Open Access

Identification of the lncRNA‑miRNA‑mRNA network associated with gastric cancer via integrated bioinformatics analysis

  • Authors:
    • Xiao‑Yu Ma
    • Yu Ma
    • Huan Zhou
    • Hui‑Jing Zhang
    • Ming‑Jun Sun
  • View Affiliations

  • Published online on: September 25, 2019     https://doi.org/10.3892/ol.2019.10922
  • Pages: 5769-5784
  • Copyright: © Ma 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 investigate the long non‑coding RNA (lncRNA)‑microRNA (miRNA)‑mRNA regulatory network in gastric cancer (GC) using bioinformatics analysis. Two mRNA gene expression profiles, GSE79973 and GSE54129, and two miRNA expression profiles, GSE93415 and GSE78091, were downloaded from the Gene Expression Omnibus database. The differentially expressed mRNAs (DEMs) and the differentially expressed miRNAs (DEMis) were merged separately. Gene ontology and pathway enrichment analysis were conducted using the Database for Annotation, Visualization and Integrated Discovery. A protein‑protein interaction (PPI) network was then constructed and the 10 top hub genes in the network were analyzed using the Search Tool for the Retrieval of Interacting Genes. The lncRNA‑miRNA‑mRNA networks were visualized using Cytoscape software. As a result, 158 shared DEMs (40 upregulated and 118 downregulated) were identified from two mRNA datasets. A total of 30 upregulated miRNAs and 1 downregulated miRNA functioned as DEMis. The PPI network consisted of 129 nodes and 572 interactions. The 10 top hub genes were selected by degree using Cytohubba, including Jun proto‑oncogene, mitogen‑activated protein kinase (MAPK)3, transforming growth factor‑β1, Fos proto‑oncogene, AP‑1 transcription factor subunit, interleukin (IL)‑8, MAPK1, RELA proto‑oncogene nuclear factor‑κB subunit, interferon regulatory factor 7, ubiquitin like modifier and vascular endothelial growth factor A. In the lncRNA‑miRNA‑mRNA network, a total of 1,215 regulatory associations were constructed using Cytoscape. In conclusion, the present study provides a novel perspective of the molecular mechanisms underlying GC by identifying the lncRNA‑miRNA‑mRNA regulatory network via bioinformatics analysis.

Introduction

Gastric cancer (GC) is one of the most common malignant diseases globally (1). Although substantial advances have been made in the diagnosis and therapy of this disease, the prognosis of GC remains poor and the 5-year survival rate is still comparatively low (2,3). Therefore, it is essential to investigate the molecular mechanism including potential biomarkers and therapeutic targets of GC.

In previous years, a large number of microarrays and bioinformatics methods have been conducted to investigate the molecular mechanism underlying cancer progression including diagnosis, treatment and prognosis (46). For example, bioinformatics analysis has been used to elucidate the potential key candidate genes and pathways in colorectal cancer from four cohort profile datasets (7). In addition, target genes and the prognostic value in non-small cell lung cancer have been discovered previously via bioinformatics analysis (8). Similarly, bioinformatics analysis has been performed to identify long non-coding RNA (lnc-RNA)-microRNA (miRNA/miR)-mRNA networks via the combination of lncRNA, miRNA and mRNA expression profiles based on competitive endogenous RNA in rheumatoid arthritis (9). In conclusion, it is necessary to perform further investigation of the molecular mechanism underlying GC using integrated bioinformatics analysis.

In the present study, differentially expressed mRNAs (DEMs) and differentially expressed miRNAs (DEMis) were screened out from the Gene Expression Omnibus (GEO) database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of DEMs were also performed. The lncRNA-miRNA-mRNA network was subsequently established, which may provide additional information on the molecular mechanism of GC.

Materials and methods

Gene expression profiles

Two human mRNA expression profiles, GSE79973 and GSE54129, were acquired from the GEO database (ncbi.nlm.nih.gov/geo/) (10), which included 121 GC and 31 normal samples. Two human miRNA expression profiles, GSE93415 and GSE78091, were also downloaded from the GEO database, which included 23 GC and 23 normal samples. FunRich version 3.1.3 was used to draw the venn diagram (11).

Identification of DEMs and DEMis

Two mRNA and two miRNA databases were analyzed using the GEO2R web tool comparing samples in the GC and control groups (12). In order to select the DEMs, an adjusted (adj.) P-value of ≤0.05 and |log2 fold change (FC)| ≥2 were selected as the cut-off values for the two mRNA databases. For DEMis, adj.P-value ≤0.05 and |log2FC| ≥1 were regarded as the cut-off criteria values.

Function analysis of DEMs

GO function and KEGG pathway enrichment analysis of the up- and downregulated genes and 10 hub genes were analyzed using the online Database for Annotation, Visualization and Integrated Discovery (DAVID; david.abcc.ncifcrf.gov/). P<0.05 was considered to indicate a statistically signicant difference.

Protein-protein interaction (PPI) network construction and sub-network identification

Search Tool for the Retrieval of Interacting Genes (STRING; string-db.org/) was used to analyze the PPI network of DEMs (13). The PPIs of DEMs were selected using a combined score of >0.9. Cytoscape version 3.5.1 (cytoscape.org/) software was utilized to construct the PPI network (14). Cytohubba in Cytoscape software was employed to identify the 10 top hub genes by degree. The sub-network in the PPI network was then visualized by MCODE with a cut-off criterion of k-score=2 (15). P<0.05 was considered to indicate a statistically signicant difference.

Survival analysis of the 10 top hub genes

The publicly available database Kaplan-Meier Plotter (www.kmplot.com) was used to identify the prognostic effect of the 10 top hub genes (16). P<0.05 was considered to indicate a statistically signicant difference.

Determining the expression level of the significantly up- and downregulated DEMs

A total of 5 significantly upregulated DEMs (inhibin bA, collagen type VIII α1, secreted frizzle related protein 4, secreted phosphoprotein 1 and thormbospondin 2) and 5 significantly downregulated DEMs [Gastrokine 2 (GKN2), GKN1, gastric intrinsic factor, Mucin like 3 and Keratin 20 (KRT20)] were selected for further analysis. The Gene Expression Profiling Interactive Analysis (GEPIA; gepia.cancer-pku.cn/) database was used to determine the expression of 10 DEMs in GC tissues (17). In order to provide more sufficient evidence to support the results, the present study investigated the data in the Cancer Cell Line Encyclopedia (CCLE) portals (broadinstitute.org/ccle/about) database, which supplied information on the expression of 10 DEMs in GC cell lines (18). P<0.05 was considered to indicate a statistically signicant difference.

Reconstruction of the lncRNA-miRNA-mRNA networks

The miRNA-targeted genes were screened out using the miRWalk database (mirwalk.umm.uni-heidelberg.de/) with a score of >0.9 (19,20). The targeted genes and DEMs mentioned above were merged to identify the number of miRNA-regulated target gene pairs. The prediction of lncRNA-miRNA interactions was based on the analysis of the lncRNASNP (www.lncRNAblog.com) database (21). The networks between miRNA-mRNA and lncRNA-miRNA were visualized using Cytoscape version 3.5.1. P<0.05 was considered to indicate a statistically signicant difference.

Statistical analysis

All statistical analysis was performed using corresponding databases. Data was presented as the mean ± standard deviation. A paired Student's t-test or one-way analysis of variance (ANOVA) or two-way ANOVA were used to evaluate the degree of differential expression. A log-rank test was applied to analyse the association between expression and prognosis. Post-hoc tests (least significant difference and Tukey's tests) were performed following ANOVA. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of DEMs and DEMis

A total of 158 DEMs (5 of the 163 genes were identified as unreconized genes) were identified based on the cut-off criteria using FunRich, including 40 upregulated DEMs and 118 downregulated DEMs (Fig. 1A). Furthermore, 31 DEMis were obtained between GC and control samples. A total of 30 upregulated miRNAs and 1 downregulated miRNA were identified as DEMis (Fig. 1B). All DEMs and DEMis are presented in Table I.

Table I.

158 DEMs were obtained from two datasets, including 40 upregulated DEMs and 118 downregulated DEMs. 31 DEMis were obtained between gastric cancer samples and control samples, including 30 upregulated miRNAs and 1 downregulated miRNA.

Table I.

158 DEMs were obtained from two datasets, including 40 upregulated DEMs and 118 downregulated DEMs. 31 DEMis were obtained between gastric cancer samples and control samples, including 30 upregulated miRNAs and 1 downregulated miRNA.

DEMsGene name
UpregulatedCOL11A1, INHBA, IGF2BP3, COL10A1, FNDC1, FAP, THBS2, SULF1, CST1, COL8A1, SFRP4, SPP1, COL1A1, ADAMTS2, WISP1, CTHRC1, COL12A1, ASPN, CRISPLD1, THY1, COL1A2, FN1, BGN, RARRES1, CAP2, MFAP2, PDPN, PRRX1, TIMP1, SPARC, COL6A3, COL4A1, THBS1, NRP2, PDLIM7, LY6E, SPOCK1, PI15, CEMIP, CXCL8
DownregulatedGKN2, GKN1, ATP4A, ATP4B, GIF, LIPF, KCNJ16, DPCR1, SOSTDC1 KCNE2, CWH43, ESRRG, PGC, SLC28A2, PSAPL1, KRT20, VSIG1, LTF, CXCL17, AKR1B10, LOC643201, GSTA1, ADH7, ADH1C, GC, CAPN9, MAL, SLC26A9, HRASLS2, MFSD4A, MUC5AC, FBP2, ALDH3A1, ADGRG2, VSTM2A, LINC00982, CAPN13, KIAA1324, CA9, TPCN2, PIK3C2G, RDH12, SLC26A7, SSTR1, VSIG2, GATA6-AS1, HPGD, UPK1B, KCNJ15, SULT1C2, LINC00675, BTNL8, AXDND1, LINC00992, KAZALD1, TMED6, UGT2B15, SCNN1B, HAPLN1, AKR1C1, LYPD6B, FCGBP, ADTRP, IGH, CA2, RFX6, ACER2, CYP2C9, PCAT18, PKIB, SH3RF2, HHIP, HEPACAM2, AADAC, CYP2C18, RAB27B, MGAM, SPINK7, CNTN3, LINC01133, BCAS1, SULT1B1, CAPN8, SMIM6, AMPD1, JCHAIN, PBLD, ATP13A4, RNASE1, PLLP, B4GALNT3, STYK1, CYP2C19, SMIM24, LRRC66, RASSF6, ADAM28, FA2H, GATA5, SCIN, SGK2, TPH1, PROM2, APOBEC1, ACKR4, ADH1A, AKR7A3, OASL, SMPD3, XK, KLHDC7A, STX19, CYP3A5, STS, VILL, ANG, S100P, DDX60

DEMismiRNA name

Upregulatedhsa-let-7i-3p, hsa-miR-100-5p, hsa-miR-106b-5p, hsa-miR-10a-5p, hsa-miR-151a-5p, hsa-miR-15a-5p, hsa-miR-195-5p, hsa-miR-199a-3p, hsa-miR-199a-5p, hsa-miR-199b-5p, hsa-miR-19a-3p, hsa-miR-20a-5p, hsa-miR-214-3p, hsa-miR-214-5p, hsa-miR-218-5p, hsa-miR-223-3p, hsa-miR-301a-3p, hsa-miR-331-3p, hsa-miR-335-5p, hsa-miR-342-3p, hsa-miR-377-3p, hsa-miR-4262, hsa-miR-4291, hsa-miR-4317, hsa-miR-454-3p, hsa-miR-455-3p, hsa-miR-4791, hsa-miR-93-5p, hsa-miR-99a-5p, hsa-miR-99b-5p
Downregulatedhsa-miR-375

[i] DEM, differentially expressed mRNA; DEMis, differentially expressed microRNA; miRNA/miR, microRNA.

Functional analysis of DEMs

According to GO functional enrichment analysis for up- and downregulated DEMs, the significantly enriched biological process (BP), cellular component (CC) and molecular function (MF) terms were selected (Fig. 2). The values in the x-axes represent the quantification of -log10 (P-value). The GO terms enriched by upregulated DEMs were mainly associated with ‘endodermal cell differentiation’, ‘proteinaceous extracellular’ and ‘extracellular matrix binding’ while the GO terms enriched by downregulated DEMs were mainly associated with ‘regulation of cell proliferation’, ‘extracellular exosome’ and ‘iron ion binding’ (Table II). Following KEGG pathway enrichment analysis, upregulated differentially expressed genes (DEGs) were revealed to be mainly involved in ‘ECM-receptor interaction’, ‘focal adhesion’ and ‘phosphoinositide-3-kinase (PI3K)-protein kinase B (Akt) signaling pathway’. Downregulated DEGs were associated with ‘gastric acid secretion’, ‘retinol metabolism’ and ‘chemical carcinogenesis’ (Table III).

Table II.

Top 5 enriched GO terms of upregulated and downregulated differentially expressed mRNAs.

Table II.

Top 5 enriched GO terms of upregulated and downregulated differentially expressed mRNAs.

UpregulatedTermCountP-value
BP
  GO:0035987Endodermal cell differentiation54.00 ×10−07
  GO:0007155Cell adhesion62.44 ×10−05
  GO:0030199Collagen fibril organization45.04 ×10−05
  GO:0016525Negative regulation of angiogenesis41.66 ×10−04
  GO:0001937Negative regulation of endothelial cell proliferation30.001248824
CC
  GO:0005578Proteinaceous extracellular matrix134.06 ×10−15
  GO:0005615Extracellular space145.60 ×10−08
  GO:0005581Collagen trimer66.88 ×10−08
  GO:0031012Extracellular matrix40.002041798
  GO:0070062Extracellular exosome130.004832505
MF
  GO:0050840Extracellular matrix binding42.15 ×10−05
  GO:0005201Extracellular matrix structural constituent49.22 ×10−05
  GO:0008201Heparin binding40.001122327
  GO:0005509Calcium ion binding60.008415225
  GO:0008191 Metalloendopeptidase inhibitor activity20.02384997
Downregulated
BP
  GO:0042127Regulation of cell proliferation60.001561961
  GO:0010107Potassium ion import30.007733402
  GO:0042552Myelination30.014660727
  GO:0051453Regulation of intracellular pH30.01645907
  GO:0046903Secretion20.017262077
CC
  GO:0070062Extracellular exosome220.00577329
  GO:0005887Integral component of plasma membrane110.014280261
  GO:0005615Extracellular space120.014702585
  GO:0009986Cell surface60.036068461
  GO:0016324Apical plasma membrane40.06075832
MF
  GO:0005506Iron ion binding71.17 ×10−04
  GO:0005242Inward rectifier potassium channel activity30.002175164
  GO:0004522Ribonuclease A activity20.016178947
  GO:0020037Heme binding40.027324696
  GO:0019911Structural constituent of myelin sheath20.032099763

[i] GO, Gene Ontology; BP, biological processes; CC, cellular component; MF, molecular function.

Table III.

Top 5 enriched Kyoto Encyclopedia of Genes and Genomes pathway analysis of upregulated and downregulated differentially expressed mRNAs.

Table III.

Top 5 enriched Kyoto Encyclopedia of Genes and Genomes pathway analysis of upregulated and downregulated differentially expressed mRNAs.

IDTermCountP-value
Upregulated
  GO:cfa04512Extracellular matrix-receptor interaction9 3.17×10−12
  GO:cfa04510Focal adhesion9 3.53×10−09
  GO:cfa04974Protein digestion and absorption7 1.38×10−08
  GO:cfa04151 Phosphoinositide-3-kinase-protein kinase B signaling pathway9 1.62×10−07
  GO:cfa05146Amoebiasis6 2.25×10−06

IDTermCountP-value

Downregulated
  GO:bta04971Gastric acid secretion7 1.29×10−06
  GO:bta00830Retinol metabolism6 8.34×10−06
  GO:bta05204Chemical carcinogenesis6 2.30×10−05
  GO:bta01100Metabolic pathways16 5.74×10−04
  GO:bta00140Steroid hormone biosynthesis40.002819276
PPI network construction and sub-network identification

The PPI network consisted of 129 nodes and 572 interactions (Fig. 3). Following the use of Cytohubba in Cytoscape software, 10 top hub genes Jun proto-oncogene (JUN), mitogen activated protein kinase (MAPK)3, transforming growth factor-β1, Fos proto-oncogene, AP-1 transcription factor subunit (FOS), interleukin (IL)-8, MAPK1, RELA proto-oncogene nuclear factor κβ (NF-κB) subunit (RELA), interferon regulatory factor 7 (IRF7), ubiquitin like modifier (ISG15) and vascular endothelial growth factor A (VEGFA) were evaluated by degree in the PPI network (Fig. 4). The sub-network in the PPI network was then visualized using MCODE with a cut-off criterion of k-score=2. The sub-network was obtained from the PPI network with 14 nodes and 86 interactions (Fig. 5).

Survival analysis of the 10 top hub genes

The present study used the Kaplan-Meier Plotter database to evaluate the prognostic effect of the 10 top hub genes by overall survival (OS; Fig. 6). The Kaplan-Meier curves indicated that a higher expression of MAPK3, TGFB1, RELA, IRF7, ISG15 and VEGFA was significantly associated with poor survival in GC. On the other hand, a lower expression of JUN, FOS, IL8 and MAPK1 was significantly associated with poor survival in GC.

Functional analysis of 10 top hub genes and DEMs

In order to elucidate the specific signaling pathways that the 158 DEMs and 10 top hub genes were involved in, the DAVID database was used for further research. The results revealed that the 158 DEMs were involved in 17 KEGG pathways (Table IV). Among these pathways, 4 KEGG pathways (bta04151: PI3K-Akt signaling pathway, bta04510: Focal adhesion, bta05146: Amoebiasis and bta05144: Malaria) were identified to be significantly associated with the 10 top hub genes (Table V).

Table IV.

Enriched Kyoto Encyclopedia of Genes and Genomes pathway analysis of 158 differentially expressed mRNAs.

Table IV.

Enriched Kyoto Encyclopedia of Genes and Genomes pathway analysis of 158 differentially expressed mRNAs.

TermCountP-valueGenes
bta04151:Phosphoinositide-3-kinase-protein kinase B signaling pathway106.79 ×10−04COL4A1, SGK2, COL6A3, COL1A2, COL1A1, THBS1, COL11A1, THBS2, FN1, SPP1
bta04510:Focal adhesion99.84 ×10−05COL4A1, COL6A3, COL1A2, COL1A1, THBS1, COL11A1, THBS2, FN1, SPP1
bta05146:Amoebiasis60.001142COL4A1, COL1A2, CXCL8, COL1A1, COL11A1, FN1
bta05144:Malaria30.054453CXCL8, THBS1, THBS2
bta01100:Metabolic pathways160.024433CYP3A5, PIK3C2G, CYP2C18, ACER2, ADH1C, ADH7, FBP2, AMPD1, ALDH3A1, RDH12, AKR1B10, MGAM, TPH1, SMPD3, LIPF
bta04512:Extracellular matrix-receptor interaction91.47 ×10−07COL4A1, COL6A3, COL1A2, COL1A1, THBS1, COL11A1, THBS2, FN1, SPP1
bta04971:Gastric acid secretion71.07 ×10−05KCNJ16, KCNJ15, ATP4A, ATP4B, SLC26A7, KCNE2, CA2
bta04974:Protein digestion and absorption72.61 ×10−05COL4A1, COL6A3, COL1A2, COL12A1, COL1A1, COL11A1, COL10A1
bta00830:Retinol metabolism64.79 ×10−05RDH12, CYP3A5, CYP2C18, ADH1C, ADH7
bta05204:Chemical carcinogenesis61.29 ×10−04CYP3A5, CYP2C18, ADH1C, ADH7, ALDH3A1
bta00140:Steroid hormone biosynthesis40.007616CYP3A5, STS, CYP2C18
bta00010:Glycolysis/Gluconeogenesis40.010031ADH1C, ADH7, FBP2, ALDH3A1
bta04966:Collecting duct acid secretion30.015591ATP4A, ATP4B, CA2
bta00591:Linoleic acid metabolism30.028276CYP3A5, CYP2C18
bta00350:Tyrosine metabolism30.032672ADH1C, ADH7, ALDH3A1
bta00982:Drug metabolism-cytochrome P45030.063878ADH1C, ADH7, ALDH3A1
bta00980:Metabolism of xenobiotics by cytochrome P45030.065826ADH1C, ADH7, ALDH3A1

Table V.

Four enriched Kyoto Encyclopedia of Genes and Genomes pathway analysis of 10 hub genes that are identical to the results of 158 differentially expressed mRNAs.

Table V.

Four enriched Kyoto Encyclopedia of Genes and Genomes pathway analysis of 10 hub genes that are identical to the results of 158 differentially expressed mRNAs.

TermCountP-valueGenes
bta04151:Phosphoinositide-3-kinase-protein kinase B signaling pathway62.16 ×10−05MAPK1, IL6, RELA, VEGFA, MAPK3
bta04510:Focal adhesion40.001531MAPK1, JUN, VEGFA, MAPK3
bta05146:Amoebiasis30.007335IL6, RELA, TGFB1
bta05144:Malaria20.061465IL6, TGFB1

[i] MAPK, mitogen-activated protein kinase; IL-6, interleukin 6; RELA, RELA proto-oncogene NF-κβ subunit; VEGFA, vascular endothelial growth factor A; JUN, Jun proto-oncogene; TGFB1, transforming growth factor-β1.

Expression levels of significantly up- and downregulated DEMs

The GEPIA database was used to reveal the expression levels of 10 DEMs in GC tissues. The results revealed that 9 DEMs presented the same trend that was previously noted and only 1 DEM (KRT20) was contrary to the aforementioned trend (Fig. 7). The CCLE database was used to obtain information on the expression levels of 10 DEMs in GC cell lines. The present study then selected 5 different GC cell lines (HGC27, HS746T, MKN1, NUGC3 and RERFGC1B) to analyze the expression levels of the 10 DEMs (Fig. 8). For the upregulated DEMs, the expression level was partially different from the aforementioned results. All downregulated DEMs exhibited the same trend observed previously in the present study.

Association analysis of lncRNAs, miRNAs and mRNAs

The network of miRNAs and their targets was composed of 85 nodes and 145 interactions, determined using Cytoscape software (Fig. 9). In the lncRNA-miRNA-mRNA network, a total of 1,215 regulatory associations were screened out (Fig. 10). Pink circles indicated lncRNAs, hexagons indicated miRNAs, and diamonds represented mRNAs. With regards to the colors, red nodes indicated upregulated mRNAs and miRNAs, and green nodes represented downregulated mRNAs and miRNAs.

Discussion

The identification of the underlying molecular mechanism of GC is necessary to detect therapeutic targets in the malignant transformation process for management strategies. To date, microarrays and bioinformatics methods have been used to analyze the process of carcinogenesis to enhance the universality and reliability of the results (48). In the present study, 158 shared DEMs (40 upregulated and 118 downregulated) were obtained from the investigation of the GSE79973 and GSE54129 datasets. One previous study used GSE79973 to identify 14 significantly downregulated genes in GC, including KRT20, cytochrome P450 family 3 subfamily A member 5, RAB27B member RAS oncogene family and sulfotransferase family 1C member 2 (22). The four genes were additionally downregulated in the present study. Another previous study using GSE54129 identified 1829 DEMs including 838 upregulated genes and 991 downregulated genes (23). The Affy and limma packages in R software were used to select the DEMs in previous studies. In the present study, 158 shared DEMs (40 upregulated and 118 downregulated) were selected. The GEO2R web tool was used in the present study to identify DEMs. Although the data was from the same database, unequal results were obtained due to the different processing methods and filtering conditions utilized. For the analysis of 10 hub nodes, the previous study using the GSE54129 dataset identified 10 top hub nodes [tumor protein p53, C-X-C motif chemokine ligand (CXCL)8, tetraspanin 4, lysophospatidic acid receptor 2, adenylate cyclase 3, phosphoinositide-3-kinase regulatory subunit 1, neuromedin U, CXCL1, FOS and sphingosine-1-phosphate receptor 1] (23). FOS was also one of 10 hub genes identified in the present study. Meanwhile, the present study performed GO function and KEGG pathway enrichment analyses with 158 DEGs. The significant GO terms enriched by upregulated DEMs included ‘endodermal cell differentiation’, ‘proteinaceous extracellular’ and ‘extracellular matrix binding’ while the terms enriched by downregulated DEMs were mainly associated with ‘regulation of cell proliferation’, ‘extracellular exosome’ and ‘iron ion binding’. The results of KEGG pathway enrichment revealed that upregulated DEGs were mainly involved in ‘ECM-receptor interaction’, ‘focal adhesion’ and ‘PI3K-Akt signaling pathway’, and downregulated DEGs were associated with ‘gastric acid secretion’, ‘retinol metabolism’ and ‘chemical carcinogenesis’. Among the enriched pathways, the ‘PI3K/Akt signaling pathway’ has an essential biological function in the development of proliferation, apoptosis and invasion in various types of human cancer, including GC (2426). Furthermore, the PI3K/Akt signaling pathway is commonly activated in advanced GC and serves an important function in resistance to chemotherapy in GC (27). LY294002 has been identified as a PI3K inhibitor and has been confirmed to suppress cell proliferation and enhance apoptosis by downregulating VEGF, matrix metalloproteinase (MMP)-2 and MMP-9 in GC (28).

In the constructed PPI network, there were 129 nodes and 572 interactions. The top 10 hub genes (JUN, MAPK3, TGFB1, FOS, IL8, MAPK1, RELA, IRF7, ISG15 and VEGFA) were evaluated by degree following Cytohubba analysis. Then, the DAVID database was used to elucidate the specific signaling pathways that the 158 DEMs and 10 top hub genes were involved in. The results revealed that 158 DEMs were involved in 17 KEGG pathways (Table IV). Amongst these pathways, 4 KEGG pathways (bta04151: PI3K-Akt signaling pathway, bta04510: Focal adhesion, bta05146: Amoebiasis and bta05144: Malaria) were also associated with the 10 hub genes (Table V). Among four common KEGG pathways, PI3K-Akt signaling pathway is closely associated with the function of miRNAs. miR-15a-5p (an upregulated DEMi) regulated granulosa cell proliferation by activating the PI3K-Akt-mechanistic target of rapamycin kinase (mTOR) signaling pathway (29). miR-4262 (an upregulated DEMi) regulates chondrocyte viability, apoptosis, autophagy by targeting sirtuin 1 and activating the PI3K/Akt/mTOR signaling pathway in rats with osteoarthritis (30). miR-375 (a downregulated DEMi) functions as a tumor suppressor in osteosarcoma and colorectal cancer by targeting phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (31,32), which indicates that miR-375 may be associated with the PI3K-Akt signaling pathway. Furthermore, survival analysis of the top 10 hub genes was performed using the publicly available database Kaplan-Meier Plotter. The Kaplan-Meier curves indicated that the higher expression of MAPK3, TGFB1, RELA, IRF7 and VEGFA were associated with poor survival in GC. By contrast, a lower expression of JUN, FOS, IL8, ISG15 and MAPK1 were associated with poor survival in GC. Similar to the present study, a previous study applied bioinformatics analysis to identify the key genes in NCI-N87 GC cells exposed to quercetin. A PPI network was constructed, and hub genes were identified according to degree level, including FOS (degree=12) and JUN (degree=11) (33). One genome-wide search was performed to identify the genes epigenetically silenced by CpG methylation in GC. Three GC cell lines (SNU-1, SNU-601 and SNU-719) treated with 5Aza-Dc were analyzed to identify 143 associated genes by microarrays. Among the associated genes, IRF7 exhibited promoter hypermethylation in one or more gastric cancer cell lines (34). As the intracellular signal transduction pathway, the MAPK cascade serves an important function in the progression of various tumor types. MAPK1 was upregulated in GC tissues and participated in the proliferation and cell migration of GC cells. In addition, miRNAs may regulate the expression of MAPK1 and were therefore involved in the development and progression of GC (35,36). In the analysis of patients with resected GC, the results of patient prognosis analysis indicated that MAPK3/1 expression was an independent prognostic marker for patients with resected GC (37). The NF-κB signaling pathway serves an important function in the biological process of GC, including cell migration, cell invasion and cell apoptosis. As a basic component of the NF-κB signaling pathway, RELA has been reported to be activated in the progression of GC. The results of a study by Huang et al (38) revealed that RELA was upregulated in GC tumor tissues and GC cell lines compared with control groups. Furthermore, the upregulation of RELA was significantly correlated with poor OS in the prognosis of 876 patients with GC (P<0.001), which was consistent with the results of the present study. As an inflammatory factor, IL-6 serves an important function in the process of various types of cancer. Previous studies have revealed that IL-6 is involved in the regulation of invasion and prognosis in GC (39,40). Mechanistically, mesenchymal stem cells promote the activation of neutrophils through the IL-6-STAT3 axis to mediate GC progression (41). IL8 has been reported to be involved in the development of GC. A previous study demonstrated that IL-8 was upregulated in GC cells and enhanced cell proliferation, invasion and migration in GC (42). In addition, IL8 served as an inflammatory cytokine serving an important function in the angiogenesis of GC (43). Furthermore, a previous study also conducted a meta-analysis to investigate the potential functions of IL-6 and IL-8 polymorphisms in the development of GC. The results revealed that IL-6 rs1800796 and IL-8 rs4073 polymorphisms may serve as genetic biomarkers of GC in an Asian population (44). A number of studies have indicated that VEGFA functions as a direct target of miRNAs to participate in the tumor progression of GC (45,46). Furthermore, VEGFA may also be a predictive biomarker for antiangiogenic therapy in GC, which is consistent with the present bioinformatics results (47). A previous study also conducted analysis to reveal the correlation between polymorphisms of TGFB1 and VEGF genes and the survival of patients with GC. The results suggested that the TGFB1+915CG/CC and VEGF-634CG genotypes were associated with short-term survival in patients with GC (48).

MiRNAs serve an important function in the downregulation of the transcription of target mRNAs by binding to complementary 3′-untranslated regions of genes (49). An accumulating body of evidence has demonstrated the association between miRNAs and targets in tumor progression in various types of cancer. For example, it has been confirmed that miR-218 is a tumor suppressor in glioblastoma cells by directly targeting E2F transcription factor 2 (50). miR-155 has also been demonstrated to enhance cell growth and invasion by regulating transforming growth factor-β receptor 2 (51). In the present study, a total of 30 upregulated miRNAs and 1 downregulated miRNA were identified between the GC and control samples using systematic analysis. Then, a network of miRNAs and targets was constructed using the miRwalk database and Cytoscape software, which consisted of 124 nodes and 490 interactions. According to the results of the present analysis, miR-375 was downregulated in GC tissues when compared with the NC group. Similarly, a number of studies have confirmed the function of miR-375 as a tumor suppressor in GC. MiR-375 has been reported to be involved in cell proliferation, migration and invasion by regulating janus kinase 2 and macrophage stimulating 1 receptor (52,53). MiR-375 serves as a controller of the Hippo signaling pathway by targeting the Yes associated protein 1-TEA domain transcription factor 4-cellular communication network factor 2 axis (54). A previous study indicated that miR19a3p is upregulated in GC and promotes epithelialmesenchymal transition via the PI3K/Akt signaling pathway (55). Furthermore, miR-19a-3P has been confirmed to mediate metastasis by directly targeting MAX dimerization protein 1 in GC cells (56). Emerging evidence has indicated that miR-214-3p serves an essential oncogenic function and is correlated with distant metastasis as a novel biomarker of GC (57). In addition, miR-214-3p negatively regulates phosphatase and tensin homolog and is involved in GC cell proliferation, migration and invasion (58,59).

Previous studies have suggested that lncRNAs participate in the development of cell growth, metastasis and invasion progression in various types of cancer (6062). LncRNAs have also been demonstrated to function as miRNA sponges that are involved in a variety of cancer types. For example, the lncRNA XIST has been demonstrated to function as a molecular sponge of miR-101 to modulate enhancer of zeste 2 polycomb repressive complex 2 subunit expression in GC (63). LncRNA-regulator of reprogramming regulates the expression of miR-145 and ADP ribosylation factor 6 by functioning as a sponge in triple-negative breast cancer (64). Therefore, it is essential to investigate the regulatory mechanism underlying the action of lncRNAs, miRNAs and mRNAs. In the present study, the lncRNASNP database was used to perform prediction analysis of DEMis-lncRNA pairs. In addition, a lncRNA-miRNA-mRNA network was constructed using the combination of DEMis-target pairs and DEMis-lncRNA pairs, which included 1,215 regulatory associations. Altogether, these results provide a better understanding of the potential functions of lncRNA, miRNA and mRNA in GC.

However, there were a number of limitations in the present analysis. Firstly, the present study lacked experimental evidence on the expression levels and biological functions of genes. Further detection and experiments on the associated genes in larger sample sizes should be validated in future studies. Secondly, lncRNAs were only predicted using the lncRNASNP database without the investigation of expression profile databases, which are different from mRNAs and miRNAs. In order to increase the credibility, GC-associated lncRNAs require further analysis in the future.

In conclusion, the present study analyzed 158 DEMs, 31 DEMis and the top 10 hub genes in GC from multiple profile datasets by integrated bioinformatical analysis. In addition, lncRNA-miRNA-mRNA regulatory networks were established to gain a better understanding of the underlying molecular mechanism of GC. These results provide an effective foundation for further research on potential target therapy strategies in GC.

Acknowledgements

Not applicable.

Funding

The present study was supported in part by the Department of Gastrointestinal Endoscopy, First Affiliated Hospital of China Medical University (Shenyang, China).

Availability of data and materials

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

Authors' contributions

MS designed the study. XM, YM, HZ and HJZ performed the research. XM analyzed the data and wrote the paper.

Ethics approval and consent to participate

Not applicable

Patient consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

BP

biological processes

CC

cellular component

DAVID

Database for Annotation, Visualization and Integrated Discovery

DEM

differentially expressed mRNA

DEMi

differentially expressed miRNA

GC

gastric cancer

GEO

Gene Expression Omnibus

GO

Gene ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes

lncRNA

long non-coding RNA

MF

molecular function

miRNA

microRNA

OS

overall survival

PPI

protein-protein interaction network

STRING

Search Tool for the Retrieval of Interacting Genes

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 

Karimi P, Islami F, Anandasabapathy S, Freedman ND and Kamangar F: Gastric cancer: Descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev. 23:700–713. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Wang J, Guo W, Wu Q, Zhang R and Fang J: Impact of combination epidural and general anesthesia on the long-term survival of gastric cancer patients: A retrospective study. Med Sci Monit. 22:2379–2385. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Long J, Zhang Z, Liu Z, Xu Y and Ge C: Identification of genes and pathways associated with pancreatic ductal adenocarcinoma by bioinformatics analyses. Oncol Lett. 11:1391–1397. 2016. View Article : Google Scholar : PubMed/NCBI

5 

He JH, Han ZP, Zou MX, Wang L, Lv YB, Zhou JB, Cao MR and Li YG: Analyzing the LncRNA, miRNA, and mRNA regulatory network in prostate cancer with bioinformatics software. J Comput Biol. 25:146–157. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Wang H, Luo J, Liu C, Niu H, Wang J, Liu Q, Zhao Z, Xu H, Ding Y, Sun J and Zhang Q: Investigating MicroRNA and transcription factor co-regulatory networks in colorectal cancer. BMC Bioinformatics. 18:3882017. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Piao J, Sun J, Yang Y, Jin T, Chen L and Lin Z: Target gene screening and evaluation of prognostic values in non-small cell lung cancers by bioinformatics analysis. Gene. 647:306–311. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Jiang H, Ma R, Zou S, Wang Y, Li Z and Li W: Reconstruction and analysis of the lncRNA-miRNA-mRNA network based on competitive endogenous RNA reveal functional lncRNAs in rheumatoid arthritis. Mol Biosyst. 13:1182–1192. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Edgar R, Domrachev M and Lash AE: Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30:207–210. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Pathan M, Keerthikumar S, Ang CS, Gangoda L, Quek CY, Williamson NA, Mouradov D, Sieber OM, Simpson RJ, Salim A, et al: FunRich: An open access standalone functional enrichment and interaction network analysis tool. Proteomics. 15:2597–2601. 2015. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al: STRING v11: Protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1): D607–D613. 2019. View Article : Google Scholar : PubMed/NCBI

14 

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

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 

Nagy Á, Lánczky A, Menyhárt O and Győrffy B: Validation of miRNA prognostic power in hepatocellular carcinoma using expression data of independent datasets. Sci Rep. 8:92272018. View Article : Google Scholar : PubMed/NCBI

17 

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 (W1). W98–W102. 2017. View Article : Google Scholar

18 

Ghandi M, Huang FW, Jané-Valbuena J, Kryukov GV, Lo CC, McDonald ER III, Barretina J, Gelfand ET, Bielski CM, Li H, et al: Next-generation characterization of the cancer cell line encyclopedia. Nature. 569:503–508. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Dweep H, Sticht C, Pandey P and Gretz N: miRWalk-database: Prediction of possible miRNA binding sites by ‘walking’ the genes of three genomes. J Biomed Inform. 44:839–847. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Dweep H and Gretz N: miRWalk2.0: A comprehensive atlas of microRNA-target interactions. Nat methods. 12:6972015. View Article : Google Scholar : PubMed/NCBI

21 

Miao YR, Liu W, Zhang Q and Guo AY: lncRNASNP2: An updated database of functional SNPs and mutations in human and mouse lncRNAs. Nucleic Acids Res. 46(D1): D276–D280. 2018. View Article : Google Scholar : PubMed/NCBI

22 

He J, Jin Y, Chen Y, Yao HB, Xia YJ, Ma YY, Wang W and Shao QS: Downregulation of ALDOB is associated with poor prognosis of patients with gastric cancer. Onco Targets Ther. 9:6099–6109. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Qi W, Sun L, Liu N, Zhao S, Lv J and Qiu W: Tetraspanin family identified as the central genes detected in gastric cancer using bioinformatics analysis. Mol Med Rep. 18:3599–3610. 2018.PubMed/NCBI

24 

Liu Q, Dong HW, Sun WG, Liu M, Ibla JC, Liu LX, Parry JW, Han XH, Li MS and Liu JR: Apoptosis initiation of β-ionone in SGC-7901 gastric carcinoma cancer cells via a PI3K-AKT pathway. Arch Toxicol. 87:481–490. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Li GQ, Xie J, Lei XY and Zhang L: Macrophage migration inhibitory factor regulates proliferation of gastric cancer cells via the PI3K/Akt pathway. World J Gastroenterol. 15:5541–5548. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Ye B, Jiang LL, Xu HT, Zhou DW and Li ZS: Expression of PI3K/AKT pathway in gastric cancer and its blockade suppresses tumor growth and metastasis. Int J Immunopathol Pharmacol. 25:627–636. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Ang KL, Shi DL, Keong WW and Epstein RJ: Upregulated Akt signaling adjacent to gastric cancers: Implications for screening and chemoprevention. Cancer Lett. 225:53–59. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Xing CG, Zhu BS, Fan XQ, Liu HH, Hou X, Zhao K and Qin ZH: Effects of LY294002 on the invasiveness of human gastric cancer in vivo in nude mice. World J Gastroenterol. 15:5044–5052. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Zhang K, Zhong W, Li WP, Chen ZJ and Zhang C: miR-15a-5p levels correlate with poor ovarian response in human follicular fluid. Reproduction. 154:483–496. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Sun W, Li Y and Wei S: miR-4262 regulates chondrocyte viability, apoptosis, autophagy by targeting SIRT1 and activating PI3K/AKT/mTOR signaling pathway in rats with osteoarthritis. Exp Ther Med. 15:1119–1128. 2018.PubMed/NCBI

31 

Shi ZC, Chu XR, Wu YG, Wu JH, Lu CW, Lü RX, Ding MC and Mao NF: MicroRNA-375 functions as a tumor suppressor in osteosarcoma by targeting PIK3CA. Tumour Biol. 36:8579–8584. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Wang Y, Tang Q, Li M, Jiang S and Wang X: MicroRNA-375 inhibits colorectal cancer growth by targeting PIK3CA. Biochem Biophys Res Commun. 444:199–204. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Zeng Y, Shen Z, Gu W and Wu M: Bioinformatics analysis to identify action targets in NCI-N87 gastric cancer cells exposed to quercetin. Pharm Biol. 56:393–398. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Jee CD, Kim MA, Jung EJ, Kim J and Kim WH: Identification of genes epigenetically silenced by CpG methylation in human gastric carcinoma. Eur J Cancer. 45:1282–1293. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Xiong HL, Zhou SW, Sun AH, He Y, Li J and Yuan X: MicroRNA-197 reverses the drug resistance of fluorouracil-induced SGC7901 cells by targeting mitogen-activated protein kinase 1. Mol Med Rep. 12:5019–5025. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Hu L, Wu H, Wan X, Liu L, He Y, Zhu L, Liu S, Yao H3 and Zhu Z: MicroRNA-585 suppresses tumor proliferation and migration in gastric cancer by directly targeting MAPK1. Biochem Biophys Res Commun. 499:52–58. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Kim JG, Lee SJ, Chae YS, Kang BW, Lee YJ, Oh SY, Kim MC, Kim KH and Kim SJ: Association between phosphorylated AMP-activated protein kinase and MAPK3/1 expression and prognosis for patients with gastric cancer. Oncology. 85:78–85. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Huang T, Kang W, Zhang B, Wu F, Dong Y, Tong JH, Yang W, Zhou Y, Zhang L, Cheng AS, et al: miR-508-3p concordantly silences NFKB1 and RELA to inactivate canonical NF-κB signaling in gastric carcinogenesis. Mol Cancer. 15:92016. View Article : Google Scholar : PubMed/NCBI

39 

Xia Y, Khoi PN, Yoon HJ, Lian S, Joo YE, Chay KO, Kim KK and Jung YD: Piperine inhibits IL-1β-induced IL-6 expression by suppressing p38 MAPK and STAT3 activation in gastric cancer cells. Mol Cell Biochem. 398:147–156. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Yin Y, Si X, Gao Y, Gao L and Wang J: The nuclear factor-κB correlates with increased expression of interleukin-6 and promotes progression of gastric carcinoma. Oncol Rep. 29:34–38. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Zhu Q, Zhang X, Zhang L, Li W, Wu H, Yuan X, Mao F, Wang M, Zhu W, Qian H and Xu W: The IL-6-STAT3 axis mediates a reciprocal crosstalk between cancer-derived mesenchymal stem cells and neutrophils to synergistically prompt gastric cancer progression. Cell Death Dis. 5:e12952014. View Article : Google Scholar : PubMed/NCBI

42 

Li W, Lin S, Li W, Wang W, Li X and Xu D: IL-8 interacts with metadherin promoting proliferation and migration in gastric cancer. Biochem Biophys Res Commun. 478:1330–1337. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Shi J, Li YJ, Yan B and Wei PK: Interleukin-8: A potent promoter of human lymphatic endothelial cell growth in gastric cancer. Oncol Rep. 33:2703–2710. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Wang X, Yang F, Xu G and Zhong S: The roles of IL-6, IL-8 and IL-10 gene polymorphisms in gastric cancer: A meta-analysis. Cytokine. 111:230–236. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Wang CQ, Chen L, Dong CL, Song Y, Shen ZP, Shen WM and Wu XD: MiR-377 suppresses cell proliferation and metastasis in gastric cancer via repressing the expression of VEGFA. Eur Rev Med Pharmacol Sci. 21:5101–5111. 2017.PubMed/NCBI

46 

Zhang X, Tang J, Zhi X, Xie K, Wang W, Li Z, Zhu Y, Yang L, Xu H and Xu Z: Correction: miR-874 functions as a tumor suppressor by inhibiting angiogenesis through STAT3/VEGF-A pathway in gastric cancer. Oncotarget. 8:295352017.PubMed/NCBI

47 

Caporarello N, Lupo G, Olivieri M, Cristaldi M, Cambria MT, Salmeri M and Anfuso CD: Classical VEGF, Notch and Ang signalling in cancer angiogenesis, alternative approaches and future directions (Review). Mol Med Rep. 16:4393–4402. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Guan X, Zhao H, Niu J, Tan D, Ajani JA and Wei Q: Polymorphisms of TGFB1 and VEGF genes and survival of patients with gastric cancer. J Exp Clin Cancer Res. 28:942009. View Article : Google Scholar : PubMed/NCBI

49 

Cho WC: OncomiRs: The discovery and progress of microRNAs in cancers. Mol Cancer. 6:602007. View Article : Google Scholar : PubMed/NCBI

50 

Zhang Y, Han D, Wei W, Cao W, Zhang R, Dong Q, Zhang J, Wang Y and Liu N: MiR-218 inhibited growth and metabolism of human glioblastoma cells by directly targeting E2F2. Cell Mol Neurobiol. 35:1165–1173. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Qu Y, Zhang H, Sun W, Han Y, Li S, Qu Y, Ying G and Ba Y: MicroRNA-155 promotes gastric cancer growth and invasion by negatively regulating transforming growth factor-β receptor 2. Cancer Sci. 109:618–628. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Ding L, Xu Y, Zhang W, Deng Y, Si M, Du Y, Yao H, Liu X, Ke Y, Si J and Zhou T: MiR-375 frequently downregulated in gastric cancer inhibits cell proliferation by targeting JAK2. Cell Res. 20:784–793. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Lian S, Park JS, Xia Y, Nguyen TT, Joo YE, Kim KK, Kim HK and Jung YD: MicroRNA-375 functions as a tumor-suppressor gene in gastric cancer by targeting recepteur d'Origine nantais. Int J Mol Sci. 17(pii): E16332016. View Article : Google Scholar : PubMed/NCBI

54 

Kang W, Huang T, Zhou Y, Zhang J, Lung RWM, Tong JHM, Chan AWH, Zhang B, Wong CC, Wu F, et al: miR-375 is involved in Hippo pathway by targeting YAP1/TEAD4-CTGF axis in gastric carcinogenesis. Cell Death Dis. 9:922018. View Article : Google Scholar : PubMed/NCBI

55 

Lu WD, Zuo Y, Xu Z and Zhang M: MiR-19a promotes epithelial-mesenchymal transition through PI3K/AKT pathway in gastric cancer. World J Gastroenterol. 21:4564–4573. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Wu Q, Yang Z, An Y, Hu H, Yin J, Zhang P, Nie Y, Wu K, Shi Y and Fan D: MiR-19a/b modulate the metastasis of gastric cancer cells by targeting the tumour suppressor MXD1. Cell Death Dis. 5:e11442014. View Article : Google Scholar : PubMed/NCBI

57 

Zhang KC, Xi HQ, Cui JX, Shen WS, Li JY, Wei B and Chen L: Hemolysis-free plasma miR-214 as novel biomarker of gastric cancer and is correlated with distant metastasis. Am J Cancer Res. 5:821–829. 2015.PubMed/NCBI

58 

Xin R, Bai F, Feng Y, Jiu M, Liu X, Bai F, Nie Y and Fan D: MicroRNA-214 promotes peritoneal metastasis through regulating PTEN negatively in gastric cancer. Clin Res Hepatol Gastroenterol. 40:748–754. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Yang TS, Yang XH, Wang XD, Wang YL, Zhou B and Song ZS: MiR-214 regulate gastric cancer cell proliferation, migration and invasion by targeting PTEN. Cancer Cell Int. 13:682013. View Article : Google Scholar : PubMed/NCBI

60 

Prensner JR and Chinnaiyan AM: The emergence of lncRNAs in cancer biology. Cancer Discov. 1:391–407. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Rinn JL and Chang HY: Genome regulation by long noncoding RNAs. Annu Rev Biochem. 81:145–166. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Gutschner T and Diederichs S: The hallmarks of cancer: A long non-coding RNA point of view. RNA Biol. 9:703–719. 2012. View Article : Google Scholar : PubMed/NCBI

63 

Chen DL, Ju HQ, Lu YX, Chen LZ, Zeng ZL, Zhang DS, Luo HY, Wang F, Qiu MZ, Wang DS, et al: Long non-coding RNA XIST regulates gastric cancer progression by acting as a molecular sponge of miR-101 to modulate EZH2 expression. J Exp Clin Cancer Res. 35:1422016. View Article : Google Scholar : PubMed/NCBI

64 

Eades G, Wolfson B, Zhang Y, Li Q, Yao Y and Zhou Q: lincRNA-RoR and miR-145 regulate invasion in triple-negative breast cancer via targeting ARF6. Mol Cancer Res. 13:330–338. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma XY, Ma Y, Zhou H, Zhang HJ and Sun MJ: Identification of the lncRNA‑miRNA‑mRNA network associated with gastric cancer via integrated bioinformatics analysis. Oncol Lett 18: 5769-5784, 2019
APA
Ma, X., Ma, Y., Zhou, H., Zhang, H., & Sun, M. (2019). Identification of the lncRNA‑miRNA‑mRNA network associated with gastric cancer via integrated bioinformatics analysis. Oncology Letters, 18, 5769-5784. https://doi.org/10.3892/ol.2019.10922
MLA
Ma, X., Ma, Y., Zhou, H., Zhang, H., Sun, M."Identification of the lncRNA‑miRNA‑mRNA network associated with gastric cancer via integrated bioinformatics analysis". Oncology Letters 18.6 (2019): 5769-5784.
Chicago
Ma, X., Ma, Y., Zhou, H., Zhang, H., Sun, M."Identification of the lncRNA‑miRNA‑mRNA network associated with gastric cancer via integrated bioinformatics analysis". Oncology Letters 18, no. 6 (2019): 5769-5784. https://doi.org/10.3892/ol.2019.10922