Open Access

Identification of key candidate genes for colorectal cancer by bioinformatics analysis

  • Authors:
    • Zhihua Chen
    • Yilin Lin
    • Ji Gao
    • Suyong Lin
    • Yan Zheng
    • Yisu Liu
    • Shao Qin Chen
  • View Affiliations

  • Published online on: October 17, 2019     https://doi.org/10.3892/ol.2019.10996
  • Pages: 6583-6593
  • Copyright: © Chen 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

Colorectal cancer (CRC) is one of the most common cancers of the digestive tract. Although numerous studies have been conducted to elucidate the cause of CRC, the exact mechanism of CRC development remains to be determined. To identify candidate genes that may be involved in CRC development and progression, the microarray datasets GSE41657, GSE77953 and GSE113513 were downloaded from the Gene Expression Omnibus database. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes were used for functional enrichment analysis of differentially expressed genes (DEGs). A protein‑protein interaction network was constructed, and the hub genes were subjected to module analysis and identification using Search Tool for the Retrieval of Interacting Genes/Proteins and Cytoscape. A total of 142 DEGs were identified, with enriched functions and pathways in the ‘cell cycle’, ‘cell proliferation’, ‘the mitotic cell cycle’ and ‘one‑carbon metabolic process’. In addition, 10 hub genes were identified, and functional analysis indicated that these genes are mainly enriched in ‘cell division’, ‘cell cycle’ and functions associated with nucleotide binding processes. Survival analysis demonstrated that DNA topoisomerase II α, cyclin‑dependent kinase 1 and CDC28 protein kinase regulatory subunit 2 may be involved in cancer invasion or recurrence. The DEGs identified in the present study may help explain the molecular mechanisms of CRC development and progression.

Introduction

Colorectal cancer (CRC) ranks third (13.5%) and second (9.5%) among the incidence of malignancies worldwide in male and female patients, respectively, and is a serious hazard to human health (1). Previous studies have demonstrated that the molecular pathogenesis of CRC is mostly caused by genetic mutations (2,3). Numerous studies over the past two decades have reported that genetic mutations are associated with the prognosis and treatment of CRC, and targeted therapies have been developed (47). The progression of CRC is usually accompanied by the activation of the KRAS and BRAF genes and the inhibition of the p53 tumour suppressor gene expression; mutations in these genes are associated with changes in the number and structure of chromosomes (810). However, >15% of sporadic CRCs occur through completely different molecular pathogenesis. For example, serrated precancerous lesions usually manifest as a result of the methylation of the CpG locus and mutation of the gene (11).

The prognosis of CRC is poor due to a lack of effective diagnostic methods at an early stage. Therefore, an effective solution can only be provided for subsequent diagnosis and treatment by better understanding the gene expression of CRC during its occurrence and development and identifying the genes that may be involved in the occurrence and progression of cancer.

With the rapid development of science and technology, microarray technology has become increasingly accurate and has been widely used to explore changes in animal and plant gene expression (1214). Microarray technology aids in the discovery of changes in gene expression during cancer development and progression (1517). However, it can be difficult to obtain reliable results with single microarray analysis. The present study aimed to identify genetic changes in CRC in three mRNA microarray datasets from the Gene Expression Omnibus (GEO) database by obtaining differentially expressed genes (DEGs) between CRC and normal tissues. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for functional enrichment analysis, and a protein-protein interaction (PPI) network was used to analyse the associations between the DEGs. A total of 142 DEGs and 10 hub genes were identified, which may be candidate biomarkers of CRC.

Materials and methods

Microarray data

GEO (http://www.ncbi.nlm.nih.gov/geo) (18) is a public functional genomic database. Three gene expression datasets (GSE41657, GSE77953, and GSE113513; analysed on Agilent-014850 Whole Human Genome Microarray 4×44K G4112F, Affymetrix Human Genome U133A Array, and Affymetrix Human Gene Expression Array platforms, respectively) were downloaded from GEO. The probes were converted to the corresponding gene symbols based on the annotation information on the platform. The GSE41657 dataset contained 25 CRC and 12 adjacent normal tissue samples. The GSE77953 dataset contained 28 CRC and 13 non-tumour samples. The GSE113513 dataset contained 14 CRC and 14 non-tumour samples.

Identification of DEGs

DEGs between CRC and non-tumour samples were screened using GEO2R (http://www.ncbi.nlm.nih.gov/geo/geo2r). GEO2R is an interactive web tool that compares two or more GEO datasets to identify DEGs across experimental conditions. Adjusted P-values (adj. P) and Benjamin and Hochberg false discovery rates were applied to balance between statistically significant findings and false-positive limits. Probe sets without corresponding gene symbols were removed, and genes with more than one probe set were averaged. Log [fold change(FC)]>1 and adj. P-value <0.01 were considered to indicate a statistically significant difference.

KEGG and GO enrichment analysis of DEGs

The Database for Annotation, Visualization and Integrated Discovery (DAVID; version 6.7; http://david.ncifcrf.gov) (19) is an online bioinformatics database that integrates biological data and analysis tools and provides gene annotation information and protein data. KEGG is a database resource for understanding advanced functions and biological systems from large-scale molecular data generated by high-throughput experimental techniques (20). GO is a major bioinformatics tool for annotating genes and analysing their biological processes (BPs), molecular functions (MFs) and cellular components (CCs) (21). To analyse the function of the DEGs, analysis was performed using DAVID. P<0.05 was considered to indicate a statistically significant difference. The results based on the top ten BPs, MFs, CCs and KEGG were visualized.

PPI network construction and module analysis

A PPI network was predicted using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version 10.0; http://string-db.org) database (22). The PPI network of the DEGs was constructed, and interactions with a combined score >0.4 were considered statistically significant. Cytoscape (version 3.4.0) is an open source bioinformatics platform for visualizing molecular interaction networks (23). Cytoscape plug-in Molecular Complex Detection (MCODE; version 1.4.2) provides topology-based clustering for a network (24). Cytoscape was used to draw a PPI network, and MCODE was used to identify the most important modules in the network. The selection criteria were as follows: MCODE score >5, degree cut-off =2, node score cut-off =0.2, maximum depth =100, and k score =2. Subsequently, KEGG and GO analysis of the genes in this module was performed using DAVID.

Hub genes selection and analysis

The selection criterion for the hub genes was degree of connectivity ≥10. The functions of the genes were identified using the GeneCards online analysis tool (https://www.genecards.org). The BP analysis and visualization of the hub genes were performed using the cBioPortal (http://www.cbioportal.org) online platform (25). The functions of the hub genes were identified using GO and KEGG analysis. Hierarchical clustering of the hub genes was constructed using the University of California Santa Cruz Cancer Genomics Browser (http://genome-cancer.ucsc.edu) (26). The overall survival analysis was performed using Kaplan-Meier curves in cBioPortal. The log-rank test was used to for statistical analysis. The expression profiles of DNA topoisomerase IIα (TOP2A) and phosphoribosylaminoimidazole carboxylase (PAICS) were analysed using the Oncomine database (http://www.oncomine.com). The expression levels of genes in normal tissues and CRC tissues were also analysed using the Oncomine database (27).

Results

Determination of DEGs in CRC

DEGs were identified by standardized microarray results (GSE41657, GSE77953 and GSE113513). A total of 142 genes overlapped in the three datasets, as demonstrated in the Venn diagram (Fig. 1A).

PPI network and module analysis

A PPI network of the 142 identified DEGs was constructed (Fig. 1B). A significantly enriched module was obtained using Cytoscape software (Fig. 1C).

GO and KEGG enrichment analysis of the DEGs

DAVID was used for the functional enrichment analysis of DEGs. The results indicated that the DEGs were mainly enriched in ‘cell proliferation’, ‘G2/M transition of mitotic cell cycle’ and ‘one-carbon metabolism’ BPs (Fig. 2A); ‘cytoplasm’, ‘cytosol’ and ‘extracellular exosome’ cellular components (CCs) (Fig. 2B); and ‘protein binding’ and ‘ATP binding’ molecular functions (MFs) (Fig. 2C). KEGG pathway analysis revealed strong enrichment in ‘biosynthesis of antibiotics’, ‘purine metabolism’, ‘pancreatic secretion’ and ‘mineral absorption’ (Fig. 2D).

The functional results of the genes in the most significant module indicated that these genes were mainly enriched in processes associated with cell cycle progression, ATP binding, nucleotide binding, the p53 signalling pathway and oocyte meiosis (Table I).

Table I.

GO and KEGG pathway enrichment analysis of DEGs in the most significant module.

Table I.

GO and KEGG pathway enrichment analysis of DEGs in the most significant module.

TermDescriptionNo. of genesP-value
GO:0007049Cell cycle11 8.42×10−10
GO:0000279M phase9 6.24×10−10
GO:0022403Cell cycle phase9 3.84×10−9
GO:0022402Cell cycle process9 4.38×10−8
GO:0043228 Non-membrane-bounded organelle11 1.83×10−5
GO:0015630Microtubule cytoskeleton9 1.17×10−8
GO:0044430Cytoskeletal part9 8.45×10−7
GO:0005524ATP binding6 2.91×10−3
GO:0032559Adenyl ribonucleotide binding6 3.10×10−3
GO:0030554Adenyl nucleotide binding6 3.91×10−3
hsa04115p53 signalling pathway3 1.72×10−3
hsa04114Oocyte meiosis3 4.44×10−3
hsa04110Cell cycle3 5.71×10−3

[i] GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Hub genes screening and analysis

Genes with a degree of connectivity ≥10 were identified as the hub genes. The names, abbreviations and functions of these genes are presented in Table II. The cBioPortal online platform was used to construct a hub gene network (Fig. 3A), and functional enrichment analysis of DEGs was performed using DAVID (Table III). The heat map constructed using the UCSC Cancer Genomics Browser indicated that the hub genes may be used to distinguish CRC tissue samples from normal intestinal cancer tissue samples (Fig. 3B). Overall and progression-free survival based on alterations in the hub genes was subsequently analysed using Kaplan-Meier curves. Alterations in TOP2A, cyclin-dependent kinase 1 (CDK1) and CDC28 protein kinase regulatory subunit 2 (CKS2) in patients with CRC were associated with a poor overall survival rate (Fig. 4A). However, patients with alterations in TOP2A and CKS2 did not exhibit significant differences in progression-free survival (Fig. 4B).

Table II.

Functional roles of the 10 hub genes with degree of connectivity ≥10.

Table II.

Functional roles of the 10 hub genes with degree of connectivity ≥10.

No.Gene symbolFull nameFunction
1TOP2ADNA Topoisomerase II αTarget for anticancer agents; mutations are associated with drug resistance
2PAICS Phosphoribosylaminoimidazole carboxylaseInvolved in de novo synthesis of purine nucleotides
3CDK1Cyclin-dependent kinase 1Regulates cell cycle progression, apoptosis and carcinogenesis of tumour cells
4CKS2CDC28 protein kinase regulatory subunit 2Binds to the catalytic subunit of cyclin-dependent kinases; essential for their biological function
5CKAP2Cytoskeleton associated protein 2Possesses microtubule stabilizing properties; involved in regulating aneuploidy, cell cycle and cell death in a p53/TP53-dependent manner
6CEP55Centrosomal protein 55Promotes the proliferation of lung, breast and thyroid cancer
7VEGFAVascular endothelial growth factor AInduces endothelial cell proliferation, promotes cell migration, inhibits apoptosis and induces blood vessel permeabilization
8PHLPP2PH domain and leucine rich repeat protein phosphatase 2Regulates Akt and PKC signalling.
9RRM2Ribonucleotide reductase M2 polypeptideCatalyses the biosynthesis of deoxyribonucleotides; inhibits Wnt signalling
10NEK2NIMA (never in mitosis gene a)-related kinase 2Involved in the regulation of mitosis

Table III.

GO and KEGG pathway enrichment analysis of hub genes.

Table III.

GO and KEGG pathway enrichment analysis of hub genes.

TermDescriptionNo. of genesP-value
GO:0007049Cell cycle5 6.24×10−4
GO:0051301Cell division4 5.30×10−4
GO:0000279M phase4 7.29×10−4
GO:0015630Microtubule cytoskeleton5 1.06×10−4
GO:0044430Cytoskeletal part5 8.92×10−4
GO:0005856Cytoskeleton5 3.63×10−3
GO:0005524ATP binding4 3.62×10−2
GO:0032559Adenyl ribonucleotide binding4 3.75×10−2
GO:0030554Adenyl nucleotide binding4 4.29×10−2
hsa04115p53 signalling pathway2 3.96×10−2

[i] GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Among these genes, TOP2A exhibited a node degree of 34, and PAICS exhibited the highest degree of connectivity among the hub genes (34 and 26, respectively). These two genes may serve important roles in the occurrence or development of CRC. Based on the survival analysis, alterations in PAICS exhibited no statistically significant differences in overall and progression-free survival in patients with CRC (overall survival P=0.986; progression-free survival P=0.918; Fig. 4). Overall survival rates were significantly different for patients with TOP2A alterations, but progression-free survival was not significantly different (overall survival P=0.002; progression-free survival P=0.703; Fig. 4). The expression profiles of TOP2A and PAICS in human tissues were analysed using the Oncomine database; TOP2A and PAICS mRNA was upregulated in various cancer tissues compared with normal tissues (Fig. 5A and B). Further analysis revealed significant increases in the expression of TOP2A and PAICS in CRC (Fig. 5C and D).

Discussion

CRC is a common malignant tumour of the digestive tract, and its mortality rate (9.2%) ranks second among all types of cancer (1). The main causes of CRC include dietary and environmental factors, as well as genetic mutations (3). The simultaneous methylation of the CpG site and mutation of the BRAF gene are also important factors in the development and progression of CRC (11). Failure to detect CRC early may be one of the reasons for poor prognosis in patients. Therefore, there is an urgent need for efficient diagnostic and therapeutic methods.

In the present study, DEGs between CRC and non-cancerous tissues were obtained from three mRNA microarray datasets. The functions of the DEGs were identified by GO and KEGG enrichment analysis. The results demonstrated that the DEGs were mainly enriched in the cell cycle, proliferation, mitotic cell cycle and carbon metabolism. Previous studies have reported that dysregulation of the cell cycle and the mitotic cell cycle serves an important role in the development and progression of CRC (2831). In a study by Chamberlain et al (32), functional B vitamin was used to assess the association of carbon metabolism with CRC, and a similar association between total B vitamin status and CRC risk was identified. Ducker (33) et al also revealed that the single-carbon metabolism of the cytosol can support tumourigenesis. These findings are consistent with the results of the present study. GO cluster and KEGG analysis in the present study also revealed that the changes in the most important module were mainly enriched in processes associated with cell cycle progression, ATP and nucleotide binding and in KEGG pathways associated with the cell cycle, progesterone-mediated oocyte maturation and oocyte cell meiosis.

A total of 10 DEGs were selected as the hub genes. Among them, TOP2A and PAICS were the top two nodes with the highest degree of connectivity. TOP2A is involved in DNA replication, transcription and chromosome segregation, and is essential for tumorigenesis and cancer development (34). TOP2A has been demonstrated to be associated with chemoresistance and tumour recurrence and has been recognized as a target for anticancer drugs (35,36). In addition, TOP2A is highly expressed in lung, colon and breast cancer, involved in the inhibition of apoptosis, proliferation and chemoresistance of CRC and may be considered an important biomarker for the diagnosis, prognosis and treatment of tumours (3739). In the present study, the PPI network indicated that TOP2A interacted directly with CDK1, ribonucleoside-diphosphate reductase subunit M2 (RRM2) and CKS2, indicating a key role for TOP2A in CRC. PAICS is involved in purine nucleotide synthesis; a previous study has demonstrated that high expression of PAICS in lung and prostate cancer is associated with an altered metabolic state of cells, apoptosis inhibition in cancer cells and enhanced cancer cell invasion (40). In addition, PAICS has been reported to promote tumourigenesis and progression in breast and bladder cancer (41,42). The results of the analysis in the Oncomine database in the present study demonstrated that TOP2A and PAICS expression was significantly upregulated in CRC compared with that in normal tissues; however, to the best of our knowledge, there are currently no studies focusing on the association between PAICS expression and CRC. The association between TOP2A and PAICS and overall and disease-free survival was also analysed; changes in TOP2A were significantly associated with overall survival but were independent of disease-free survival. Changes in PAICS exhibited a reduction in overall and disease-free survival; however, these observations were not statistically significant. These results may require further research for verification.

CDK1 is a non-redundant cyclin-dependent kinase that serves an important role in mitosis (43,44). Perturbations in chromosomal stability and aspects of S phase and G2/M control mediated by CDK2 and CDK1 are pivotal tumorigenic events (45). A previous study has demonstrated that CDK1 is required for the survival of cells overexpressing MYC, and CDK1 has a therapeutic effect in the treatment of human malignancies that overexpress MYC (46). The combination of CKS2 and CDK is essential for promoting cancer cell metastasis in diseases such as colon cancer (47,48). Vascular endothelial growth factor A (VEGFA) is a class of cytokines called antigen growth factors that stimulate the formation of new blood vessels; tumour angiogenesis is primarily dependent on VEGFA-driven responses (49). Anti-VEGF-A treatment can reduce Treg proliferation in CRC patients, and VEGFA inhibitors have been used to treat CRC (50,51). In the present study revealed, the expression levels of CDK11, CSK2 and VEGFA in CRC were analysed; the results revealed high expression of these genes in CRC, which was consistent with previous studies. In addition, high expression levels of CDK1 and CKS2 were associated with poor survival.

CKAP2 is a spindle-associated protein that is degraded during mitosis (52,53). Upregulation of CEP55 promotes the metastasis of several cancers, such as lung adenocarcinoma, breast and anaplastic thyroid cancer (5456). RRM2, also known as the ribonucleotide reductase small subunit, promotes cyclin F degradation to maintain a balanced dNTP pool and genomic stability (57). PHLPP2 is a protein phosphatase involved in the regulation of Akt and PKC signalling (58,59). Previous studies have reported that microRNA-224 promotes the proliferation and tumour growth of human CRC cells by inhibiting PHLPP1 and PHLPP2 (60) and acts as a tumour suppressor for lung and breast cancer (61,62). The results of the present study also demonstrated that it is downregulated in CRC tissues. NEK2 is a serine/threonine kinase that is a key regulator of mitosis in cells (63). Upregulation of NEK2 is associated with the development of cancer, such as breast (64), ovarian (65) and colorectal (66,67) cancer. Overexpression of NEK2 promotes the invasion and metastasis of tumour cells and is considered a potential biomarker in nasopharyngeal carcinoma and liver cancer (68,69). The results of the present study indicated that the aforementioned hub genes may be distinguishing factors between CRC and non-tumour tissue samples and candidate tumour biomarkers. In addition, alterations in TOP2A, CDK1 and CKS2 are associated with poor survival, suggesting that these genes may serve an important role in the development, progression or recurrence of cancer.

In summary, the aim of the present study was to identify DEGs that may be involved in the development or progression of CRC. The results demonstrated that TOP2A and CDK1 may be involved in the survival prognosis of patients with CRC. The present study also revealed that these genes may be biomarkers for the diagnosis of CRC. However, the present study had limitations, and other databases such as The Cancer Genome Atlas, as well as in vivo and in vitro experiments may be needed to clarify the biological functions of these genes in CRC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Fujian Provincial Key Medical Specialist Construction Project (grant no. 2016-SLCZD), the Training Project of Young Talents in the Health System of Fujian Province (grant no. 2016-ZQN-45) and the Fujian Medical University Start up Fund for Scientific Research (grant no. 2017-XQ1061).

Availability of data and materials

The datasets used and analysed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

SQC and ZHC designed the study and obtained funding YLL drafted the manuscript and supervised the study. YLL participates in the design of the study, obtaining data and writing manuscripts. YZ, YSL, JG and SYL collected and analysed the data. All authors read and approved the final manuscript.

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 

Van Cutsem E, Lenz HJ, Kohne CH, Heinemann V, Tejpar S, Melezinek I, Beier F, Stroh C, Rougier P, van Krieken JH and Ciardiello F: Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. 33:692–700. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Tong K, Pellón-Cárdenas O, Sirihorachai VR, Warder BN, Kothari OA, Perekatt AO, Fokas EE, Fullem RL, Zhou A, Thackray JK, et al: Degree of tissue differentiation dictates susceptibility to BRAF-driven colorectal cancer. Cell Rep. 21:3833–3845. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Sanz-Garcia E, Argiles G, Elez E and Tabernero J: BRAF mutant colorectal cancer: Prognosis, treatment, and new perspectives. Ann Oncol. 28:2648–2657. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Corcoran RB, André T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, et al: Combined BRAF, EGFR, and MEK inhibition in patients with BRAF(V600E)-mutant colorectal cancer. Cancer Discov. 8:428–443. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Hamzehzadeh L, Khadangi F, Ghayoor Karimiani E, Pasdar A and Kerachian MA: Common KRAS and NRAS gene mutations in sporadic colorectal cancer in Northeastern Iranian patients. Curr Probl Cancer. 42:572–581. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Reggiani Bonetti L, Barresi V, Bettelli S, Caprera C, Manfredini S and Maiorana A: Analysis of KRAS, NRAS, PIK3CA, and BRAF mutational profile in poorly differentiated clusters of KRAS-mutated colon cancer. Hum Pathol. 62:91–98. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, et al: Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 486:532–536. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Cooks T, Pateras IS, Jenkins LM, Patel KM, Robles AI, Morris J, Forshew T, Appella E, Gorgoulis VG and Harris CC: Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun. 9:7712018. View Article : Google Scholar : PubMed/NCBI

10 

Fu X, Huang Y, Fan X, Deng Y, Liu H, Zou H, Wu P, Chen Z, Huang J, Wang J, et al: Demographic trends and KRAS/BRAFV600E mutations in colorectal cancer patients of South China: A single-site report. Int J Cancer. 144:2109–2117. 2019.PubMed/NCBI

11 

O'Brien MJ, Yang S, Mack C, Xu H, Huang CS, Mulcahy E, Amorosino M and Farraye FA: Comparison of microsatellite instability, CpG island methylation phenotype, BRAF and KRAS status in serrated polyps and traditional adenomas indicates separate pathways to distinct colorectal carcinoma end points. Am J Surg Pathol. 30:1491–1501. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Saito M, Momma T and Kono K: Targeted therapy according to next generation sequencing-based panel sequencing. Fukushima J Med Sci. 64:9–14. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Deshiere A, Berthet N, Lecouturier F, Gaudaire D and Hans A: Molecular characterization of Equine Infectious Anemia Viruses using targeted sequence enrichment and next generation sequencing. Virology. 537:121–129. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Meng H, Wang L, You H, Huang C and Li J: Circular RNA expression profile of liver tissues in an EtOH-induced mouse model of alcoholic hepatitis. Eur J Pharmacol. 862:1726422019. View Article : Google Scholar : PubMed/NCBI

15 

Harada K, Okamoto W, Mimaki S, Kawamoto Y, Bando H, Yamashita R, Yuki S, Yoshino T, Komatsu Y, Ohtsu A, et al: Comparative sequence analysis of patient-matched primary colorectal cancer, metastatic, and recurrent metastatic tumors after adjuvant FOLFOX chemotherapy. BMC Cancer. 19:2552019. View Article : Google Scholar : PubMed/NCBI

16 

Hu Y, He C, Liu JP, Li NS, Peng C, Yang-Ou YB, Yang XY, Lu NH and Zhu Y: Analysis of key genes and signaling pathways involved in Helicobacter pylori-associated gastric cancer based on the cancer genome atlas database and RNA sequencing data. Helicobacter. 23:e125302018. View Article : Google Scholar : PubMed/NCBI

17 

Nakagawa H and Fujita M: Whole genome sequencing analysis for cancer genomics and precision medicine. Cancer Sci. 109:513–522. 2018. View Article : Google Scholar : PubMed/NCBI

18 

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:D991–D995. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Huang DW, Sherman BT, Tan Q, Collins JR, Alvord WG, Roayaei J, Stephens R, Baseler MW, Lane HC and Lempicki RA: The DAVID gene functional classification tool: A novel biological module-centric algorithm to functionally analyze large gene lists. Genome Biol. 8:R1832007. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

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

22 

Franceschini A, Szklarczyk D, Frankild S, Kuhn M, Simonovic M, Roth A, Lin J, Minguez P, Bork P, von Mering C and Jensen LJ: STRING v9.1: Protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41:D808–D815. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: New features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Bandettini WP, Kellman P, Mancini C, Booker OJ, Vasu S, Leung SW, Wilson JR, Shanbhag SM, Chen MY and Arai AE: MultiContrast Delayed Enhancement (MCODE) improves detection of subendocardial myocardial infarction by late gadolinium enhancement cardiovascular magnetic resonance: A clinical validation study. J Cardiovasc Magn Reson. 14:832012. View Article : Google Scholar : PubMed/NCBI

25 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Haeussler M, Zweig AS, Tyner C, Speir ML, Rosenbloom KR, Raney BJ, Lee CM, Lee BT, Hinrichs AS, Gonzalez JN, et al: The UCSC genome browser database: 2019 update. Nucleic Acids Res. 47:D853–D858. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, Barrette T, Pandey A and Chinnaiyan AM: ONCOMINE: A cancer microarray database and integrated data-mining platform. Neoplasia. 6:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Wang ZZ, Yang J, Jiang BH, Di JB, Gao P, Peng L and Su XQ: KIF14 promotes cell proliferation via activation of Akt and is directly targeted by miR-200c in colorectal cancer. Int J Oncol. 53:1939–1952. 2018.PubMed/NCBI

29 

Wu J, Yi J, Wu Y, Chen X, Zeng J, Wu J and Peng W: 3, 3′-dimethylquercetin inhibits the proliferation of human colon cancer RKO cells through Inducing G2/M cell cycle arrest and apoptosis. Anticancer Agents Med Chem. 19:402–409. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Cheng J, Dwyer M, Okolotowicz KJ, Mercola M and Cashman JR: A novel inhibitor targets both wnt signaling and ATM/p53 in colorectal cancer. Cancer Res. 78:5072–5083. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Li J, Liu YY, Yang XF, Shen DF, Sun HZ, Huang KQ and Zheng HC: Effects and mechanism of STAT3 silencing on the growth and apoptosis of colorectal cancer cells. Oncol Lett. 16:5575–5582. 2018.PubMed/NCBI

32 

Chamberlain JA, Dugué PA, Bassett JK, Hodge AM, Brinkman MT, Joo JE, Jung CH, Makalic E, Schmidt DF, Hopper JL, et al: Dietary intake of one-carbon metabolism nutrients and DNA methylation in peripheral blood. Am J Clin Nutr. 108:611–621. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Ducker GS, Chen L, Morscher RJ, Ghergurovich JM, Esposito M, Teng X, Kang Y and Rabinowitz JD: Reversal of cytosolic one-carbon flux compensates for loss of the mitochondrial folate pathway. Cell Metab. 23:1140–1153. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Nitiss JL: DNA topoisomerase II and its growing repertoire of biological functions. Nat Rev Cancer. 9:327–337. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Pommier Y, Leo E, Zhang H and Marchand C: DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 17:421–433. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Deweese JE and Osheroff N: The DNA cleavage reaction of topoisomerase II: Wolf in sheep's clothing. Nucleic Acids Res. 37:738–748. 2009. View Article : Google Scholar : PubMed/NCBI

37 

McLeod HL, Douglas F, Oates M, Symonds RP, Prakash D, van der Zee AG, Kaye SB, Brown R and Keith WN: Topoisomerase I and II activity in human breast, cervix, lung and colon cancer. Int J Cancer. 59:607–611. 1994. View Article : Google Scholar : PubMed/NCBI

38 

Shibao K, Takano H, Nakayama Y, Okazaki K, Nagata N, Izumi H, Uchiumi T, Kuwano M, Kohno K and Itoh H: Enhanced coexpression of YB-1 and DNA topoisomerase II alpha genes in human colorectal carcinomas. Int J Cancer. 83:732–737. 1999. View Article : Google Scholar : PubMed/NCBI

39 

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

40 

Goswami MT, Chen G, Chakravarthi BV, Pathi SS, Anand SK, Carskadon SL, Giordano TJ, Chinnaiyan AM, Thomas DG, Palanisamy N, et al: Role and regulation of coordinately expressed de novo purine biosynthetic enzymes PPAT and PAICS in lung cancer. Oncotarget. 6:23445–23461. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Meng M, Chen Y, Jia J, Li L and Yang S: Knockdown of PAICS inhibits malignant proliferation of human breast cancer cell lines. Biol Res. 51:242018. View Article : Google Scholar : PubMed/NCBI

42 

Chakravarthi BVSK, Rodriguez Pena MDC, Agarwal S, Chandrashekar DS, Hodigere Balasubramanya SA, Jabboure FJ, Matoso A, Bivalacqua TJ, Rezaei K, Chaux A, et al: A role for de novo purine metabolic enzyme PAICS in bladder cancer progression. Neoplasia. 20:894–904. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Santamaría D, Barrière C, Cerqueira A, Hunt S, Tardy C, Newton K, Cáceres JF, Dubus P, Malumbres M and Barbacid M: Cdk1 is sufficient to drive the mammalian cell cycle. Nature. 448:811–815. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Brown NR, Korolchuk S, Martin MP, Stanley WA, Moukhametzianov R, Noble MEM and Endicott JA: CDK1 structures reveal conserved and unique features of the essential cell cycle CDK. Nat Commun. 6:67692015. View Article : Google Scholar : PubMed/NCBI

45 

Asghar U, Witkiewicz AK, Turner NC and Knudsen ES: The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov. 14:130–146. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Goga A, Yang D, Tward AD, Morgan DO and Bishop JM: Inhibition of CDK1 as a potential therapy for tumors over-expressing MYC. Nat Med. 13:820–827. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Martinsson-Ahlzén HS, Liberal V, Grünenfelder B, Chaves SR, Spruck CH and Reed SI: Cyclin-dependent kinase-associated proteins Cks1 and Cks2 are essential during early embryogenesis and for cell cycle progression in somatic cells. Mol Cell Biol. 28:5698–5709. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Li M, Lin YM, Hasegawa S, Shimokawa T, Murata K, Kameyama M, Ishikawa O, Katagiri T, Tsunoda T, Nakamura Y and Furukawa Y: Genes associated with liver metastasis of colon cancer, identified by genome-wide cDNA microarray. Int J Oncol. 24:305–312. 2004.PubMed/NCBI

49 

Claesson-Welsh L and Welsh M: VEGFA and tumour angiogenesis. J Intern Med. 273:114–127. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Ferrara N and Adamis AP: Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov. 15:385–403. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Terme M, Pernot S, Marcheteau E, Sandoval F, Benhamouda N, Colussi O, Dubreuil O, Carpentier AF, Tartour E and Taieb J: VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 73:539–549. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Seki A and Fang G: CKAP2 is a spindle-associated protein degraded by APC/C-Cdh1 during mitotic exit. J Biol Chem. 282:15103–15113. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Tsuchihara K, Lapin V, Bakal C, Okada H, Brown L, Hirota-Tsuchihara M, Zaugg K, Ho A, Itie-Youten A, Harris-Brandts M, et al: Ckap2 regulates aneuploidy, cell cycling, and cell death in a p53-dependent manner. Cancer Res. 65:6685–6691. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Weinberger P, Ponny SR, Xu H, Bai S, Smallridge R, Copland J and Sharma A: Cell cycle M-phase genes are highly upregulated in anaplastic thyroid carcinoma. Thyroid. 27:236–252. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Wang Y, Jin T, Dai X and Xu J: Lentivirus-mediated knockdown of CEP55 suppresses cell proliferation of breast cancer cells. Biosci Trends. 10:67–73. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Jiang W, Wang Z, Chen G and Jia Y: Prognostic significance of centrosomal protein 55 in stage I pulmonary adenocarcinoma after radical resection. Thorac Cancer. 7:316–322. 2016. View Article : Google Scholar : PubMed/NCBI

57 

D'Angiolella V, Donato V, Forrester FM, Jeong YT, Pellacani C, Kudo Y, Saraf A, Florens L, Washburn MP and Pagano M: Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair. Cell. 149:1023–1034. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Brognard J and Newton AC: PHLiPPing the switch on Akt and protein kinase C signaling. Trends Endocrinol Metab. 19:223–230. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Brognard J, Sierecki E, Gao T and Newton AC: PHLPP and a second isoform, PHLPP2, differentially attenuate the amplitude of Akt signaling by regulating distinct Akt isoforms. Mol Cell. 25:917–931. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Liao WT, Li TT, Wang ZG, Wang SY, He MR, Ye YP, Qi L, Cui YM, Wu P, Jiao HL, et al: microRNA-224 promotes cell proliferation and tumor growth in human colorectal cancer by repressing PHLPP1 and PHLPP2. Clin Cancer Res. 19:4662–4672. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Cai J, Fang L, Huang Y, Li R, Yuan J, Yang Y, Zhu X, Chen B, Wu J and Li M: miR-205 targets PTEN and PHLPP2 to augment AKT signaling and drive malignant phenotypes in non-small cell lung cancer. Cancer Res. 73:5402–5415. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Santarpia L, Qi Y, Stemke-Hale K, Wang B, Young EJ, Booser DJ, Holmes FA, O'Shaughnessy J, Hellerstedt B, Pippen J, et al: Mutation profiling identifies numerous rare drug targets and distinct mutation patterns in different clinical subtypes of breast cancers. Breast Cancer Res Treat. 134:333–343. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Hu CM, Zhu J, Guo XE, Chen W, Qiu XL, Ngo B, Chien R, Wang YV, Tsai CY, Wu G, et al: Novel small molecules disrupting Hec1/Nek2 interaction ablate tumor progression by triggering Nek2 degradation through a death-trap mechanism. Oncogene. 34:1220–1230. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Hayward DG, Clarke RB, Faragher AJ, Pillai MR, Hagan IM and Fry AM: The centrosomal kinase Nek2 displays elevated levels of protein expression in human breast cancer. Cancer Res. 64:7370–7376. 2004. View Article : Google Scholar : PubMed/NCBI

65 

Hawkins SM, Loomans HA, Wan YW, Ghosh-Choudhury T, Coffey D, Xiao W, Liu Z, Sangi-Haghpeykar H and Anderson ML: Expression and functional pathway analysis of nuclear receptor NR2F2 in ovarian cancer. J Clin Endocrinol Metab. 98:E1152–E1162. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Neal CP, Fry AM, Moreman C, McGregor A, Garcea G, Berry DP and Manson MM: Overexpression of the Nek2 kinase in colorectal cancer correlates with beta-catenin relocalization and shortened cancer-specific survival. J Surg Oncol. 110:828–838. 2014. View Article : Google Scholar : PubMed/NCBI

67 

Lu L, Zhai X and Yuan R: Clinical significance and prognostic value of Nek2 protein expression in colon cancer. Int J Clin Exp Pathol. 8:15467–15473. 2015.PubMed/NCBI

68 

Xu H, Zeng L, Guan Y, Feng X, Zhu Y, Lu Y, Shi C, Chen S, Xia J, Guo J, et al: High NEK2 confers to poor prognosis and contributes to cisplatin-based chemotherapy resistance in nasopharyngeal carcinoma. J Cell Biochem. 120:3547–3558. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Zhang Y, Wang W, Wang Y, Huang X, Zhang Z, Chen B, Xie W, Li S, Shen S and Peng B: NEK2 promotes hepatocellular carcinoma migration and invasion through modulation of the epithelial-mesenchymal transition. Oncol Rep. 39:1023–1033. 2018.PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Z, Lin Y, Gao J, Lin S, Zheng Y, Liu Y and Chen SQ: Identification of key candidate genes for colorectal cancer by bioinformatics analysis. Oncol Lett 18: 6583-6593, 2019
APA
Chen, Z., Lin, Y., Gao, J., Lin, S., Zheng, Y., Liu, Y., & Chen, S.Q. (2019). Identification of key candidate genes for colorectal cancer by bioinformatics analysis. Oncology Letters, 18, 6583-6593. https://doi.org/10.3892/ol.2019.10996
MLA
Chen, Z., Lin, Y., Gao, J., Lin, S., Zheng, Y., Liu, Y., Chen, S. Q."Identification of key candidate genes for colorectal cancer by bioinformatics analysis". Oncology Letters 18.6 (2019): 6583-6593.
Chicago
Chen, Z., Lin, Y., Gao, J., Lin, S., Zheng, Y., Liu, Y., Chen, S. Q."Identification of key candidate genes for colorectal cancer by bioinformatics analysis". Oncology Letters 18, no. 6 (2019): 6583-6593. https://doi.org/10.3892/ol.2019.10996