Open Access

Differentially expressed genes ASPN, COL1A1, FN1, VCAN and MUC5AC are potential prognostic biomarkers for gastric cancer

  • Authors:
    • Kaiyuan Jiang
    • Hongmei Liu
    • Dongyi Xie
    • Qiang Xiao
  • View Affiliations

  • Published online on: January 21, 2019     https://doi.org/10.3892/ol.2019.9952
  • Pages: 3191-3202
  • Copyright: © Jiang 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

Gastric cancer (GC) is one of the most common malignancies worldwide. To the best of our knowledge, no biomarkers have been widely accepted for the early diagnosis and prognostic prediction of GC. This study aimed to identify potential novel prognostic biomarkers for GC. The dataset GSE29272, which originates from the public database Gene Expression Omnibus, was employed in the present study. The online tool GEO2R was used to calculate the differentially expressed genes (DEGs) in GSE29272 between tumour tissues and adjacent tissues. CytoHubba and MCODE plugins of Cytoscape software were used to obtain hub genes and modules of DEGs. The online tools Database for Annotation, Visualisation and Integrated Discovery and Search Tool for the Retrieval of Interacting Genes were employed to conduct Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis, and to construct protein‑protein interaction networks. A total of 117 DEGs were extracted from GSE29272. In addition, 15 hub genes and seven modules were identified in the 117 DEGs. The enrichment analysis revealed that they were mainly enriched in GO biological process and cellular component domains, and the ‘ECM‑receptor interaction’, ‘focal adhesion’, ‘metabolism of xenobiotics by cytochrome P450’ and ‘drug metabolism’ pathways. The hub genes asporin (ASPN), collagen type I α1 chain (COL1A1), fibronectin 1 (FN1), versican (VCAN) and mucin 5AC (MUC5AC) were demonstrated to have prognostic value for patients with GC. The ASPN and VCAN genes were significantly associated with overall survival and disease‑free survival (log‑rank P=0.025, 0.038, 0.0014 and 0.015, respectively). COL1A1 and FN1 were significantly associated with overall survival (log‑rank P=0.013 and 0.05, respectively), and MUC5AC was significantly associated with disease‑free survival (log‑rank P=0.027). Results from the present study suggested that ASPN, COL1A1, FN1, VCAN and MUC5AC may represent novel prognostic biomarkers for GC.

Introduction

Gastric cancer (GC) is one of the most common causes of tumour-associated mortality worldwide (1). GC in East Asia represents ~50% of all GC cases (2). The high incidence of GC is partly due to the popular application of endoscopy (3). Despite advances in the diagnosis and treatment options of GC, the prognosis remains poor, and the 5-year survival rate of patients with GC is <20% (4). The commonly used biomarkers, carcinoembryonic antigen and cancer antigen 19-9, possess limited sensitivity and specificity in clinical application, which results in unsatisfactory levels of early diagnosis of GC (5).

Some molecules have been recently documented for their prognostic value in the screening and diagnosis of patients with GC. For instance, carbohydrate antigen 72-4 (CA72-4), an independent prognostic marker, has a good prognostic value for overall and relapse-free survival for patients with GC (6). As a prognostic marker, CA72-4 is widely used in various types of tumour, including pancreatic cancer, lung cancer and ovarian carcinoma (79). The sensitivity of CA72-4 in ovarian carcinoma is ~47% at the time of primary diagnosis (7). In addition, after analysis of the receiver operating characteristic curve of CA72-4, the area under the curve is ~88.4% in lung cancer (8). The sensitivity of CA72-4 is 25.5% in pancreatic cancer, which is higher than that in benign pancreatic disease (9). Although several proteins have recently been reported to be associated with prognosis for patients with GC, specific biomarkers for early diagnosis and prognostic assessment of GC are still lacking (1012). Therefore, establishing novel tumour markers with sufficient sensitivity and specificity is therefore crucial in order to improve the value of early diagnosis and prognostic prediction for patients with GC.

High-throughput sequencing (HTS) is an increasingly widely used tool that has significant roles in numerous life science fields, including early diagnosis, tumour grading and prognostic assessment (13). Databases containing HTS datasets are well acknowledged and have an increasingly significant role in the early diagnosis and prognostic prediction of various types of malignancy. The Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/) is a public functional genomics data source supporting Minimum Information About a Microarray Experiment-compliant data submissions. The GEO database contains array- and sequence-based data, providing users with experimental and curated gene expression information. In the present study, the GEO database was employed to identify novel prognostic biomarkers for patients with GC; these novel insights may aid in the development of individual treatments.

Materials and methods

Patient data collection

The GC expression profile dataset GSE29272 [GPL96, (HG-U133A), Affymetrix Human Genome U133A Array; Affymetrix; Thermo Fisher Scientific, Inc., Waltham, MA, USA] in the GEO database (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE29272, accessed January 10, 2018) was used in the present study. This dataset includes 134 normal gastric tissue samples and 134 GC tissue samples (14,15).

Data processing

The online tool, GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r/, accessed January 10, 2018) was applied to determine the differentially expressed genes (DEGs) in normal gastric tissues and GC tissues (16). Adjusted P-values were used to reduce the false positive rate using the Benjamini and Hochberg false discovery rate method by default. Adjusted P≤0.05 and |log fold change (FC)|≥1.5 were set as cut-off values. A total of 117 DEGs were then identified, including 43 upregulated and 74 downregulated genes. Eventually, the top 15 genes were determined as hub genes ranked by the Degree method in cytoHubba, a plugin in Cytoscape 3.6.0 software (17,18).

Enrichment analysis of tissue expression, gene ontology (GO) terms and kyoto encyclopedia of genes and genomes (KEGG) pathways of DEGs

GO analysis, including biological process (BP), cellular component (CC) and molecular function (MF) domains, is a tool widely used for annotating specific genes and gene products, and for assembling biological features for high-throughput genome and transcriptome data (19). KEGG is a database resource used to understand high-level functions and utilities of a biological system from molecular-level information by genome sequencing and other high-throughput experimental technologies (20). The Database for Annotation, Visualisation and Integrated Discovery (DAVID) version 6.7 (https://david-d.ncifcrf.gov/, accessed January 16, 2018) was used to identify detailed tissue expression, GO terms, including BP, CC, and MF, and KEGG pathways associated with the 117 DEGs (21,22).

Protein-protein interaction (PPI) network and modules analysis

The online resource Search Tool for the Retrieval of Interacting Genes (STRING' http://string-db.org/cgi/input.pl) was used to construct relationships for hub proteins. Subsequently, the Molecular Complex Detection (MCODE) plugin in Cytoscape 3.6.0 software was used to screen modules of the PPI network with the following default settings: Degree cut-off, 2; node score cut-off, 0.2; K-core, 2; maximum depth, 100. Eventually, enrichment analysis of GO terms and KEGG pathways was performed on DAVID using the 117 DEGs and the genes in the different modules.

Expression levels, correlation and survival analysis of hub genes

The online resource Gene Expression Profiling Interactive Analysis (GEPIA, http://gepia.cancer-pku.cn/index.html, accessed January 18, 2018), which originated from The Cancer Genome Atlas database, was used to determine the overall survival (OS) and disease-free survival (DFS) outcomes of hub genes (23). Furthermore, the genes associated with OS and/or DFS were applied for further analysis, including Pearson correlation analysis and analysis of expression levels in tumour and normal tissues.

Results

Identification of DEGs and hub genes

There were 134 GC tissues and 134 normal gastric tissue samples analysed in this study. Firstly, the GEO2R tool was employed to identify DEGs using the following cut-off values: Adjusted P≤0.05 and |logFC|≥1.5. As a result, a total of 117 DEGs were identified, including 43 upregulated and 74 downregulated genes. Subsequently, a PPI network of 117 DEGs was constructed using STRING (Fig. 1). Furthermore, 15 hub genes out of the 117 DEGs were determined by the Degree method using the following criteria: P≤0.05 and |log fold change (FC)|≥1.5. The 15 hub genes were as follows: Fibronectin 1 (FN1), TIMP metallopeptidase inhibitor 1, secreted phosphoprotein 1, matrix metalloproteinase 7, carbonic anhydrase 2, collagen type I α2 chain, secreted protein acidic and cysteine rich, mucin 5AC (MUC5AC), versican (VCAN), apolipoprotein E, collagen type III α1 chain, collagen type I α1 chain (COL1A1), thrombospondin 2, asporin (ASPN) and biglycan (Table I).

Table I.

Top 15 hub genes of differentially expressed genes, with high degrees.

Table I.

Top 15 hub genes of differentially expressed genes, with high degrees.

GeneDegreeAdjusted P-valueLog FC
FN139 7.92×10431.752388
TIMP136 1.87×10632.109944
SPP135 1.77×10412.640095
MMP733 2.05×10171.616738
CA233 1.22×1029−2.8229
COL1A231 4.10×10391.578489
SPARC30 7.63×10441.887773
MUC5AC30 2.12×1040−3.4951
VCAN30 7.31×10261.559823
APOE29 7.79×10351.561487
COL3A129 7.42×10421.625067
COL1A129 8.31×10331.51057
THBS227 8.67×10502.163794
ASPN27 4.46×10372.303273
BGN27 2.91×10501.728095

[i] FC, fold change.

Enrichment analysis of tissue expression, GO terms and KEGG pathways of the 117 DEGs

After importing the 43 upregulated and 74 downregulated genes into DAVID, the enriched results of tissue expression, GO term and KEGG pathway analyses were revealed. The 43 upregulated genes were enriched in tissues including colon endothelium, saliva, placenta, bone, plasma, fibroblasts, skin, cartilage, peripheral nervous system, liver and endometrial tumours. The 74 downregulated genes were enriched in tissues including stomach, liver, colon, pancreas, small intestine, stomach mucosa, prostate, foetal liver and erythrocytes (Table II).

Table II.

Enrichment analysis of 117 differentially expressed genes in different tissues.

Table II.

Enrichment analysis of 117 differentially expressed genes in different tissues.

A, Upregulated genes

TermCountPercent (%)P-valueFDR
Colon endothelium37.317073 1.99×10−040.217355
Saliva37.3170730.0057546.109932
Placenta1536.585370.0059726.334224
Bone37.3170730.0078018.200374
Plasma49.7560980.02313522.56379
Fibroblast37.3170730.03025528.51154
Skin921.951220.03173429.69333
Cartilage24.8780490.05428145.64904
PNS37.3170730.05677647.19527
Liver921.951220.0630350.89721
Endometrial tumour24.8780490.08820863.53566

B, Downregulated genes

TermCountPercent (%)P-valueFDR

Stomach1523.07692 1.02×10−14 1.10×10−11
Liver2233.84615 2.40×10−060.002588
Colon1421.53846 1.32×10−040.141746
Pancreas1116.923080.0016171.729784
Small intestine69.2307690.0054475.720101
Stomach mucosa34.6153850.0065186.809155
Prostate710.769230.01369713.81931
Foetal liver46.1538460.03371730.9166
Erythrocyte23.0769230.08308660.75539

[i] FDR, false discovery rate; PNS, peripheral nervous system; UP_TISSUE, tissue expression.

The top GO terms of the 43 upregulated genes were extracellular, including ‘proteinaceous extracellular matrix’, ‘extracellular matrix’, ‘collagen’, ‘extracellular matrix part’, ‘extracellular region’, ‘extracellular matrix structural constituent’, ‘fibrillar collagen’, ‘platelet-derived growth factor binding’ and ‘collagen fibril organization’. The top GO terms of the 74 downregulated genes were ‘digestion’, ‘cadmium ion binding’, ‘extracellular region’, ‘copper ion binding’, ‘response to inorganic substance’, ‘cellular aldehyde metabolic process’, ‘oxidation-reduction’, ‘extracellular space’, ‘response to metal ion’ and ‘aldo-keto reductase activity’ (Table III). Detailed results of the enriched GO terms included various terms from the BP, CC and MF domains (data not shown).

Table III.

Top 10 enriched Gene Ontology terms of upregulated and downregulated genes.

Table III.

Top 10 enriched Gene Ontology terms of upregulated and downregulated genes.

A, Upregulated genes

CategoryTermCountPercent (%)P-valueFDR
GOTERM_CC_FATExtracellular region part2356.097564.38E-164.88E-13
GOTERM_CC_FATProteinaceous extracellular matrix1639.024393.89E-154.27E-12
GOTERM_CC_FATExtracellular matrix1639.024391.19E-141.31E-11
GOTERM_CC_FATCollagen921.951224.88E-145.37E-11
GOTERM_CC_FATExtracellular matrix part1126.829277.91E-138.69E-10
GOTERM_CC_FATExtracellular region2663.414631.92E-122.11E-09
GOTERM_MF_FATExtracellular matrix structural constituent921.951221.85E-112.07E-08
GOTERM_CC_FATFibrillar collagen614.634151.20E-101.32E-07
GOTERM_MF_FATPlatelet-derived growth factor binding512.195128.67E-099.68E-06
GOTERM_BP_FATCollagen fibril organization614.634159.77E-091.46E-05

B, Downregulated genes

CategoryTermCountPercent (%)P-valueFDR

GOTERM_BP_FATDigestion1218.461546.44E-149.14E-11
GOTERM_MF_FATCadmium ion binding69.2307694.04E-105.11E-07
GOTERM_CC_FATExtracellular region2335.384621.89E-060.002038
GOTERM_MF_FATCopper ion binding69.2307691.47E-050.018561
GOTERM_BP_FATResponse to inorganic substance710.769231.76E-040.249357
GOTERM_BP_FATCellular aldehyde metabolic process46.1538461.94E-040.274926
GOTERM_BP_FAT Oxidation-reduction1015.384620.0010021.412983
GOTERM_CC_FATExtracellular space1015.384620.0011991.282203
GOTERM_BP_FATResponse to metal ion57.6923080.0018482.59269
GOTERM_MF_FATAldo-keto reductase activity34.6153850.0020522.565994

[i] BP, biological process; CC, cellular component; MF, molecular function; FDR, false discovery rate.

With regards to the KEGG pathway analysis of the 43 upregulated genes, three pathways were enriched: ‘ECM-receptor interaction’, ‘focal adhesion’ and ‘leukocyte transendothelial migration’. With regards to the KEGG pathway analysis of the 74 downregulated genes, four pathways were enriched: ‘metabolism of xenobiotics by cytochrome P450’, ‘drug metabolism’, ‘retinol metabolism’ and ‘nitrogen metabolism’. Detailed results are displayed in Table IV.

Table IV.

Enriched KEGG pathways of differentially expressed genes.

Table IV.

Enriched KEGG pathways of differentially expressed genes.

A, Upregulated genes

TermCountPercent (%)P-valueGenesFDR
ECM-receptor interaction1126.82927 2.66×10−13COL4A1, COL6A3, COL3A1, COL1A2, COL1A1, COL5A2, THBS2, COL11A1, COL5A1, SPP1, FN1   1.89×10−10
Focal adhesion1126.82927 1.80×10−09COL4A1, COL6A3, COL3A1, COL1A2, COL1A1, COL5A2, THBS2, COL11A1, COL5A1, SPP1, FN1   1.28×10-06
Leukocyte transendothelial migration  3   7.3170730.084213CLDN7, CLDN3, THY146.45857

B, Downregulated genes

TermCountPercent (%)P-valueGenesFDR

Metabolism of xenobiotics by cytochrome P450  5   7.692308 9.02×10−05GSTA1, CYP2C18, ADH1C, ADH1B, UGT2B15, ALDH3A10.082042
Drug metabolism  5   7.692308 1.03×10−04GSTA1, CYP2C18, ADH1C, ADH1B, UGT2B15, ALDH3A10.093347
Retinol metabolism  4   6.1538460.001315ALDH1A1, CYP2C18, ADH1C, ADH1B, UGT2B151.189785
Nitrogen metabolism  2   3.0769230.090981CA9, CA257.99834

[i] FDR, false discovery rate.

PPI network of hub genes and modules using MCODE

Following import of the 15 hub genes, the STRING website highlighted an interaction network. This network contains known interactions from curated databases and those that were experimentally determined; predicted interactions containing gene neighbourhood, gene fusions and gene co-occurrence; and text-mining, co-expression and protein homology (Fig. 2).

After importing the PPI network of 117 DEGs, Cytoscape displayed modules in the default MCODE settings. A total of seven modules were presented in the results (Fig. 3). Genes in these modules were then assembled for enrichment analysis using DAVID. Only two gave rise to KEGG pathways (Fig. 3B and G). Two pathways, ‘the ECM-receptor interaction’ and ‘focal adhesion’ pathways, were enriched by genes presented in Fig. 3A. Four pathways, ‘metabolism of xenobiotics by cytochrome P450’, ‘drug metabolism’, ‘tyrosine metabolism’, and ‘glycolysis/gluconeogenesis’, were enriched by the genes presented in Fig. 3F.

Survival curves, expression levels and correlation analysis of hub genes

All aforementioned 15 hub genes were analysed using the prognostic values of OS and DFS via the GEPIA website. ASPN and VCAN were significantly associated with OS and DFS (log-rank P=0.025, 0.038, 0.0014 and 0.015, respectively, Fig. 4A-D). COL1A1 and FN1 were significantly associated with OS (Log-rank P=0.013 and 0.05) (Fig. 4E and F). MUC5AC was significantly associated with DFS (Log-rank P=0.027, Fig. 4G). The analysis of these five genes revealed that low expression levels led to better survival status. The other hub genes did not exhibit statistical significance.

The genes ASPN, VCAN, COL1A1, FN1 and MUC5AC were then subjected to further analysis. Expression levels of these five genes are displayed in Fig. 5A-E. With the exception of MUC5AC, which exhibited low expression levels in GC tissues, the other four genes presented high expression levels in GC tissues. Furthermore, ASPN, VCAN, COL1A1 and FN1 had lower expression levels in normal gastric tissues. In addition, Pearson correlation analyses between the genes are presented in Fig. 5F-O. Results revealed that MUC5AC was negatively correlated with the four other genes: ASPN (R=−0.14, P=0.0042); COL1A1 (R=−0.092, P=0.062); FN1 (R=−0.15, P=0.0029); VCAN (R=−0.12, P=0.017). However, among the four other genes, each gene was positively correlated with the three other genes (all R>0, P<0.05).

Discussion

In the present study, the potential prognostic associations between GC and DEGs in GSE29272 were investigated. The results highlighted 117 DEGs, including 43 upregulated and 74 downregulated genes, between the 134 gastric normal tissues and the 134 GC tissues. A total of 15 hub genes were selected and seven modules were identified from the 117 DEGs. Some of these hub genes exhibited potential prognostic values for patients with GC.

GC is one of the most common types of tumour and ranks sixth among all tumours (24). Approximately 60% of newly diagnosed cases originate from Eastern Asia, particularly from China (25). There is currently a lack of sensitive biomarkers for the early diagnosis of GC. Therefore, many patients are in an advanced stage or have distant metastases at the time of diagnosis, resulting in poor prognosis (26). With a median OS of <1 year and a poor 5-year OS the prognosis remains unsatisfactory for patients with advanced stages of GC, and surgical resection is a common palliative treatment (27). Many studies have focused on the identification of novel biomarkers for early diagnosis and recurrence prediction of GC (2831). However, no widely accepted biomarkers have yet been discovered. Therefore, identifying novel and effective biomarkers for GC is crucial.

The present study revealed that some genes were differentially expressed between GC and normal tissues. A total of 43 genes were upregulated and 74 genes were downregulated. The DEGs then underwent tissue expression, GO term and KEGG pathway analyses. Both upregulated and downregulated genes were enriched in multiple organs. Notably, the stomach was the first organ highlighted in the enrichment analysis of the downregulated genes, and 15 genes were associated with this result. These genes were ATPase H+/K+ transporting subunit α, cholecystokinin B receptor, progastricsin, calpain 9, gastric intrinsic factor, mucin 6, aldehyde dehydrogenase 3 family member A1, chromogranin A, trefoil factor 2, cathepsin E, sulfotransferase family 1C member 2, MUC5AC, trefoil factor 1, gastrokine 1 and lipase F.

The majority of GO terms were enriched in CC terms in the top 10 upregulated genes. The enriched CC terms contained the ‘extracellular region part’, ‘proteinaceous extracellular matrix’, ‘extracellular matrix’, ‘collagen’, extracellular matrix part’, ‘extracellular region’ and ‘fibrillar collagen’. Furthermore, the majority of GO terms were enriched in BP terms in the top 10 downregulated genes. The enriched BP terms contained ‘digestion’, ‘response to inorganic substance’, ‘cellular aldehyde metabolic process’, ‘oxidation-reduction’ and ‘response to metal ions’. In the KEGG pathway analysis, upregulated and downregulated genes were enriched in four pathways. In the module analysis, a total of seven modules were identified by MCODE analysis of the 117 DEGs. The genes in these seven modules were then analysed for KEGG pathway enrichment. Six KEGG pathways were highlighted in only two modules. Notably, four pathways were presented in the results of both DEG enrichment and module analyses; these were ‘ECM-receptor interaction’, ‘focal adhesion’, ‘metabolism of xenobiotics by cytochrome P450’ and ‘drug metabolism’.

In the prognostic survival analysis, five of the 15 hub genes had prognostic values. ASPN and VCAN were associated with OS and DFS. COL1A1 and FN1 were associated with OS, whereas MUC5AC was associated with DFS. With the exception of MUC5AC, which exhibited low expression in GC, the four other genes exhibited high expression in GC tissues, and low expression in normal gastric tissues. These results suggest that MUC5AC, ASPN, COL1A1, FN1 and VCAN may serve oncogenic roles in gastric cancer. These genes also serve numerous functions, possibly via BP, CC and the aforementioned four pathways.

Secreted MUC5AC is commonly expressed in microsatellite instability (MSI) cancers (32). Expression of MUC5AC usually occurs in mucinous and MSI carcinomas (33). Renaud et al (34) reported that abnormal expression levels of MUC5AC in high CpG island methylation phenotype/MSI colorectal cancer (CRC) is closely associated with altered methylation of their promoters. Notably, MUC5AC is associated with the absence of tumour protein 53 mutations, and when combined with mucin 2, is associated with poor differentiation and MSI status (34). In addition, the hypomethylation of the proximal region of the MUC5AC promoter (MUC5AC-I) is not associated with MUC5AC protein expression (14,32,34). MUC5AC hypomethylation is a highly predictive biomarker and a specific regulatory mechanism of MSI cancers (34), whereas the determination of MUC5AC methylation status may be important for understanding and predicting the natural history of CRC (34). Renaud et al (35), suggested that MUC5AC hypomethylation can serve as a biomarker to identify serrated pathway neoplasia-associated precursors. Numerous studies strongly suggested that MUC5AC generates the major receptor for Helicobacter pylori in the human stomach (36,37) and that infection with H. pylori can modify expression levels of MUC5AC (38). Zhou et al (39) reported that common polymorphisms of MUC5AC are associated with the risk of non-cardia GC in a Chinese population. MUC5AC expression is also associated with Notch3 signalling, which provides an encouraging prognosis in patients with small intestine adenocarcinomas (40). The present study hypothesized that MUC5AC may serve a oncogenic role, which was inconsistent with the findings of Kim et al (41), who stated that the decreased expression of MUC5AC is associated with poor prognosis in GC. This inconsistency may be due to the small sample size of the present study; therefore, further investigations regarding to the role of MUC5AC are required.

ASPN has been widely explored in osteoarthritis, and is finely regulated in cartilage (42). ASPN is also implicated in the mechanisms of local invasion of breast ductal carcinoma (43). In addition, ASPN is overexpressed in pancreatic ductal adenocarcinoma, suggesting that it is a good candidate for diagnostic and therapeutic application (44). ASPN also participates in GC cell growth and migration by influencing epidermal growth factor receptor (EGFR) receptor signalling (45). The present study hypothesized that ASPN may have an oncogenic role in gastric tumours, which is consistent with a previous study reporting that ASPN serves an oncogenic role in GC progression and metastasis via the EGFR signalling pathway (45).

Two mutations (c.3235G>A and c.3247G>A) occur simultaneously in COL1A1 and lead to type IV osteogenesis imperfecta (46). In addition, a novel mutation in the start codon of COL1A1 causes osteogenesis imperfecta type I in a Korean family (47). COL1A1 C-propeptide cleavage site mutation also leads to high bone mass, bone fragility and jaw lesions (48). Furthermore, COL1A1 has been incorporated in fibroblasts as a molecular signature by hCellMarkerPlex, which indicates COL1A1 is associated with fibroblasts and functions as a molecular signature (49). Previous studies have suggested that COL1A1 is a candidate survival-related factor in hepatocellular carcinoma (50). In addition, COL1A1 polymorphism is associated with an elevated risk of osteosarcoma susceptibility and mortality (51).

FN1 is a novel fusion partner of anaplastic lymphoma kinase in inflammatory myofibroblastic tumours (52). FN1-EGF gene fusions are recurrent in calcifying aponeurotic fibroma (53). In addition, the FN1-fibroblast growth factor receptor 1 genetic fusion is a frequent event in phosphaturic mesenchymal tumours (54). A single nucleotide polymorphism in FN1 has also been reported to be associated with tumour shape in CRCs (55). In addition, FN1 may interact with vascular endothelial growth factor A and serve important roles in non-small cell lung cancer (NSCLC), and the corresponding proteins can serve as targets for the diagnosis or treatment of patients with NSCLC (56). The overexpression of FN1 is also associated with latent membrane protein 1 expression and has an independent prognostic value for nasopharyngeal cancer (57).

VCAN has potential prognostic value in multiple myeloma (58). VCAN expression levels are also associated with OS in CRC (59), and it has been identified as a potential biomarker for oral squamous cell carcinoma (60). The present study hypothesized that VCAN may have an oncogenic role in GC, which is not consistent with a report by Kim et al (61), stating that VCAN expression predicts a good prognosis for patients with GC.

The present study presented limitations. Firstly, a larger population is required in order to increase the credibility of the present findings. Secondly, other influencing factors, including pathological types, tumour size, tumour numbers and microvascular invasion, should be included, in order to better evaluate the association between DEGs and GC prognosis. Thirdly, a better-designed study focusing on the functional validation of these genes and including more ethnicities is required, combined with a greater number of research centres. Finally, future studies should increase the amount of data obtained from databases, include pathological types of GC and analyze the correlation of clinical stage classification.

In the present study, the plugins cytoHubba and MCODE of Cytoscape were used to obtain hub genes, which may represent predictive biomarkers for GC. Furthermore, DAVID and STRING were used to determine the biological processes and metabolic pathways in which these hub genes were involved. The expression levels and prognostic values of the hub genes were eventually analyzed. The present study aimed to determine potential predictive biomarkers for GC. The results demonstrated that some hub genes possessed some prognostic value for GC. Further studies focusing on the functional validation of these genes are required (via reverse transcription-quantitative polymerase chain reaction and western blotting) and should include a larger number of medical centres and more ethnicities.

In conclusion, the present study identified 117 DEGs in patients with GC and identified 15 hub genes. In addition, some hub genes had prognostic value for patients with GC. The present study suggested that ASPN, COL1A1, FN1, VCAN and MUC5AC may represent potential prognostic biomarkers for GC. In addition, ASPN, COL1A1, FN1 and VCAN may serve oncogenic roles in gastric tumours, whereas MUC5AC may act as a tumour suppressor. The genes may act via BP and CC domains, and via ‘ECM-receptor interaction’, ‘focal adhesion’, ‘metabolism of xenobiotics by cytochrome P450’ and ‘drug metabolism’ pathways. In the present study, some hub genes were differentially expressed and had prognostic value for GC. Further studies are required to explore the functional roles of these genes, particularly in the development of metastases and cancer progression, in order to guide clinical direction.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. 81660511) and The Guangxi Natural Science Foundation of Key Projects (grant no. 2015GXNSFDA227001).

Availability of data and materials

The datasets generated and/or/analyzed during the present study are available in the GEO (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE29272) and GEPIA (http://gepia.cancer-pku.cn/index.html) repositories.

Authors' contributions

KJ and QX were responsible for study design. KJ, HL and DX conducted the study and analysed the data. KJ wrote the manuscript and QX guided the writing.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Conflicts of interest

The authors declare they have no competing interests.

References

1 

Heinebröring RC, Winkels RM, Renkema JM, Kragt L, van Orten-Luiten AC, Tigchelaar EF, Chan DS, Norat T and Kampman E: Dietary supplement use and colorectal cancer risk: A systematic review and meta-analyses of prospective cohort studies. Int J Cancer. 136:2388–2401. 2015. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Leung WK, Wu MS, Kakugawa Y, Kim JJ, Yeoh KG, Goh KL, Wu KC, Wu DC, Sollano J, Kachintorn U, et al: Screening for gastric cancer in Asia: Current evidence and practice. Lancet Oncol. 9:279–287. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Shen W, Li J, Cui J, Xi H, Liu S, Wei B and Chen L: Meta-analysis of prognosis after surgical treatment in GC patients with liver metastasis. Zhonghua Wei Chang Wai Ke Za Zhi. 17:128–132. 2014.(In Chinese). PubMed/NCBI

5 

Xu H, Zhang M and Qiao H: Diagnostic significance of miR-106a in GC. Int J Clin Exp Pathol. 8:13096–13101. 2015.PubMed/NCBI

6 

Yoon SK, Yoo C, Ryu MH, Ryoo BY, Chang HM, Kim TW, Lee JL, Yook JH, Oh ST, Kim BS and Kang YK: 6525 Prognostic significance of preoperative serum tumor markers in the patients with curatively resected advanced gastric cancer patients. J Clin Oncol. 7:3692009.

7 

Hasholzner U, Baumgartner L, Stieber P, Meier W, Reiter W, Pahl H and Fateh-Moghadam A: Clinical significance of the tumour markers CA 125 II and CA 72-4 in ovarian carcinoma. Int J Cancer. 69:329–334. 1996. View Article : Google Scholar : PubMed/NCBI

8 

Miédougé M, Rouzaud P, Salama G, Pujazon MC, Vincent C, Mauduyt MA, Reyre J, Carles P and Serre G: Evaluation of seven tumour markers in pleural fluid for the diagnosis of malignant effusions. Br J Cancer. 81:1059–1065. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Luo G, Liu C, Guo M, Cheng H, Lu Y, Jin K, Liu L, Long J, Xu J, Lu R, et al: Potential biomarkers in lewis negative patients with pancreatic cancer. Ann Surg. 265:800–805. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Bian Y, Wang Z, Xu J, Zhao W, Cao H and Zhang Z: Elevated Rictor expression is associated with tumor progression and poor prognosis in patients with gastric cancer. Biochem Biophys Res Commun. 464:534–540. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Gui C, Min L, Jun W, Ji M, Fang C, Shi H, Zhu D, Chen L, Zhao J, Shi L, et al: Expression of Tim-3 in gastric cancer tissue and its relationship with prognosis. Int J Clin Exp Pathol. 8:9452–9457. 2015.PubMed/NCBI

12 

Jiang W, Jin Z, Zhou F, Cui J and Wang L and Wang L: High co-expression of Sp1 and HER-2 is correlated with poor prognosis of gastric cancer patients. Surg Oncol. 24:220–225. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Kulasingam V and Diamandis EP: Strategies for discovering novel cancer biomarkers through utilization of emerging technologies. Nat Clin Pract Oncol. 5:588–599. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Wang G, Hu N, Yang HH, Wang L, Su H, Wang C, Clifford R, Dawsey EM, Li JM, Ding T, et al: Comparison of global gene expression of gastric cardia and noncardia cancers from a high-risk population in china. PLoS One. 8:e638262013. View Article : Google Scholar : PubMed/NCBI

15 

Li WQ, Hu N, Burton VH, Yang HH, Su H, Conway CM, Wang L, Wang C, Ding T, Xu Y, et al: PLCE1 mRNA and protein expression and survival of patients with esophageal squamous cell carcinoma and gastric adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 23:1579–1588. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Davis S and Meltzer PS: GEOquery: A bridge between the gene expression omnibus (GEO) and BioConductor. Bioinformatics. 23:1846–1847. 2007. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

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

19 

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

20 

Minoru K and Susumu G: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 27:27–30. 2000.

21 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:(Web Server Issue). W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Cao GD, Xu XY, Zhang JW, Chen B and Xiong MM: Phosphorylated mammalian target of rapamycin p-mTOR is a favorable prognostic factor than mTOR in gastric cancer. PLoS One. 11:e01680852016. View Article : Google Scholar : PubMed/NCBI

25 

Sasako M, Sano T, Yamamoto S, Kurokawa Y, Nashimoto A, Kurita A, Hiratsuka M, Tsujinaka T, Kinoshita T, Arai K, et al: D2 Lymphadenectomy Alone or with Para-aortic Nodal Dissection for gastric cancer. N Engl J Med. 359:453–462. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Zou WB, Yang F and Li ZS: How to improve the diagnosis rate of early gastric cancer in China. Zhejiang Da Xue Xue Bao Yi Xue Ban. 44:9–14. 2015.(In Chinese). PubMed/NCBI

27 

Kamangar F, Dores GM and Anderson WF: Patterns of cancer incidence, mortality, and prevalence across five continents: Defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol. 24:2137–2150. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Tsai MM, Wang CS, Tsai CY, Huang HW, Chi HC, Lin YH, Lu PH and Lin KH: Potential diagnostic, prognostic and therapeutic targets of MicroRNAs in human gastric cancer. Int J Mol Sci. 17(pii): E9452016. View Article : Google Scholar : PubMed/NCBI

29 

Azarbarzin S, Feizi MAH, Safaralizadeh R, Kazemzadeh M and Fateh A: The value of MiR-383, an intronic MiRNA, as a diagnostic and prognostic biomarker in intestinal-type gastric cancer. Biochem Genet. 55:244–252. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Singh P, Toom S and Huang Y: Anti-claudin 18.2 antibody as new targeted therapy for advanced gastric cancer. J Hematol Oncol. 10:1052017. View Article : Google Scholar : PubMed/NCBI

31 

Chen SL, Li JP, Li LF, Zeng T and He X: Elevated preoperative serum alanine aminotransferase/aspartate aminotransferase (ALT/AST) ratio is associated with better prognosis in patients undergoing curative treatment for gastric adenocarcinoma. Int J Mol Sci. 17(pii): E9112016. View Article : Google Scholar : PubMed/NCBI

32 

Biemer-Hüttmann AE, Walsh MD, McGuckin MA, Simms LA, Young J, Leggett BA and Jass JR: Mucin core protein expression in colorectal cancers with high levels of microsatellite instability indicates a novel pathway of morphogenesis. Clin Cancer Res. 6:1909–1916. 2000.PubMed/NCBI

33 

Kim DH, Kim JW, Cho JH, Baek SH, Kakar S, Kim GE, Sleisenger MH and Kim YS: Expression of mucin core proteins, trefoil factors, APC and p21 in subsets of colorectal polyps and cancers suggests a distinct pathway of pathogenesis of mucinous carcinoma of the colorectum. Int J Oncol. 27:957–964. 2005.PubMed/NCBI

34 

Renaud F, Vincent A, Mariette C, Crépin M, Stechly L, Truant S, Copin MC, Porchet N, Leteurtre E, Van Seuningen I and Buisine MP: MUC5AC hypomethylation is a predictor of microsatellite instability independently of clinical factors associated with colorectal cancer. Int J Cancer. 136:2811–2821. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Renaud F, Mariette C, Vincent A, Wacrenier A, Maunoury V, Leclerc J, Coppin L, Crépin M, Van Seuningen I, Leteurtre E and Buisine MP: The serrated neoplasia pathway of colorectal tumors: Identification of MUC5AC hypomethylation as an early marker of polyps with malignant potential. Int J Cancer. 138:1472–1481. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Lindén SK, Wickström C, Lindell G, Gilshenan K and Carlstedt I: Four modes of adhesion are used during Helicobacter pylori binding to human mucins in the oral and gastric niches. Helicobacter. 13:81–93. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Van de Bovenkamp JH, Mahdavi J, Korteland-Van Male AM, Büller HA, Einerhand AW, Borén T and Dekker J: The MUC5AC glycoprotein is the primary receptor for Helicobacter pylori in the human stomach. Helicobacter. 8:521–532. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Kocer B, Ulas M, Ustundag Y, Erdogan S, Karabeyoglu M, Yldrm O, Unal B, Cengiz O and Soran A: A confirmatory report for the close interaction of Helicobacter pylori with gastric epithelial MUC5AC expression. J Clin Gastroenterol. 38:496–502. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Zhou CJ, Zhang LW, Gao F, Zhang B, Wang Y, Chen DF and Jia YB: Association analysis of common genetic variations in MUC5AC gene with the risk of non-cardia gastric cancer in a Chinese population. Asian Pac J Cancer Prev. 15:4207–4210. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Eom DW, Hong SM, Kim J, Kim G, Bae YK, Jang KT and Yu E: Notch3 signaling is associated with MUC5AC expression and favorable prognosis in patients with small intestinal adenocarcinomas. Pathol Res Pract. 210:501–507. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Kim SM, Kwon CH, Shin N, Park DY, Moon HJ, Kim GH and Jeon TY: Decreased Muc5AC expression is associated with poor prognosis in gastric cancer. Int J Cancer. 134:114–124. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Duval E, Bigot N, Hervieu M, Kou I, Leclercq S, Galéra P, Boumediene K and Baugé C: Asporin expression is highly regulated in human chondrocytes. Mol Med. 17:816–823. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Castellana B, Escuin D, Peiró G, Garcia-Valdecasas B, Vázquez T, Pons C, Pérez-Olabarria M, Barnadas A and Lerma E: ASPN and GJB2 are implicated in the mechanisms of invasion of ductal breast carcinomas. J Cancer. 3:175–183. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Turtoi A, Musmeci D, Wang Y, Dumont B, Somja J, Bevilacqua G, De Pauw E, Delvenne P and Castronovo V: Identification of novel accessible proteins bearing diagnostic and therapeutic potential in human pancreatic ductal adenocarcinoma. J Proteome Res. 10:4302–4313. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Ding Q, Zhang M and Liu C: Asporin participates in gastric cancer cell growth and migration by influencing EGF receptor signaling. Oncol Rep. 33:1783–1790. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Ju M, Zhang T, Bai X, Ren X, Li K and Li G: Analysis of type IV osteogenesis imperfecta caused by two mutations occurred simultaneously in COL1A1 gene in a Chinese child. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 33:140–144. 2016.(In Chinese). PubMed/NCBI

47 

Cho SY, Lee JH, Ki CS, Chang MS, Jin DK and Han HS: Osteogenesis imperfecta Type I caused by a novel mutation in the start codon of the COL1A1 gene in a Korean family. Ann Clin Lab Sci. 45:100–105. 2015.PubMed/NCBI

48 

McInerney-Leo AM, Duncan EL, Leo PJ, Gardiner B, Bradbury LA, Harris JE, Clark GR, Brown MA and Zankl A: COL1A1 C-propeptide cleavage site mutation causes high bone mass, bone fragility and jaw lesions: A new cause of gnathodiaphyseal dysplasia? Clin Genet. 88:49–55. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Drew JE, Farquharson AJ, Mayer CD, Vase HF, Coates PJ, Steele RJ and Carey FA: Predictive gene signatures: Molecular markers distinguishing colon adenomatous polyp and carcinoma. PLoS One. 9:e1130712014. View Article : Google Scholar : PubMed/NCBI

50 

Hayashi M, Nomoto S, Hishida M, Inokawa Y, Kanda M, Okamura Y, Nishikawa Y, Tanaka C, Kobayashi D, Yamada S, et al: Identification of the collagen type 1 α 1 gene (COL1A1) as a candidate survival-related factor associated with hepatocellular carcinoma. BMC Cancer. 14:1082014. View Article : Google Scholar : PubMed/NCBI

51 

He M, Wang Z, Zhao J, Chen Y and Wu Y: COL1A1 polymorphism is associated with risks of osteosarcoma susceptibility and death. Tumour Biol. 35:1297–1305. 2014. View Article : Google Scholar : PubMed/NCBI

52 

Ouchi K, Miyachi M, Tsuma Y, Tsuchiya K, Iehara T, Konishi E, Yanagisawa A and Hosoi H: FN1: A novel fusion partner of ALK in an inflammatory myofibroblastic tumor. Pediatr Blood Cancer. 62:909–911. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Puls F, Hofvander J, Magnusson L, Nilsson J, Haywood E, Sumathi VP, Mangham DC, Kindblom LG and Mertens F: FN1-EGF gene fusions are recurrent in calcifying aponeurotic fibroma. J Pathol. 238:502–507. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Lee JC, Jeng YM, Su SY, Wu CT, Tsai KS, Lee CH, Lin CY, Carter JM, Huang JW, Chen SH, et al: Identification of a novel FN1-FGFR1 genetic fusion as a frequent event in phosphaturic mesenchymal tumour. J Pathol. 235:539–545. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Kida H, Takano Y, Yamamoto K, Mori M, Yanaga K, Tanaka J, Kudo SE and Mimori K: A single nucleotide polymorphism in fibronectin 1 determines tumor shape in colorectal cancer. Oncol Rep. 32:548–552. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Wang Y, Huang L, Wu S, Jia Y, Yang Y, Luo L, Bi A and Fang M: Bioinformatics analyses of the role of vascular endothelial growth factor in patients with non-small cell lung cancer. PLoS One. 10:e01392852015. View Article : Google Scholar : PubMed/NCBI

57 

Ma LJ, Lee SW, Lin LC, Chen TJ, Chang IW, Hsu HP, Chang KY, Huang HY and Li CF: Fibronectin overexpression is associated with latent membrane protein 1 expression and has independent prognostic value for nasopharyngeal carcinoma. Tumour Biol. 35:1703–1712. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Gupta N, Khan R, Kumar R, Kumar L and Sharma A: Versican and its associated molecules: Potential diagnostic markers for multiple myeloma. Clin Chim Acta. 442:119–124. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Sluiter N, de Cuba E, Kwakman R, Meijerink WJ, Delis-van Diemen PM, Coupé VM, Beliën JA, Meijer GA, de Hingh IH and te Velde EA: Versican and vascular endothelial growth factor expression levels in peritoneal metastases from colorectal cancer are associated with survival after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Clin Exp Metastasis. 33:297–307. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Yen CY, Huang CY, Hou MF, Yang YH, Chang CH, Huang HW, Chen CH and Chang HW: Evaluating the performance of fibronectin 1 (FN1), integrin α4β1 (ITGA4), syndecan-2 (SDC2), and glycoprotein CD44 as the potential biomarkers of oral squamous cell carcinoma (OSCC). Biomarkers. 18:63–72. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Kim NS, Lee HH, Jung CK and Jeon HM: Versican expression in tumor epithelial cells is correlated with a good prognosis in gastric cancer. Anticancer Res. 34:5613–5619. 2014.PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 17 Issue 3

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
Jiang K, Liu H, Xie D and Xiao Q: Differentially expressed genes ASPN, COL1A1, FN1, VCAN and MUC5AC are potential prognostic biomarkers for gastric cancer. Oncol Lett 17: 3191-3202, 2019
APA
Jiang, K., Liu, H., Xie, D., & Xiao, Q. (2019). Differentially expressed genes ASPN, COL1A1, FN1, VCAN and MUC5AC are potential prognostic biomarkers for gastric cancer. Oncology Letters, 17, 3191-3202. https://doi.org/10.3892/ol.2019.9952
MLA
Jiang, K., Liu, H., Xie, D., Xiao, Q."Differentially expressed genes ASPN, COL1A1, FN1, VCAN and MUC5AC are potential prognostic biomarkers for gastric cancer". Oncology Letters 17.3 (2019): 3191-3202.
Chicago
Jiang, K., Liu, H., Xie, D., Xiao, Q."Differentially expressed genes ASPN, COL1A1, FN1, VCAN and MUC5AC are potential prognostic biomarkers for gastric cancer". Oncology Letters 17, no. 3 (2019): 3191-3202. https://doi.org/10.3892/ol.2019.9952