Open Access

Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis

  • Authors:
    • Feng He
    • Bo Ai
    • Lei Tian
  • View Affiliations

  • Published online on: July 25, 2018     https://doi.org/10.3892/br.2018.1134
  • Pages: 305-312
  • Copyright: © He 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

Esophageal adenocarcinoma (EAC) is one of the most common subtypes of esophageal cancer, and is associated with a low 5-year survival rate. The present study aimed to identify key genes and pathways associated with EAC using bioinformatics analysis. The gene expression profiles of GSE92396, which includes 12 EAC samples and 9 normal esophageal samples, were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between the EAC and normal samples were identified using the limma package in R language. Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the identified DEGs were conducted using the online analysis tool, the Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction (PPI) network of the DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes (STRING) database and Cytoscape software. Finally, module analysis was conducted for the PPI network using the MCODE plug-in in Cytoscape. Of the 386 DEGs identified, the 150 upregulated genes were mainly enriched in the KEGG pathways of complement and coagulation cascades, maturity onset diabetes of the young and protein digestion and absorption; and the 236 downregulated genes were mainly enriched in amoebiasis, retinol metabolism and drug metabolism-cytochrome P450. Based on information from the STRING database, a PPI network comprising of 369 nodes and 534 edges was constructed in Cytoscape. The top 10 hub nodes with the highest degrees were determined as interleukin-8, involucrin, tissue inhibitor of metalloproteinase 1, fibronectin 1, serpin family E member 1, serpin family A member 1, cystic fibrosis transmembrane conductance regulator, secreted phosphoprotein 1, collagen type I alpha 1 chain and angiotensinogen. A total of 6 modules were detected from the PPI network that satisfied the criteria of MCODE score >4 and number of nodes >4. KEGG pathways enriched for the module DEGs were mainly within arachidonic acid metabolism, complement and coagulation cascades and rheumatoid arthritis. In conclusion, identification of these key genes and pathways may improve understanding of the mechanisms underlying the development of EAC, and may be used as diagnostic and therapeutic targets in EAC.

Introduction

Esophageal cancer is among the most common malignancies worldwide, and in the United States has a 5-year survival rate following diagnosis of only ~19% (1). Squamous cell carcinoma and adenocarcinoma are the two main subtypes of esophageal cancer. The incidence of esophageal adenocarcinoma (EAC) has increased substantially in the United States, Western Europe, Australia and other developed countries over the past four decades (2). It is generally accepted that gastroesophageal reflux disease and obesity are explanations for the increased incidence of EAC (3). However, the underlying mechanism remains unclear.

Several genes have been reported to serve important roles in the development of EAC. The P53 gene has been found to be dysregulated in most cancer types (4). Furthermore, it is considered that P53 may be involved in the development of different cancers. For instance, a cohort study of chemoradiotherapy-naive surgically treated EAC reported that p53 expression was significantly correlated with disease-free survival and overall survival, independent of tumor stage (5). Meanwhile, a genome-wide association study of 2,515 EAC cases and 3,207 controls provided data to suggest that germline variations at the cyclin-dependent kinase inhibitor 2A locus may influence susceptibility to EAC (6). In addition, Gli and epithelial-mesenchymal transition-related protein expression was previously examined by western blot analysis in paired EAC patient tissues and cell lines. The results suggested that Gli may be critical for the metastasis and recurrence of esophageal adenocarcinomas (7). Osteopontin (OPN) isoforms have also been investigated in EAC, where results indicated that all OPN isoforms were frequently co-overexpressed in primary EACs, and that isoforms OPNb and OPNc enhanced invasion and dissemination through collective yet distinct mechanisms (8). However, despite these in-depth studies to identify novel targets for the treatment of EAC, there lacks a comprehensive presentation of the key genes and pathways implicated in EAC.

Gene expression profile analysis is a high-throughput method for detecting messenger RNA expression in tissue or cell samples. By analyzing the different gene expression between cancer patients and normal controls, an improved understanding of the molecular pathogenesis of a tumor can be obtained, facilitating the identification of the potential target genes and pathways for therapy (9,10).

The present study aimed to investigate the pathogenesis of EAC by a computational bioinformatics analysis of gene expression. Data from the Gene Expression Omnibus (GEO) database was extracted, and differentially expressed genes (DEGs) between EAC and normal samples were identified. The possible functions of the DEGs were predicted using enrichment analysis. Furthermore, protein-protein interaction (PPI) networks were visualized and module analysis was conducted using Cytoscape software to search for key genes that may be involved in the development of EAC.

Materials and methods

Affymetrix microarray data

The gene expression profiles of GSE92396, contributed by Peng et al (11), were downloaded from the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/). The platform was GPL6244, HuGene-1_0-st Affymetrix Human Gene 1.0 ST Array. The dataset included 12 esophageal adenocarcinoma samples and 9 normal esophageal samples; 9 were tumor-normal pairs.

Identification of DEGs

The data were pre-processed in R language (version 3.4.3; http://www.r-project.org/) using the oligo package (version 1.32.0; http://www.bioconductor.org/packages/release/bioc/html/oligo.html) (12,13). Probe levels were calculated and converted into the gene expression levels according to the annotation files in the GEO database. The DEGs of GSE92396 between the normal tissues and the tumor samples were analyzed with limma package (version 3.34.8) in R language (14). Fold-changes (FCs) in the gene expression values were calculated. |log2 FC|>2 and adjusted P-values <0.05 were considered to be the cut-off criteria for the identification of DEGs. A volcano plot was drawn using the gplots package (version 3.0.1).

Gene ontology (GO) and pathway enrichment analysis of the DEGs

The online analysis tool, the Database for Annotation, Visualization and Integrated Discovery (DAVID; version 6.8; http://david.abcc.ncifcrf.gov/) was used to analyse the DEGs for GO term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Enriched terms with >2 genes and a P-value <0.05 were considered to be statistically significant.

Construction of PPI network and screening of modules

The online analysis tool, the Search Tool for the Retrieval of Interacting Genes (STRING version 10.0; http://string-db.org/) was used to assess the PPI network of the DEGs, with the required confidence (combined score) >0.4. Visualization of the network and module analysis were performed with Cytoscape software (version 3.6.0; http://www.cytoscape.org/) and the MCODE plug-in (version 1.5.1) (15). The degree was statistically analysed using the CentiScaPe plug-in (version 2.2) to obtain hub nodes or genes in the PPI network (16). An MCODE computed node score >4 and node number >4 were considered as the cut-off criteria. Subsequent GO function and KEGG pathway enrichment analyses of the DEGs in the modules were performed using DAVID.

Results

Identification of DEGs

To identify DEGs between EAC samples and normal controls, the microarray dataset GSE92396, obtained from the GEO database, was screened. DEGs with |log2 FC|>2 and a P-value <0.05 were determined. A total of 386 DEGs were identified in EAC samples compared with in the normal controls, including 150 upregulated and 236 downregulated DEGs. The volcano plot is presented in Fig. 1.

GO and pathway enrichment analysis of DEGs

To categorize the representation of DEGs and the involved pathways, GO and KEGG pathway enrichment analyses were performed using the online tool DAVID. The upregulated DEGs were enriched in 60 GO terms and 3 KEGG pathways. The GO functions enriched for the upregulated DEGs were mainly within the extracellular exosome (P=1.09×10−14), extracellular space (P=5.44×10−15) and extracellular region (P=1.61×10−10). KEGG pathways enriched for the upregulated DEGs were mainly in complement and coagulation cascades (P=6.84×10−5), maturity onset diabetes of the young (P=0.002282) and protein digestion and absorption (P=0.012483).

The downregulated DEGs were enriched in 67 GO terms and 5 KEGG pathways. The GO functions enriched for the downregulated DEGs were mainly within the extracellular exosome (P=8.36×10−12), epidermis development (P=3.26×10−22) and keratinocyte differentiation (P=2.62×10−20). KEGG pathways enriched for the downregulated DEGs were mainly in amoebiasis (P=3.46×10−4), retinol metabolism (P=0.020258) and drug metabolism-cytochrome P450 (P=0.022809). The top 10 terms of the GO enrichment analysis for up- and downregulated genes are presented respectively in Table I. The results of KEGG enrichment analysis for up- and downregulated genes are presented respectively in Table II.

Table I.

GO enrichment analysis of the differentially expressed genes.

Table I.

GO enrichment analysis of the differentially expressed genes.

CategoryTermCountP-value
Upregulated genes
GOTERM_CC_DIRECTGO:0005615 extracellular space43 5.44×10−15
GOTERM_CC_DIRECTGO:0070062 extracellular exosome62 1.09×10−14
GOTERM_CC_DIRECTGO:0005576 extracellular region40 1.61×10−10
GOTERM_CC_DIRECTGO:0005578 proteinaceous extracellular matrix15 3.09×10−8
GOTERM_BP_DIRECTGO:0022617 extracellular matrix disassembly9 2.36×10−7
GOTERM_CC_DIRECTGO:0031012 extracellular matrix14 7.17×10−7
GOTERM_CC_DIRECTGO:0016324 apical plasma membrane13 3.74×10−6
GOTERM_BP_DIRECTGO:0030198 extracellular matrix organization11 6.13×10−6
GOTERM_CC_DIRECTGO:0005796 Golgi lumen8 1.15×10−5
GOTERM_BP_DIRECTGO:0030574 collagen catabolic process7 1.58×10−5
Downregulated genes
GOTERM_BP_DIRECTGO:0008544 epidermis development22 3.26×10−22
GOTERM_BP_DIRECTGO:0030216 keratinocyte differentiation20 2.62×10−20
GOTERM_CC_DIRECTGO:0001533 cornified envelope15 1.04×10−16
GOTERM_BP_DIRECTGO:0018149 peptide cross-linking14 1.69×10−14
GOTERM_BP_DIRECTGO:0031424 keratinization13 2.97×10−13
GOTERM_MF_DIRECTGO:0005198 structural molecule activity21 7.55×10−12
GOTERM_CC_DIRECTGO:0070062 extracellular exosome74 8.36×10−12
GOTERM_CC_DIRECTGO:0030057 desmosome8 8.15×10−9
GOTERM_MF_DIRECTGO:0004867 serine-type endopeptidase inhibitor activity10 1.66×10−6
GOTERM_BP_DIRECTGO:0061436 establishment of skin barrier6 1.76×10−6

[i] GO, gene ontology; _BP, biological process; _CC, cellular component; _MF, molecular function.

Table II.

KEGG pathway enrichment analysis of the differentially expressed genes.

Table II.

KEGG pathway enrichment analysis of the differentially expressed genes.

CategoryTermCountP-value
Upregulated genes
KEGG_PATHWAYhsa04610 Complement and coagulation cascades7 6.84×10−5
KEGG_PATHWAYhsa04950 Maturity onset diabetes of the young40.002282
KEGG_PATHWAYhsa04974 Protein digestion and absorption50.012483
Downregulated genes
KEGG_PATHWAYhsa05146 Amoebiasis7 3.46×10−4
KEGG_PATHWAYhsa00830 Retinol metabolism40.020258
KEGG_PATHWAYhsa00982 Drug metabolism - cytochrome P45040.022809
KEGG_PATHWAYhsa05204 Chemical carcinogenesis40.034682
KEGG_PATHWAYhsa00350 Tyrosine metabolism30.038991

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; hsa, homo sapiens.

Construction of PPI network and screening of modules

Based on information from the STRING database, a PPI network comprising of 369 nodes and 534 edges was constructed using the Cytoscape software (Fig. 2). The top 10 hub nodes with the highest degrees were interleukin (IL)-8, involucrin (IVL), tissue inhibitor of metalloproteinase 1 (TIMP1), fibronectin 1 (FN1), serpin family E member 1 (SERPINE1), serpin family A member 1 (SERPINA1), cystic fibrosis transmembrane conductance regulator (CFTR), secreted phosphoprotein 1 (SPP1), collagen type I alpha 1 chain (COL1A1) and angiotensinogen (AGT). A total of 6 modules from the PPI network satisfied the criteria of an MCODE computed node score >4 and number of nodes >4. The results are presented in Fig. 3. The functional annotation of the DEGs involved in the modules was determined using DAVID. The results showed that the module DEGs were enriched in 66 GO terms and 9 KEGG pathways. The GO functions enriched for the module DEGs were mainly within the extracellular exosome (P=1.27×10−8), extracellular region (P=8.63×10−11) and extracellular space (P=1.21×10−7). KEGG pathways enriched for the module DEGs were mainly within arachidonic acid metabolism (P=1.02×10−4), complement and coagulation cascades (P=1.56×10−4), and rheumatoid arthritis (P=3.98×10−4). The top 10 terms of the GO and KEGG enrichment analyses for module DEGs are presented in Table III.

Table III.

GO and KEGG enrichment analyses of the differentially expressed genes in the modules.

Table III.

GO and KEGG enrichment analyses of the differentially expressed genes in the modules.

CategoryTermCountP-value
GOTERM_CC_DIRECTGO:0005576 extracellular region22 8.63×10−11
GOTERM_BP_DIRECTGO:0018149 peptide cross-linking7 5.00×10−9
GOTERM_CC_DIRECTGO:0070062 extracellular exosome25 1.27×10−8
GOTERM_BP_DIRECTGO:0030216 keratinocyte differentiation7 6.52×10−8
GOTERM_CC_DIRECTGO:0005615 extracellular space17 1.21×10−7
GOTERM_BP_DIRECTGO:0031424 keratinization6 2.16×10−7
GOTERM_BP_DIRECTGO:0016266 O-glycan processing6 6.72×10−7
GOTERM_BP_DIRECTGO:0022617 extracellular matrix disassembly6 2.20×10−6
GOTERM_MF_DIRECTGO:0005198 structural molecule activity8 3.19×10−6
GOTERM_CC_DIRECTGO:0005796 Golgi lumen6 4.70×10−6
KEGG_PATHWAYhsa00590 Arachidonic acid metabolism5 1.02×10−4
KEGG_PATHWAYhsa04610 Complement and coagulation cascades5 1.56×10−4
KEGG_PATHWAYhsa05323 Rheumatoid arthritis5 3.98×10−4
KEGG_PATHWAYhsa00982 Drug metabolism - cytochrome P45040.002504
KEGG_PATHWAYhsa00980 Metabolism of xenobiotics by cytochrome P45040.003187
KEGG_PATHWAYhsa00350 Tyrosine metabolism30.008675
KEGG_PATHWAYhsa00830 Retinol metabolism30.028145
KEGG_PATHWAYhsa00010 Glycolysis/Gluconeogenesis30.02977
KEGG_PATHWAYhsa05204 Chemical carcinogenesis30.041201

[i] GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; _BP, biological process; _CC, cellular component; _MF, molecular function; hsa, homo sapiens.

Discussion

EAC is one of the most common subtypes of esophageal cancer (17), and only ~19% of patients survive 5 year after diagnosis in the United States (1). Therefore, there is a need to screen for key genes and pathways that are associated with the progression of EAC, with the aim of improving its diagnosis and treatment.

The present study used bioinformatics analysis to identify the DEGs between EAC and normal tissue expression profiles. The results revealed that the expression of 386 genes was significantly altered in EAC samples (150 upregulated and 236 downregulated genes) compared with in the normal controls. A PPI network was constructed to reveal the associations between these genes. The top 10 genes with the highest degrees were identified. Furthermore, 6 modules were selected according to their respective MCODE computed node scores (>4), and their functions were determined by GO and KEGG pathway analyses.

The GO functions enriched for the upregulated DEGs were mainly within the extracellular exosome, extracellular space and extracellular region. KEGG pathways enriched for the upregulated DEGs were mainly within complement and coagulation cascades, maturity onset diabetes of the young and protein digestion and absorption. The GO functions enriched for the downregulated DEGs were mainly within the extracellular exosome, epidermis development and keratinocyte differentiation. KEGG pathways enriched for the downregulated DEGs were mainly within amoebiasis, retinol metabolism and drug metabolism-cytochrome P450. Previous study has demonstrated that activation of the coagulation cascade affected tumor development (18). The underlying mechanism through which coagulation cascade proteins promote tumorigenesis remains unclear. Therefore, investigating these identified signaling pathways may aid to elucidate the carcinogenic mechanism behind EAC.

Based on the results of PPI network construction for the DEGs, a number of hub nodes were identified. The top 10 hub nodes with the highest degrees were IL8, IVL, TIMP1, FN1, SERPINE1, SERPINA1, CFTR, SPP1, COL1A1 and AGT. IL8, also named C-X-C motif (CXC) chemokine ligand 8, is a chemokine that mainly attracts inflammatory leukocyte infiltrate by acting on CXC chemokine receptor 1/2. Recent speculations propose that IL8 serves important roles in angiogenesis and survival signaling for cancer stem cells, and that the interleukin may stimulate the secretion of local growth factors in malignant tumors (19). IL8 stimulation on endothelial cells has been reported to begin angiogenic processes characterized by secretion of matrix metalloproteinases (MMPs), which can break down the extracellular matrix and stimulate the formation of new vessels (20). One study reported that IL8 was significantly upregulated in esophageal carcinogenesis, being detected in the serum of patients with esophageal adenocarcinoma (21). IVL is a squamous cell differentiation marker, and is associated with terminal differentiation of epithelial cells (22,23). Upon IL4 stimulation, the overall esophageal epithelia still contained stratified morphology. However, IVL was significantly decreased in esophageal basal and suprabasal layers, which was associated with a disorganized morphology of stratified layers on the basal side (24). TIMP1 is an inhibitor of matrix metalloproteinases, which has a key role in cancer cell dissemination and endothelial cell migration in angiogenesis (25). High serum levels of TIMP1 have been associated with tumor progression and poor prognosis in esophageal cancer patients (26). FN1, a mesenchymal marker (27), is an extracellular matrix glycoprotein that serves key roles in cell differentiation, growth and migration; through which it is associated with certain processes including wound healing, embryonic development and carcinogenesis (28). SERPINE1, also known as plasminogen activator inhibitor-1, is involved in the inhibition of urokinase-type plasminogen activator (29). It serves important roles in increasing tumor invasion and angiogenesis, and has been correlated with a poor prognosis (30). A high tumor level of plasminogen activator inhibitor-1 in patients with primary breast cancer is reportedly suggestive of poor prognosis (31), through this association requires verification in EAC. SERPINA1 encodes for α1-antitrypsin, which targets several proteases, including elastase, plasmin, thrombin, trypsin, chymotrypsin, and plasminogen activator (32). One study suggested that α1-antitrypsin may be involved in lung adenocarcinoma metastasis by targeting fibronectin (33). CFTR encodes an ATP-binding cassette membrane protein that functions as a chloride channel, and is mutated in cystic fibrosis (34,35). A previous large-scale meta-analysis suggested that the novel single nucleotide polymorphism (SNP) rs17451754, which is located within intron 21 of the CFTR gene, markedly associates with the risk of Barrett's esophagus and EAC (36). This region is reportedly involved with the enhancer histone modifications in the gastrointestinal tract mucosa and DNAse hypersensitivity (37). SPP1, also known as OPN, encodes a protein that binds hydroxyapatite, and is a cytokine that upregulates interferon-γ and IL12 (38). A previous study reported that SPP1 isoforms collectively enhanced tumor cell invasion and dissemination in EAC (8). COL1A1 encodes the pro-α1 chains of type I collagen. It has been reported COL1A1 is overexpressed in many tumours, including gastric cancer, hepatocellular carcinoma, non-small cell lung cancer, and colorectal cancer (3942). One study suggested that COL1A1 may promote metastasis in colorectal cancer by regulating the Wnt/planar cell polarity pathway (39). AGT is involved in the renin-angiotensin system (RAS). Previous study reported that RAS participated in the physiological control of esophageal motor activity (43). Together with these previous findings, the present results are suggestive that RAS may be involved in the contraction disorder of esophageal adenocarcinoma.

A total of 6 modules from the PPI network satisfied the criteria of MCODE score >4 and number of nodes >4. The GO functions enriched for the module DEGs were mainly within the extracellular exosome, extracellular region and extracellular space. KEGG pathways enriched for the module DEGs were mainly in arachidonic acid metabolism, complement and coagulation cascades and rheumatoid arthritis. A previous study suggested that the activated arachidonic acid metabolism pathway serves an important role in tumorigenesis (44). The enzymes activated by this pathway and their products promote the inflammatory response and have been implicated in multiple cellular processes, including cell proliferation, invasion and metastasis, and thus may promote tumorigenesis. Additionally, previous study has demonstrated that activation of the coagulation cascade affected tumor development (18). Therefore, the arachidonic acid metabolism and complement and coagulation cascades pathways may be involved in the development of EAC.

In conclusion, the present study identified the genes differentially expressed between EAC and normal samples. The top most altered DEGs included IL8, IVL, TIMP1, FN1, SERPINE1, SERPINA1, CFTR, SPP1, COL1A1 and AGT, and the pathways of arachidonic acid metabolism, complement and coagulation cascades, and rheumatoid arthritis may potentially be used as diagnostic and therapeutic targets in EAC. However, the present study is limited to an extent due to the small sample size and lack of experimental validation. Further experimental confirmation of the expression profile in EAC by immunoblotting or immunohistochemical staining is therefore required to validate the current findings.

Acknowledgements

Not applicable.

Funding

Not applicable.

Availability of data and materials

The datasets used during the current study are available in the Gene Expression Omnibus database (accession no. GSE92396; http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE92396).

Authors' contributions

FH and BA designed the study. FH and LT analyzed and interpreted the data. FH was primarily responsible for writing the manuscript. All authors reviewed and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EAC

esophageal adenocarcinoma

GEO

Gene Expression Omnibus

DEGs

differentially expressed genes

PPI

protein-protein interaction

GO

gene ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

FC

fold-change

DAVID

Database for Annotation, Visualization and Integrated Discovery

IL

interleukin

IVL

involucrin

TIMP1

tissue inhibitor of metalloproteinase 1

FN1

fibronectin 1

SERPINE1

serpin family E member 1

SERPINA1

serpin family A member 1

CFTR

cystic fibrosis transmembrane conductance regulator

SPP1

secreted phosphoprotein 1

COL1A1

collagen type I alpha 1 chain

AGT

angiotensinogen

OPN

osteopontin

CXC

C-X-C motif

RAS

renin-angiotensin system

References

1 

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

2 

Reid BJ, Li X, Galipeau PC and Vaughan TL: Barrett's oesophagus and oesophageal adenocarcinoma: Time for a new synthesis. Nat Rev Cancer. 10:87–101. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Cowie A, Noble F and Underwood T: Strategies to improve outcomes in esophageal adenocarcinoma. Expert Rev Anticancer Ther. 14:677–687. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Kalatskaya I: Overview of major molecular alterations during progression from Barrett's esophagus to esophageal adenocarcinoma. Ann NY Acad Sci. 1381:74–91. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Ten Kate FJ, Suzuki L, Dorssers LC, Dinjens WN, Jones DT, Nieboer D, Doukas M, Van Lanschot JJ, Wijnhoven BP, Looijenga LH, et al: Pattern of p53 protein expression is predictive for survival in chemoradiotherapy-naive esophageal adenocarcinoma. Oncotarget. 8:104123–104135. 2017.PubMed/NCBI

6 

Buas MF, Levine DM, Makar KW, Utsugi H, Onstad L, Li X, Galipeau PC, Shaheen NJ, Hardie LJ, Romero Y, et al: Integrative post-genome-wide association analysis of CDKN2A and TP53 SNPs and risk of esophageal adenocarcinoma. Carcinogenesis. 35:2740–2747. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Wang L, Jin JQ, Zhou Y, Tian Z, Jablons DM and He B: Gli is activated and promotes epithelial-mesenchymal transition in human esophageal adenocarcinoma. Oncotarget. 9:853–865. 2017.PubMed/NCBI

8 

Lin J, Myers AL, Wang Z, Nancarrow DJ, Ferrer-Torres D, Handlogten A, Leverenz K, Bao J, Thomas DG, Wang TD, et al: Osteopontin (OPN/SPP1) isoforms collectively enhance tumor cell invasion and dissemination in esophageal adenocarcinoma. Oncotarget. 6:22239–22257. 2015.PubMed/NCBI

9 

Han DY, Fu D, Xi H, Li QY, Feng LJ, Zhang W, Ji G, Xiao JC and Wei Q: Genomic expression profiling and bioinformatics analysis of pancreatic cancer. Mol Med Rep. 12:4133–4140. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Zhang C, Peng L, Zhang Y, Liu Z, Li W, Chen S and Li G: The identification of key genes and pathways in hepatocellular carcinoma by bioinformatics analysis of high-throughput data. Med Oncol. 34:1012017. View Article : Google Scholar : PubMed/NCBI

11 

Peng D, Guo Y, Chen H, Zhao S, Washington K, Hu T, Shyr Y and El-Rifai W: Integrated molecular analysis reveals complex interactions between genomic and epigenomic alterations in esophageal adenocarcinomas. Sci Rep. 7:407292017. View Article : Google Scholar : PubMed/NCBI

12 

Gentleman R: Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Springer Science+Business Media; New York: 2005, View Article : Google Scholar

13 

Carvalho B, Bengtsson H, Speed TP and Irizarry RA: Exploration, normalization, and genotype calls of high-density oligonucleotide SNP array data. Biostatistics. 8:485–499. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:1–25. 2004. View Article : Google Scholar

15 

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

16 

Scardoni G, Tosadori G, Faizan M, Spoto F, Fabbri F and Laudanna C: Biological network analysis with CentiScaPe: Centralities and experimental dataset integration. F1000Res. 3:1392014.PubMed/NCBI

17 

Coleman HG, Xie SH and Lagergren J: The epidemiology of esophageal adenocarcinoma. Gastroenterology. 154:390–405. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Guglietta S and Rescigno M: Hypercoagulation and complement: Connected players in tumor development and metastases. Semin Immunol. 28:578–586. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Alfaro C, Sanmamed MF, Rodríguez-Ruiz ME, Teijeira Á, Oñate C, González Á, Ponz M, Schalper KA, Pérez-Gracia JL and Melero I: Interleukin-8 in cancer pathogenesis, treatment and follow-up. Cancer Treat Rev. 60:24–31. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Li A, Varney ML, Valasek J, Godfrey M, Dave BJ and Singh RK: Autocrine role of interleukin-8 in induction of endothelial cell proliferation, survival, migration and MMP-2 production and angiogenesis. Angiogenesis. 8:63–71. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Shrivastava MS, Hussain Z, Giricz O, Shenoy N, Polineni R, Maitra A and Verma A: Targeting chemokine pathways in esophageal adenocarcinoma. Cell Cycle. 13:3320–3327. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Xiaoyun X, Chaofei H, Weiqi Z, Chen C, Lixia L, Queping L, Cong P, Shuang Z, Juan S and Xiang C: Possible involvement of F1F0-ATP synthase and intracellular ATP in keratinocyte differentiation in normal skin and skin lesions. Sci Rep. 7:426722017. View Article : Google Scholar : PubMed/NCBI

23 

Bernard BA, Asselineau D, Schaffar-Deshayes L and Darmon MY: Abnormal sequence of expression of differentiation markers in psoriatic epidermis: Inversion of two steps in the differentiation program? J Invest Dermatol. 90:801–805. 1988. View Article : Google Scholar : PubMed/NCBI

24 

Shan J, Oshima T, Farre R, Fukui H, Watari J and Miwa H: IL-4 induces columnar-like differentiation of esophageal squamous epithelium through JAK/PI3K pathway: Possible role in pathogenesis of Barrett's esophagus. Am J Physiol Gastrointest Liver Physiol. 306:G641–G649. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Jackson HW, Defamie V, Waterhouse P and Khokha R: TIMPs: Versatile extracellular regulators in cancer. Nat Rev Cancer. 17:38–53. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Kozłowski M, Laudański W, Mroczko B, Szmitkowski M, Milewski R and Łapuć G: Serum tissue inhibitor of metalloproteinase 1 (TIMP-1) and vascular endothelial growth factor A (VEGF-A) are associated with prognosis in esophageal cancer patients. Adv Med Sci. 58:227–234. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Tian L, Lu ZP, Cai BB, Zhao LT, Qian D, Xu QC, Wu PF, Zhu Y, Zhang JJ, Du Q, et al: Activation of pancreatic stellate cells involves an EMT-like process. Int J Oncol. 48:783–792. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Pankov R and Yamada KM: Fibronectin at a glance. J Cell Sci. 115:3861–3863. 2002. View Article : Google Scholar : PubMed/NCBI

29 

McMahon B and Kwaan HC: The plasminogen activator system and cancer. Pathophysiol Haemost Thromb. 36:184–194. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Placencio VR and DeClerck YA: Plasminogen activator inhibitor-1 in cancer: Rationale and insight for future therapeutic testing. Cancer Res. 75:2969–2974. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Jänicke F, Schmitt M and Graeff H: Clinical relevance of the urokinase-type and tissue-type plasminogen activators and of their type 1 inhibitor in breast cancer. Semin Thromb Hemost. 17:303–312. 1991. View Article : Google Scholar : PubMed/NCBI

32 

Ortiz G, Salica JP, Chuluyan EH and Gallo JE: Diabetic retinopathy: Could the alpha-1 antitrypsin be a therapeutic option? Biol Res. 47:582014. View Article : Google Scholar : PubMed/NCBI

33 

Li Y, Miao L, Yu M, Shi M, Wang Y, Yang J, Xiao Y and Cai H: α1-antitrypsin promotes lung adenocarcinoma metastasis through upregulating fibronectin expression. Int J Oncol. 50:1955–1964. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Mutesa L, Azad AK, Verhaeghe C, Segers K, Vanbellinghen JF, Ngendahayo L, Rusingiza EK, Mutwa PR, Rulisa S, Koulischer L, et al: Genetic analysis of Rwandan patients with cystic fibrosis-like symptoms: Identification of novel cystic fibrosis transmembrane conductance regulator and epithelial sodium channel gene variants. Chest. 135:1233–1242. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Kerem B, Rommens JM, Buchanan JA, Markiewicz D, Cox TK, Chakravarti A, Buchwald M and Tsui LC: Identification of the cystic fibrosis gene: Genetic analysis. Science. 245:1073–1080. 1989. View Article : Google Scholar : PubMed/NCBI

36 

Gharahkhani P, Fitzgerald RC, Vaughan TL, Palles C, Gockel I, Tomlinson I, Buas MF, May A, Gerges C, Anders M, et al: Barrett's and Esophageal Adenocarcinoma Consortium (BEACON); Esophageal Adenocarcinoma GenEtics Consortium (EAGLE); Wellcome Trust Case Control Consortium 2 (WTCCC2): Genome-wide association studies in oesophageal adenocarcinoma and Barrett's oesophagus: A large-scale meta-analysis. Lancet Oncol. 17:1363–1373. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Ward LD and Kellis M: HaploReg: A resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 40:D930–D934. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Rittling SR and Singh R: Osteopontin in immune-mediated diseases. J Dent Res. 94:1638–1645. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Zhang Z, Wang Y, Zhang J, Zhong J and Yang R: COL1A1 promotes metastasis in colorectal cancer by regulating the WNT/PCP pathway. Mol Med Rep. 17:5037–5042. 2018.PubMed/NCBI

40 

Tian ZQ, Li ZH, Wen SW, Zhang YF, Li Y, Cheng JG and Wang GY: Identification of commonly dysregulated genes in non-small-cell lung cancer by integrated analysis of microarray data and qRT-PCR validation. Hai. 193:583–592. 2015.

41 

Song Y, Kim SH, Kim KM, Choi EK, Kim J and Seo HR: Activated hepatic stellate cells play pivotal roles in hepatocellular carcinoma cell chemoresistance and migration in multicellular tumor spheroids. Sci Rep. 6:367502016. View Article : Google Scholar : PubMed/NCBI

42 

Li J, Ding Y and Li A: Identification of COL1A1 and COL1A2 as candidate prognostic factors in gastric cancer. World J Surg Oncol. 14:2972016. View Article : Google Scholar : PubMed/NCBI

43 

Casselbrant A, Edebo A, Wennerblom J, Lönroth H, Helander HF, Vieth M, Lundell L and Fändriks L: Actions by angiotensin II on esophageal contractility in humans. Gastroenterology. 132:249–260. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Wang D and Dubois RN: Eicosanoids and cancer. Nat Rev Cancer. 10:181–193. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 9 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He F, Ai B and Tian L: Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis. Biomed Rep 9: 305-312, 2018
APA
He, F., Ai, B., & Tian, L. (2018). Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis. Biomedical Reports, 9, 305-312. https://doi.org/10.3892/br.2018.1134
MLA
He, F., Ai, B., Tian, L."Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis". Biomedical Reports 9.4 (2018): 305-312.
Chicago
He, F., Ai, B., Tian, L."Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis". Biomedical Reports 9, no. 4 (2018): 305-312. https://doi.org/10.3892/br.2018.1134