Open Access

Whole-genome re-sequencing for the identification of high contribution susceptibility gene variants in patients with type 2 diabetes

  • Authors:
    • Xiaojuan Sun
    • Weiguo Sui
    • Xiaobing Wang
    • Xianliang Hou
    • Minglin Ou
    • Yong Dai
    • Yueying Xiang
  • View Affiliations

  • Published online on: March 18, 2016     https://doi.org/10.3892/mmr.2016.5014
  • Pages: 3735-3746
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

There is increasing evidence that several genes are associated with an increased risk of type 2 diabetes (T2D); genome-wide association investigations and whole-genome re‑sequencing investigations offer a useful approach for the identification of genes involved in common human diseases. To further investigate which polymorphisms confer susceptibility to T2D, the present study screened for high‑contribution susceptibility gene variants Chinese patients with T2D using whole‑genome re‑sequencing with DNA pooling. In total, 100 Chinese individuals with T2D and 100 healthy Chinese individuals were analyzed using whole‑genome re‑sequencing using DNA pooling. To minimize the likelihood of systematic bias in sampling, paired‑end libraries with an insert size of 500 bp were prepared for in T2D in all samples, which were then subjected to whole‑genome sequencing. Each library contained four lanes. The average sequencing depth was 35.70. In the present study, 1.36 GB of clean sequence data were generated, and the resulting calculated T2D genome consensus sequence covered 99.88% of the hg19 sequence. A total of 3,974,307 single nucleotide polymorphisms were identified, of which 99.88% were in the dbSNP database. The present study also found 642,189 insertions and deletions, 5,590 structure variants (SVs), 4,713 copy number variants (CNVs) and 13,049 single nucleotide variants. A total of 1,884 somatic CNVs and 74 somatic SVs were significantly different between the cases and controls. Therefore, the present study provided validation of whole‑genome re‑sequencing using the DNA pooling approach. It also generated a whole-genome re-sequencing genotype database for future investigations of T2D.

Introduction

Type 2 diabetes (T2D) is a complex, multifactorial disorder characterized by chronic hyperglycemia due to the interplay of multiple genetic variants and several environmental factors. As a result of aging populations, and the increasing prevalence of obesity and physical inactivity, the number of patients with T2D has markedly increased worldwide (1). The disease is considered 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, insulin resistance syndromes, mitochondrial diabetes and neonatal diabetes (2). Examining T2D susceptibility genes may be useful for the prediction, prevention and early treatment of the disease.

Following previous genome-wide association studies (GWAS), the number of replicated common genetic variants associated with T2D has rapidly increased (39). In addition, >40 T2D-associated genetic loci have been identified, however, these loci have been revealed primarily on the basis of investigations of European individuals (10). The identified genomes only explain a small proportion of the estimated heritability of T2D, suggesting that additional genetic factors remain to be identified. One limitation of GWAS is the large number of hypotheses and the high economic cost of these investigations (11). Several studies have addressed the feasibility and effectiveness of pooling-based GWAS, with considerable savings in time and cost (1113). Additionally, whole-genome sequencing across multiple samples in a population provides an unprecedented opportunity for comprehensively characterizing the polymorphic variants in the population (14).

Although the genetic contribution to T2D is well recognized, there are now at least 19 loci containing genes, which are known to increase the risk of T2D, including PPARG, KCNJ11, KCNQ1, CDKAL1, CDKN2A-2B, CDC123-CAMK1D, MTNR1B, TCF7L2, TCF2 (HNF1B), HHEX-KIF11-IDE, JAZF1, IGF2BP2, SLC30A8, THADA, ADAMTS9, WFS1, FTO, NOTCH2 and TSPAN8 (2). To date, the current set of 66 established susceptibility loci, identified primarily through large-scale GWAS (2,8,1522), encompasses, at most, 10% of the familial aggregation of the disease. Of the currently established susceptibility loci, nine of the loci are contained in the 19 loci-containing genes. In the present study, the genomes of 100 Chinese patients with T2D and 100 non-diabetic Chinese individuals were examined using high throughput genome-wide re-sequencing and DNA pooling with Illumina HiSeq 2000 (Illumina, San Diego, CA, USA). The aim of the present study was to determine the rates of susceptibility genes in T2D in the Chinese population.

Materials and methods

Study populations

The present study was performed between August 2012 and the end of June 2013 at the 181st Hospital of People's Liberation Army, (Guilin, China). A total of 200 Chinese individuals were recruited, of which half were diagnosed with T2D. All participants with T2D were unrelated, and their disease was defined by World Health Organization criteria (23). The healthy individuals had a fasting plasma glucose <5.6 mmol/l, a 2-h oral glucose tolerance test-based plasma glucose <7.8 mmol/l, a body mass index <27.5 kg/m2 and blood pressure <140/90 mmHg, with no antihypertensive treatment. The clinical and biochemical characteristics of the 200 individuals are presented in Table I.

Table I

Clinical and biochemical characteristics of the 200 individuals recruited for re-sequencing.

Table I

Clinical and biochemical characteristics of the 200 individuals recruited for re-sequencing.

CharacteristicND (n=100)T2D (n=100)
Gender (males/females)63/3755/45
Age (years)60.1±11.85.4±0.6
HbA1c (%)5.4±0.69.6±2.3
Fasting plasma glucose (mmol/l)4.9±0.611.4±3.9
2-h OGTT-based plasma glucose (mmol/l)6.0±0.413.5±2.1
Body mass index (kg/m2)24.0±1.531.7±5.0
Waist circumference (cm)82.3±6.7105.0±9.8
Systolic blood pressure (mmHg)125.0±7.0152.0±9.0
Diastolic blood pressure (mmHg)84.0±5.092.0±6.0

[i] Data are presented as the mean ± standard deviation for normally distributed traits, or the median. ND, non-diabetic; T2D, type 2 diabetes; OGTT, oral glucose tolerance test.

Ethics

The present study was approved by the Medical and Health Research Ethics Committee of the 181st Hospital of the People's Liberation Army (Guilin, China). All participants provided informed consent for the use of their biological samples for genetic investigation.

Experimental procedure

Peripheral blood samples from the 200 volunteers were collected for genomic DNA extraction. DNA preparation followed the manufacturer's protocol (Illumina). Genomic DNA was extracted and then randomly fragmented. Following electrophoresis, DNA fragments of desired length (90 bp) were gel purified using QIAquick PCR Purification kit (Qiagen GmbH, Hilden, Germany). Adapter ligation and DNA cluster preparation were performed as part of Solexa sequencing by Beijing Genomics Institute (Shenzhen, China) (2426).

Bioinformatics analysis

The bioinformatics analysis used the sequencing data (raw data) generated from the Illumina HiSeq 2000. First, the adapter sequence in the raw data was removed, and low quality reads, which contained too many unknown bases (N) or low quality bases were discarded. This step produced 'clean data'. Secondly, Burrows-Wheeler Aligner (BWA) (27) was used to align the reads to the reference sequence. The alignment information was stored in BAM format files, which were further processed during subsequent steps, including fixing mate-pair information, adding read group information and marking duplicate reads caused by polymerase chain reaction (PCR). Following these processes, the final BAM files were ready for variant calling. Single nucleotide polymorphisms (SNPs) were detected using SOAPsnp (28), small insertion/deletions (InDels) were detected using SAMtools (29)/GATK (30,31), copy number variants (CNVs) were detected using CNVnator (32,33), single nucleotide variants (SNVs) were detected using Varscan (34) and somatic InDels were detected using GATK. Structure variants (SVs) and somatic CNVs were identified using BreakDancer (35)/CREST/SeekSV (self-method) and a self-method based on the SegSeq (36) algorithm, respectively. Virus integration sites were identified using a self-method based on unmapped reads. The procedure also included purity estimation. Subsequently, filters were applied to obtain variant results of higher confidence and, based on which subsequent advanced analysis could be performed, ANNOVAR (37) was used to annotate the variant results. Quality control was required at each stage of analysis to ensure clean data, alignment and variants. SIFT (38) was used to assess the likely phenotypic effect of identified missense mutations. PolyPhen-2 (39) analysis was performed to calculate the probability of an identified mutation as deleterious for disease pathogenesis.

Sequencing quality control

The raw reads, which contained the adapter sequence, a high content of unknown bases and low quality reads were removed prior to data analysis. The filtering steps were as follows: i) Removal of adapter reads. An adapter read was defined as a read that included the adapter bases, which were removed from the raw FASTQ data; ii) removal of low-quality reads. If more than half of the bases in a read were low-quality, defined as a base quality ≤5, the read was treated as a low-quality read and removed from the raw FASTQ data; iii) removal of reads in which unknown bases were >10%. The 'clean reads' were then used for downstream bioinformatics analysis. Finally statistical analysis was performed for data interpretation. The quality of the clean data is shown in Figs. 1 and 2.

Alignment quality control

The human genome build37 (hg19) was used as the reference genome in the present study. The whole-genome size of hg19 is 3,137,161,264 bp, whereas the effective size is 2,861,327,131 bp, following exclusion of the N bases, random regions, hap regions, and chromosome Un and chromosome M in the reference sequence. BWA was used to align the reference genome sequence for sequencing reads. Picard (broadinstitute.github.io/picard) was used to mark duplicated reads, which were redundant information produced by PCR.

Results

Quality control of sequencing data

To minimize the likelihood of systematic bias during sampling, two paired-end libraries with insert sizes of 500 bp were prepared for all samples and were subjected to whole-genome sequencing. Each library comprised four lanes, resulting in at least 30-fold haploid coverage for each sample. The raw image files were processed using the Illumina pipeline for base calling; default parameters and the sequences of each individual were generated as 90-bp-paired-end reads. A total of 144.3 GB raw sequence data were generated in a sequencing depth of ~30-fold. As shown in Table II, comparison was performed between the raw and clean data, which were detected using whole-genome re-sequencing.

Table II

Quality control of sequencing data.

Table II

Quality control of sequencing data.

CategoryData
Discarded reads (n)
RawClean
Reads (n)1,442,754,0241,367,776,414
Data size (bp) 129,847,862,160 123,099,877,260
N of fq1 (n)41,142,1581,257,883
N of fq2 (n)130,903,2223,072,470
GC of fq1 (%)39.62–39.8239.47–39.7
GC of fq2 (%)39.69–39.9739.56–39.78
Q20 of fq1 (%)96.16–97.0697.12–97.76
Q20 of fq2 (%)90.02–93.3393.88–95.97
Q30 of fq1 (%)90.13–92.3091.28–93.20
Q30 of fq2 (%)82.31–87.6186.04–90.21
Discarded reads associated with N4,639,892
Discarded reads due to low quality bases69,293,920
Discarded reads associated with the adapter1,043,798
Clean data/raw data (%)94.80

[i] N, unknown bases more than 10%; Q20, recognition reliability of a base is equal to 99.0%; Q30, recognition reliability of a base is equal to 99.99%; fq1/2, file 1/2 of pair-end sequencing data; GC, the combination of bases G and C.

Alignment of quality control data

The resulting calculated T2D genome consensus sequence covered 99.88% of the hg19 sequence. At a depth of 10-fold, the assembled consensus covered 97.98% of the reference genome using two paired-end reads. Thus, increased sequencing depth provided only a marginal increase in genome coverage. The alignment quality control results are shown in Table III. The distribution of per-base sequencing depth and cumulative depth distribution in the target regions are presented in Fig. 3. The data approximately followed a Poisson distribution, which showed that the exome-capturing target region was evenly sampled.

Table III

Alignment of quality control data.

Table III

Alignment of quality control data.

Whole-genome statisticValue
Clean reads (n)1,367,776,414
Clean bases (bp) 123,099,877,260
Mapped reads1,325,654,972
Mapped bases (bp) 117,572,810,280
Mapping rate (%)96.92
Unique reads (n)1,271,136,561
Unique bases (bp) 112,742,935,221
Unique rate (%)95.89
Duplicate reads (n)157,244,383
Duplicate rate (%)11.86
Mismatch bases (bp)481,007,764
Mismatch rate (%)0.41
Average sequencing depth35.70
Coverage (%)99.88
Coverage of at least 4X (%)99.38
Coverage of at least 10X (%)97.98
Coverage of at least 20X (%)93.02
SNP analysis

The genotype with the highest probability at a given locus was identified for each individual sequencing sample, and the consensus sequence of the sample was assembled and saved in CNS format. Using the consensus sequence, the polymorphic loci between the identified genotype and the reference were filtered and highlighted; this constitutes the high confident SNP dataset. Following the identification of the SNPs, ANNOVAR was used to perform annotation and classification.

The results revealed that 99.88% of the T2D SNPs were present in dbSNP, and there were 3,010 novel SNPs (Table IV). A total of 485 SNPs were screened, for which the SIFT score was <0.05, and the PolyPhen score was >0.85. These features suggested the pathological nature of the identified genetic variation. Of these 485 SNPs, 480 SNPs were found at exonic regions. The remaining SNPs were at exonic and splicing regions. All the SNPs were nonsynonymous genes. Compared with the 76 loci-containing genes causing an increased risk of T2D, 77 SNP loci were identified in 37 genes (Table V).

Table IV

Single nucleotide polymorphism data.

Table IV

Single nucleotide polymorphism data.

CategoryValue
Total (n)3,974,307
1000 genome and dbSNP135 (n)3,911,119
1000 genome-specific (n)1,712
dbSNP135-specific (n)58,466
dbSNP rate (%)99.88
Novel (n)3,010
Homozygous (n)475,874
Heterozygous (n)3,498,433
Synonymous (n)11,723
Missense (n)9,897
Stopgain (n)76
Stoploss (n)31
Exonic (n)21,422
Exonic and splicing (n)305
Splicing (n)155
ncRNA (n)97,213
UTR5 (n)4,043
UTR5 and UTR3 (n)14
UTR3 (n)25,860
Intronic (n)1,382,366
Upstream (n)18,977
Upstream and downstream (n)582
Downstream (n)22,165
Intergenic (n)2,401,205
Sorting intolerant from tolerant (n)1,201
Ti/Tv (n)2.1030
dbSNP Ti/Tv (n)2.1043
Novel Ti/Tv (n)1.1923

[i] UTR, untranslated region; ncRNA, non-coding RNA; dbSNP, SNP database; Ti/Tv, ratio of transition to transversion.

Table V

List of 77 single nucleotide polymorphism loci in 37 genes identified in the present study.

Table V

List of 77 single nucleotide polymorphism loci in 37 genes identified in the present study.

GeneFunctionExonic functiondbSNP135SIFTPolyPhen2ChrRefObsHet/hom
ANK1ExonicSynonymous SNVrs23048808GAHet
ExonicSynonymous SNVrs2304873CTHet
ExonicSynonymous SNVrs2304871GAHet
ANKRD55ExonicSynonymous SNVrs3217755TCHet
ExonicNonsynonymous SNVrs32177610CTHet
BCAR1ExonicSynonymous SNVrs316933016AGHom
exonicSynonymous SNVrs3743613CTHet
GRB14ExonicNonsynonymous SNVrs617482450.270.0092ATHet
CAMK1DExonicSynonymous SNVrs175705110CGHet
TSPAN8ExonicNonsynonymous SNVrs10513341012ACHet
ExonicSynonymous SNVrs2270587GAHet
ExonicNonsynonymous SNVrs37639780.080.981CGHet
ExonicNonsynonymous SNVrs794438920.730CTHet
THADAExonicNonsynonymous SNVrs170310560.342CTHet
ExonicSynonymous SNVrs11899823AGHet
ExonicSynonymous SNVrs13021894TCHet
ADAMTS9ExonicNonsynonymous SNVrs170709050.0573CTHet
ExonicNonsynonymous SNVrs67876330GCHet
BCL11AExonicSynonymous SNVrs75699462AGHom
KCNQ1ExonicSynonymous SNVrs105712811GAHet
HNF1AExonicSynonymous SNVrs116928912CGHet
ExonicNonsynonymous SNVrs11692880.090.052ACHet
ExonicSynonymous SNVrs2259820CTHet
ExonicNonsynonymous SNVrs24641960.060.053GAHet
ExonicNonsynonymous SNVrs11693050.40.423999AGHom
PRC1ExonicNonsynonymous SNVrs71727581015GTHom
ExonicSynonymous SNVrs2301826CTHet
MADDExonicSynonymous SNVrs32621411GAHet
ExonicSynonymous SNVrs326217TCHet
ExonicNonsynonymous SNVrs10510060.190GAHet
ExonicSynonymous SNVrs1017594TCHom
ADRA2AExonicSynonymous SNVrs180003810CAHet
GLIS3ExonicNonsynonymous SNVrs8060520.3809AGHom
SLC2A2ExonicSynonymous SNVrs53983GAHet
C2CD4BExonicNonsynonymous SNVrs80407120.34015ACHet
PTPRDExonicSynonymous SNVrs22797769CGHet
ExonicSynonymous SNVrs2281747AGHet
ExonicNonsynonymous SNVrs359294280.090.016GAHet
ExonicSynonymous SNVrs7026388TCHet
ExonicSynonymous SNVrs3763653GAHet
C2CD4BExonicNonsynonymous SNVrs80407120.34015ACHet
GRB14ExonicNonsynonymous SNVrs617482450.270.0092ATHet
GLIS3ExonicNonsynonymous SNVrs8060520.3809AGHom
PEPDExonicSynonymous SNVrs1756919GAHet
FITM2ExonicSynonymous SNVrs607340120TCHom
KCNK16ExonicNonsynonymous SNVrs117560910.0306GTHet
ExonicSynonymous SNVrs11753141GAHet
ExonicNonsynonymous SNVrs15355000.12GTHet
ExonicSynonymous SNVrs3734618AGHet
ExonicSynonymous SNVrs3734619CTHet
MAEAExonicSynonymous SNVrs11284270.134TCHet
PAX4ExonicNonsynonymous SNVrs712701107TGHet
GCC1ExonicSynonymous SNVrs37356447GAHet
ExonicSynonymous SNVrs3735642AGHet
KCNJ11ExonicNonsynonymous SNVrs52150.310.00211CTHet
ExonicSynonymous SNVrs5218GAHet
ExonicNonsynonymous SNVrs52190.360TCHet
KCNQ1ExonicSynonymous SNVrs105712811GAHet
CDKAL1ExonicSynonymous SNVrs93502696CTHet
ExonicSynonymous SNVrs9465994GAHet
HHEXExonicSynonymous SNVrs11312194210GAHet
SLC30A8ExonicNonsynonymous SNVrs132666340.0408CTHet
WFS1ExonicNonsynonymous SNVrs1801212104GAHom
ExonicSynonymous SNVrs1801206CTHom
ExonicSynonymous SNVrs1801214CTHom
ExonicNonsynonymous SNVrs7343120.020.99GAHet
ExonicSynonymous SNVrs1046314GAHom
TCF7L2ExonicNonsynonymous SNVrs779616540.150.99610CAHet
THADAExonicNonsynonymous SNVrs170310560.342CTHet
ExonicSynonymous SNVrs11899823AGHet
ExonicSynonymous SNVrs13021894TCHet
ADAMTS9ExonicNonsynonymous SNVrs170709050.0573CTHet
ExonicNonsynonymous SNVrs67876330GCHet
TSPAN8ExonicNonsynonymous SNVrs10513341012ACHet
ExonicSynonymous SNVrs2270587GAHet
ExonicNonsynonymous SNVrs37639780.080.981CGHet
ExonicNonsynonymous SNVrs794438920.730CTHet

[i] SNV, single nucleotide variant; Chr, chromosome.; dbSNP, SNP database; Ref, reference genotype; Het, heterozygous; Hom, homozygous; Obs, observed.

InDel analysis

To detect the InDels, the present study used pair-end reads for gap alignment using the mpileup program in SAM tools. Following identification of the InDels, ANNOVAR was used for annotation and classification. Of the 642,189 identified InDels, the percentage that overlapped the dbSNP InDels was 68.52% (Table VI). The length distribution of the InDels in the whole target region and CDS region were also plotted (Fig. 4). The length distribution of InDel in CDS region indicated that peaks are present at 3, 6 and 9 bp length, the InDels with this periodicity are non-framenshift InDels, they have relatively small effect on the genome comparing with frameshift InDels.

Table VI

Insertion/deletion data.

Table VI

Insertion/deletion data.

CategoryValue
Total642,189
1000 genome and dbSNP135314,143
1000 genome specific81,476
dbSNP135 specific125,867
dbSNP rate (%)68.52
Novel120,703
Homozygous103,137
Heterozygous539,052
Frameshift insertion120
Non-frameshift insertion88
Frameshift deletion99
Non-frameshift deletion110
Frameshift block substitution0
Non-frameshift block substitution0
Stopgain2
Stoploss1
Exonic415
Exonic and splicing5
Splicing77
ncRNA16,036
UTR5457
UTR5 and UTR33
UTR35,172
Intronic225,732
Upstream3,326
Upstream and downstream102
Downstream4,324
Intergenic386,540

[i] SNP, single nucleotide polymorphism; UTR, untranslated region; dbSNP, dbSNP database; ncRNA, non-coding RNA.

SV, CNV and SNV analyses

When aligning the paired-end reads, if a structure variation existed between the sequencing and the reference sequences, the requirements for pair-end alignment, also termed the PE map, may not be met; thus, these anomalous read pairs and soft clip reads were used in the present study to detect SVs. The resulting list of the SVs, that were detected at the whole-genome level are listed in Table VII.

Table VII

Structure variant data.

Table VII

Structure variant data.

CategoryValue
Total5,590
Insertion348
Deletion5,002
Inversion14
ITX122
CTX104
Exonic3
Exonic and splicing3
Splicing7
ncRNA133
UTR53
UTR5 and UTR30
UTR39
Intronic1,875
Upstream15
Upstream and downstream0
Downstream31
Intergenic3,511

[i] UTR, untranslated region; ncRNA, non-coding RNA; ITX, inversion; CTX, translocation.

The CNVs of each sample were detected using CNVnator. Following identification of the CNVs, ANNOVAR was used for annotation and classification (Table VIII). Varscan was used to identify specific SNVs by simultaneously comparing read counts, base quality and allele frequency between the healthy individuals and patients with type 2 diabetes. Following identification of the SNVs, ANNOVAR was again used for annotation and classification (Table IX and Figs. 5 and 6).

Table VIII

Copy number variant data.

Table VIII

Copy number variant data.

CategoryValue
Total4,713
Exonic930
Exonic and splicing0
Splicing242
ncRNA165
UTR51
UTR5 and UTR30
UTR39
Intronic1,026
Upstream56
Upstream and downstream6
Downstream36
Intergenic2,242
Amplification size13,445,200
Deletion size84,646,400

[i] UTR, untranslated region, ncRNA, non-coding RNA.

Table IX

Single nucleotide variant statistics (healthy control, vs. T2D).

Table IX

Single nucleotide variant statistics (healthy control, vs. T2D).

CategoryValue
Total13,049
1000 genome and dbSNP13512,655
1000 genome specific11
dbSNP135 specific282
dbSNP rate (%)99.14
Novel101
Hom0
Het13,049
Synonymous52
Missense36
Stopgain0
Stoploss0
Exonic88
Exonic and splicing0
Splicing1
ncRNA305
UTR515
UTR5 and UTR30
UTR3112
Intronic4,638
Upstream73
Upstream and downstream0
Downstream74
Intergenic7,743
Sorting Intolerant from Tolerant7
Ti/Tv2.1188
dbSNP Ti/Tv2.1324
Novel Ti/Tv1.0612

[i] UTR, untranslated region; ncRNA, non-coding RNA; dbSNP, SNP database.

Analyses of somatic InDels, somatic CNVs and somatic SVs

In the sufficiently covered sites, the initial call was produced in the type 2 diabetes sample and then compared with the normal sample to detect evidence for the event. If there was no evidence to support the InDel event in the normal sample, the site was considered to be a putative somatic InDel. In total, there were 1,249 somatic InDels in 1,000 genome and dbSNP135, 688 in 1,000 genome specific, and 310 dbSNP135 specific. The results of the statistical analyses are provided in Table X. The dbSNP rate was 42.19%, and without heterozygous.

Table X

Somatic insertion and deletion statistics (healthy control, vs. T2D).

Table X

Somatic insertion and deletion statistics (healthy control, vs. T2D).

CategoryValue
1000 genome and dbSNP1351,249
1000 genome specific688
dbSNP135 specific310
dbSNP rate (%)42.19
Novel1,448
Hom3,695
Het0
Frameshift insertion0
Non-frameshift insertion0
Frameshift deletion1
Non-frameshift deletion3
Frameshift block substitution0
Non-frameshift block substitution0
Stopgain0
Stoploss0
Exonic4
Exonic and splicing0
Splicing1
ncRNA93
UTR54
UTR5 and UTR30
UTR332
Intronic1,242
Upstream16
Upstream and downstream0
Downstream27
Intergenic2,276

[i] UTR, untranslated region; ncRNA, non-coding RNA; dbSNP, SNP database.

Somatic CNVs correspond to relatively large regions of the genome, which are either deleted and fewer than the normal number, or duplicated and more than the normal number, on certain chromosomes. The results of the somatic CNVs analyses are shown in Table XI, and the somatic CNV overview is plotted in Fig. 7.

Table XI

Somatic copy number variant analysis (healthy control, vs, T2D).

Table XI

Somatic copy number variant analysis (healthy control, vs, T2D).

CategoryValue
Total1,884
Exonic185
Exonic and splicing0
Splicing21
ncRNA41
UTR50
UTR5 and UTR30
UTR36
Intronic538
Upstream14
Upstream and downstream0
Downstream17
Intergenic1,062
Amplification size1,372,716
Deletion size1,879,767

[i] UTR, untranslated region; ncRNA, non-coding RNA.

In the sufficiently covered sites, the initial call was produced in the type 2 diabetes sample and then compared with the normal sample to detect evidence for the event. If there was no evidence to support the SV event in the normal sample, this event was considered to be a putative somatic SV. The results of the somatic SV statistical analyses are presented in Table XII.

Table XII

Somatic structure variant statistics (healthy control, vs. T2D).

Table XII

Somatic structure variant statistics (healthy control, vs. T2D).

CategoryValue
Total74
Insertion6
Deletion58
Inversion0
ITX2
CTX8
Exonic0
Exonic and splicing0
Splicing0
ncRNA0
UTR50
UTR5 and UTR30
UTR30
Intronic24
Upstream0
Upstream and downstream0
Downstream0
Intergenic50

[i] UTR, untranslated region, ncRNA, non-coding RNA.

Discussion

In the present study, whole-genome re-sequencing was performed with DNA pooling to investigate T2D in Chinese individuals. In total, 1.44 GB of raw data were generated in a short period of time. Among the data obtained, 3,010 novel SNPs and 120,703 novel InDels were found. In addition, 5,590 SVs, 4,713 CNVs and 13,049 SNVs were identified. There was a significant difference between cases and controls in 1,884 somatic CNVs and 74 somatic SVs. These findings improve current understanding of the genetic basis of T2D and offer insight for future investigations.

Among the identified genes, only rs734312 in WFS1 (with a SIFT score 0.02 and a PolyPhen score of 0.99) suggested a pathologic nature. It was also found that, even in the same genes, the associated loci were different in the present study. Although >30 genetic susceptibility loci have been found in the comparison of 76 genes, the most frequently reported variants have small to moderate effects, and account for only a small proportion of the heritability of T2D, suggesting that the majority of inter-person genetic variation in this disease remains to be elucidated (20).

KCNQ1 (40), UBE2E2 and C2CD4A-C2CD4B (19) have been identified as T2D susceptibility genes in three GWA scans in Japanese individuals. The combined analyses identified GLIS3, PEPD, FITM2-R3HDML-HNF4A, KCNK16, MAEA, GCC1-PAX4, PSMD6 and ZFAND3 as T2D loci reaching genome-wide significance in East Asia (22). PTPRD and SRR have been identified as diabetes susceptibility loci in a study of a Han Chinese population (2). In the present study, the SNP loci in the UBE2E2, PSMD6, ZFAND3 and SRR genes were not found. The results of the present study suggested that, in different patient populations, different genes may confer risks for diabetes, which may lead to more complex study designs for investigating the molecular pathogenesis of T2D.

A simple, but important observation was that DNA pooling provides a highly effective approach for examining the genetic underpinnings of common familial diseases. DNA pooling has been confirmed to be an effective and efficient method to select candidate susceptibility loci for follow-up by individual genotyping (12,13,41). This indicates that the use of GWAS for a large number of cases and controls are technically and financially feasible. Additional findings of particular interest include the large-scale examination of possible genetic variants. The present study demonstrated novel, significant associations, including SNPs, CNVs, InDels and SNVs.

The present study indicated general recommendations, which are relevant to whole-genome re-sequencing using DNA pooling. The first recommendation is associated with the importance of careful quality control. In the present study, 144.3 GB of raw data were generated from the Illumina pipeline, which contained too many Ns or low quality bases. Small systematic differences can readily produce effects capable of obscuring true associations from being identified (42,43). The present study implemented extensive quality control checks to minimize differences in the clean data, alignment and called variants.

The sequencing method used in the present study also resulted in sequence redundancy reaching an average of 35.70-fold. Thus, the consensus sequence accuracy was higher and particularly suitable for calling heterozygous alleles. Whole-genome re-sequencing with DNA pooling technologies is high throughput technique, as one hundred million DNA fragments can be sequenced in parallel on one chip. The Illumina HiSeq 2000 platform from Illumina used in the present study can provide up to 55 GB of high-quality data per day. In this regard, it was possible to undertake comprehensive assessments of the variants within the regions of interest using this high-throughput and time efficient method.

Thirty years ago, James V. Neel (44) labeled T2D as 'the geneticist's nightmare', describing the identification of genetic factors in T2D as challenging. Numerous investigations on candidate genes for T2D have been published; however, the various approaches, including high-throughput gene scanning and gene and pedigree analysis have not been entirely successful in identifying robustly replicating T2D-susceptibility loci. Ultimately, with large samples and worldwide collaboration, novel risk factors for diabetes are likely be identified using whole-genome re-sequencing technology.

Acknowledgments

The authors would like to thank Professor Yong Dai (Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China) and Professor Yueying Xiang (Department of Health Management Center, 181st Hospital, Guilin, Guangxi, China) for their helpful comments, and Dr Minglin Ou and Mr Xianliang Hou from the Nephrology Department of 181st Hospital and Guangxi Key Laboratory of Metabolic Diseases Research (Guilin, China) for their technical assistance.

References

1 

Chen L, Magliano DJ and Zimmet PZ: The worldwide epidemiology of type 2 diabetes mellitus-present and future perspectives. Nat Rev Endocrinol. 8:228–236. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Tsai FJ, Yang CF, Chen CC, Chuang LM, Lu CH, Chang CT, Wang TY, Chen RH, Shiu CF, Liu YM, et al: A genome-wide association study identifies susceptibility variants for type 2 diabetes in Han Chinese. PLoS Genet. 6:e10008472010. View Article : Google Scholar : PubMed/NCBI

3 

Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, et al: A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science. 316:1341–1345. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Diabetes Genetics Initiative of Broad Institute of Harvard and MIT; Lund University; Novartis Institutes of BioMedical Research; Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ, Kathiresan S, Hirschhorn JN, et al: Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science. 316:1331–1336. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, Boutin P, Vincent D, Belisle A, Hadjadj S, et al: A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature. 445:881–885. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Zeggini E, Weedon MN, Lindgren CM, Frayling TM, Elliott KS, Lango H, Timpson NJ, Perry JR, Rayner NW, Freathy RM, et al: Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes. Science. 316:1336–1341. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Wellcome Trust Case Control Consortium: Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 447:661–678. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Zeggini E, Scott LJ, Saxena R, Voight BF, Marchini JL, Hu T, de Bakker PI, Abecasis GR, Almgren P, Andersen G, et al: Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet. 40:638–645. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Gudmundsson J, Sulem P, Steinthorsdottir V, Bergthorsson JT, Thorleifsson G, Manolescu A, Rafnar T, Gudbjartsson D, Agnarsson BA, Baker A, et al: Two variants on chromosome 17 confer prostate cancer risk and the one in TCF2 protects against type 2 diabetes. Nat Genet. 39:977–983. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Saxena R, Elbers CC, Guo Y, Peter I, Gaunt TR, Mega JL, Lanktree MB, Tare A, Castillo BA, Li YR, et al: Large-scale gene-centric meta-analysis across 39 studies identifies type 2 diabetes loci. Am J Hum Genet. 90:410–425. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Baum AE, Akula N, Cabanero M, Cardona I, Corona W, Klemens B, Schulze TG, Cichon S, Rietschel M, Nöthen MM, et al: A genome-wide association study implicates diacylglycerol kinase eta (DGKH) and several other genes in the etiology of bipolar disorder. Mol Psychiatry. 13:197–207. 2008. View Article : Google Scholar :

12 

Galvan A, Falvella FS, Frullanti E, Spinola M, Incarbone M, Nosotti M, Santambrogio L, Conti B, Pastorino U, Gonzalez-Neira A and Dragani TA: Genome-wide association study in discordant sibships identifies multiple inherited susceptibility alleles linked to lung cancer. Carcinogenesis. 31:462–465. 2010. View Article : Google Scholar

13 

Forstbauer LM, Brockschmidt FF, Moskvina V, Herold C, Redler S, Herzog A, Hillmer AM, Meesters C, Heilmann S, Albert F, et al: Genome-wide pooling approach identifies SPATA5 as a new susceptibility locus for alopecia areata. Eur J Hum Genet. 20:326–332. 2012. View Article : Google Scholar :

14 

Wong LP, Ong RT, Poh WT, Liu X, Chen P, Li R, Lam KK, Pillai NE, Sim KS, Xu H, et al: Deep whole-genome sequencing of 100 southeast asian malays. Am J Hum Genet. 92:52–66. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Kong A, Steinthorsdottir V, Masson G, Thorleifsson G, Sulem P, Besenbacher S, Jonasdottir A, Sigurdsson A, Kristinsson KT, Jonasdottir A, et al: Parental origin of sequence variants associated with complex diseases. Nature. 462:868–874. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Voight BF, Scott LJ, Steinthorsdottir V, Morris AP, Dina C, Welch RP, Zeggini E, Huth C, Aulchenko YS, Thorleifsson G, et al: Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat Genet. 42:579–589. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Dupuis J, Langenberg C, Prokopenko I, Saxena R, Soranzo N, Jackson AU, Wheeler E, Glazer NL, Bouatia-Naji N, Gloyn AL, et al: New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet. 42:105–116. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Qi L, Cornelis MC, Kraft P, Stanya KJ, Linda Kao WH, Pankow JS, Dupuis J, Florez JC, Fox CS, Paré G, et al: Genetic variants at 2q24 are associated with susceptibility to type 2 diabetes. Hum Mol Genet. 19:2706–2715. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Yamauchi T, Hara K, Maeda S, Yasuda K, Takahashi A, Horikoshi M, Nakamura M, Fujita H, Grarup N, Cauchi S, et al: A genome-wide association study in the Japanese population identifies susceptibility loci for type 2 diabetes at UBE2E2 and C2CD4A-C2CD4B. Nat Genet. 42:864–868. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Shu XO, Long J, Cai Q, Qi L, Xiang YB, Cho YS, Tai ES, Li X, Lin X, Chow WH, et al: Identification of new genetic risk variants for type 2 diabetes. PLoS Genet. 6:e10011272010. View Article : Google Scholar : PubMed/NCBI

21 

Kooner JS, Saleheen D, Sim X, Sehmi J, Zhang W, Frossard P, Been LF, Chia KS, Dimas AS, Hassanali N, et al: Genome-wide association study in individuals of South Asian ancestry identifies six new type 2 diabetes susceptibility loci. Nat Genet. 43:984–989. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Cho YS, Chen CH, Hu C, Long J, Ong RT, Sim X, Takeuchi F, Wu Y, Go MJ, Yamauchi T, et al: Meta-analysis of genome-wide association studies identifies eight new loci for type 2 diabetes in east Asians. Nat Genet. 44:67–72. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Alberti KG and Zimmet PZ: Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet Med. 15:539–553. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Mardis ER: Next-generation DNA sequencing methods. Annu Rev Genomics Hum Genet. 9:387–402. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Wang J, Wang W, Li R, Li Y, Tian G, Goodman L, Fan W, Zhang J, Li J, Zhang J, et al: The diploid genome sequence of an Asian individual. Nature. 456:60–65. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Wheeler DA, Srinivasan M, Egholm M, Shen Y, Chen L, McGuire A, He W, Chen YJ, Makhijani V, Roth GT, et al: The complete genome of an individual by massively parallel DNA sequencing. Nature. 452:872–876. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Li H and Durbin R: Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 25:1754–1760. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Li R, Li Y, Fang X, Yang H and Wang J, Kristiansen K and Wang J: SNP detection for massively parallel whole-genome resequencing. Genome Res. 19:1124–1132. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G and Durbin R: 1000 Genome project data processing subgroup: The sequence alignment/Map format and SAMtools. Bioinformatics. 25:2078–2079. 2009. View Article : Google Scholar : PubMed/NCBI

30 

McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, Garimella K, Altshuler D, Gabriel S, Daly M and DePristo MA: The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20:1297–1303. 2010. View Article : Google Scholar : PubMed/NCBI

31 

DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, Philippakis AA, del Angel G, Rivas MA and Hanna M: A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet. 43:491–498. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Abyzov A, Urban AE, Snyder M and Gerstein M: CNVnator: An approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res. 21:974–984. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Mills RE, Walter K, Stewart C, Handsaker RE, Chen K, Alkan C, Abyzov A, Yoon SC, Ye K, Cheetham RK, et al: Mapping copy number variation by population-scale genome sequencing. Nature. 470:59–65. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Koboldt DC, Chen K, Wylie T, Larson DE, McLellan MD, Mardis ER, Weinstock GM, Wilson RK and Ding L: VarScan: Variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics. 25:2283–2285. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Chen K, Wallis JW, McLellan MD, Larson DE, Kalicki JM, Pohl CS, McGrath SD, Wendl MC, Zhang Q, Locke DP, et al: BreakDancer: An algorithm for high-resolution mapping of genomic structural variation. Nat Methods. 6:677–681. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Chiang DY, Getz G, Jaffe DB, O'Kelly MJ, Zhao X, Carter SL, Russ C, Nusbaum C, Meyerson M and Lander ES: High-resolution mapping of copy-number alterations with massively parallel sequencing. Nat Methods. 6:99–103. 2009. View Article : Google Scholar :

37 

Wang K, Li M and Hakonarson H: ANNOVAR: Functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38:e1642010. View Article : Google Scholar : PubMed/NCBI

38 

Kumar P, Henikoff S and Ng PC: Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 4:1073–1081. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS and Sunyaev SR: A method and server for predicting damaging missense mutations. Nat Methods. 7:248–249. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Yasuda K, Miyake K, Horikawa Y, Hara K, Osawa H, Furuta H, Hirota Y, Mori H, Jonsson A, Sato Y, et al: Variants in KCNQ1 are associated with susceptibility to type 2 diabetes mellitus. Nat Genet. 40:1092–1097. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Vélez JI, Chandrasekharappa SC, Henao E, Martinez AF, Harper U, Jones M, Solomon BD, Lopez L, Garcia G, Aguirre-Acevedo DC, et al: Pooling/bootstrap-based GWAS (pbGWAS) identifies new loci modifying the age of onset in PSEN1 p.Glu280Ala Alzheimer's disease. Mol Psychiatry. 18:568–575. 2013. View Article : Google Scholar :

42 

Clayton DG, Walker NM, Smyth DJ, Pask R, Cooper JD, Maier LM, Smink LJ, Lam AC, Ovington NR, Stevens HE, et al: Population structure, differential bias and genomic control in a large-scale, case-control association study. Nat Genet. 37:1243–1246. 2005. View Article : Google Scholar : PubMed/NCBI

43 

Zondervan KT and Cardon LR: The complex interplay among factors that influence allelic association. Nat Rev Genet. 5:89–100. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Neel JV: The Genetics of Diabetes Mellitus. Creutzfeldt W, Köbberling J and Neel JV: Springer; Berlin: 1976, pp. 1–11. View Article : Google Scholar

Related Articles

Journal Cover

May-2016
Volume 13 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun X, Sui W, Wang X, Hou X, Ou M, Dai Y and Xiang Y: Whole-genome re-sequencing for the identification of high contribution susceptibility gene variants in patients with type 2 diabetes. Mol Med Rep 13: 3735-3746, 2016
APA
Sun, X., Sui, W., Wang, X., Hou, X., Ou, M., Dai, Y., & Xiang, Y. (2016). Whole-genome re-sequencing for the identification of high contribution susceptibility gene variants in patients with type 2 diabetes. Molecular Medicine Reports, 13, 3735-3746. https://doi.org/10.3892/mmr.2016.5014
MLA
Sun, X., Sui, W., Wang, X., Hou, X., Ou, M., Dai, Y., Xiang, Y."Whole-genome re-sequencing for the identification of high contribution susceptibility gene variants in patients with type 2 diabetes". Molecular Medicine Reports 13.5 (2016): 3735-3746.
Chicago
Sun, X., Sui, W., Wang, X., Hou, X., Ou, M., Dai, Y., Xiang, Y."Whole-genome re-sequencing for the identification of high contribution susceptibility gene variants in patients with type 2 diabetes". Molecular Medicine Reports 13, no. 5 (2016): 3735-3746. https://doi.org/10.3892/mmr.2016.5014