Open Access

Identification of recurrent and novel mutations by whole‑genome sequencing of colorectal tumors from the Han population in Shanghai, eastern China

  • Authors:
    • Hongfei Teng
    • Renyuan Gao
    • Nan Qin
    • Xun Jiang
    • Min Ren
    • Yu Wang
    • Shouxin Wu
    • Ning Li
    • Jiangman Zhao
    • Huanlong Qin
  • View Affiliations

  • Published online on: October 17, 2018     https://doi.org/10.3892/mmr.2018.9563
  • Pages: 5361-5370
  • Copyright: © Teng 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

Previous studies have identified recurrent oncogenic mutations in colorectal cancer (CRC), but there is limited CRC genomic data from the Chinese Han population. Whole‑genome sequencing was performed on 10 primary CRC tumors and matched adjacent normal tissues from patients from the Han population in Shanghai, at an average of 27.8x and 27.9x coverage, respectively. In the 10 tumor samples, 32 significant somatic mutated genes were identified, 13 of which were also reported as CRC mutations in The Cancer Genome Atlas Network. All the mutated genes were enriched in functions associated with channel activity, which has rarely been reported in previous studies investigating CRC. Furthermore, 21 chromosomal rearrangements were detected and 4 rearrangements encoded predicted in‑frame fusion proteins, including a fusion of phosphorylase kinase regulatory subunit b and NOTCH2 demonstrated in 2 out of 10 tumors. Chromosome 8 was amplified in 1 tumor and chromosome 20 was amplified in 2 out of 10 CRC patients. The present study produced a genomic mutation profile of CRC, which provides a valuable resource for further insight into the mutations that characterize CRC in patients from the Han population in Shanghai, eastern China.

Introduction

Colorectal cancer (CRC) is the third most common malignancy and one of the leading causes of cancer-associated mortality worldwide (1). For tumor molecular profiling of CRC, several organizations have completed large-scale sequencing projects, including The Cancer Genome Atlas (TCGA) (2), Dana-Farber Cancer Institute (DFCI) (3), Memorial Sloan Kettering Cancer Center (4) and Genentech, Inc. (5). Exome-wide sequencing has identified recurrent gene mutations and dysregulated signaling pathways that may contribute to carcinogenesis, including APC, WNT signaling pathway regulator (APC), tumor protein p53 (TP53), KRAS proto-oncogene, GTPase (KRAS) and titin (TTN) genes, and the WNT, tumor growth factor-β, phosphoinositide 3-kinase and P53 signaling pathways (2). TCGA has detailed subgroups of tumors characterized by hypermutation (16%), or by a high degree of microsatellite instability (MSI-H) and non-hypermutated (84%). Together with clinical annotations, these molecular profiling methods can be used to identify potentially actionable tumor biomarkers that may be useful for clinical practice.

Over the past decades, the 5-year relative survival rate of patients with CRC has increased markedly (6). In China, ~376,000 people are diagnosed with CRC per year, which is 2.5 times higher than in the United States (7,8). Additionally, 5-year relative survival is substantially lower in China (~47.2%) compared with in the United States (~66%) (9). In China, survival of patients with CRC in urban areas, including Shanghai, is markedly higher than in rural areas, due to differences in socioeconomics and healthcare standards (9). Next-generation sequencing enables precision medicine, the tailoring of treatments based on unique genomic variations of each patient's tumor. Sequencing a panel of CRC-associated genes may identify actionable genomic driver mutations and further determine mutational burden in CRC, which is more cost effective, efficient and achieves higher sequencing depth than whole-exome sequencing. CRC panel design is mainly based on the TCGA database. Sequencing data generated from TCGA (2) or previous studies (35) are essential sources, but tumors from Asian populations have not been the subject of comprehensive evaluation. Furthermore, in the previous studies (1013) almost all exome-wide sequencing in CRC, including whole-exome sequencing and target sequencing, were limited to the detection of single nucleotide variations (SNVs) and small insertion and deletions (InDels) in genes. While structure variations, including copy number variation (CNV) and chromosomal rearrangement, are also key factors in the process of cancer development.

In the present study, 10 CRC patients were recruited at the Shanghai Tenth People's Hospital (Shanghai, China) as a representative sample of CRC patients in the Shanghai Han population and Chinese urban population. The whole genomes of the 10 CRC patient tumors and matched normal tissues were sequenced. A comprehensive analysis was performed, including identification of SNVs, InDels, CNVs and chromosomal rearrangement, which not only validated results from TCGA to a certain extent, but also resulted in novel findings.

Patients and methods

Patients and samples

Fresh primary colorectal tumor tissues and matched adjacent normal tissues were collected from 10 patients with pathologically confirmed CRC at the Shanghai Tenth People's Hospital Affiliated to Tongji University between March and May 2015. Patient clinical characteristics are presented in Table I. Patients were numbered CRC-1 to 10. The median age was 62 years (range, 43–82 years), 6 cases were female, 8 cases exhibited colon cancer and 2 were rectal cancer. No patients had received therapeutic procedures, including chemotherapy or radiotherapy. Samples were frozen immediately in liquid nitrogen and stored at −80°C prior to analysis.

Table I.

Clinical characteristics of ten colorectal cancer patients.

Table I.

Clinical characteristics of ten colorectal cancer patients.

PatientsSexAgeClinical diagnosisMSITNM stage DifferentiationLocationSize (cm)Lymphatic metastasisDistant metastasis
CRC-1Female62Colon cancerMSST3N0M0ModeratelySigmoid colon6×4×10/12No
CRC-2Female68Colon cancerMSST3N1M0ModeratelyRight colon7×4.5×41/9No
CRC-3Female58Rectal cancerMSST4N1M0ModeratelyRectum4.5×3×31/5No
CRC-4Male80Colon cancerMSST3N1M0 Poorly-moderatelyAscending colon4×3×1.53/13No
CRC-5Male57Colon cancerMSST1N0M0 Moderately-wellColon1×1.5×10/5No
CRC-6Female75Colon cancerMSST4aN1M1ModeratelyAscending colon9.5×7×2.51/14Yes
CRC-7Male82Rectal cancerMSI-LT4bN0M0ModeratelyRectum6×5.5×1.50/15No
CRC-8Male62Colon cancerMSST3N0M0ModeratelyTransverse colon4×3.5×20/12No
CRC-9Female60Colon cancerMSI-HT4bN0M0 Poorly-moderatelyTransverse colon6×4.5×40/17No
CRC-10Female43Colon cancerMSST1N1M0 Moderately-wellColon5.5×5×41/15No

[i] MSI, microsatellite instability; MSS, microsatellite stability; MSI-L, microsatellite instability-low; MSI-H, microsatellite instability-high.

DNA extraction

DNA was extracted from fresh frozen tissue using QIAamp DNA Minikit (Qiagen GmbH, Hilden, Germany) according to the manufacturers' protocol. DNA was quantified using the Qubit Fluorometer (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Library preparation

A total of 0.5 µg DNA per sample was used as input material for the DNA library preparations. A sequencing library was generated using Truseq Nano DNA HT Sample Prep kit (Illumina, Inc., San Diego, CA, USA) following the manufacturer's recommendations and index codes were added to each sample. Briefly, genomic DNA samples were fragmented by sonication to a size of ~350 bp (duty factor 10%, peak incident power 175, cycles per burst 200, treatment time 180 seconds, bath temperature 4–8°C). Then, DNA fragments were end polished, A-tailed and ligated with the full-length adapter for Illumina sequencing, followed by further polymerase chain reaction (PCR) amplification using KAPA HiFi HotStart ReadyMix (Kapa Biosystems, Inc., Wilmington, MA, USA). Primers are based on the P5 and P7 Illumina flow cell sequences, and are suitable for the amplification of libraries prepared with full-length adapters (P5: 5′-AATGATACGGCGACCACCGAGATC-3′, P7: 5′-CAAGCAGAAGACGGCATACGA-3′). Thermocycling conditions: Initial denaturation 98°C for 45 sec, denaturation 98°C for 15 sec, annealing 60°C for 30 sec, extension 72°C for 30 sec, library amplification with 3 cycles and final extension 72°C for 1 min, hold at 4°C. Subsequently, PCR products were purified by the AMPure XP system (Beckman Coulter, Inc., Brea, CA, USA), libraries were analyzed for size distribution by Agilent2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA) and quantified by quantitative PCR (3 nM) using KAPA Library Quantification kits (Kapa Biosystems, Inc.) according to the manufacturer's protocol. The primers were the same as for the amplification procedure (P5: 5′-AATGATACGGCGACCACCGAGATC-3′, P7: 5′-CAAGCAGAAGACGGCATACGA-3′). qPCR protocol for library quantification consists of an initial denaturation step at 95°C for 5 min followed by 35 cycles of denaturation at 95°C for 30 sec and combined annealing/extension at 60°C for 45 sec. A total of 6 pre-diluted DNA Standards and appropriately diluted NGS libraries are amplified at the same time. The average Cq value for each DNA standard was plotted against its known concentration to generate a standard curve. The standard curve is used to convert the average Cq values for diluted libraries to concentration, from which the working concentration of each library is calculated.

Clustering and sequencing

The clustering of the index-coded samples was performed on a cBot Cluster Generation System using HiseqX PE Cluster kit V2.5 (Illumina, Inc.) according to the manufacturer's protocol. Following cluster generation, the DNA libraries were sequenced using the IlluminaHiseq platform and 150 bp paired-end reads were generated.

Bioinformatics analysis

For whole-genome sequencing, clean data was obtained following filtering adapter, low quality reads and reads with proportion of N>10%. Reads were aligned to the reference human genome (UCSC hg19; http://genome.ucsc.edu/) (14) using the Burrows-Wheeler Aligner v. 0.7.12 (15). Next, the Picard and Genome Analysis Toolkit (GATK v.3.2) (16) method was adopted for duplicate removal, local realignment and Base Quality Score Recalibration, and generated quality statistics, including mapped reads, mean mapping quality and mean coverage. Finally, the GATK HaplotypeCaller was used for SNV and InDel identification.

Variants were annotated using the ANNOVAR software tool (17). Annotations for mutation function (including frameshift insertion/deletion, non-frameshift insertion/deletion, synonymous SNV, nonsynonymous SNV and stopgain stoploss), mutation location [including exonic, intronic, splicing, upstream, downstream, 3′untranslated region (UTR) and 5′UTR], amino acid changes, 1000 Genomes Project data and dbSNP reference number were performed.

Somatic SNVs and InDels of tumors compared with matched normal tissue were named and functionally annotated using MuTect v. 1.1.4 (16) and Varscan2 v. 2.3.9 (18) software. Somatic mutations in coding regions were filtered (Frame_Shift_Del, Frame_Shift_Ins, In_Frame_Del, In_Frame_Ins, Missense_Mutation, Nonsense_Mutation and Nonstop_Mutation) along with challenging regions (3′Flank, 3′UTR, 5′Flank, 5′UTR, intergenic region, Splice_Site, Translation_Start_Site, RNA, Splice_Site, Translation_Start_Site). The mutations with variant allele frequency >5% were defined as high confidence mutations. MutSigCV v.0.9 (19) was used to identify significantly mutated genes (q≤0.1). Then, gene mutation data were downloaded from TCGA database (https://tcga-data.nci.nih.gov/docs/publications/coadread_2012/) (2) and comparative analysis was performed using the sequencing data produced in the present study.

Control-FREEC v.8.7 (20) software was used for identifying and annotating CNVs, including gain, loss or copy neutral loss of heterozygosity. Structural variations, including inversion, intra-chromosomal translocation and inter-chromosomal translocation, were identified using Factera (21) software.

The mutation landscape across a cohort, including SNVs, InDels and mutational burden, were created by Genomic Visualizations in R (GenVisR) (22). The custom mutation lists of proteins were visualized by MutationMapper tool from cBioPortal (http://www.cbioportal.org/mutation_mapper.jsp) and structural variants, and copy number data were visualized using CIRCOS version 0.69 (http://www.circos.ca/) (23). Gene ontology (GO: http://www.geneontology.org/) and Kyoto Encyclopedia of Genes and Genomes (KEGG: http://www.kegg.jp/) enrichment analysis was performed to investigate the biological importance of the somatic mutated genes using the ClusterProfiler in R software (10.18129/B9.bioc.clusterProfiler).

Results

Clinical and sequencing data

The 10 CRC samples were analyzed (Table I). The microsatellite instability status of the CRC-9 tumor was high (MSI-H), the CRC-7 tumor was low (MSI-L) and others were microsatellite stable.

Whole-genome sequencing achieved an average of 27.8× coverage of the tumor genomes and 27.9× coverage of the germline genome (Table II). Somatic DNA alterations were analyzed, including SNVs, InDels, CNVs and chromosomal rearrangements. An overall mutation rate of ~7.78 per Mb with a range of 2.11–29.79 mutations per Mb was calculated (Table II; Fig. 1A). In study of TCGA (2), cases with mutation rates >12 per Mb were designated as hypermutated. The mutation rate of CRC-9 was 29.79 per Mb in the present study, which was the only hypermutated case (Fig. 1A).

Table II.

Summary of whole-genome sequencing results from each patients' tissues.

Table II.

Summary of whole-genome sequencing results from each patients' tissues.

PatientsTumor coverageNormal coverageMutationsMutations per Mb
CRC-129.929.410923.64
CRC-227.929.06342.11
CRC-326.928.27902.63
CRC-425.727.021807.27
CRC-526.628.87162.39
CRC-626.727.220216.74
CRC-729.025.7359711.99
CRC-829.929.112884.29
CRC-927.827.6893729.79
CRC-1028.126.620716.90
Average27.827.923327.78
Significantly mutated somatic genes

The MutSigCV tool was used to define significantly mutated genes and identified 32 significantly mutated genes (Fig. 1B). Fig. 1 presents the significantly mutated genes (Fig. 1B), mutation type (Fig. 1B), frequency and tumor mutation burden (Fig. 1A). The five most frequently mutated genes were TP53 (4/10), transmembrane protein 128 (TMEM128; 4/10), KRAS (4/10), FAM47C (4/10) and BAGE family member 2 (BAGE2; 4/10).

The mutation frequency of the 32 significantly mutated genes was compared with TCGA data (Fig. 1C). Of these 32 genes, 13 were detected by TCGA, including TP53, KRAS, FAM47C, MUC7, SHC adaptor protein 4, keratin associated protein 5–5 (KRTAP5-5), AKT serine/threonine kinase 1, taste 2 receptor member 10 (TAS2R10), β-2-microglobulin (B2M), potassium voltage-gated channel interacting protein 2 (KCNIP2), cluster of differentiation (CD)58, FK506 binding protein 3 (FKBP3) and INO80 complex subunit E (INO80E; Fig. 1C). Among these 13 genes, the top three genes with the highest mutation frequency in the present study and TCGA data were TP53, KRAS and FAM47C (4/10). As expected, the mutated KRAS gene had oncogenic codon 12 mutations (3/10 samples; Fig. 2A) and another mutation was in codon 146 of KRAS (1/10 samples; Fig. 2A), which was in accordance with a previous study of CRC in the Chinese population (24). FAM47C was mutated in 4 out of 10 tumor samples including 4 missense mutations (p.Q225E, p.P502P, p.Q225E and p.R701H), 1 silent mutation (p.P502P) and 1 mutation in the 3′flank region, as presented in Fig. 2B. The mutation frequency of FAM47C in the COSMIC and TCGA databases was 5.71 and 5.41%, respectively (2). FAM47C encodes a product belonging to a family of proteins with unknown function. Additionally, FAM47C was mutated exclusively in KRAS wild-type tumors. Specific ones out of the 13 genes were mutated only in the tumor from patient CRC-9, including TAS2R10, B2M, KCNIP2, CD58 and INO80E. B2M had two mutations (a frame shift deletion and an insertion in 3′flank region) in the CRC-9 tumor. Previous studies reported B2M mutations in CRC and melanoma resulting in loss of expression of HLA class 1 complexes (25), suggesting these mutations benefit tumor growth by reducing antigen presentation to the immune system (26). CD58 is reported to be a surface marker that promotes self-renewal of tumor-initiating cells in CRC (27).

Of the 32 significantly mutated genes, 19 were not listed in TCGA data, including BAGE2, TMEM128, spermatogenesis associated 3 (SPATA3), CD1B, RAB40A like, cysteine rich protein 3 (CRIP3), crystallin β B2, EBP like, guanidinoacetate N-methyltransferase, hes family bHLH transcription factor 3, olfactory receptor family 2 subfamily A member 7, proline rich nuclear receptor coactivator 2, small proline rich protein 2B (SPRR2B), sushi repeat containing protein X-linked 2 (SRPX2), translocase of inner mitochondrial membrane 17A (TIMM17A), TMEM179, vesicle associated membrane protein 8 (VAMP8), WD repeat domain 61 (WDR61) and zinc finger protein 124 (ZNF124). Notably, CRIP3 demonstrated a nonstop_mutation (p.*205Rext*51) and CD1B had a nonsense_mutation (p.Q221*) in the CRC-9 tumor, which was the hypermutated case (29.79 mutations per Mb).

Considering all somatic mutations in coding regions, the top 11 most frequently mutated genes were mucin 4 (MUC4; 8/10), immunoglobulin-like and fibronectin type III domain containing 1 (IGFN1; 5/10), ALMS1, centrosome and basal body associated protein (4/10), APC (4/10), family with sequence similarity 47 member C (FAM47C; 4/10), KRAS (4/10), mucin like 3 (DPCR1; 3/10), family with sequence similarity 186 member A (FAM186A; 3/10), polycystin 1, transient receptor potential channel interacting (3/10), and TTN (3/10). The most frequent mutated gene was MUC4 (8/10), with a mutation frequency that was higher compared with that reported in a previous study (2). The mutation frequency of MUC4 has been previously reported as 9.72% (Genentech) (5), 5.33% (DFCI) (3) and 2.23% (TCGA) (2). MUC4 is a major constituent of mucus that has important roles in the protection of epithelial cells and has been implicated in epithelial renewal and differentiation. The mucin gene MUC4 is reported to be a transcriptional and post-transcriptional target of the oncogene KRAS in pancreatic cancer (28). However, MUC4 was not defined as a driver gene by MutSigCV in the current study, which may due to the positions of MUC4 mutations, which were not in functional regions (Fig. 2C).

Functional enrichment analysis of mutated genes

To better understand the biological function of mutated genes, GO and KEGG enrichment analysis were performed. All mutated genes were categorized into 16 functional categories by GO enrichment (adjusted P<0.05; Table III). Notably, 11 functional categories were associated with transporter/channel activity. House et al (29) reported that voltage-gated Na+ channel activity increases colon cancer transcriptional activity and invasion via persistent mitogen-activated protein kinase signaling. All mutated genes were categorized into three pathways by KEGG enrichment, including ‘neuroactive ligand-receptor interaction’, ‘alanine, aspartate and glutamate metabolism’ and ‘nicotine addiction’ (adjusted P<0.05).

Table III.

Functional enrichment of mutated genes by GO.

Table III.

Functional enrichment of mutated genes by GO.

IDDescriptionP adjustGene count
GO:0022803Passive transmembrane transporter activity0.0004191209
GO:0015267Channel activity0.0004191208
GO:0022838Substrate-specific channel activity0.0009566193
GO:0001227Transcriptional repressor activity, RNA polymerase II transcription regulatory region sequence-specific binding0.001371187
GO:0005216Ion channel activity0.0015965185
GO:0000987Core promoter proximal region sequence-specific DNA binding0.0118789159
GO:0022836Gated channel activity0.0118789144
GO:0000982Transcription factor activity, RNA polymerase II core promoter proximal region sequence-specific binding0.0150299146
GO:0001159Core promoter proximal region DNA binding0.0150299159
GO:0005267Potassium channel activity0.018679860
GO:0046873Metal ion transmembrane transporter activity0.0198865176
GO:0022843Voltage-gated cation channel activity0.019886565
GO:0000978RNA polymerase II core promoter proximal region sequence-specific DNA binding0.0308052148
GO:0005261Cation channel activity0.0330241129
GO:0005244Voltage-gated ion channel activity0.048034485
GO:0022832Voltage-gated channel activity0.048034485
Chromosomal rearrangement

Chromosomal structural variation (SV) was also analyzed using whole-genome sequencing of 10 tumors with matched normal samples. There were 21 candidate-chromosomal rearrangements detected by filtering criterion of above 20 supporting reads, including 2 inversions and 19 translocations (Table IV). Among these, the fusion sites of 4 SVs were in gene regions, which were termed fusion genes, including, EF-hand domain family member B-mannosidase α class 1A member 1, phosphorylase kinase regulatory subunit β (PHKB)-NOTCH2 (2 samples) and polyamine modulated factor 1-FAM182B. A fusion of PHKB and NOTCH2 was identified in 2 out of 10 CRCs and the fusion occurred downstream of PHKB exon 5 and upstream of NOTCH2 exon 4 (Fig. 3A). This appears likely to enable translation of the fusion protein (the glycosyl hydrolases family 15 domain of PHKβ linked with the calcium-binding epidermal growth factor-like domain of Notch2; Fig. 3A). PHKB encodes a member of the PHKβ regulatory subunit family. PHKB was reported to promote glycogen breakdown and cancer cell survival by interacting with cell migration inducing hyaluronidase 1 (30). NOTCH2 encodes a member of the Notch family with a role in a variety of developmental processes by controlling cell fate decisions. Previous studies reported that Notch2 is a crucial regulator of self-renewal and tumorigenicity in human hepatocellular carcinoma cells (31,32) and contributes to cell growth, invasion and migration in salivary adenoid cystic carcinoma (33).

Table IV.

Predicted chromosomal rearrangement detected by Factera.

Table IV.

Predicted chromosomal rearrangement detected by Factera.

SampleTypeRegion1 (gene, position)Region2 (gene, position)Fusion sites
CRC-1TRATMEM194BIntergenicHFM1Intronicchr2:191402786 to chr1:91852783
CRC-1TRAROCK1IntergenicCTD-2144E22.5Intergenicchr18:18519930 to chr16:35239604
CRC-1TRATRIM48IntergenicMTRNR2L9Intergenicchr11:55021850 to chr6:61902202
CRC-2INVFAM27E3IntergenicAL445665.1Intergenicchr9:66971068 to chr9:69710933
CRC-2TRAUNC5BIntergenicMAN1A1Intronicchr10:72814597 to chr6:119558701
CRC-2TRAANO3IntergenicMAN1A1Intronicchr11:26173964 to chr6:119558701
CRC-2TRAAL445665.1IntergenicCTD-2144E22.5Intergenicchr9:69711250 to chr16:35239606
CRC-2TRAPPAP2CIntergenicPLEKHG4BIntergenicchr19:249186 to chr5:15867
CRC-3TRAEFHBIntronicMAN1A1Intronicchr3:19950145 to chr6:119558701
CRC-3TRATRIM48IntergenicMTRNR2L9Intergenicchr11:55021850 to chr6:61902202
CRC-3TRASOX14IntergenicZNF92Intergenicchr3:137265780 to chr7:64879411
CRC-3TRAPHKBIntronicNOTCH2Intronicchr16:47538780 to chr1:120544074
CRC-4INVRP11-146D12.2IntergenicCNTNAP3BIntergenicchr9:42416106 to chr9:44070790
CRC-4TRACSNK1G3IntergenicDLG2Intronicchr5:122990837 to chr11:85195011
CRC-4TRAPHKBIntronicNOTCH2Intronicchr16:47538780 to chr1:120544074
CRC-8TRAMTRNR2L1IntergenicOR4C46Intergenicchr17:22253139 to chr11:51568509
CRC-8TRASOX14IntergenicZNF92Intergenicchr3:137265780 to chr7:64879411
CRC-8TRATRIM48IntergenicMTRNR2L9Intergenicchr11:55021850 to chr6:61902202
CRC-9TRAPMF1IntronicFAM182BUpstreamchr1:156186653 to chr20:26190511
CRC-9TRAWDR74IntergenicPPP4R2Intergenicchr11:62609281 to chr3:73160133
CRC-9TRASOX14IntergenicZNF92Intergenicchr3:137265780 to chr7:64879411

[i] TRA, translocation; INV, inversion.

Copy number variations

Somatic CNVs in the 10 tumor tissues were analyzed using Control-FREEC software. Chromosomes 8 and 20 were amplified in CRC-4, and chromosome 20 was amplified in CRC-7 (Fig. 3B). GNAScomplex locus (GNAS) was detected in 2 out of 10 tumors (CRC-4 and CRC-7) and the GNAS copy number was 3. GNAS is located at 20q13.32. A previous study reported that amplification of the GNAS locus may contribute to the pathogenesis of breast cancer (34). In TCGA, GNAS was amplified in 8.17% tumor samples (2). In the CRC-7 tumor, a subset of genes located at chromosome 20 was amplified (copy number 3), including teashirt zinc finger homeobox 2, aurora kinase A, GNAS, SS18L1 nBAF chromatin remodeling complex subunit, regulator of telomere elongation helicase 1 and ADP ribosylation factor related protein 1. SVs of 10 tumors are presented in Fig. 3C, including chromosomal rearrangements and CNVs, displayed as CIRCOS plots.

Discussion

In the present study, whole-genome sequencing was performed on tumor and matched adjacent normal tissues from 10 patients with CRC in Shanghai. A comprehensive analysis including SNVs, InDels, CNVs and chromosomal rearrangements was performed, which identified certain recurrent and novel variations in CRC patients from the Han population in Shanghai, eastern China.

Among the significantly mutated genes, certain previously reported driver genes in CRC were identified, including TP53, KRAS, FAM47C and MUC7. Additionally, a group of driver genes were identified that have rarely been reported in CRC, including BAGE2, TMEM128, SPATA3 and CD1B. Certain well-established mutated genes, including APC and TTN, which were defined as driver genes in a previous study (2), were not significantly mutated genes in the present study. Signaling pathway analysis indicated that the mutated genes may alter pathways associated with channel activity. Notably, PHKB-NOTCH2 fusion was detected in 2 out of 10 tumors, which has not been previously reported in CRC, to the best of our knowledge. The structure of fusion proteins was also predicted. Although, further study will be required to fully understand the role of PHKB-NOTCH2 fusion.

The tumor mutation burden of the tumor of CRC-9 was 29.79 per Mb, which indicates hypermutation according to TCGA (2). TCGA identified 16% of CRCs to be hypermutated, three quarters of which are due to a mismatch repair defect phenotype, otherwise known as MSI-H. In CRC-9, 23 significantly mutated genes were identified, while the other 9 cases harbored a mean of 3.3 significantly mutated genes (range, 3–4). Somatic mutations have the potential to encode ‘non-self’ immunogenic antigens. Evidence demonstrated improved responses to programmed cell death protein 1 (PD-1) blockade in CRC with MSI-H (35,36). The tumor of patient CRC-9 was a locally advanced colon cancer (T4bN0M0) with poor-moderate differentiation and MSI-H. Cancer antigen (CA)12-5 and CA15-3 were at high levels following surgery, which indicated a high risk of recurrence. CRC-9 may benefit from PD-1 blockade to treat recurrence.

There are large differences in diet, living conditions and genetic background between the Han population and ethnic minorities, which are associated with CRC risk. For example, the Uyghur population in Xinjiang is predominantly Caucasian, while the Han population is mainly Mongoloid. Uyghur CRC patients have a lower age of onset, larger tumor size, more advanced stage and higher proportion of signet-ring cell carcinoma and mucinous adenocarcinoma compared with Han patients (37). A previous study reported that CRC patients in the Uyghur population exhibited a higher rate of KRAS mutation compared with the Han population (46.2% vs. 28.8%) and the mutation rate in KRAS codon 12 is higher in the Uygur population than in the Han population (38.5% vs. 17.3%) (38). In the present study, the KRAS mutation rate was 4/10 and 3/10 of mutations were in codon 12, which was at a comparable level to the Uyghur population (38).

There are limitations in this study. Firstly, the sample size was small, which lead to low statistical power to identify significantly mutated genes and could not well represent Chinese Han population. Secondly, the sequencing depth was not high enough to detect mutations with low variant allele frequency. Thirdly, further validation in samples and functional studies were not performed. Finally, due to the short time from patient enrollment, survival analysis was not performed. In future studies of a panel of CRC-associated genes, including reported recurrent genes and novel mutated genes in the present study, will be analyzed in a cohort of patients with CRC. In addition, survival analysis with genomic variations should be performed following long term follow-up for CRC patients.

In conclusion, in the present study, reported mutated genes were validated to a certain extent and novel mutations were identified, including fusion gene PHKB-NOTCH2. In addition, mutated genes were enriched in functions associated with channel activity, which has rarely been reported by previous CRC studies (2,4). The present study produced a CRC genomic mutation profile, which provides a valuable resource for further insight into CRC within the eastern Chinese Han population.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets generated during the current study are available from the corresponding author on reasonable request.

Authors' contributions

HT and JZ wrote the manuscript; HT and HQ contributed to the study design; HT collected and interpreted the clinical data of patients; RG, NQ and XJ collected samples and performed experiments; JZ, NL and SW developed the analysis methods and provided the figures and tables. YW and MR analyzed the sequencing data. NL and HQ contributed to manuscript revision.

Ethics approval and consent to participate

Ethics approval for the recruitment of human subjects was obtained from the Ethics Committee of Shanghai Tenth People's Hospital, Tongji University School of Medicine and was consistent with ethical guidelines provided by the Declaration of Helsinki (1975). Written informed consent was obtained from each patient.

Patient consent for publication

All individuals whose data were used provided informed consent for publication.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A and Jemal A: Colorectal cancer statistics, 2017. CA Cancer J Clin. 67:177–193. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Cancer Genome Atlas Network: Comprehensive molecular characterization of human colon and rectal cancer. Nature. 487:330–337. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Giannakis M, Mu XJ, Shukla SA, Qian ZR, Cohen O, Nishihara R, Bahl S, Cao Y, Amin-Mansour A, Yamauchi M, et al: Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep. 17:12062016. View Article : Google Scholar : PubMed/NCBI

4 

Brannon AR, Vakiani E, Sylvester BE, Scott SN, McDermott G, Shah RH, Kania K, Viale A, Oschwald DM, Vacic V, et al: Comparative sequencing analysis reveals high genomic concordance between matched primary and metastatic colorectal cancer lesions. Genome Biol. 15:4542014. View Article : Google Scholar : PubMed/NCBI

5 

Seshagiri S, Stawiski EW, Durinck S, Modrusan Z, Storm EE, Conboy CB, Chaudhuri S, Guan Y, Janakiraman V, Jaiswal BS, et al: Recurrent R-spondin fusions in colon cancer. Nature. 488:660–664. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Zeng H, Zheng R, Guo Y, Zhang S, Zou X, Wang N, Zhang L, Tang J, Chen J, Wei K, et al: Cancer survival in China, 2003–2003: A population-based study. Int J Cancer. 136:1921–1930. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Cajuso T, Hanninen UA, Kondelin J, Gylfe AE, Tanskanen T, Katainen R, Pitkänen E, Ristolainen H, Kaasinen E, Taipale M, et al: Exome sequencing reveals frequent inactivating mutations in ARID1A, ARID1B, ARID2 and ARID4A in microsatellite unstable colorectal cancer. Int J Cancer. 135:611–623. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Ashktorab H, Daremipouran M, Devaney J, Varma S, Rahi H, Lee E, Shokrani B, Schwartz R, Nickerson ML and Brim H: Identification of novel mutations by exome sequencing in African American colorectal cancer patients. Cancer. 121:34–42. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Oh BY, Cho J, Hong HK, Bae JS, Park WY, Joung JG and Cho YB: Exome and transcriptome sequencing identifies loss of PDLIM2 in metastatic colorectal cancers. Cancer Manag Res. 9:581–589. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Nagahashi M, Wakai T, Shimada Y, Ichikawa H, Kameyama H, Kobayashi T, Sakata J, Yagi R, Sato N, Kitagawa Y, et al: Genomic landscape of colorectal cancer in Japan: Clinical implications of comprehensive genomic sequencing for precision medicine. Genome Med. 8:1362016. View Article : Google Scholar : PubMed/NCBI

14 

Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al: Initial sequencing and analysis of the human genome. Nature. 409:860–921. 2001. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

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

17 

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

18 

Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, Miller CA, Mardis ER, Ding L and Wilson RK: VarScan 2: Somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22:568–576. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, Carter SL, Stewart C, Mermel CH, Roberts SA, et al: Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 499:214–218. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Boeva V, Popova T, Bleakley K, Chiche P, Cappo J, Schleiermacher G, Janoueix-Lerosey I, Delattre O and Barillot E: Control-FREEC: A tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics. 28:423–425. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Newman AM, Bratman SV, Stehr H, Lee LJ, Liu CL, Diehn M and Alizadeh AA: FACTERA: A practical method for the discovery of genomic rearrangements at breakpoint resolution. Bioinformatics. 30:3390–3393. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Skidmore ZL, Wagner AH, Lesurf R, Campbell KM, Kunisaki J, Griffith OL and Griffith M: GenVisR: Genomic Visualizations in R. Bioinformatics. 32:3012–3014. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Krzywinski M, Schein J, Birol I, Connors J, Gascoyne R, Horsman D, Jones SJ and Marra MA: Circos: An information aesthetic for comparative genomics. Genome Res. 19:1639–1645. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Zhang J, Zheng J, Yang Y, Lu J, Gao J, Lu T, Sun J, Jiang H, Zhu Y, Zheng Y, et al: Molecular spectrum of KRAS, NRAS, BRAF and PIK3CA mutations in chinese colorectal cancer patients: Analysis of 1,110 cases. Sci Rep. 5:186782015. View Article : Google Scholar : PubMed/NCBI

25 

Bernal M, Ruiz-Cabello F, Concha A, Paschen A and Garrido F: Implication of the β2-microglobulin gene in the generation of tumor escape phenotypes. Cancer Immunol Immunother. 61:1359–1371. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Cancer Genome Atlas Research Network: Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Xu S, Wen Z, Jiang Q, Zhu L, Feng S, Zhao Y, Wu J, Dong Q, Mao J and Zhu Y: CD58, a novel surface marker, promotes self-renewal of tumor-initiating cells in colorectal cancer. Oncogene. 34:1520–1531. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Vasseur R, Skrypek N, Duchêne B, Renaud F, Martínez-Maqueda D, Vincent A, Porchet N, Van Seuningen I and Jonckheere N: The mucin MUC4 is a transcriptional and post-transcriptional target of K-ras oncogene in pancreatic cancer. Implication of MAPK/AP-1, NF-κB and RalB signaling pathways. Biochim Biophys Acta. 1849:1375–1384. 2015. View Article : Google Scholar : PubMed/NCBI

29 

House CD, Wang BD, Ceniccola K, Williams R, Simaan M, Olender J, Patel V, Baptista-Hon DT, Annunziata CM, Gutkind JS, et al: Voltage-gated Na+ channel activity increases colon cancer transcriptional activity and invasion via persistent MAPK signaling. Sci Rep. 5:115412015. View Article : Google Scholar : PubMed/NCBI

30 

Terashima M, Fujita Y, Togashi Y, Sakai K, De Velasco MA, Tomida S and Nishio K: KIAA1199 interacts with glycogen phosphorylase kinase beta-subunit (PHKB) to promote glycogen breakdown and cancer cell survival. Oncotarget. 5:7040–7050. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Hayashi Y, Osanai M and Lee GH: NOTCH2 signaling confers immature morphology and aggressiveness in human hepatocellular carcinoma cells. Oncol Rep. 34:1650–1658. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Wu WR, Zhang R, Shi XD, Yi C, Xu LB and Liu C: Notch2 is a crucial regulator of self-renewal and tumorigenicity in human hepatocellular carcinoma cells. Oncol Rep. 36:181–188. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Qu J, Song M, Xie J, Huang XY, Hu XM, Gan RH, Zhao Y, Lin LS, Chen J, Lin X, et al: Notch2 signaling contributes to cell growth, invasion, and migration in salivary adenoid cystic carcinoma. Mol Cell Biochem. 411:135–141. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Garcia-Murillas I, Sharpe R, Pearson A, Campbell J, Natrajan R, Ashworth A and Turner NC: An siRNA screen identifies the GNAS locus as a driver in 20q amplified breast cancer. Oncogene. 33:2478–2486. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al: PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 372:2509–2520. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, et al: Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 357:409–413. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Roukeyan K, Yue N, Liang LP and Zhao F: Clinicopathological features and expression of hMLH1 and hMSH2 in Uygur and Han patients with colorectal carcinoma. Shijie Huaren Xiaohua Zazhi. 23:2382–2388. 2015.(In Chinese).

38 

Eli M, Mollayup A, Muattar, Liu C, Zheng C and Bao YX: K-ras genetic mutation and influencing factor analysis for Han and Uygur nationality colorectal cancer patients. Int J Clin Exp Med. 8:10168–10177. 2015.PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 18 Issue 6

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
Teng H, Gao R, Qin N, Jiang X, Ren M, Wang Y, Wu S, Li N, Zhao J, Qin H, Qin H, et al: Identification of recurrent and novel mutations by whole‑genome sequencing of colorectal tumors from the Han population in Shanghai, eastern China. Mol Med Rep 18: 5361-5370, 2018
APA
Teng, H., Gao, R., Qin, N., Jiang, X., Ren, M., Wang, Y. ... Qin, H. (2018). Identification of recurrent and novel mutations by whole‑genome sequencing of colorectal tumors from the Han population in Shanghai, eastern China. Molecular Medicine Reports, 18, 5361-5370. https://doi.org/10.3892/mmr.2018.9563
MLA
Teng, H., Gao, R., Qin, N., Jiang, X., Ren, M., Wang, Y., Wu, S., Li, N., Zhao, J., Qin, H."Identification of recurrent and novel mutations by whole‑genome sequencing of colorectal tumors from the Han population in Shanghai, eastern China". Molecular Medicine Reports 18.6 (2018): 5361-5370.
Chicago
Teng, H., Gao, R., Qin, N., Jiang, X., Ren, M., Wang, Y., Wu, S., Li, N., Zhao, J., Qin, H."Identification of recurrent and novel mutations by whole‑genome sequencing of colorectal tumors from the Han population in Shanghai, eastern China". Molecular Medicine Reports 18, no. 6 (2018): 5361-5370. https://doi.org/10.3892/mmr.2018.9563