Open Access

Identification of key genes and pathways for esophageal squamous cell carcinoma by bioinformatics analysis

  • Authors:
    • Xiaohua Chen
    • Sina Cai
    • Baoxia Li
    • Xiaona Zhang
    • Wenhui Li
    • Henglun Linag
    • Xiaolong Cao
  • View Affiliations

  • Published online on: June 15, 2018     https://doi.org/10.3892/etm.2018.6316
  • Pages: 1121-1130
  • 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

The aim of the present study was to identify the differentially expressed genes (DEGs) in esophageal squamous‑cellcarcinoma (ESCC) and provide potential therapeutic targets. The microarray dataset GSE20347 was downloaded from the Gene Expression Omnibus (GEO) database, and included 17 tissue samples and 13 normal adjacent tissue samples from patients with ESCC. A total of 22,277 DEGs were identified. A heat map for the DEGs was constructed with the Morpheus online tool and the top 200 genes (100 upregulated and 100 downregulated) were selected for further bioinformatics analysis, including analysis of gene ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, protein‑protein interaction networks and Spearman's correlation tests. The results of the GO analysis indicated that the upregulated DEGs were most significantly enriched in membrane‑bounded vesicles in the cellular component (CC) category, but were not significantly enriched in any GO terms of the categories biological process (BP) or molecular function (MF); furthermore, the downregulated DEGs were most significantly enriched in regulation of DNA metabolic processes, nucleotide binding and chromosomes in the categories BP, MF and CC, respectively. The KEGG analysis indicated that the downregulated DEGs were enriched in the regulation of cell cycle pathways. The top 10 hub proteins in the protein‑protein interaction network were cyclin‑dependent kinase 4, budding uninhibited by benzimidazoles 1, cyclin B2, heat shock protein 90AA1, aurora kinase A, H2A histone family member Z, replication factor C subunit 4, and minichromosome maintenance complex component 2, ‑4 and ‑7. These proteins are mainly involved in regulating tumor progression. The genes in the four top modules were mainly implicated in regulating cell cycle pathways. Secreted Ly‑6/uPAR‑related protein (SLURP) was the hub gene, and SLURP and its interacting genes were most enriched in the chromosomal part in the CC category, organelle organization in the BP category and protein binding in the MF category, and were involved in pathways including DNA replication, cell cycle and P53 signaling. The expression of SLURP‑1 in fifteen patients with esophageal carcinoma was detected using quantitative polymerase chain reaction analysis, and the results indicated that SLURP‑1 expression was significantly decreased in the tumor samples relative to that in normal adjacent tissues. These results suggest that several hub proteins and the hub gene SLURP‑1 may serve as potential therapeutic targets, and that gene dysfunction may be involved in the tumorigenesis of ESCC.

Introduction

Esophageal cancer (EC) has the sixth highest mortality and the eighth highest incidence rate worldwide (1). Its incidence rate in China is the highest in the world (2). The primary histological type is esophageal squamous-cell carcinoma (ESCC), which accounts for ~90% of all EC cases in China. Patients with ESCC are usually diagnosed at an advanced stage, and their 5-year survival rate is therefore low (~10-20%) (3,4). It has been reported that smoking and alcohol consumption are major causative factors of ESCC, as they promote gene mutations associated with processes including tumor initiation, progression and even metastasis. Thus, it is important to understand the molecular mechanisms of the tumorigenesis process to identify targets and develop novel treatments for ESCC.

Various genes, mRNAs and micro (mi)RNAs have been reported to form a network regulating the tumorigenesis and development of EC. Numerous studies have indicated that certain genes act as tumor suppressors, and that several genes inhibit cancer cell migration, invasion and tumor progression in ESCC (58). High-throughput sequencing technologies, including microarrays, which are able to detect changes in the expression of a vast amount of genes, have been widely used in cancer diagnosis and cancer research. In a previous study, numerous differentially expressed genes (DEGs) were detected in the tumor tissues of patients with ESCC relative to those in normal tissue or normal epithelial cells by microarrays (9). These hundreds of DEGs are involved in signalling pathways in ESCC, which encompass biological processes (BP), molecular functions (MF) and cellular components (CC). Hu et al (10) examined DEGs in tumor and matched normal adjacent tissue samples from patients with ESCC using microarrays. However, the regulatory roles of these DEGs, including the pathways in their interaction network, have remained to be elucidated (10).

Therefore, in the present study, bioinformatics methods were used to analyze the DEGs and their interaction networks. Original data were downloaded from the Gene Ontology (GEO) database (http://www.ncbi.nlm.nih.gov/geo/). The DEGs were identified from tumor tissues of patients with ESCC compared with those in matched normal adjacent tissues. The 200 top DEGs were then selected for further bioinformatics analysis, including analysis of Gene Ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, protein-protein interaction (PPI) networks and Spearman's correlation tests (11). In general, the present study may help identify potential therapeutic targets and provide valuable information to further illuminate the molecular mechanisms of ESCC.

Materials and methods

Microarray data

Gene expression profiles of GSE20347 were downloaded from the GEO repository collated by Hu et al (10). These data were based on the AgilentGPL571 platform (Affymetrix Human Genome U133A 2.0 Array, HG-U133A_2; Affymetrix; Thermo Fisher Scientific, Inc., Waltham, MA, USA), which included 13 samples of normal adjacent esophageal tissues (with the ID nos. GSM509787-GSM509803) and 17 samples of tumor tissues (with the ID nos. GSM509804-GSM509820) from patients with ESCC. Total RNA had been extracted using the PureLink Micro-to-Midi RNA Purification System (Invitrogen; Thermo Fisher Scientific, Inc.) and was detected by Affymetrix HG-U133A 2.0 gene expression arrays (Affymetrix; Thermo Fisher Scientific, Inc.). R (Bioconductor; http://www.bioconductor.org/) was used for background correction and normalization of the data.

Identification of DEGs

The raw data files used for analysis included TXT files (Agilent platform). The files were used to create heat maps with the Morpheus online software (https://software.broadinstitute.org/morpheus/). The data were classified into two groups, namely the normal and tumor groups. The top 200 DEGs (100 upregulated and 100 downregulated genes) were screened by their signal-to-noise ratio (SNR).

GO and pathway enrichment analysis of DEGs

The top 200 DEGs were analyzed using with the database for annotation, visualization, integrated discovery (DAVID) online tool (https://david.ncifcrf.gov/; DAVID bioinformatics resources 6.8 of the National Institute of Allergy and Infectious Diseases/National Institutes of Health), which may be used for the high-throughput functional analysis of genes (12). The database includes GO and KEGG pathway analyses. GO is a useful tool for identifying characteristic biological information by using high-throughput genome transcriptome data (13) and was used in the present study for GO enrichment analysis in the categories BP, cellular component (CC) and MF. KEGG pathway analysis was also performed to gain insight into the signaling pathways regulated by the DEGs (1416).

PPI network and module analysis

The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; http://version10.string-db.org/; version 10.0) database is a powerful online tool that overlays 9.6 million proteins from 2,034 organisms and 184 million interactions. The top 200 DEGs were analyzed by STRING for the PPI. Subsequently, interactions with a combined score of >0.4 (the value that was considered statistically significant) were selected to construct the PPI network using Cytoscape software 3.4.0 (http://www.cytoscape.org). Finally, the plug-in Molecular Complex Detection (MCODE) of Cytoscape was used to construct the modules of the PPI network. The top modules with an MCODE score of >3 and a node number of >4 were selected for further pathway analysis with KEGG.

Spearman correlation test of the top 200DEGs in patients with ESCC

To further determine DEGs that has the most connections to the other top DEG, a Spearman's correlation test was performed in Excel 2007 (Microsoft Corp., Redmond, WA, USA). Subsequently, the genes were screened with the following settings: 0.95<Pearson correlation coefficient (PCC) <1 and −1<PCC<-0.95. The gene with the highest correlation was selected as the hub gene to construct the co-expression network in Cytoscape. Finally, GO enrichment analyses in the categories BP, CC and MF for the hub gene and their associated genes were further performed by (Biological Networks Gene Ontology) BiNGO tool (17) and the KEGG analysis was performed with the ClueGO plug-in of Cytoscape.

Sample collection

ESCC and normal tissue samples were collected from the Panyu Central Hospital and the Third Affiliated Hospital of Southern Medical University (Guangzhou, China) from June 30, 2015 to May 2, 2017. All patients were male, with a mean age of 62.20±5.98 years, and diagnosed by clinical pathology (14 cases were squamous carcinoma and 1 was adenocarcinoma). All samples were stored at −80°C after collection. A total of 30 samples, which included 15 tumor samples and 15 normal adjacent tissue samples (>3 cm from the tumor tissue) as controls, were used to detect the gene expression of SLURP by reverse transcription-quantitative polymerase chain reaction (RT-qPCR).

RT-qPCR assay

Total RNA was extracted from all samples using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. First-strand complementary (c)DNA was synthesized with 1 µg total RNA per sample using the All-in-One™ First-Strand cDNA Synthesis kit (Gene Copoeia, Inc., Rockville, MD, USA). Subsequently, the cDNA sample was amplified using All-in-One qPCR mix (Gene Copoeia, Inc.) in a final volume of 20 µl in an ABI Vii7 dx reactor (Applied Biosystems; Thermo Fisher Scientific, Inc.). The amplifications were performed as follows: Initial incubation for 2 min at 50°C, denaturation for 30 sec at 95°C, and 45 cycles of 95°C for 5 sec and 65.6°C for 34 sec. The experiments were performed in triplicate and quantified using melting curve analysis. β-actin was used as an endogenous reference control. The relative gene expression levels were calculated using the 2−ΔΔCq method (18). The primer pairs for SLURP-1 and β-actin were as follows: SLURP-1 forward, 5′-GCTCCTGTGTGGCCACCGAC-3′ and reverse, 5′-GAGCCAGGCCCCGTCAGAGA-3′; β-actin forward, 5′-ACTCTTCCAGCCTTCCTTCC-3′ and reverse, 5′-GCGGCGCAATACGAATGCCCC-3′.

Statistical analysis

P<0.05 was considered to indicate a statistically significant difference. The P-value was adjusted by using the false discovery rate (FDR) method for multiple hypothesis testing. FDR <0.05 was established as the threshold (1517). The data are expressed as mean ± standard deviation. Independent t-tests were used to analyze the PCR results and performed using SPSS software (version 16.0; SPSS, Inc., Chicago, IL, USA).

Results

Identification of DEGs

A total of 22,277 DEGs were identified. The expression of these genes was analyzed with the Morpheus online tool to form a heat map (top 100 upregulated and 100 downregulated genes), which were selected according to their SNR value. The heat map is presented in Fig. 1.

GO term enrichment and KEGG pathway analysis of DEGs

GO term enrichment analysis indicated that the upregulated DEGs were most significantly enriched in membrane-bound vesicles in the category CC, but no significant enrichment was identified in the categories BP and MF. The downregulated DEGs were most significantly enriched in the regulation of DNA metabolic processes, nucleotide binding and chromosomes in the categories BP, MF and CC, respectively (Table I). The KEGG analysis indicated that the downregulated DEGs were enriched in the regulation of cell cycle pathways (Table II).

Table I.

GO analysis of differentially expressed genes in esophageal squamous cell carcinoma.

Table I.

GO analysis of differentially expressed genes in esophageal squamous cell carcinoma.

A, Upregulated genes in the category cellular component
GO termFunctionN (%)P-value
GO:0031988Membrane-bound vesicle33 (0.25) 4.02×10−8
GO:0070062Extracellular exosome29 (0.22) 1.27×10−7
GO:1903561Extracellular vesicle29 (0.22) 1.45×10−7
GO:0043230Extracellular organelle29 (0.22) 1.46×10−7
GO:0044421Extracellular region part30 (0.23) 3.16×10−5

B, Downregulated genes

GO term FunctionN (%)P-value

Biological process
  GO:0006259DNA metabolic process17 (0.11) 1.81×10−7
  GO:0051276Chromosome organization17 (0.11) 1.33×10−6
  GO:0010564Regulation of cell cycle process12 (0.08) 7.36×10−6
  GO:1903047Mitotic cell cycle process12 (0.08) 2.68×10−5
Cellular component
  GO:0005694Chromosome20 (0.13) 2.80×10−8
  GO:0044427Chromosomal part19 (0.12) 4.98×10−8
  GO:0098687Chromosomal region11 (0.07) 1.35×10−6
  GO:0000228Nuclear chromosome14 (0.09) 1.78×10−6
  GO:0000793Condensed chromosome8 (0.05) 1.00×10−5
  GO:0044454Nuclear chromosome part12 (0.08) 3.14×10−5
Molecular function
  GO:0000166Nucleotide binding27 (0.17) 6.38×10−6
  GO:1901265Nucleoside phosphate binding27 (0.17) 6.38×10−6
  GO:0035639Purine ribonucleoside triphosphate binding23 (0.15) 1.03×10−5
  GO:0032550Purine ribonucleoside binding23 (0.15) 1.08×10−5
  GO:0001883Purine nucleoside binding23 (0.15) 1.09×10−5
  GO:0032549Ribonucleoside binding23 (0.15) 1.12×10−5
  GO:0001882Nucleoside binding23 (0.15) 1.18×10−5
  GO:0032555Purine ribonucleotide binding23 (0.15) 1.41×10−5
  GO:1901363Heterocyclic compound binding44 (0.28) 1.48×10−5
  GO:0017076Purine nucleotide binding23 (0.15) 1.50×10−5
  GO:0032553Ribonucleotide binding23 (0.15) 1.62×10−5
  GO:0005524ATP binding20 (0.13) 1.70×10−5
  GO:0036094Small molecule binding27 (0.17) 1.97×10−5
  GO:0097159Organic cyclic compound binding44 (0.28) 2.02×10−5
  GO:0032559Adenyl ribonucleotide binding20 (0.13) 2.15×10−5
  GO:0030554Adenyl nucleotide binding20 (0.13) 2.28×10−5

[i] GO, Gene Ontology.

Table II.

Kyoto Encyclopedia of Genes and genomes pathway analysis of differentially expressed genes in esophageal squamous cell carcinoma.

Table II.

Kyoto Encyclopedia of Genes and genomes pathway analysis of differentially expressed genes in esophageal squamous cell carcinoma.

ExpressionTermFunctionN (%)P-value
Downregulatedcfa04110Cell cycle pathway8 (0.05) 1.62×10−5
PPI network and top module construction

Based on the STRING database, the PPI network of the top 200 DEGs was constructed with Cytoscape software (Fig. 2). In the PPI networks, nodes with a high degree of connectivity were defined as hub proteins. The top 10 hub proteins included cyclin-dependent kinase 4 (CDK4), budding uninhibited by benzimidazoles 1 (BUB1) and cyclin B2 (CCNB2). The degree of connectivity of CDK4 was 30, and it was therefore the most highly connected node. The network consisted of 110 nodes and 262 edges. The top four significant modules were selected for further pathway enrichment analysis (MCODE score, >3; number of nodes, >4) and the results indicated that the genes were significantly enriched in cell cycle pathways (Table III).

Table III.

The enriched pathway of module 2.

Table III.

The enriched pathway of module 2.

PathwayP-valueFalse discovery rateNodes
Cell cycle 1.71×10−6 9.35×10−4Cyclin-dependent kinases regulatory subunit 1, Cyclin-dependent kinase 1, MCM7, Nuclear pore complex protein Nup107, MCM4, MCM2, Mitotic checkpoint serine/threonine-protein kinase BUB1, KAT8 regulatory NSL complex subunit 1, Myb-related protein B

[i] MCM, DNA replication licensing factor MCM.

Spearman correlation analysis of the top 200 DEGs

The results of the Spearman correlation analysis indicated that the most connected gene was secreted LY6/PLAUR domain (SLURP). SLURP was selected as a hub gene and further analysis was performed on SLURP and its associated genes (Fig. 3). The results of the bioinformatics analysis suggested that these genes were most significantly enriched in the chromosomal part, organelle organization and protein binding in the categories CC, BP and MF, respectively (Figs. 46). KEGG pathway analysis revealed that the genes were involved in DNA replication, cell cycle and P53 signaling pathways (Fig. 7).

PCR analysis of SLURP

The results indicated that the expression of the hub gene SLURP-1 was significantly decreased in the tumor samples relative to that in the normal adjacent tissues in patients with esophageal carcinoma (P<0.05; Fig. 8).

Discussion

ESCC is caused by external factors leading to gene mutations. Thus, understanding the underlying molecular mechanisms is of pivotal importance for ESCC diagnosis and treatment. In the present study, a dataset downloaded from the GEO database was analyzed and the bioinformatics tools Morpheus, DAVID and STRING were used to obtain DEGs, hub proteins, hub genes and major deregulated pathways in ESCC.

A total of 22,277 DEGs were identified from the dataset GSE20347. To better understand the interactions of DEGs, the top 200 DEGs were selected for further analysis. The results of the GO term enrichment revealed that these DEGs were most highly involved in membrane-bound vesicles, DNA metabolic processes, nucleotide binding and chromosomes. KEGG analysis indicated that the DEGs were enriched in cell cycle pathways. These results suggest that the DEGs may be mainly involved in regulating the cell cycle (19,20) and organogenesis, which are closely associated with tumorigenesis (21) and tumor progression (2229). The results from a study by Lin et al (30) confirmed that the dysregulation of genes that regulate the G1/S transition is common in ESCC. Reduced expression of the protein p21WAF1/Cip1 was reported to predict a shorter overall survival time of patients with ESCC (27,28). The present results indicated that the deregulation of certain genes is involved in ESCC, and that various DEGs may be associated with the genesis of ESCC.

The analysis of PPI networks indicated that the top 10 hub proteins included CDK4, BUB1, CCNB2, heat shock protein (HSP)90AA1, aurora kinase (AURK)A, H2A histone family member Z (H2AFZ), replication factor C subunit 4 (RFC4), as well as minichromosome maintenance complex component 7 (MCM7), MCM4 and MCM2. These proteins are closely associated with the cell cycle, tumorigenesis (31), transferase signaling pathway (32,33), transforming growth factor β-mediated cell cycle control (34,35), embryonic development (36), DNA-dependent ATPase activity (37) and DNA unwinding enzymes (3840). CDK4 was the node with the highest degree of interaction and was the most connected hub protein, interacting with 30 genes in the regulatory network. This result was consistent with that by Su et al (41), which also indicated that CDK4 was the most significantly upregulated gene by analyzing 5 mRNA expression datasets of EC tissues/cell lines from GEO. A recent study revealed that CDK4 had a negative association with EC-related gene 4, which has a tumor suppressor function in ESCC (42). CDK4/6 inhibitor-SHR6390 was reported to exert an antitumor effect against ESCC (43). AURKA, MCM7 and MCM4 were closely associated with cell proliferation and migration in ESCC (4446). BUB1-related protein kinase was significantly higher in cancerous tissue than in adjacent normal tissue, and after radiochemotherapy, it was significantly decreased in the tissue of patients with ESCC (47). HSP90A and CCNB1 protein were reported to be associated with tumor malignancy and prognosis in patients with ESCC (48). It was demonstrated that abnormal levels of H2AF may be associated with poor survival of ESCC patients (49). However, as the involvement of RFC4 and MCM2 in ESCC has been rarely investigated, further study is necessary. Analysis of Hub protein functions indicated that these proteins have a key role in the regulation of ESCC, including the genesis development and progression of tumors, and that these hub proteins may serve as therapeutic targets in ESCC.

The present study used the plug-in MCODE to construct the modules. The results indicated that the functions of genes in the top 4 modules were mainly associated with cell cycle pathways. Li et al (50) indicated that overall, the DNA repair pathways were significantly associated with a risk of ESCC. Roncalli et al (51) studied cell cycle-associated genes in patients with EC, and their results demonstrated that these were significantly associated lymph node metastasis and unfavorable survival rates. These results indicate that the pathways associated with the top modules mainly regulate tumor progression in ESCC and that certain genes in theses pathways may serve as potential prognostic biomarkers.

In the present study, Spearman's correlation test was used to analyze the correlation between the top 200 DEGs. The results revealed that SLURP was the hub gene that was most highly connected to the other genes. The results of the bioinformatics analysis indicated that this hub gene and its associated genes are significantly enriched in the chromosomal part, organelle organization and protein binding in the GO categories CC, BP and MF, respectively. These genes are also involved in DNA replication, cell cycle and P53 signaling pathways. The expression of SLURP-1 was the assessed in 15 patients with EC, and the results revealed that SLURP-1was significantly decreased in the tumor samples relative to that in the normal adjacent tissues. SLURP-1 has been reported to participate in signal transduction, immune activation and cell adhesion to exert its antitumor activity (52). This was consistent with the results of recent studies, which indicated that DEGs screened by RNA-sequencing data or The Cancer Genome Atlas analysis have important roles in regulating growth, invasion and metastasis of tumors as well as immune responses, and the DEGs included collagen type I α 1, matrix metallopeptidases, keratin 4, cysteine-rich secretory protein2 and 3, mucin 21 and cyclin D1 (53,54). The present results indicated that the hub gene SLURP-1 may have a key role in regulating the tumorigenesis of ESCC and that it may serve as a potential biomarker in tumor diagnosis.

In conclusion, the present study identified DEGs and the hub proteins (CDK4, BUB1, CCNB2, HSP90AA1, AURKA, H2AFZ, RFC4, MCM7, MCM4 and MCM2) and a hub gene (SLURP) in ESCC. These genes are primarily involved in regulating the tumorigenesis and progression of ESCC. The hub proteins and gene may be considered as candidate therapeutic targets and may provide information for further studies on the molecular biological functions and mechanisms of ESCC. However, the present study had certain limitations, including the fact that only GEO 1 dataset of microarray data was used and that the sample size was relatively small.

Acknowledgements

The authors would like to thank Professor Edward I. Wong (Milton International Education Group, Hong Kong) for revising the manuscript.

Funding

This work was funded by grants of the Technical New Star of Zhujiang, Pan Yu district, Guangzhou (grant no. 2013-special-15-6.10), the Science and Technology program of Pan Yu (grant no. 2015-Z03-09), the Traditional Chinese Medicine Bureau of Guangdong Province (grant no. 20161186) and the Science and Technology Program of Guangzhou (grant no. 201804010012).

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

XC and SC performed the analysis of the data and writing of the paper. BL, XZ, WL and HL collected the samples and performed the PCR. XC performed the data analysis of GEO.

Ethical approval and consent to participate

This study was approved by the Ethics Committee of Panyu Central Hospital (ethical approval no. 20180001). All patients provided informed consent to participate in this study.

Patient consent for publication

All patients gave consent for the publication on their data in the current study.

Competing interests

The authors declared they have no competing conflicts.

References

1 

Chen W, Zheng R, Zhang S, Zeng H, Xia C, Zuo T, Yang Z, Zou X and He J: Cancer incidence and mortality in China, 2013. Cancer Lett. 401:63–71. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Tran GD, Sun XD, Abnet CC, Fan JH, Dawsey SM, Dong ZW, Mark SD, Qiao YL and Taylor PR: Prospective study of risk factors for esophageal and gastric cancers in the Linxian general population trial cohort in China. Int J Cancer. 113:456–463. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Yang Q, Wang YX, He M, Li J, Qi Z, Zhu SC and Qiao XY: Factors affecting on long-time survival in patients with stageIII thoracic esophageal carcinoma after esophagectomy. Zhonghua Zhong Liu Za Zhi. 38:530–537. 2016.(In Chinese). PubMed/NCBI

4 

Yu S, Zhang W, Ni W, Xiao Z, Wang X, Zhou Z, Feng Q, Chen D, Liang J, Fang D, et al: Nomogram and recursive partitioning analysis to predict overall survival in patients with stage IIB-III thoracic esophageal squamous cell carcinoma after esophagectomy. Oncotarget. 7:55211–55221. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Yi Y, Lu X, Chen J, Jiao C, Zhong J, Song Z, Yu X and Lin B: Downregulated miR-486-5p acts as a tumor suppressor in esophageal squamous cell carcinoma. Exp Ther Med. 12:3411–3416. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Osako Y, Seki N, Kita Y, Yonemori K, Koshizuka K, Kurozumi A, Omoto I, Sasaki K, Uchikado Y, Kurahara H, et al: Regulation of MMP13 by antitumor microRNA-375 markedly inhibits cancer cell migration and invasion in esophageal squamous cell carcinoma. Int J Oncol. 49:2255–2264. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Zuo J, Wang D, Shen H, Liu F, Han J and Zhang X: MicroRNA-153 inhibits tumor progression in esophageal squamous cell carcinoma by targeting SNAI1. Tumour Biol. Oct 13–2016.(Epub ahead of print). View Article : Google Scholar

8 

Jing C, Ma G, LiX, Wu X, Huang F, Liu K and Liu Z: MicroRNA-17/20a impedes migration and invasion via TGF-β/ITGB6 pathway in esophageal squamous cell carcinoma. Am J Cancer Res. 6:1549–1562. 2016.PubMed/NCBI

9 

Hu N, Wang C, Clifford RJ, Yang HH, Su H, Wang L, Wang Y, Xu Y, Tang ZZ, Ding T, et al: Integrative genomics analysis of genes with biallelic loss and its relation to the expression of mRNA and micro-RNA in esophageal squamous cell carcinoma. BMC Genomics. 16:7322015. View Article : Google Scholar : PubMed/NCBI

10 

Hu N, Clifford RJ, Yang HH, Wang C, Goldstein AM, Ding T, Taylor PR and Lee MP: Genome wide analysis of DNA copy number neutral loss of heterozygosity (CNNLOH) and its relation to gene expression in esophageal squamous cell carcinoma. BMC Genomics. 11:5762010. View Article : Google Scholar : PubMed/NCBI

11 

Garner E, Wallace JS, Argoty GA, Wilkinson C, Fahrenfeld N, Heath LS, Zhang L, Arabi M, Aga DS and Pruden A: Metagenomic profiling of historic Colorado Front Range flood impact on distribution of riverine antibiotic resistance genes. Sci Rep. 6:384322016. View Article : Google Scholar : PubMed/NCBI

12 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization, and integrated discovery. GenomeBiol. 4:P32003.

13 

Torto-Alalibo T, Purwantini E, Lomax J, Setubal JC, Mukhopadhyay B and Tyler BM: Genetic resources for advanced biofuel production described with the Gene Ontology. Front Microbiol. 5:5282014. View Article : Google Scholar : PubMed/NCBI

14 

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.

15 

Du J, Yuan Z, Ma Z, Song J, Xie X and Chen Y: KEGG-PATH: Kyotoencydopedia of genes and genomes-based pathway analysis using a path analysis model. MolBiosyst. 10:2441–14447. 2014.

16 

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

17 

Maere S, Heymans K and Kuiper M: BiNGO: A Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics. 21:3448–9344. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Ren K, Li Y, Lu H, Li Z, Li Z, Wu K, Li Z and Han X: Long Noncoding RNA HOTAIR Controls Cell Cycle by Functioning as a Competing Endogenous RNA in Esophageal Squamous Cell Carcinoma. Transl Oncol. 9:489–497. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Zhang HF, Alshareef A, Wu C, Jiao JW, Sorensen PH, Lai R, Xu LY and Li EM: miR-200b induces cell cycle arrest and represses cell growth in esophageal squamous cell carcinoma. Carcinogenesis. 37:858–869. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Cao J, Ge MH and Ling ZQ: Fbxw7 tumor suppressor: A vital regulator contributes to human tumorigenesis. Medicine (Baltimore). 95:e24962016. View Article : Google Scholar : PubMed/NCBI

22 

Costa C, Santos M, Martínez-Fernández M, Lorz C, Lázaro S and Paramio JM: Deregulation of the pRb-E2F4 axis alters epidermal homeostasis and favors tumor development. Oncotarget. 7:75712–75728. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Lambot MA, Peny MO, Fayt I, Haot J and Noël JC: Overexpression of 27-kDa heat shock protein relates to poor histological differentiation in human oesophageal squamous cell carcinoma. Histopathology. 36:326–330. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Xia W, Qiu M, Chen R, Wang S, Leng X, Wang J, Xu Y, Hu J, Dong G, Xu PL, et al: CCircular RNA has_circ_0067934 is upregulated in esophageal squamous cell carcinoma and promoted proliferation. Sci Rep. 6:355762016. View Article : Google Scholar : PubMed/NCBI

25 

Xu Y, Qiu M, Chen Y, Wang J, Xia W, Mao Q, Yang L, Li M, Jiang F, Xu L and Yin R: Long noncoding RNA, tissue differentiation-inducing nonprotein coding RNA is upregulated and promotes development of esophageal squamous cell carcinoma. Dis Esophagus. 29:950–958. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Zhi H, Zhang J, Hu G, Lu J, Wang X, Zhou C, Wu M and Liu Z: The deregulation of arachidonic acid metabolism-related genes in human esophageal squamous cell carcinoma. Int J Cancer. 106:327–333. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Mao Y, Li L, Liu J, Wang L and Zhou Y: MiR-495 inhibits esophageal squamous cell carcinoma progression by targeting Akt1. Oncotarget. 7:51223–51236. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Zeng LS, Yang XZ, Wen YF, Mail SJ, Wang MH, Zhang MY, Zheng XF and Wang HY: Overexpressed HDAC4 is associated with poor survival and promotes tumor progression in esophageal carcinoma. Aging (Albany NY). 8:1236–1249. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Luo J, Zhang C, Wang C, Li L, Li C, Li Q, Zhang M and Wu Q: Miz-1 promotes the proliferation of esophageal cancer cells via suppression of p21 and release of p21-arrested cyclin D1. Oncol Rep. 35:3532–3540. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Lin DC, Shi ZZ, Xue LY, Chen W, Xu X, Han YL, Lv N and Wang MR: Expression of cell cycle related proteins cyclin D1, p53 and p21WAF1/Cip1 in esophageal squamous cell carcinoma. Yi Chuan. 32:455–460. 2010.(In Chinese). View Article : Google Scholar : PubMed/NCBI

31 

Lang E, Zelenak C, Eberhard M, Bissinger R, Rotte A, Ghashghaeinia M, Lupescu A, Lang F and Qadri SM: Impact of cyclin-dependent kinase CDK4 inhibition on eryptosis. Cell Physiol Biochem. 37:1178–1186. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Breit C, Bange T, Petrovic A, Weir JR, Müller F, Vogt D and Musacchio A: Role of intrinsic and extrinsic factors in the regulation of the mitotic checkpoint kinase Bub1. PLoS One. 10:e01446732015. View Article : Google Scholar : PubMed/NCBI

33 

Takashima S, Saito H, Takahashi N, Imai K, Kudo S, Atari M, Saito Y, Motoyama S and Minamiya Y: Strong expression of cyclin B2 mRNA correlates with a poor prognosis in patients with non-small cell lung cancer. Tumour Biol. 35:4257–4265. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Chen JT, Younusi A, Cao L, Tian Z, Zhou YJ and Song XH: Potential role of heat-shock proteins in giant cell tumors. Genet Mol Res. 14:19144–19154. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Kim SR, Kim KB, Chae YC, Park JW and Seo SB: H3S10 phosphorylation-mediated transcriptional regulation by Aurora kinase A. Biochem Biophys Res Commun. 469:22–28. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Kusakabe M, Oku H, Matsuda R, Hori T, Muto A, Igarashi K, Fukagawa T and Harata M: Genetic complementation analysis showed distinct contributions of the N-terminal tail of H2A.Z to epigenetic regulations. Genes Cells. 21:122–135. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Arai M, Kondoh N, Imazeki N, Hada A, Hatsuse K, Matsubara O and Yamamoto M: The knockdown of endogenous replication factor C4 decreases the growth and enhances the chemosensitivity of hepatocellular carcinoma cells. Liver Int. 29:55–62. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Ishibashi Y, Kinugasa T, Akagi Y, Ohchi T, Gotanda Y, Tanaka N, Fujino S, Yuge K, Kibe S, Yoshida N, et al: Minichromosome maintenance protein 7 is a risk factor for recurrence in patients with Dukes C colorectal cancer. Anticancer Res. 34:4569–4575. 2014.PubMed/NCBI

39 

Watanabe E, Ohara R and Ishimi Y: Effect of an MCM4 mutation that causes tumours in mouse on human MCM4/6/7 complex formation. J Biochem. 152:191–198. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Razavi SM, Jafari M, Heidarpoor M and Khalesi S: Minichromosome maintenance-2 (MCM2) expression differentiates oral squamous cell carcinoma from pre-cancerous lesions. Malays J Pathol. 37:253–258. 2015.PubMed/NCBI

41 

Su P, Wen S, Zhang Y, Li Y, Xu Y, Zhu Y, Lv H, Zhang F, Wang M and Tian Z: Identification of the key genes and pathways in esophageal carcinoma. Gastroenterol Res Pract. 2016:29681062016. View Article : Google Scholar : PubMed/NCBI

42 

Li L, Wang W, Li X and Gao T: Association of ECRG4 with PLK1, CDK4, PLOD1 and PLOD2 in esophageal squamous cell carcinoma. Am J Transl Res. 9:3741–3748. 2017.PubMed/NCBI

43 

Wang J, Li Q, Yuan J, Wang J, Chen Z, Liu Z, Li Z, Lai Y, Gao J and Shen L: CDK4/6 inhibitor-SHR6390 exerts potent antitumor activity in esophageal squamous cell carcinoma by inhibiting phosphorylated Rb and inducing G1 cell cycle arrest. J Transl Med. 15:1272017. View Article : Google Scholar : PubMed/NCBI

44 

Wang X, Li X, Li C, He C, Ren B, Deng Q, Gao W and Wang B: Aurora-A modulates MMP-2 expression via AKT/NF-κB pathway in esophageal squamous cell carcinoma cells. Acta Biochim Biophys Sin (Shanghai). 48:520–527. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Qiu YT, Wang WJ, Zhang B, Mei LL and Shi ZZ: MCM7 amplification and overexpression promote cell proliferation, colony formation and migration in esophageal squamous cell carcinoma by activating the AKT1/mTOR signaling pathway. Oncol Rep. 37:3590–3596. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Choy B, LaLonde A, Que J, Wu T and Zhou Z: MCM4 and MCM7, potential novel proliferation markers, significantly correlated with Ki-67, Bmi1, and cyclin E expression in esophageal adenocarcinoma, squamous cell carcinoma, and precancerous lesions. Hum Pathol. 57:126–135. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Tanaka K, Mohri Y, Ohi M, Yokoe T, Koike Y, Morimoto Y, Miki C, Tonouchi H and Kusunoki M: Mitotic checkpoint genes, hsMAD2 and BubR1, in oesophageal squamous cancer cells and their association with 5-fluorouracil and cisplatin-based radiochemotherapy. Clin Oncol (R Coll Radiol). 20:639–646. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Huang T, Chen S, Han H, Li H, Huang Z, Zhang J, Yin Q, Wang X, Ma X, Dai P, et al: Expression of Hsp90α and cyclin B1 were related to prognosis of esophageal squamous cell carcinoma and keratin pearl formation. Int J Clin Exp Pathol. 7:1544–1552. 2014.PubMed/NCBI

49 

Zhang K, Li L, Zhu M, Wang G, Xie J, Zhao Y, Fan E, Xu L and Li E: Comparative analysis of histone H3 and H4 post-translational modifications of esophageal squamous cell carcinoma with different invasive capabilities. J Proteomics. 112:180–189. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Li WQ, Hu N, Hyland PL, Gao Y, Wang ZM, Yu K, Su H, Wang CY, Wang LM, Chanock SJ, et al: Genetic variants in DNA repair pathway genes and risk of esophageal squamous cell carcinoma and gastric adenocarcinoma in a Chinese population. Carcinogenesis. 34:1536–1542. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Roncalli M, Bosari S, Marchetti A, Buttitta F, Bossi P, Graziani D, Peracchia A, Bonavina L, Viale G and Coggi G: Cell cycle-related gene abnormalities and product expression in esophageal carcinoma. Lab Invest. 78:1049–1057. 1998.PubMed/NCBI

52 

Pettersson A, Nylund G, Khorram-Manesh A, Nordgren S and Delbro DS: Nicotine induced modulation of SLURP-1 expression in human colon cancer cells. Auton Neurosci. 148:97–100. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Fu JH, Wang LQ, Li T and Ma GJ: RNA-sequencing based identification of crucial genes for esophageal squamous cell carcinoma. J Cancer Res Ther. 11:240–425. 2015.

54 

Cancer Genome Atlas Network; Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 517:576–582. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 16 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Cai S, Li B, Zhang X, Li W, Linag H and Cao X: Identification of key genes and pathways for esophageal squamous cell carcinoma by bioinformatics analysis. Exp Ther Med 16: 1121-1130, 2018
APA
Chen, X., Cai, S., Li, B., Zhang, X., Li, W., Linag, H., & Cao, X. (2018). Identification of key genes and pathways for esophageal squamous cell carcinoma by bioinformatics analysis. Experimental and Therapeutic Medicine, 16, 1121-1130. https://doi.org/10.3892/etm.2018.6316
MLA
Chen, X., Cai, S., Li, B., Zhang, X., Li, W., Linag, H., Cao, X."Identification of key genes and pathways for esophageal squamous cell carcinoma by bioinformatics analysis". Experimental and Therapeutic Medicine 16.2 (2018): 1121-1130.
Chicago
Chen, X., Cai, S., Li, B., Zhang, X., Li, W., Linag, H., Cao, X."Identification of key genes and pathways for esophageal squamous cell carcinoma by bioinformatics analysis". Experimental and Therapeutic Medicine 16, no. 2 (2018): 1121-1130. https://doi.org/10.3892/etm.2018.6316