Open Access

Screening key genes and signaling pathways in colorectal cancer by integrated bioinformatics analysis

  • Authors:
    • Chang Yu
    • Fuqiang Chen
    • Jianjun Jiang
    • Hong Zhang
    • Meijuan Zhou
  • View Affiliations

  • Published online on: June 4, 2019     https://doi.org/10.3892/mmr.2019.10336
  • Pages: 1259-1269
  • Copyright: © Yu 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

The aim of the present study was to identify potential key genes associated with the progression and prognosis of colorectal cancer (CRC). Differentially expressed genes (DEGs) between CRC and normal samples were screened by integrated analysis of gene expression profile datasets, including the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was conducted to identify the biological role of DEGs. In addition, a protein‑protein interaction network and survival analysis were used to identify the key genes. The profiles of GSE9348, GSE22598 and GSE113513 were downloaded from the GEO database. A total of 405 common DEGs were identified, including 236 down‑ and 169 upregulated. GO analysis revealed that the downregulated DEGs were mainly enriched in ‘detoxification of copper ion’ [biological process, (BP)], ‘oxidoreductase activity, acting on CH‑OH group of donors, NAD or NADP as acceptor’ [molecular function, (MF)] and ‘brush border’ [cellular component, (CC)]. Upregulated DEGs were mainly involved in ‘nuclear division’ (BP), ‘snoRNA binding’ (MF) and ‘nucleolar part’ (CC). KEGG pathway analysis revealed that DEGs were mainly involved in ‘mineral absorption’, ‘nitrogen metabolism’, ‘cell cycle’ and ‘caffeine metabolism’. A PPI network was constructed with 268 nodes and 1,027 edges. The top one module was selected, and it was revealed that module‑related genes were mainly enriched in the GO terms ‘sister chromatid segregation’ (BP), ‘chemokine activity’ (MF), and ‘condensed chromosome (CC)’. The KEGG pathway was mainly enriched in ‘cell cycle’, ‘progesterone‑mediated oocyte maturation’, ‘chemokine signaling pathway’, ‘IL‑17 signaling pathway’, ‘legionellosis’, and ‘rheumatoid arthritis’. DNA topoisomerase II‑α (TOP2A), mitotic arrest deficient 2 like 1 (MAD2L1), cyclin B1 (CCNB1), checkpoint kinase 1 (CHEK1), cell division cycle 6 (CDC6) and ubiquitin conjugating enzyme E2 C (UBE2C) were indicated as hub genes. Furthermore, survival analysis revealed that TOP2A, MAD2L1, CDC6 and CHEK1 may serve as prognostic biomarkers in CRC. The present study provided insights into the molecular mechanism of CRC that may be useful in further investigations.

Introduction

Colorectal cancer (CRC) is a global burden ranking third in terms of incidence and second in terms of mortality, with >1.8 million new CRC cases and 881,000 estimated deaths in 2018 (1). The main reason for the poor 5-year overall survival in CRC is late detection, when the opportunity for treatment has passed. Despite progress in novel therapies, early detection remains a challenge (2). A variety of tests are available for screening and detecting CRC; all, however, have their disadvantages (35). It is therefore urgent to identify novel diagnostic and prognostic biomarkers for CRC.

In recent years, high-quality microarray and high- throughput sequencing have been effective in detecting the development and progression of CRC, and even in screening biomarkers for CRC diagnosis, therapy and prognosis.

A number of gene profiles can be obtained from public databases such as Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), both of which can expand our understanding of cancer. Limitations and inconsistent results may exist, due to different microarray platforms and small sample sizes, but integrated bioinformatics methods may overcome these limitations.

Several recent studies have identified certain key genes and pathways in CRC using bioinformatics analysis (6,7). Based on these articles, updated datasets were selected and a gene classification method (clusterProfiler package in R) was used for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis; the prognostic role of hub genes was then ascertained using the TCGA dataset. In the present study, integrated analysis was first performed to identify the common DEGs from multiple microarrays (GSE113513, GSE9348 and GSE22598) and TCGA CRC RNA-seq data. Next, GO and KEGG enrichment analysis was conducted to identify the potential biological role of DEGs. A protein-protein interaction (PPI) network created based on the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version 10.5; http://string-db.org/) database, and the significant modules and hub genes were then selected from the network. The prognostic roles of hub genes were analyzed using TCGA.

Materials and methods

Gene expression profile data

The raw microarray data of CRC [GSE9348 (8), GSE22598 (9), GSE113513 (unpublished, 2018)] were downloaded from the GEO database (10) (Table I). All datasets fulfilled the following criteria: i) Human CRC tissue samples were used for the profiles; ii) normal samples were matched to the tumor tissues, when data could not be matched, matching was based on patient information, such as age and sex; and iii) The sample size per dataset was >10. The level 3 RNA sequencing data of CRC and normal samples as well as the CRC clinical information were downloaded from the Genomic Data Commons (GDC) which were retrieved from TCGA (https://tcga-data.nci.nih.gov/tcga/) database.

Table I.

Information on the datasets included in the current study.

Table I.

Information on the datasets included in the current study.

DatasetReferencePlatformNo. of samples (normal/tumor)
GSE9348Hong et al, 2010 (8)[HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array12/12
GSE22598Okazaki et al, 2011 (9)[HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array17/17
GSE113513Peng et al, 2018 (unpublished)[PrimeView] Affymetrix Human Gene Expression Array14/14
TCGA_CRCThe Cancer Genome Atlas (TCGA) data portalIlluminaHiseq (Illumina, San Diego, CA)51/647
Data pre-processing and DEG identification

CEL files from three Affymetrix microarrays were downloaded from GEO, and pre-processed using the Affy package (version 1.60.0; http://bioconductor.org/packages/release/bioc/html/affy.html) in R software (version 3.4.3; http://www.r-project.org/). The Robust Multi-array Average (RMA) method (11) was used for the pre-processing, which included background correcting, normalizing and calculating expression. The latest annotation files were downloaded for re-annotation. The Limma package (version 3.34.8) (12) in R software was subsequently used to screen DEGs between CRC and matched normal tissues in the microarray. The RNA sequencing data were obtained from TCGA and Ensembl Release 93 (http://jul2018.archive.ensembl.org/index.html) files were used for annotation and all the data processing and normalization were finished using the Perl (version 5.28.2; http://www.perl.org/) and R scripts. The edgeR package (version 3.24.3) (13) was used for DEG screening and the trimmed mean of M-values normalization method in edgeR was used to normalize the raw data. Notably, |log2FC|>1, P<0.05 and adjusted P<0.05 were considered the cut-off criteria. Intersect function was used to identify the common DEGs, and a Venn diagram was created using Venny (version 2.1) (14). All common DEGs in these datasets were selected for further study.

GO and KEGG pathway analyses

To elucidate the potential gene functional annotation and pathway enrichment associated with the common DEGs. GO (15,16) and KEGG (1719) analyses were performed using the clusterProfiler (version 3.10.1) package (20). The enrichplot and DOSE (21) packages were used to supply enrichment result visualization to help interpretation. P<0.05 and adjusted P<0.05 were set as the threshold values.

PPI network construction and module selection

The online STRING database was used to identify potential interaction among the common DEGs, and a confidence score of ≥0.4 was set as the threshold. Cytoscape (version 3.6.1) (22) was used to visualize the PPI network of common DEGs. The MCODE plug-in (23) was used to search sub-networks of the PPI network and the default parameters (Degree cutoff ≥10, node score cutoff ≥0.4, K-core ≥4, and max depth=100.) were set in the functional interface of Cytoscape software. GO and KEGG enrichment analyses of cluster modules were performed using ClueGO plug-in (24) with default parameters. The Cytohubba plug-in (25) was used to explore hub genes, and the top ten were generated using Maximal Clique Centrality (MCC), closeness and degree methods. The intersect function was used to identify the common hub genes.

Survival analysis of hub genes

In order to identify the potential prognostic role of hub genes, Kaplan-Meier analysis was performed based on the expression and clinical data of TCGA, and Perl was used to merge data. Hub genes were divided into two strata based on expression level and median value. The survival (version 2.44–1.1; http://cran.r-project.org/web/packages/survival/index.html) and survminer package (version 0.4.3; http://cran.r-project.org/web/packages/survminer/index.html) in R were used.

Results

Identification of DEGs in GEO and TCGA

Following pre-processing of the raw data, DEGs were identified by R package. In total, 931 DEGs (543 down- and 388 upregulated genes), 1,420 DEGs (759 down- and 661 upregulated genes), 1,324 DEGs (659 down- and 665 upregulated genes) and 12,237 DEGs (3,776 down- and 8,461 upregulated genes) were screened from the GSE113513, GSE9348, GSE22598 and TCGA datasets, respectively. Heatmaps of DEGs are presented in Fig. S1, and volcano plots in Fig. S2. The intersect function revealed 405 common DEGs, including 236 down- and 169 upregulated DEGs from four independent datasets (Fig. 1). The top 20 down- and upregulated DEGs from the four datasets are listed in Table II.

Table II.

Top 20 down- and upregulated overlapping DEGs in GSE113513, GSE9348, GSE22598 and TCGA were screened by intersected analysis.

Table II.

Top 20 down- and upregulated overlapping DEGs in GSE113513, GSE9348, GSE22598 and TCGA were screened by intersected analysis.

GenesGSE113513GSE9348GSE22598TCGARegulation
AQP8−4.00−6.74−2.75−6.87Down
CLCA4−2.30−7.61−4.91−5.35Down
GUCA2B−4.82−5.08−3.37−6.06Down
MS4A12−3.68−6.04−3.54−5.36Down
GUCA2A−4.10−5.00−3.72−5.11Down
CA2−3.68−5.32−3.76−4.81Down
ABCG2−2.39−6.03−3.93−4.68Down
CLDN8−3.69−4.40−3.65−5.07Down
GCG−4.57−3.92−4.15−4.13Down
ZG16−3.18−4.70−4.45−4.44Down
PKIB−3.21−4.99−3.85−3.95Down
CA4−3.39−4.54−3.07−4.76Down
BEST4−3.50−2.72−3.66−5.85Down
CA1−3.33−3.20−1.94−6.51Down
MT1M−3.29−3.90−3.35−4.30Down
CD177−2.84−4.29−2.35−5.22Down
HSD17B2−2.63−5.14−3.04−3.39Down
INSL5−3.31−2.03−3.00−5.82Down
ADH1C−3.26−3.50−3.10−4.01Down
CLCA1−3.45−3.77−3.62−2.54Down
FOXQ14.475.165.556.47Up
KRT233.694.254.287.37Up
LY6G6D3.504.093.635.42Up
MMP73.153.112.387.03Up
CDH32.772.312.765.77Up
MMP32.912.942.774.83Up
CST11.252.421.008.33Up
CRNDE3.172.362.734.60Up
DPEP12.973.064.142.61Up
MMP12.462.593.144.56Up
EPHX43.012.322.954.43Up
CTHRC11.214.473.153.78Up
CLDN12.182.812.744.84Up
CEL1.562.532.376.10Up
CLDN21.542.572.755.57Up
SLC35D31.792.973.244.35Up
COL11A11.592.491.936.33Up
CXCL33.223.602.342.92Up
SLCO1B31.431.642.556.44Up
CKMT23.332.552.683.46Up

[i] Each column represents a dataset and each row a gene. The values in the table represent the log2FC in the datasets. DEGs, differentially expressed genes; TCGA, The Cancer Genome Atlas; FC, fold change.

GO and KEGG enrichment analysis

To explore the potential biological function of common DEGs, GO and KEGG pathways enrichment analyses were conducted using the ClusterProfiler package. In the present study, downregulated DEGs were mainly enriched in ‘detoxification of copper ion’ (BP), ‘oxidoreductase activity, acting on CH-OH group of donors, NAD or NADP as acceptor’ (MF) and ‘brush border’ (CC) respectively. Upregulated DEGs were mainly involved in ‘nuclear division’ (BP), ‘snoRNA binding’ (MF) and ‘nucleolar part’ (CC).'The GO distribution of down- and upregulated DEGs is presented in Fig. 2A and B and details of the top 15 GO terms in Table III.

Table III.

GO analysis of down- and upregulated overlapping DEGs associated with CRC.

Table III.

GO analysis of down- and upregulated overlapping DEGs associated with CRC.

ExpressionTermsIDDescriptionq-valueGenes
UpregulatedBPGO:0000280Nuclear division 4.33×10−7 RRS1/CKS2/TRIP13/CCND1/FIGNL1/TPX2/ANLN/UBE2C/CHEK1/NUF2/RAD54B/DLGAP5/CDC6/MAD2L1/TOP2A/TTK/CCNB1/ASPM/BUB1B/AURKA
BPGO:0048285Organelle fission 1.16×10−6 RRS1/CKS2/TRIP13/CCND1/FIGNL1/TPX2/ANLN/UBE2C/CHEK1/NUF2/RAD54B/DLGAP5/CDC6/MAD2L1/TOP2A/TTK/CCNB1/ASPM/BUB1B/AURKA
BPGO:0140014Mitotic nuclear division 6.51×10−6 RRS1/TRIP13/CCND1/TPX2/ANLN/UBE2C/CHEK1/NUF2/DLGAP5/CDC6/MAD2L1/TTK/CCNB1/BUB1B/AURKA
BPGO:0007088Regulation of mitotic nuclear division 1.18×10−5 TRIP13/CCND1/ANLN/UBE2C/CHEK1/DLGAP5/CDC6/MAD2L1/TTK/CCNB1/BUB1B/AURKA
BPGO:0000075Cell cycle checkpoint 1.18×10−5 TRIP13/CCND1/ARID3A/CHEK1/CDC6/MAD2L1/TOP2A/TTK/CCNB1/SOX4/BUB1B/AURKA/PROX1
CCGO:0044452Nucleolar part 4.66×10−5 TAF1D/DKC1/UTP4/WDR43/NOP58/RRS1/RPP40/NUFIP1/ORC6/POLR1D/E2F5
CCGO:0000793Condensed chromosome 4.66×10−5 RRS1/SKA3/CHEK1/NUF2/CENPA/MAD2L1/TOP2A/CCNB1/SPC25/ERCC6L/BUB1B/AURKA
CCGO:0000779Condensed chromosome, centromeric region 5.94×10−5 SKA3/NUF2/CENPA/MAD2L1/CCNB1/SPC25/ERCC6L/BUB1B/AURKA
CCGO:0000780Condensed nuclear Chromosome, centromeric region 1.13×10−4 NUF2/CENPA/CCNB1/BUB1B/AURKA
CCGO:0000776Kinetochore 1.13×10−4 SKA3/NUF2/CENPA/MAD2L1/TTK/CCNB1/SPC25/ERCC6L/BUB1B
MFGO:0030515snoRNA binding 2.99×10−3 DKC1/NOP58/DDX21/NUFIP1
MFGO:0009982Pseudouridine synthase activity 3.70×10−3PUS1/PUS7/DKC1
MFGO:0035173Histone kinase activity 7.05×10−3 CHEK1/CCNB1/AURKA
MFGO:0001664G protein-coupled receptor binding 8.09×10−3 CXCL3/RNF43/HOMER1/NMU/CXCL8/ZNRF3/CXCL1/CTHRC1/CXCL5
MFGO:0140098Catalytic activity, acting on RNA 1.15×10−2 NOB1/PUS1/EXOSC5/NOP2/RPP40/DDX21/RNASEH2A/POLR1D/RAD54B/AZGP1
DownregulatedBPGO:0010273Detoxification of copper ion 1.22×10−5 MT1M/MT1X/MT1E/MT1H/MT1F/MT1G
BPGO:1990169Stress response to copper ion 1.22×10−5 MT1M/MT1X/MT1E/MT1H/MT1F/MT1G
BPGO:0061687Detoxification of inorganic compound 1.22×10−5 MT1M/MT1X/MT1E/MT1H/MT1F/MT1G
BPGO:0071280Cellular response to copper ion 1.22×10−5 MT1M/MT1X/MT1E/MT1H/MT1F/MT1G/AOC1
BPGO:0097501Stress response to metal ion 1.35×10−5 MT1M/MT1X/MT1E/MT1H/MT1F/MT1G
CCGO:0005903Brush border 6.96×10−5 CDHR5/TRPM6/SCIN/CDHR2/CA4/LIMA1/CYBRD1/MYO1A/SLC26A3/SI
CCGO:0045177Apical part of cell 3.15×10−4 ABCG2/CA2/RAB27A/CDHR5/PTPRH/TRPM6/AQP8/CDHR2/CA4/SCNN1B/CLCA4/CEACAM1/FABP1/CEACAM7/MYO1A/SLC26A3/SI
CCGO:0031526Brush border membrane 3.15×10−4 CDHR5/TRPM6/CDHR2/CA4/LIMA1/CYBRD1/SLC26A3
CCGO:0005902Microvillus 6.78×10−4 CA2/CDHR5/PTPRH/CDHR2/CEACAM1/CLCA1/AOC3/MYO1A
CCGO:0098862Cluster of actin-based cell projections 8.31×10−4 CDHR5/TRPM6/SCIN/CDHR2/CA4/LIMA1/CYBRD1/MYO1A/SLC26A3/SI
MFGO:0016616Oxidoreductase activity, acting on the CH-OH group of donors, NAD or NADP as acceptor 6.48×10−5 UGDH/ADH1B/HPGD/LDHD/ADH1C/DHRS11/DHRS9/HSD17B2/AKR1B10/HSD11B2/BMP2
MFGO:0016614Oxidoreductase activity, acting on CH-OH group of donors 1.09×10−4 UGDH/ADH1B/HPGD/LDHD/ADH1C/DHRS11/DHRS9/HSD17B2/AKR1B10/HSD11B2/BMP2
MFGO:0004089Carbonate dehydratase activity 1.26×10−4 CA7/CA2/CA12/CA1/CA4
MFGO:0048038Quinone binding 4.90×10−3 ETFDH/TP53I3/AOC3/AOC1
MFGO:0033764Steroid dehydrogenase activity, acting on the CH-OH group of donors, NAD or NADP as acceptor 1.52×10−2 DHRS11/DHRS9/HSD17B2/HSD11B2

[i] Adjusted P<0.05 was considered to be significant. DEGs, differentially expressed genes; CRC, colorectal cancer; q-value, adjusted P-value; GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function.

According to KEGG enrichment analysis, four significant pathways of common DEGs were identified, including: ‘Mineral absorption’, ‘nitrogen metabolism’, ‘cell cycle’ and ‘caffeine metabolism’ (Fig. 2C). The significant genes in these pathways are presented in Fig. 2D.

PPI network and module selection

The PPI network of DEGs was constructed with 268 nodes and 1,027 edges (Fig. 3A). The Degree, MCC and Closeness methods were performed to calculate the top 10 ranking hub genes. The results revealed 6 genes identified as hub genes, including DNA topoisomerase II-α (TOP2A), mitotic arrest deficient 2 like 1 (MAD2L1), cyclin B1 (CCNB1), checkpoint kinase 1 (CHEK1), cell division cycle 6 (CDC6), ubiquitin conjugating enzyme E2 C (UBE2C) (Fig. 3B). MCODE was used to identify the significant cluster modules in the PPI network and the top module was selected (Fig. 3C). Following GO annotation screening, the module (44 nodes and 462 edges) was revealed to be associated with ‘sister chromatid segregation’ (BP), ‘chemokine activity’ (MF), ‘condensed chromosome (CC)’ (Table IV) and KEGG pathway enrichment analysis revealed that the top module was mainly enriched in ‘cell cycle’, ‘progesterone-mediated oocyte maturation’, ‘chemokine signaling pathway’, ‘IL-17 signaling pathway’, ‘legionellosis’ and ‘rheumatoid arthritis’; related information were represent in Table V.

Table IV.

GO analysis of selected module-associated DEGs.

Table IV.

GO analysis of selected module-associated DEGs.

IDGO TermsOntologyq-valueGenes
GO:0000819Sister chromatid segregationBP 5.54×10−13 BUB1B/CCNB1/CDC6/CENPA/DLGAP5/ERCC6L/MAD2L1/NUF2/SPC25/TOP2A/TRIP13/TTK/UBE2C
GO:0007088Regulation of mitotic nuclear divisionBP 8.77×10−12 ANLN/AURKA/BUB1B/CCNB1/CDC6/CHEK1/DLGAP5/MAD2L1/TRIP13/TTK/UBE2C
GO:0098813Nuclear chromosomeBP 1.24×10−11 segregation BUB1B/CCNB1/CDC6/CENPA/DLGAP5/ERCC6L/MAD2L1/NUF2/SPC25/TOP2A/TRIP13/TTK/UBE2C
GO:0051304Chromosome separationBP 1.37×10−11 BUB1B/CCNB1/CDC6/DLGAP5/MAD2L1/TOP2A/TRIP13/TTK/UBE2C
GO:0030071Regulation of mitotic metaphase/anaphase transitionBP 1.46×10−11 BUB1B/CCNB1/CDC6/DLGAP5/MAD2L1/TRIP13/TTK/UBE2C
GO:0000793Condensed chromosomeCC 2.53×10−10 AURKA/BUB1B/CCNB1/CENPA/CHEK1/ERCC6L/MAD2L1/NUF2/SKA3/SPC25/TOP2A
GO:0000779Condensed chromosome/centromeric regionCC 5.40×10−10 AURKA/BUB1B/CCNB1/CENPA/ERCC6L/MAD2L1/NUF2/SKA3/SPC25
GO:0000775 Chromosome/centromeric regionCC 1.59×10−9 AURKA/BUB1B/CCNB1/CENPA/ERCC6L/MAD2L1/NUF2/SKA3/SPC25/TTK
GO:0000776KinetochoreCC 1.80×10−9 BUB1B/CCNB1/CENPA/ERCC6L/MAD2L1/NUF2/SKA3/SPC25/TTK
GO:0000777Condensed chromosome kinetochoreCC 8.17×10−9 BUB1B/CCNB1/CENPA/ERCC6L/MAD2L1/NUF2/SKA3/SPC25
GO:0008009Chemokine activityMF 1.41×10−9 CCL19/CCL28/CXCL1/CXCL12/CXCL3/CXCL5/CXCL8
GO:0042379Chemokine receptor bindingMF 8.09×10−9 CCL19/CCL28/CXCL1/CXCL12/CXCL3/CXCL5/CXCL8
GO:0045236CXCR chemokine receptor bindingMF 1.46×10−8 CXCL1/CXCL12/CXCL3/CXCL5/CXCL8
GO:0035173Histone kinase activityMF 1.99×10−4 AURKA/CCNB1/CHEK1

[i] Adjusted P<0.05 was considered as significanct. DEGs, differentially expressed genes; CRC, colorectal cancer; q-value, adjusted P-value; GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function.

Table V.

KEGG pathway analysis of selected module-associated DEGs.

Table V.

KEGG pathway analysis of selected module-associated DEGs.

KEGGCountq-valueGenes
Cell cycle6 1.88×10−5 BUB1B/CCNB1/CDC6/CHEK1/MAD2L1/TTK
Progesterone-mediated oocyte maturation4 7.64×10−4 AURKA/CCNB1/GNAI1/MAD2L1
Chemokine signaling pathway9 4.69×10−8 CCL19/CCL28/CXCL1/CXCL12/CXCL3/CXCL5/CXCL8/GNAI1/GNG4
IL-17 signaling pathway4 9.02×10−4 CXCL1/CXCL3/CXCL5/CXCL8
Legionellosis3 9.37×10−4 CXCL1/CXCL3/CXCL8
Rheumatoid arthritis4 1.06×10−3 CXCL1/CXCL12/CXCL5/CXCL8

[i] DEGs, differentially expressed genes; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Survival analysis

The prognostic role of hub genes was analyzed using Kaplan-Meier method based on the TCGA data. Among these hub genes, a low expression of CDC6, CHEK1, MAD2L1 and TOP2A was associated with poor prognosis in CRC patients (Fig. 4).

Discussion

In the present study, three GEOs and TCGA data were integrated, and non-paired GEO data were manually matched to increase accuracy and stabilization. In total, 236 down- and 169 upregulated DEGs were identified. The common downregulated DEGs were mostly enriched in ‘detoxification of copper ion’ (BP), ‘oxidoreductase activity, acting on CH-OH group of donors, NAD or NADP as acceptor’ (MF) and ‘brush border’ (CC), and the upregulated DEGs were mainly associated with ‘nuclear division’ (BP), ‘snoRNA binding’ (MF) and ‘nucleolar part’ (CC). Moreover, KEGG enrichment analysis identified that the common DEGs were largely involved in ‘mineral absorption’, ‘nitrogen metabolism’, ‘cell cycle’ and ‘caffeine metabolism’. Investigating these significant pathways may promote the understanding of CRC development.

Minerals are one of the five fundamental groups of nutrients; regular mineral absorption plays a vital role in sustaining life. Deficiency and insufficiency of minerals may be associated with and increase the risk of cancer; for example, efficient absorption of vitamin D may prevent CRC (26). The cell cycle is controlled by various mechanisms, which ensure correct cell division; loss of normal cell cycle control is a hallmark of cancer (27). An increasing number of studies has revealed that targeting the deregulation of the cell cycle in cancer is a potential therapeutic strategy (28). Therefore, investigating the cell cycle pathway may promote the understanding of carcinogenic mechanisms and insights into CRC treatment options. Nitrogen is an essential component of life that is involved in processes of both proteins and nucleic acids. Aberrant expression of nitrogen species could affect the risk of cancer (29). Certain studies have reported that caffeine could decrease the risk of CRC (30), inhibit colon cancer cell proliferation (31,32). Metabolic profiling revealed that caffeine metabolism differed significantly between colorectal adenoma polyps and CRC patients which may influence CRC development and outcome (33). Investigating these significant pathways may elucidate the mechanism of CRC progression.

Recently, certain important biomarkers, including CCL19, CXCL1, CXCL5, CXCL11, CXCL12, GNG4, INSL5, NMU, PYY and SST, were identified using integrated bioinformatics analysis. Furthermore, a prognostic gene signature consisting of 9 genes was also identified (34). In the present study, data from three GEO datasets (GSE22598, GSE113513 and GSE9348) and TCGA were combined for screening stable DEGs, then different calculation methods and intersect function analysis were used, and potential biomarkers were revealed that had not been previously screened out. A PPI network was constructed with DEGs, and then 6 hub genes (TOP2A, MAD2L1, CCNB1, CHEK1, CDC6 and UBE2C) were identified. Following survival analysis based on the TCGA data, the low expression of CDC6, CHEK1, MAD2L1 and TOP2A was revealed to be associated with poor prognosis. TOP2A was an important nuclear enzyme involved in DNA transcription and replication. Aberrant TOP2A expression has been identified in several types of cancer, such as CRC, breast carcinomas, gastric cancer and bladder carcinomas (3538). In addition, the high expression of TOP2A was revealed to be correlated with a worse survival in breast cancer, small-cell lung cancer and malignant melanoma, indicating that TOP2A can serve as a prognostic biomarker (3941). TOP2A was also revealed to be associated with advanced CRC and chemotherapeutic resistance via the inhibition of apoptosis (35). Zhou et al (42) revealed that TOP2A involvement in T-cell factor transcription may transmit a mechanism of multidrug resistance to TOP2A inhibitors, which can be an effective treatment option for CRC. Therefore, further investigation is required to elucidate the mechanism of TOP2A in the development, progression and treatment of CRC. CDC6 is essential for the initiation of DNA replication and contains ATP-binding and hydrolytic activities, which are required for the formation of the pre-replicative complex (43,44). It is also involved in the cell cycle by localizing to the centrosomes during the S and G2 phases (45). It has been reported that CDC6 affects proliferation during the early differentiation stages (46), and that the overexpression of CDC6 in tumors could signify that it may be an oncogenic target. CHEK1 is a serine/threonine kinase involved in delaying cell cycle progression; it is also required for the activation of DNA repair in response to the presence of DNA damage or unreplicated DNA (47). MAD2L1 (also termed MAD2) plays an important role in maintaining the spindle checkpoint function. MAD2L1 was revealed to be correlated with disease outcome in patients with breast cancer (48), and an increased MAD2L1 expression in neuroblastoma cells to be associated with poor prognosis (49).

The PPI network module analysis revealed that the development of CRC was mainly associated with the cell cycle pathway. It has been reported that cell cycle signaling pathways play a vital role in controlling normal progression, and regulating cell proliferation and apoptosis; an uncontrolled cell cycle may result in cancer (27). Therefore, cell cycle regulation is a useful way of interfering with the development of CRC.

Recent studies have indicated that mRNAs, miRNAs and lncRNAs play a crucial role in a variety of biological processes associated with human diseases (50,51). Chen et al (5255), have constructed powerful computational models to predict potential associations between miRNAs/lncRNAs and human diseases, providing a reliable and powerful tool for disease-association prediction. With the application of computational models, more stable and effective biomarkers (miRNAs/lncRNAs) would be revealed, and the pathogenesis of CRC would be explained at different molecular levels.

The present results provided useful information for early diagnosis and prevention, and supplied an effective therapeutic target for CRC. However, the study has certain limitations: i) By mixing all cancer types together, the focused insights in genetic characteristics of different subtypes of tumors may not be revealed, therefore, a stratification analysis of major clinical information should be performed; ii) more databases should be used for validation of the DEGs in future research; iii) in the present study, all tumor samples were included for the evaluation of the prognositic role of hub genes, however, it would be better to exclude CRC without matching normal controls, which would reduce the difference arising from different clinical information, and iv) intersect function analysis of GEO and TCGA was performed to screen DEGs, however, among these data sets, GSE22598 included colon cancer treated with chemotherapy which may affect the results, therefore, sample information should be more focused in future research. In addition, biological experiments are required to confirm these results. In conclusion, in the present study DEGs were identified by integrated bioinformatics analysis, in order to explore the role of DEGs in the progression and prognosis of CRC. Consequently, 405 DEGs were screened out, and hub genes CDC6, CHEK1, MAD2L1 and TOP2A were revealed to be promising prognostic biomarkers among CRC patients.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81472922 and 81673105).

Availability of data and materials

All data generated or analyzed during the present study are included in this article, and the R codes are available at https://github.com/Yuchang66/CRC-project.

Authors' contributions

All authors conceived and designed the experiments. CY contributed to the bioinformatics analysis and drafted the manuscript. CY, FC and JJ contributed to the data mining. HZ and MZ contributed to the statistical analysis and modified the manuscript. All authors read and approved the final version of the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

O'Connell JB, Maggard MA and Ko CY: Colon cancer survival rates with the new American joint committee on cancer sixth edition staging. J Natl Cancer Inst. 96:1420–1425. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Aslinia F, Uradomo L, Steele A, Greenwald BD and Raufman JP: Quality assessment of colonoscopic cecal intubation: An analysis of 6 years of continuous practice at a university hospital. Am J Gastroenterol. 101:721–731. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Uraoka T, Ramberan H, Matsuda T, Fujii T and Yahagi N: Cold polypectomy techniques for diminutive polyps in the colorectum. Dig Endosc. 26 (Suppl 2):S98–S103. 2014. View Article : Google Scholar

5 

Martín-López JE, Beltrán-Calvo C, Rodríguez-López R and Molina-López T: Comparison of the accuracy of CT colonography and colonoscopy in the diagnosis of colorectal cancer. Colorectal Dis. 16:O82–O89. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Liang B, Li C and Zhao J: Identification of key pathways and genes in colorectal cancer using bioinformatics analysis. Med Oncol. 33:1112016. View Article : Google Scholar : PubMed/NCBI

7 

Guo Y, Bao Y, Ma M and Yang W: Identification of key candidate genes and pathways in colorectal cancer by integrated bioinformatical analysis. Int J Mol Sci. 18(pii): E7222017. View Article : Google Scholar : PubMed/NCBI

8 

Hong Y, Downey T, Eu KW, Koh PK and Cheah PY: A ‘metastasis-prone’ signature for early-stage mismatch-repair proficient sporadic colorectal cancer patients and its implications for possible therapeutics. Clin Exp Metastasis. 27:83–90. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Okazaki S, Ishikawa T, Iida S, Ishiguro M, Kobayashi H, Higuchi T, Enomoto M, Mogushi K, Mizushima H, Tanaka H, et al: Clinical significance of UNC5B expression in colorectal cancer. Int J Oncol. 40:209–216. 2012.PubMed/NCBI

10 

Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, Marshall KA, Phillippy KH, Sherman PM, Holko M, et al: NCBI GEO: Archive for functional genomics data sets-update. Nucleic Acids Res 41 (Database Issue). D991–D995. 2013.

11 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

13 

Robinson MD, McCarthy DJ and Smyth GK: EdgeR: A bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 26:139–140. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Oliveros JC: VENNY. An interactive tool for comparing lists with Venn Diagrams. 2007.

15 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

16 

The Gene Ontology Consortium: The gene ontology resource: 20 years and still GOing strong. Nucleic Acids Res. 47(D1): D330–D338. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Kanehisa M, Sato Y, Furumichi M, Morishima K and Tanabe M: New approach for understanding genome variations in KEGG. Nucleic Acids Res. 47(D1): D590–D595. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Kanehisa M, Furumichi M, Tanabe M, Sato Y and Morishima K: KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45(D1): D353–D361. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Yu G, Wang LG, Yan GR and He QY: DOSE: An R/Bioconductor package for disease ontology semantic and enrichment analysis. Bioinformatics. 31:608–609. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

24 

Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pagès F, Trajanoski Z and Galon J: ClueGO: A cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 25:1091–1093. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Chin CH, Chen SH, Wu HH, Ho CW, Ko MT and Lin CY: CytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 8 (Suppl 4):S112014. View Article : Google Scholar : PubMed/NCBI

26 

Takada I and Makishima M: Control of inflammatory bowel disease and colorectal cancer by synthetic vitamin D receptor ligands. Curr Med Chem. 24:868–875. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Vermeulen K, Van Bockstaele DR and Berneman ZN: The cell cycle: A review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif. 36:131–149. 2010. View Article : Google Scholar

28 

Aarts M, Linardopoulos S and Turner NC: Tumour selective targeting of cell cycle kinases for cancer treatment. Curr Opin Pharmacol. 13:529–535. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Rieder G, Hofmann JA, Hatz RA, Stolte M and Enders GA: Up-regulation of inducible nitric oxide synthase in helicobacter pylori-associated gastritis may represent an increased risk factor to develop gastric carcinoma of the intestinal type. Int J Med Microbiol. 293:403–412. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Hashibe M, Galeone C, Buys SS, Gren L, Boffetta P, Zhang ZF and La Vecchia C: Coffee, tea, caffeine intake, and the risk of cancer in the PLCO cohort. Br J Cancer. 113:809–816. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Guertin KA, Loftfield E, Boca SM, Sampson JN, Moore SC, Xiao Q, Huang WY, Xiong X, Freedman ND, Cross AJ and Sinha R: Serum biomarkers of habitual coffee consumption may provide insight into the mechanism underlying the association between coffee consumption and colorectal cancer. Am J Clin Nutr. 101:1000–1011. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Merighi S, Benini A, Mirandola P, Gessi S, Varani K, Simioni C, Leung E, Maclennan S, Baraldi PG and Borea PA: Caffeine inhibits adenosine-induced accumulation of hypoxia-inducible factor-1alpha, vascular endothelial growth factor, and interleukin-8 expression in hypoxic human colon cancer cells. Mol Pharmacol. 72:395–406. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Long Y, Sanchez-Espiridion B, Lin M, White L, Mishra L, Raju GS, Kopetz S, Eng C, Hildebrandt MAT, Chang DW, et al: Global and targeted serum metabolic profiling of colorectal cancer progression. Cancer. 123:4066–4074. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Chen L, Lu D, Sun K, Xu Y, Hu P, Li X and Xu F: Identification of biomarkers associated with diagnosis and prognosis of colorectal cancer patients based on integrated bioinformatics analysis. Gene. 692:119–125. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Coss A, Tosetto M, Fox EJ, Sapetto-Rebow B, Gorman S, Kennedy BN, Lloyd AT, Hyland JM, O'Donoghue DP, Sheahan K, et al: Increased topoisomerase IIalpha expression in colorectal cancer is associated with advanced disease and chemotherapeutic resistance via inhibition of apoptosis. Cancer Lett. 276:228–238. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Bofin AM, Ytterhus B and Hagmar BM: TOP2A and HER-2 gene amplification in fine needle aspirates from breast carcinomas. Cytopathology. 14:314–319. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Kanta SY, Yamane T, Dobashi Y, Mitsui F, Kono K and Ooi A: Topoisomerase IIalpha gene amplification in gastric carcinomas: Correlation with the HER2 gene. An immunohistochemical, immunoblotting, and multicolor fluorescence in situ hybridization study. Hum Pathol. 37:1333–1343. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Kruger S, Lange I, Kausch I and Feller AC: Protein expression and gene copy number analysis of topoisomerase 2alpha, HER2 and P53 in minimally invasive urothelial carcinoma of the urinary bladder-a multitissue array study with prognostic implications. Anticancer Res. 25:263–271. 2005.PubMed/NCBI

39 

Depowski PL, Rosenthal SI, Brien TP, Stylos S, Johnson RL and Ross JS: Topoisomerase IIalpha expression in breast cancer: Correlation with outcome variables. Mod Pathol. 13:542–547. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Dingemans AM, Witlox MA, Stallaert RA, van der Valk P, Postmus PE and Giaccone G: Expression of DNA topoisomerase IIalpha and topoisomerase IIbeta genes predicts survival and response to chemotherapy in patients with small cell lung cancer. Clin Cancer Res. 5:2048–2058. 1999.PubMed/NCBI

41 

Mu XC, Tran TA, Ross JS and Carlson JA: Topoisomerase II-alpha expression in melanocytic nevi and malignant melanoma. J Cutan Pathol. 27:242–248. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Zhou Q, Abraham AD, Li L, Babalmorad A, Bagby S, Arcaroli JJ, Hansen RJ, Valeriote FA, Gustafson DL, Schaack J, et al: Topoisomerase IIα mediates TCF-dependent epithelial-mesenchymal transition in colon cancer. Oncogene. 35:4990–4999. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Fernandez-Cid A, Riera A, Tognetti S, Herrera MC, Samel S, Evrin C, Winkler C, Gardenal E, Uhle S, Speck C, et al: An ORC/Cdc6/MCM2-7 complex is formed in a multistep reaction to serve as a platform for MCM double-hexamer assembly. Mol Cell. 50:577–588. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Randell JC, Bowers JL, Rodriguez HK and Bell SP: Sequential ATP hydrolysis by Cdc6 and ORC directs loading of the Mcm2-7 helicase. Mol Cell. 21:29–39. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Kim GS, Kang J, Bang SW and Hwang DS: Cdc6 localizes to S- and G2-phase centrosomes in a cell cycle-dependent manner. Biochem Biophys Res Commun. 456:763–767. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Barkley LR, Hong HK, Kingsbury SR, James M, Stoeber K and Williams GH: Cdc6 is a rate-limiting factor for proliferative capacity during HL60 cell differentiation. Exp Cell Res. 313:3789–3799. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Huang M, Miao ZH, Zhu H, Cai YJ, Lu W and Ding J: Chk1 and Chk2 are differentially involved in homologous recombination repair and cell cycle arrest in response to DNA double-strand breaks induced by camptothecins. Mol Cancer Ther. 7:1440–1449. 2008. View Article : Google Scholar : PubMed/NCBI

48 

van't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 415:530–536. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Hernando E, Nahle Z, Juan G, Diaz-Rodriguez E, Alaminos M, Hemann M, Michel L, Mittal V, Gerald W, Benezra R, et al: Rb inactivation promotes genomic instability by uncoupling cell cycle progression from mitotic control. Nature. 430:797–802. 2004. View Article : Google Scholar : PubMed/NCBI

50 

Chen X, Yan CC, Zhang X and You ZH: Long non-coding RNAs and complex diseases: From experimental results to computational models. Brief Bioinform. 18:558–576. 2017.PubMed/NCBI

51 

Chen X, Xie D, Zhao Q and You ZH: MicroRNAs and complex diseases: From experimental results to computational models. Brief Bioinform. 20:515–539. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Chen X, Wang L, Qu J, Guan NN and Li JQ: Predicting miRNA-disease association based on inductive matrix completion. Bioinformatics. 34:4256–4265. 2018.PubMed/NCBI

53 

Chen X, Xie D, Wang L, Zhao Q, You ZH and Liu H: BNPMDA: Bipartite network projection for MiRNA-disease association prediction. Bioinformatics. 34:3178–3186. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Chen X, Yin J, Qu J and Huang L: MDHGI: Matrix decomposition and heterogeneous graph inference for miRNA-disease association prediction. PLoS Computat Biol. 14:e10064182018. View Article : Google Scholar

55 

Chen X and Huang L: LRSSLMDA: Laplacian regularized sparse subspace learning for MiRNA-disease association prediction. PLoS Comput Biol. 13:e10059122017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

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
Yu C, Chen F, Jiang J, Zhang H and Zhou M: Screening key genes and signaling pathways in colorectal cancer by integrated bioinformatics analysis. Mol Med Rep 20: 1259-1269, 2019
APA
Yu, C., Chen, F., Jiang, J., Zhang, H., & Zhou, M. (2019). Screening key genes and signaling pathways in colorectal cancer by integrated bioinformatics analysis. Molecular Medicine Reports, 20, 1259-1269. https://doi.org/10.3892/mmr.2019.10336
MLA
Yu, C., Chen, F., Jiang, J., Zhang, H., Zhou, M."Screening key genes and signaling pathways in colorectal cancer by integrated bioinformatics analysis". Molecular Medicine Reports 20.2 (2019): 1259-1269.
Chicago
Yu, C., Chen, F., Jiang, J., Zhang, H., Zhou, M."Screening key genes and signaling pathways in colorectal cancer by integrated bioinformatics analysis". Molecular Medicine Reports 20, no. 2 (2019): 1259-1269. https://doi.org/10.3892/mmr.2019.10336