollinTI

KnowlerWC

2003 Genome-wide and fine-mapping linkage studies of type 2 diabetes and glucose traits in the Old Order Amish: evidence for a new diabetes locus on chromosome 14q11 and confirmation of a locus on chromosome 1q21-q24. Diabetes 52 550 557

14. BowdenDW

SaleM

HowardTD

QadriA

SprayBJ

1997 Linkage of genetic markers on human chromosomes 20 and 12 to NIDDM in Caucasian sib pairs with a history of diabetic nephropathy. Diabetes 46 882 886

15. XiangK

WangY

ZhengT

JiaW

LiJ

2004 Genome-wide search for type 2 diabetes/impaired glucose homeostasis susceptibility genes in the Chinese: significant linkage to chromosome 6q21-q23 and chromosome 1q21-q24. Diabetes 53 228 234

16. JiL

MaleckiM

WarramJH

YangY

RichSS

1997 New susceptibility locus for NIDDM is localized to human chromosome 20q. Diabetes 46 876 881

17. ZoualiH

HaniEH

PhilippiA

VionnetN

BeckmannJS

1997 A susceptibility locus for early-onset non-insulin dependent (type 2) diabetes mellitus maps to chromosome 20q, proximal to the phosphoenolpyruvate carboxykinase gene. Hum Mol Genet 6 1401 1408

18. GhoshS

WatanabeRM

HauserER

ValleT

MagnusonVL

1999 Type 2 diabetes: evidence for linkage on chromosome 20 in 716 Finnish affected sib pairs. Proc Natl Acad Sci U S A 96 2198 2203

19. KlupaT

MaleckiMT

PezzolesiM

JiL

CurtisS

2000 Further evidence for a susceptibility locus for type 2 diabetes on chromosome 20q13.1-q13.2. Diabetes 49 2212 2216

20. AltshulerD

HirschhornJN

KlannemarkM

LindgrenCM

VohlMC

2000 The common PPARgamma Pro12Ala polymorphism is associated with decreased risk of type 2 diabetes. Nat Genet 26 76 80

21. FlorezJC

BurttN

de BakkerPI

AlmgrenP

TuomiT

2004 Haplotype structure and genotype-phenotype correlations of the sulfonylurea receptor and the islet ATP-sensitive potassium channel gene region. Diabetes 53 1360 1368

22. GloynAL

WeedonMN

OwenKR

TurnerMJ

KnightBA

2003 Large-scale association studies of variants in genes encoding the pancreatic beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1 (ABCC8) confirm that the KCNJ11 E23K variant is associated with type 2 diabetes. Diabetes 52 568 572

23. NielsenEM

HansenL

CarstensenB

EchwaldSM

DrivsholmT

2003 The E23K variant of Kir6.2 associates with impaired post-OGTT serum insulin response and increased risk of type 2 diabetes. Diabetes 52 573 577

24. GudmundssonJ

SulemP

SteinthorsdottirV

BergthorssonJT

ThorleifssonG

2007 Two variants on chromosome 17 confer prostate cancer risk, and the one in TCF2 protects against type 2 diabetes. Nat Genet 39 977 983

25. WincklerW

WeedonMN

GrahamRR

McCarrollSA

PurcellS

2007 Evaluation of common variants in the six known maturity-onset diabetes of the young (MODY) genes for association with type 2 diabetes. Diabetes 56 685 693

26. SandhuMS

WeedonMN

FawcettKA

WassonJ

DebenhamSL

2007 Common variants in WFS1 confer risk of type 2 diabetes. Nat Genet 39 951 953

27. ScottLJ

MohlkeKL

BonnycastleLL

WillerCJ

LiY

2007 A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science 316 1341 1345

28. YasudaK

MiyakeK

HorikawaY

HaraK

OsawaH

2008 Variants in KCNQ1 are associated with susceptibility to type 2 diabetes mellitus. Nat Genet 40 1092 1097

29. UnokiH

TakahashiA

KawaguchiT

HaraK

HorikoshiM

2008 SNPs in KCNQ1 are associated with susceptibility to type 2 diabetes in East Asian and European populations. Nat Genet 40 1098 1102

30. SteinthorsdottirV

ThorleifssonG

ReynisdottirI

BenediktssonR

JonsdottirT

2007 A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet 39 770 775

31. 2007 Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447 661 678

32. SaxenaR

VoightBF

LyssenkoV

BurttNP

de BakkerPI

2007 Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science 316 1331 1336

33. ZegginiE

WeedonMN

LindgrenCM

FraylingTM

ElliottKS

2007 Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science 316 1336 1341

34. ZegginiE

ScottLJ

SaxenaR

VoightBF

MarchiniJL

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

35. Bouatia-NajiN

BonnefondA

Cavalcanti-ProencaC

SparsoT

HolmkvistJ

2009 A variant near MTNR1B is associated with increased fasting plasma glucose levels and type 2 diabetes risk. Nat Genet 41 89 94

36. LyssenkoV

NagornyCL

ErdosMR

WierupN

JonssonA

2009 Common variant in MTNR1B associated with increased risk of type 2 diabetes and impaired early insulin secretion. Nat Genet 41 82 88

37. ProkopenkoI

LangenbergC

FlorezJC

SaxenaR

SoranzoN

2009 Variants in MTNR1B influence fasting glucose levels. Nat Genet 41 77 81

38. SladekR

RocheleauG

RungJ

DinaC

ShenL

2007 A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature 445 881 885

39. GrantSF

ThorleifssonG

ReynisdottirI

BenediktssonR

ManolescuA

2006 Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. Nat Genet 38 320 323

40. TakeuchiF

SerizawaM

YamamotoK

FujisawaT

NakashimaE

2009 Confirmation of multiple risk Loci and genetic impacts by a genome-wide association study of type 2 diabetes in the Japanese population. Diabetes 58 1690 1699

41. OmoriS

TanakaY

TakahashiA

HiroseH

KashiwagiA

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

42. HuC

WangC

ZhangR

MaX

WangJ

2009 Variations in KCNQ1 are associated with type 2 diabetes and beta cell function in a Chinese population. Diabetologia 52 1322 1325

43. WuY

LiH

LoosRJ

YuZ

YeX

2008 Common variants in CDKAL1, CDKN2A/B, IGF2BP2, SLC30A8, and HHEX/IDE genes are associated with type 2 diabetes and impaired fasting glucose in a Chinese Han population. Diabetes 57 2834 2842

44. ChangYC

ChangTJ

JiangYD

KuoSS

LeeKC

2007 Association study of the genetic polymorphisms of the transcription factor 7-like 2 (TCF7L2) gene and type 2 diabetes in the Chinese population. Diabetes 56 2631 2637

45. RonnT

WenJ

YangZ

LuB

DuY

2009 A common variant in MTNR1B, encoding melatonin receptor 1B, is associated with type 2 diabetes and fasting plasma glucose in Han Chinese individuals. Diabetologia 52 830 833

46. WangC

HuC

ZhangR

BaoY

MaX

2009 Common variants of hepatocyte nuclear factor 1beta are associated with type 2 diabetes in a Chinese population. Diabetes 58 1023 1027

47. ZhouD

ZhangD

LiuY

ZhaoT

ChenZ

2009 The E23K variation in the KCNJ11 gene is associated with type 2 diabetes in Chinese and East Asian population. J Hum Genet 54 433 435

48. PurcellS

NealeB

Todd-BrownK

ThomasL

FerreiraMA

2007 PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet 81 559 575

49. RenJM

LiPM

ZhangWR

SweetLJ

ClineG

1998 Transgenic mice deficient in the LAR protein-tyrosine phosphatase exhibit profound defects in glucose homeostasis. Diabetes 47 493 497

50. ChagnonMJ

ElcheblyM

UetaniN

DombrowskiL

ChengA

2006 Altered glucose homeostasis in mice lacking the receptor protein tyrosine phosphatase sigma. Can J Physiol Pharmacol 84 755 763

51. BattJ

AsaS

FladdC

RotinD

2002 Pituitary, pancreatic and gut neuroendocrine defects in protein tyrosine phosphatase-sigma-deficient mice. Mol Endocrinol 16 155 169

52. WrightS

1951 The genetical structure of populations. Annals of Eugenics 15 323 354

53. UetaniN

KatoK

OguraH

MizunoK

KawanoK

2000 Impaired learning with enhanced hippocampal long-term potentiation in PTPdelta-deficient mice. Embo J 19 2775 2785

54. PulidoR

Serra-PagesC

TangM

StreuliM

1995 The LAR/PTP delta/PTP sigma subfamily of transmembrane protein-tyrosine-phosphatases: multiple human LAR, PTP delta, and PTP sigma isoforms are expressed in a tissue-specific manner and associate with the LAR-interacting protein LIP.1. Proc Natl Acad Sci U S A 92 11686 11690

55. SatoM

TakahashiK

NagayamaK

AraiY

ItoN

2005 Identification of chromosome arm 9p as the most frequent target of homozygous deletions in lung cancer. Genes Chromosomes Cancer 44 405 414

56. ChagnonMJ

UetaniN

TremblayML

2004 Functional significance of the LAR receptor protein tyrosine phosphatase family in development and diseases. Biochem Cell Biol 82 664 675

57. ZabolotnyJM

KimYB

PeroniOD

KimJK

PaniMA

2001 Overexpression of the LAR (leukocyte antigen-related) protein-tyrosine phosphatase in muscle causes insulin resistance. Proc Natl Acad Sci U S A 98 5187 5192

58. WoloskerH

BlackshawS

SnyderSH

1999 Serine racemase: a glial enzyme synthesizing D-serine to regulate glutamate-N-methyl-D-aspartate neurotransmission. Proc Natl Acad Sci U S A 96 13409 13414

59. WoloskerH

ShethKN

TakahashiM

MothetJP

BradyROJr

1999 Purification of serine racemase: biosynthesis of the neuromodulator D-serine. Proc Natl Acad Sci U S A 96 721 725

60. De MirandaJ

PanizzuttiR

FoltynVN

WoloskerH

2002 Cofactors of serine racemase that physiologically stimulate the synthesis of the N-methyl-D-aspartate (NMDA) receptor coagonist D-serine. Proc Natl Acad Sci U S A 99 14542 14547

61. MothetJP

ParentAT

WoloskerH

BradyROJr

LindenDJ

2000 D-serine is an endogenous ligand for the glycine site of the N-methyl-D-aspartate receptor. Proc Natl Acad Sci U S A 97 4926 4931

62. WoloskerH

DuminE

BalanL

FoltynVN

2008 D-amino acids in the brain: D-serine in neurotransmission and neurodegeneration. Febs J 275 3514 3526

63. ImaiK

FukushimaT

SantaT

HommaH

HuangY

1998 Whole body autoradiographic study on the distribution of 14C-D-serine administered intravenously to rats. Amino Acids 15 351 361

64. GonoiT

MizunoN

InagakiN

KuromiH

SeinoY

1994 Functional neuronal ionotropic glutamate receptors are expressed in the non-neuronal cell line MIN6. J Biol Chem 269 16989 16992

65. InagakiN

KuromiH

GonoiT

OkamotoY

IshidaH

1995 Expression and role of ionotropic glutamate receptors in pancreatic islet cells. Faseb J 9 686 691

66. BertrandG

GrossR

PuechR

Loubatieres-MarianiMM

BockaertJ

1993 Glutamate stimulates glucagon secretion via an excitatory amino acid receptor of the AMPA subtype in rat pancreas. Eur J Pharmacol 237 45 50

67. BednarekAK

LaflinKJ

DanielRL

LiaoQ

HawkinsKA

2000 WWOX, a novel WW domain-containing protein mapping to human chromosome 16q23.3-24.1, a region frequently affected in breast cancer. Cancer Res 60 2140 2145

68. RiedK

FinnisM

HobsonL

MangelsdorfM

DayanS

2000 Common chromosomal fragile site FRA16D sequence: identification of the FOR gene spanning FRA16D and homozygous deletions and translocation breakpoints in cancer cells. Hum Mol Genet 9 1651 1663

69. IliopoulosD

GulerG

HanSY

JohnstonD

DruckT

2005 Fragile genes as biomarkers: epigenetic control of WWOX and FHIT in lung, breast and bladder cancer. Oncogene 24 1625 1633

70. KurokiT

YendamuriS

TrapassoF

MatsuyamaA

AqeilanRI

2004 The tumor suppressor gene WWOX at FRA16D is involved in pancreatic carcinogenesis. Clin Cancer Res 10 2459 2465

71. PaigeAJ

TaylorKJ

TaylorC

HillierSG

FarringtonS

2001 WWOX: a candidate tumor suppressor gene involved in multiple tumor types. Proc Natl Acad Sci U S A 98 11417 11422

72. KurokiT

TrapassoF

ShiraishiT

AlderH

MimoriK

2002 Genetic alterations of the tumor suppressor gene WWOX in esophageal squamous cell carcinoma. Cancer Res 62 2258 2260

73. YendamuriS

KurokiT

TrapassoF

HenryAC

DumonKR

2003 WW domain containing oxidoreductase gene expression is altered in non-small cell lung cancer. Cancer Res 63 878 881

74. SakaiM

ImakiJ

YoshidaK

OgataA

Matsushima-HibayaY

1997 Rat maf related genes: specific expression in chondrocytes, lens and spinal cord. Oncogene 14 745 750

75. KimJI

LiT

HoIC

GrusbyMJ

GlimcherLH

1999 Requirement for the c-Maf transcription factor in crystallin gene regulation and lens development. Proc Natl Acad Sci U S A 96 3781 3785

76. ImakiJ

TsuchiyaK

MishimaT

OnoderaH

KimJI

2004 Developmental contribution of c-maf in the kidney: distribution and developmental study of c-maf mRNA in normal mice kidney and histological study of c-maf knockout mice kidney and liver. Biochem Biophys Res Commun 320 1323 1327

77. AgnelloD

LankfordCS

BreamJ

MorinobuA

GadinaM

2003 Cytokines and transcription factors that regulate T helper cell differentiation: new players and new insights. J Clin Immunol 23 147 161

78. SerriaMS

IkedaH

OmoteyamaK

HirokawaJ

NishiS

2003 Regulation and differential expression of the c-maf gene in differentiating cultured cells. Biochem Biophys Res Commun 310 318 326

79. TsuchiyaM

TaniguchiS

YasudaK

NittaK

MaedaA

2006 Potential roles of large mafs in cell lineages and developing pancreas. Pancreas 32 408 416

80. KataokaK

ShiodaS

AndoK

SakagamiK

HandaH

2004 Differentially expressed Maf family transcription factors, c-Maf and MafA, activate glucagon and insulin gene expression in pancreatic islet alpha- and beta-cells. J Mol Endocrinol 32 9 20

81. GosmainY

AvrilI

MaminA

PhilippeJ

2007 Pax-6 and c-Maf functionally interact with the alpha-cell-specific DNA element G1 in vivo to promote glucagon gene expression. J Biol Chem 282 35024 35034

82. MeyreD

DelplanqueJ

ChevreJC

LecoeurC

LobbensS

2009 Genome-wide association study for early-onset and morbid adult obesity identifies three new risk loci in European populations. Nat Genet 41 157 159

83. SanguinettiMC

CurranME

ZouA

ShenJ

SpectorPS

1996 Coassembly of K(V)LQT1 and minK (IsK) proteins to form cardiac I(Ks) potassium channel. Nature 384 80 83

84. BarhaninJ

LesageF

GuillemareE

FinkM

LazdunskiM

1996 K(V)LQT1 and lsK (minK) proteins associate to form the I(Ks) cardiac potassium current. Nature 384 78 80

85. NeyroudN

TessonF

DenjoyI

LeiboviciM

DongerC

1997 A novel mutation in the potassium channel gene KVLQT1 causes the Jervell and Lange-Nielsen cardioauditory syndrome. Nat Genet 15 186 189

86. WangQ

CurranME

SplawskiI

BurnTC

MillhollandJM

1996 Positional cloning of a novel potassium channel gene: KVLQT1 mutations cause cardiac arrhythmias. Nat Genet 12 17 23

87. ChenYH

XuSJ

BendahhouS

WangXL

WangY

2003 KCNQ1 gain-of-function mutation in familial atrial fibrillation. Science 299 251 254

88. LeeMP

RavenelJD

HuRJ

LustigLR

TomaselliG

2000 Targeted disruption of the Kvlqt1 gene causes deafness and gastric hyperplasia in mice. J Clin Invest 106 1447 1455

89. DemolombeS

FrancoD

de BoerP

KuperschmidtS

RodenD

2001 Differential expression of KvLQT1 and its regulator IsK in mouse epithelia. Am J Physiol Cell Physiol 280 C359 372

90. ChouabeC

NeyroudN

GuicheneyP

LazdunskiM

RomeyG

1997 Properties of KvLQT1 K+ channel mutations in Romano-Ward and Jervell and Lange-Nielsen inherited cardiac arrhythmias. Embo J 16 5472 5479

91. UllrichS

SuJ

RantaF

WittekindtOH

RisF

2005 Effects of I(Ks) channel inhibitors in insulin-secreting INS-1 cells. Pflugers Arch 451 428 436

92. CasimiroMC

KnollmannBC

EbertSN

VaryJCJr

GreeneAE

2001 Targeted disruption of the Kcnq1 gene produces a mouse model of Jervell and Lange-Nielsen Syndrome. Proc Natl Acad Sci U S A 98 2526 2531

93. BoiniKM

GrafD

HennigeAM

KokaS

KempeDS

2009 Enhanced insulin sensitivity of gene-targeted mice lacking functional KCNQ1. Am J Physiol Regul Integr Comp Physiol 296 R1695 1701

94. PanWH

FannCS

WuJY

HungYT

HoMS

2006 Han Chinese cell and genome bank in Taiwan: purpose, design and ethical considerations. HumHered 61 27 30

Štítky
Genetika Reprodukční medicína

Článek vyšel v časopise

PLOS Genetics


2010 Číslo 2

Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Hypertenze a hypercholesterolémie – synergický efekt léčby
nový kurz
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Multidisciplinární zkušenosti u pacientů s diabetem
Autoři: Prof. MUDr. Martin Haluzík, DrSc., prof. MUDr. Vojtěch Melenovský, CSc., prof. MUDr. Vladimír Tesař, DrSc.

Úloha kombinovaných preparátů v léčbě arteriální hypertenze
Autoři: prof. MUDr. Martin Haluzík, DrSc.

Halitóza
Autoři: MUDr. Ladislav Korábek, CSc., MBA

Terapie roztroušené sklerózy v kostce
Autoři: MUDr. Dominika Šťastná, Ph.D.

Všechny kurzy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#