Open Access

Co-expressed differentially expressed genes and long non-coding RNAs involved in the celecoxib treatment of gastric cancer: An RNA sequencing analysis

  • Authors:
    • Bin Song
    • Juan Du
    • Ye Feng
    • Yong‑Jian Gao
    • Ji‑Sheng Zhao
  • View Affiliations

  • Published online on: September 1, 2016     https://doi.org/10.3892/etm.2016.3648
  • Pages: 2455-2468
  • Copyright: © Song 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 mechanisms of long non‑coding RNAs (lncRNAs) in a gastric cancer cell line treated with celecoxib. The human gastric carcinoma cell line NCI‑N87 was treated with 15 µM celecoxib for 72 h (celecoxib group) and an equal volume of dimethylsulfoxide (control group), respectively. Libraries were constructed by NEBNext Ultra RNA Library Prep kit for Illumina. Paired‑end RNA sequencing reads were aligned to a human hg19 reference genome using TopHat2. Differentially expressed genes (DEGs) and lncRNAs were identified using Cuffdiff. Enrichment analysis was performed using GO‑function package and KEGG profile in Bioconductor. A protein-protein interaction network was constructed using STRING database and module analysis was performed using ClusterONE plugin of Cytoscape. ATP5G1, ATP5G3, COX8A, CYC1, NDUFS3, UQCRC1, UQCRC2 and UQCRFS1 were enriched in the oxidative phosphorylation pathway. CXCL1, CXCL3, CXCL5 and CXCL8 were enriched in the chemokine signaling and cytokine‑cytokine receptor interaction pathways. ITGA3, ITGA6, ITGB4, ITGB5, ITGB6 and ITGB8 were enriched in the integrin‑mediated signaling pathway. DEGs co‑expressed with lnc‑SCD‑1:13, lnc‑LRR1‑1:2, lnc‑PTMS‑1:3, lnc‑S100P‑3:1, lnc‑AP000974.1‑1:1 and lnc‑RAB3IL1‑2:1 were enriched in the pathways associated with cancer, such as the basal cell carcinoma pathway in cancer. In conclusion, these DEGs and differentially expressed lncRNAs may be important in the celecoxib treatment of gastric cancer.

Introduction

Despite the mortality rate for gastric carcinoma reducing 3.1% annually and the overall 5-year relative survival rate increasing to 28% over the past 10 years, the mortality rate for gastric carcinoma remains >50% worldwide (1). The most effective treatment for resectable gastric cancer is surgery, which presents good survival rates. The majority of cases of gastric cancer are diagnosed at an advanced stage or as a relapse after surgery (2). Therefore, a further understanding of the molecular mechanisms of gastric cancer is of clinical importance and it is required in order to improve the early diagnosis and therapeutic strategies of gastric cancer.

Over the last decade, the majority of the potential therapeutic targets reported and the diagnostic markers for gastric cancer are protein-coding genes identified from large-scale DNA microarray analysis, including the novel genes KLF5, FAT4, KMT2C, GATA4, MLL and GATA6 (36). The majority of studies on non-coding RNAs (ncRNAs) are focused on short ncRNAs called microRNAs, while alterations in the structure, expression levels and cognate RNA-binding proteins of long ncRNAs (lncRNAs) with a length of >200 nucleotides (nt) have been associated with cancer, and appear to be gaining prominence as further studies are conducted (7). In addition, growing evidence has confirmed that lncRNAs that are capable of regulating tumor suppression or that exhibit oncogenic effects may be considered as novel biomarkers and therapeutic targets for cancer (8,9). Furthermore, it has been demonstrated that differentially expressed long non-coding RNAs (DE-lncRNAs), including H19 and uc001lsz, may present potential roles in the development and occurrence of gastric cancer (10). In a study by Hu et al (11), a novel lncRNA GAPLINC (924 bp) was highly expressed in gastric cancer specimens and it was capable of controlling the expression levels of CD44 to regulate cell invasion by competing for miR211-3p.

A previous study demonstrated that celecoxib induced apoptosis and autophagy of gastric cancer SGC-7901 cells via the PI3K/Akt signaling pathway (12). According to a study by Lan et al (13), celecoxib inhibited Helicobacter pylori-induced invasion in gastric cancer via the adenine nucleotide translocator-dependent pathways. Furthermore, the activated Notch1 signaling pathway may contribute to the pathogenesis of gastric cancer, at least partly through COX-2 (14). Treatment with celecoxib, a COX-2 inhibitor, can significantly reduce the incidence of gastric cancer in rats (15). In addition, an elevated COX-2 expression level is an independent prognostic factor indicative of poor prognosis and it is associated with reduced survival in patients with gastric cancer (16). Pang et al (17) reported that the Akt/GSK3β/NAG-1 signaling pathway may be considered as the major mechanism of the COX-2-independent effects of celecoxib on gastric cancer cells. COX-2 has been indicated to regulate E-cadherin expression via the NF-κB and Snail signaling pathway in gastric cancer (18). It has also been reported that celecoxib has the potential for clinical use in gastric cancer treatment by the mechanism of activating miR-29c (19). Although various advances have been made in the study of mechanisms of lncRNAs in gastric cancer, the understanding of the expression patterns and functional roles of lncRNAs in gastric cancer treated with celecoxib requires further investigation.

In the present study, the RNA sequencing data of NCI-N87 human gastric carcinoma cells treated with or without celecoxib were prepared and analyzed using bioinformatics methods. Briefly, differentially expressed genes (DEGs) and lncRNAs were identified for pathway enrichment analysis. A protein-protein interaction (PPI) network for DEGs was constructed and module analysis was performed. Finally, co-expression analysis of DEGs and lncRNAs was performed. The results of the data in the present study may provide novel insight into the roles of celecoxib in gastric cancer.

Materials and methods

Cell culture and celecoxib treatment

The human gastric carcinoma cell line NCI-N87 was obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). Cells were cultured in RPMI-1640 medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin (Thermo Fisher Scientific, Inc.) in a humidified air incubator (Thermo Fisher Scientific, Inc.) at 37°C and with 5% CO2. The cells were passaged at 80–90% confluence with 0.25% trypsin (Thermo Fisher Scientific, Inc.).

Cells at the exponential growth phase with a density of 1×106 were seeded in a cell culture dish (Corning Inc., NY, USA) with a diameter of 6 cm and incubated in 5 ml serum-free Dulbecco's modified Eagle medium (Thermo Fisher Scientific, Inc.) overnight. Celecoxib (Sigma-Aldrich, St. Louis, MO, USA) was dissolved in dimethylsulfoxide (DMSO; Sigma-Aldrich), and the cells were treated with 15 µM celecoxib for 72 h (celecoxib group). Cells treated with an equal volume of DMSO were used as a control group.

RNA sequencing data

The total RNA was extracted using TRIzol (Thermo Fisher Scientific, Inc.) following the manufacturer's protocol, and were quantified with a 721 spectrophotometer (Shanghai Precision Instrument Co., Ltd., Shanghai, China). Next, libraries were prepared by the NEBNext Ultra RNA Library Prep kit for Illumina (#E7530; New England BioLabs, Inc., Ipswich, MA, USA) according to the manufacturer's instructions. Briefly, RNA fragments ~200 nt in length were generated and then double-stranded cDNA was synthesized and end-repaired. Following the adaptor ligation, PCR amplification was performed as follows: A library was added with 10 µl 5X HF Buffer, 1 µl 10 µM reverse PCR primer 2–1: 5′-CAAGCAGAAGACGGCATACGAGATCGTGATGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3′ and primer 2–2: 5′-CAAGCAGAAGACGGCATACGAGATACATCGGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3′, primer 2–3: 5′-CAAGCAGAAGACGGCATACGAGATGCCTAAGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3′, primer 2–4: 5′-CAAGCAGAAGACGGCATACGAGATTGGTCAGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3′, 1.5 µl dNTP, 0.5 µl Phusion High-Fidelity DNA Polymerase (2 U/µl) and 5 µl ddH2O, and then incubated at 98°C for 40 sec, 65°C for 30 sec and 72°C for 30 sec. Next, 1 µl of 10 µM forward PCR primer (5′-AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACACGACGCTCTTCCGATCT-3′) was added and incubated at 98°C for 10 sec, 10 cycles at 65°C for 30 sec, 72°C for 30 sec, and 72°C for 3 min. Finally, the library was dissolved in 20 µl ddH2O after being purified by 50 µl AMPure XP magnetic beads. A 1 µg input for 15 cycles and a 5 µg input for 12 cycles was used and the library quality was assessed on a 2100 Electrophoresis Bioanalyzer instrument (Agilent Technologies, Inc., Santa Clara, CA, USA). Finally, sequencing was conducted on a HiSeq 2500 System (Illumina, Inc., San Diego, CA, USA).

Data preprocessing and sequence alignment

Quality control (QC) of obtained next generation sequencing (NGS) data was conducted with an NGS QC Toolkit (version 2.3.3; www.nipgr.res.in/ngsqctoolkit.html) in order to remove low quality reads with default parameters (20). Reads with ≥10% low quality bases (Phred quality score <20) were filtered.

The paired-end RNA sequencing reads were aligned to the human hg19 reference genome using TopHat2 (ccb.jhu.edu/software/tophat) (21), and the human hg19 reference genome and its annotation files were obtained from the University of California Santa Cruz Genome Browser (genome.ucsc.edu) (22). The ‘-no-mixed’ option was handled and other parameters were set to default.

Identification of DEGs and lncRNAs

Following sequence alignment and refseq annotation, Cuffdiff (23) was applied to screen DEGs with a cut-off criteria of q<0.05. DE-lncRNAs were identified with the combination of lncRNA annotation by LNCipedia 3.0 (www.lncipedia.org) (24). q<0.05 was considered as the threshold value.

Functional and pathway enrichment analysis for DEGs

Gene ontology (GO) terms in the biological process (BP), cellular component (CC) and molecular function (MF) categories were enriched for DEGs using the GO-function package in Bioconductor (www.bioconductor.org) (25). KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis was also conducted by the KEGG profile in Bioconductor. The enrichment thresholds were P<0.05 and the gene counts ≥2.

Construction of the PPI network and module analysis

The Search Tool for the Retrieval of Interacting Genes (STRING; www.string-db.org) database not only provides uniquely comprehensive coverage but also contains predicted, experimental, transferred and text-mined interactions (26). The PPIs for DEGs were predicted using version 9.1 of the STRING database with a combined score >0.7 (26). Cytoscape software version 2.8 (27) was used to visualize the PPI network (www.cytoscape.org).

The ClusterONE plugin of Cytoscape (28) was used to perform module analysis for the PPI network with default parameters. In addition, functional and pathway enrichment analysis of DEGs in the two modules with the highest significance was performed with the cut-off criteria of P<0.05 and gene counts ≥2.

Co-expression analysis of DEGs and lncRNAs

Pearson correlation coefficients between DEGs and lncRNAs were calculated. The co-expressed genes and lncRNA pairs were selected with a Pearson correlation coefficient >0.98. Pathway enrichment analysis was conducted for the DEGs co-expressed with each DE-lncRNA, with thresholds of P<0.05 and gene counts ≥2.

Results

DEGs and lncRNAs

A total of 490 DEGs, of which 302 were upregulated and 188 downregulated genes, were identified in the celecoxib and the control groups. A total of 37 DE-lncRNAs, of which 19 were upregulated and 18 downregulated, were screened (Table I).

Table I.

Differentially expressed lncRNAs in the celecoxib and the control groups.

Table I.

Differentially expressed lncRNAs in the celecoxib and the control groups.

lncRNAs IDCelecoxibControlFold changeq value
Upregulated
  lnc-IGFL3-2:118.4344.601.27 1.07×10−7
  lnc-PTMS-1:3427.83613.560.52 1.71×10−6
  lnc-SCD-1:13105.06152.800.54 1.71×10−6
  lnc-TNS4-2:16.2815.941.34 1.71×10−6
  lnc-TTLL10-3:17.7111.670.60 2.91×10−5
  lnc-CKMT1A-1:150.3797.920.96 5.80×10−5
  lnc-LRR1-1:24422.955914.510.42 6.36×10−5
  lnc-RAB3IL1-2:12279.253231.600.50 7.18×10−4
  lnc-JUNB-1:1372.34562.080.59 2.00×10−3
  lnc-RP11-259P6.1.1–2:129.7839.570.41 3.20×10−3
  lnc-IGFL2-2:1105.84167.350.66 5.76×10−3
  lnc-S100P-3:1142.85240.910.75 6.65×10−3
  lnc-SRGAP3-1:2901.731.80e+308 1.19×10−2
  lnc-RAB44-3:18.4314.880.82 1.25×10−2
  lnc-GLTSCR2-2:718.5826.750.53 1.26×10−2
  lnc-PDZD7-3:203.411.80e+308 2.41×10−2
  lnc-CEACAM6-1:133.2057.590.79 2.51×10−2
  lnc-SPNS3-1:318.5827.110.54 4.89×10−2
  lnc-UNC5B-1:112.6218.200.53 4.89×10−2
Downregulated
  lnc-C9orf16-2:1875.46425.36−1.040
  lnc-C9orf16-3:1352.54156.14−1.170
  lnc-TRIM31-1:247.1721.45−1.14 4.10×10−8
  lnc-DDX47-3:1211.38145.19−0.54 1.46×10−7
  lnc-PCK1-3:113.925.48−1.35 8.52×10−7
  lnc-MYO16-7:1349.12200.98−0.80 1.71×10−6
  lnc-YPEL5-5:171.8244.54−0.69 1.71×10−6
  lnc-TNK2-8:116.602.33−2.83 4.54×10−6
  lnc-AC069257.9.1-4:73124.4066.92−0.90 6.69×10−5
  lnc-CCDC80-1:418.793.17−2.57 1.74×10−3
  lnc-AC069257.9.1-4:72151.5881.26−0.90 5.89×10−3
  lnc-KRT36-1:145.0018.61−1.27 6.65×10−3
  lnc-CCDC33-1:132.4519.88−0.71 8.64×10−3
  lnc-CXCL3-1:15.061.51−1.74 2.51×10−2
  lnc-PDZK1IP1-3:125.2611.57−1.13 3.28×10−2
  lnc-SUSD3-4:219.489.17−1.09 3.37×10−2
  lnc-AC069257.9.1-4:5399.1857.43−0.79 3.59×10−2
  lnc-AP000974.1-1:136.0916.13−1.16 4.79×10−2

[i] Celecoxib and control columns indicate the average expression values of the lncRNAs in the celecoxib and the control group, respectively. lncRNA, long non-coding ribonucleic acids.

Functional and pathway enrichment analysis for DEGs

GO enrichment analysis demonstrated that 672, 108 and 120 terms in the BP, CC and MF categories, respectively, were identified as upregulated genes (Table II), and 453, 45 and 67 terms were identified for downregulated genes (Table III). The most enriched GO terms in the categories for upregulated genes were as follows: BP, CC and MF categories for upregulated genes were small molecule metabolic processes (P=1.87×10−9), extracellular region (P=3.64×10−23) and protein binding (P=7.34×10−7), respectively (Table II). The most enriched GO terms in the BP, CC and MF categories for downregulated genes were tissue development (P=4.66×10−8); extracellular region (P=1.02×10−10) and protein kinase C binding (P=1.31×10−3), respectively (Table III).

Table II.

Top five enriched gene ontology terms in biological process, cellular component and molecular function categories for upregulated DEGs.

Table II.

Top five enriched gene ontology terms in biological process, cellular component and molecular function categories for upregulated DEGs.

A, Biological process

GO_IDTermCountP-valueDEGs
GO:0044281Small molecule metabolic process99 1.87×10−9ABCC3, ACAA1, B3GNT3, CD320, DDX11, ECHS1, FA2H, GAPDH, UQCRFS1, WNT11a
GO:0055114Oxidation-reduction process44 9.79×10−9ACAA1, ACSS2, COX8A, ECHS1, FA2H, HMOX1, UQCRC1, UQCRC2, UQCRFS1, VAT1a
GO:0044710Single-organism metabolic process137 3.01×10−8ABCC3, ACAA1, B3GNT3, BMP4, PCBD1, PSMD8, RHOB, VAT1, WNT11, XRCC6a
GO:0043436Oxoacid metabolic process44 6.07×10−8ABCC3, ACAA1, B3GNT3, CKMT1A, ECHS1, SOD1, SULT2B1, TPI1, TST, UGT1A6a
GO:0006082Organic acid metabolic process44 9.50×10−8ABCC3, ACAA1, B3GNT3, CKMT1A, GOT1, SERINC2, SLC2A1, TPI1, TST, UGT1A6a

B, Cellular component

GO_IDTermCountP-valueDEGs

GO:0005576Extracellular region138 3.64×10−23ADIRF, BMP4, CAPG, IL1RN, ITGA3, ITGA6, ITGB4, ITGB5, VAT1, VDAC1a
GO:0031982Vesicle129 5.68×10−20ADIRF, AHNAK2, ENO1, ITGA3, ITGB4, ITGB5, SFN, UQCRC2, VASP, VAT1a
GO:0031988Membrane-bounded vesicle126 4.25×10−18ADIRF, ATP6AP1, BAIAP2L2, CAPG, EPS8L1, FTH1, FURIN, GAPDH, UQCRC2, VASPa
GO:0043230Extracellular organelle118 2.21×10−18ADIRF, GOT1, ITGA3, ITGB4, ITGB5, KLK14, UGT1A6, UPK3B, UQCRC2, VASPa
GO:0044421Extracellular region131 8.27×10−13ADIRF, HMOX1, IL1RN, ITGA3, ITGA6, ITGB4, ITGB5, KLK14, TXN, WNT11a

C, Molecular function

GO_IDTermCountP-valueDEGs

GO:0005515Protein binding182 7.34×10−7AATK, HSP90AA1, IRF2BP1, ITGA3, ITGA6, ITGB4, ITGB5, UQCRFS1, VASP, VDAC1a
GO:0016491Oxidoreductase activity31 9.29×10−7ACAA1, GAPDH, HMOX1, HPDL, HR, LDHA, MAOB, NDUFS3, PCBD1, PIRa
GO:0043236Laminin binding6 7.96×10−6ECM1, GPC1, ITGA3, ITGA6, LGALS1, LYPD3
GO:0050840Extracellular matrix binding7 2.07×10−5ECM1, GPC1, GPR56, ITGA3, ITGA6, LGALS1, LYPD3
GO:0008106Alcohol dehydrogenase (NADP+) activity4 3.68×10−5AKR1B1, AKR1C2, AKR1C3, ALDH3A1

{ label (or @symbol) needed for fn[@id='tfn2-etm-0-0-3648'] } GO, gene ontology; DEGs, differentially expressed genes; NADP, nicotinamide adenine dinucleotide phosphate.

a Not all of the gene names were included in the table.

Table III.

Top five enriched gene ontology terms in the biological process, cellular component and molecular function categories for downregulated DEGs.

Table III.

Top five enriched gene ontology terms in the biological process, cellular component and molecular function categories for downregulated DEGs.

A, Biological process

GO_IDTermCountP-valueDEGs
GO:0009888Tissue development42 4.66×10−8ADAM9, ALDH1A3, FNDC3B, NTN4, PKP2, RIPK4, TNFRSF19, TRIM16, TSC22D3, WNT7Ba
GO:0048513Organ development58 1.90×10−7ADAM9, EGLN1, LTBP3, MAP3K1, MDK, NRIP1, TNFRSF19, TNS3, TRIM16, TSC22D3a
GO:0048731System development70 6.10×10−7ADAM9, SGPL1, TNFAIP2, TNFRSF19, TNS3, TRIM16, TRIO, TSC22D3, WNT7B, ZSWIM6a
GO:0048518Positive regulation of biological process74 6.85×10−7ADAM9, GLIS3, HSPB1, IGFBP3, IRF1, ITGB8, KLK6, TRIM16, TRIO, WNT7Ba
GO:0009653Anatomical structure morphogenesis49 7.77×10−7ADAM9, MAP1B, MAP2, NTN4, PKP2, PTPRJ, RIPK4, SAT1, SEMA7A, SGPL1a

B, Cellular component

GO_IDTermCountP-valueDEGs

GO:0044421Extracellular region73 1.02×10−10ADAM9, CCDC80, CLIC5, FRAS1, SNX18, SOSTDC1, ST6GAL1, SULF2, TNFAIP2, VWA2a
GO:0005615Extracellular space35 1.02×10−8ADAM9, HSPG2, IGFBP3, MUC4, PLAT, POTEF, SERPINA3, TNFAIP2, VWA2, WNT7Ba
GO:0005576Extracellular region77 1.78×10−8ADAM9, KRT15, LCN2, SLC7A5, SNX18, SOSTDC1, ST6GAL1, SULF2, TACSTD2, TGM2a
GO:0043230Extracellular organelle56 1.92×10−8ADAM9, IVL, KRT13, MYOF, PLAT, POTEF, SLC7A5, SNX18, ST6GAL1, TACSTD2a
GO:0065010Extracellular organelle, membrane-bound56 1.92×10−8ADAM9, IGFBP3, LTBP3, MARCKS, SELENBP1, SNX18, ST6GAL1, TGM2, THSD4, VWA2a

C, Molecular function

GO_IDTermCountP-valueDEGs

GO:0005080Protein kinase C binding4 1.31×10−3ADAM9, HSPB1, MARCKS, PKP2
GO:0008009Chemokine activity4 1.42×10−3CXCL1, CXCL3, CXCL5, CXCL8
GO:0019838Growth factor binding6 1.43×10−3BMPR2, CTGF, IGFBP3, IGFBP6, LTBP3, TRIM16
GO:0031994Insulin-like growth factor I binding2 2.28×10−3IGFBP3, IGFBP6
GO:0055106Ubiquitin-protein transferase regulator activity2 2.28×10−3CDKN2A, TRIB1

{ label (or @symbol) needed for fn[@id='tfn4-etm-0-0-3648'] } GO, gene ontology; DEGs, differentially expressed genes.

a Not all of the gene names were included in the table.

According to the pathway enrichment analysis, 28 and 7 pathways were identified for the upregulated and downregulated genes, respectively (Table IV). The upregulated genes were significantly enriched in the glycolysis/gluconeogenesis (P=1.03×10−6), metabolic pathways (P=6.04×10−5), phenylalanine metabolism (P=4.00×10−4), oxidative phosphorylation (P=2.11×10−2) and the metabolism of xenobiotics by cytochrome P450 (P=4.14×10−3) (Table IV).

Table IV.

Top ten enriched pathways for upregulated differentially expressed genes and seven enriched pathways for downregulated DEGs.

Table IV.

Top ten enriched pathways for upregulated differentially expressed genes and seven enriched pathways for downregulated DEGs.

PathwayCountP-valueGene symbol
Upregulated
  Glycolysis/gluconeogenesis10 1.03×10−6ACSS2, ALDH3A1, ALDOA, ENO1, ENO2, GAPDH, LDHA, PGM1, PKM, TPI1
  Metabolic pathways43 6.04×10−5ACAA1, ACSL5, ACSS2, AGPAT2, AK1, AKR1B1, ALDH1A1, ALDH3A1, ALDOA, ALPP, ALPPL2, ATP5G1, ATP5G3, ATP6AP1, B3GNT3, CKMT1A, CKMT1B, COX8A, CYC1, ECHS1, ENO1, ENO2, GAPDH, GOT1, ITPK1, LDHA, MAOB, MGAT3, NDUFS3, NT5E, PGM1, PGP, PIK3C2B, PKM, PLA2G4B, PLCE1, PRDX6, TPI1, TST, UGT1A6, UQCRC1, UQCRC2, UQCRFS1
  Phenylalanine metabolism4 4.00×10−4ALDH3A1, GOT1, MAOB, PRDX6
  Parkinson's disease10 4.67×10−4ATP5G1, ATP5G3, COX8A, CYC1, NDUFS3, SLC25A5, UQCRC1, UQCRC2, UQCRFS1, VDAC1
  Huntington's disease12 5.52×10−4ATP5G1, ATP5G3, CLTB, COX8A, CYC1, NDUFS3, SLC25A5, SOD1, UQCRC1, UQCRC2, UQCRFS1, VDAC1
  Prion diseases5 8.46×10−4EGR1, HSPA1A, MAPK3, SOD1, STIP1
  Oxidative phosphorylation9 2.11×10−3ATP5G1, ATP5G3, ATP6AP1, COX8A, CYC1, NDUFS3, UQCRC1, UQCRC2, UQCRFS1
  Alzheimer's disease10 3.16×10−3ATP5G1, ATP5G3, COX8A, CYC1, GAPDH, MAPK3, NDUFS3, UQCRC1, UQCRC2, UQCRFS1
  Metabolism of xenobiotics by cytochrome P4506 4.14×10−3AKR1C2, AKR1C3, ALDH3A1, CYP1B1, EPHX1, UGT1A6
  Cardiac muscle contraction6 6.17×10−3ATP1A1, COX8A, CYC1, UQCRC1, UQCRC2, UQCRFS1
Downregulated
  Epithelial cell signaling in H. pylori infection3 2.92×10−2CXCL1, CXCL8, MAP3K14
  Complement and coagulation cascades3 3.03×10−2C3, PLAT, SERPINA1
  Histidine metabolism2 3.29×10−2ALDH1A3, AOC1
  Arrhythmogenic right ventricular cardiomyopathy3 3.62×10−2ITGB6, ITGB8, PKP2
  Axon guidance4 3.80×10−2EFNB2, NFAT5, NTN4, SEMA7A
  Chemokine signaling pathway5 3.80×10−2BCAR1, CXCL1, CXCL3, CXCL5, CXCL8
  Cytokine-cytokine receptor interaction6 4.55×10−2BMPR2, CXCL1, CXCL3, CXCL5, CXCL8, TNFRSF19

[i] DEGs, differentially expressed genes.

The downregulated genes were enriched in epithelial cell signaling in Helicobacter pylori infection (involving, CXCL1 and CXCL8; P=2.92×10−2), complement and coagulation cascades (P=3.03×10−2), arrhythmogenic right ventricular cardiomyopathy (P=3.62×10−2), chemokine signaling pathway (involving CXCL1, CXCL3, CXCL5 and CXCL8; P=3.80×10−2) and cytokine-cytokine receptor interaction (involving CXCL1, CXCL3, CXCL5 and CXCL8; P=4.55×10−2) (Table IV).

PPI network and module analysis

After the PPIs of DEGs were predicted using the STRING database, the PPI network was visualized (Fig. 1). Based on the ClusterONE plugin, two modules with the highest significance (module 1, P=9.96×10−5, nodes=10; module 2, P=8.98×10−4, nodes=7) were selected (Fig. 2).

The DEGs in module 1 (including, ITGB6, ITGA6, ITGB4, ITGB5, ITGA3 and ITGB8) were most significantly associated with functions of the integrin complex (CC, P=3.33×10−15), the protein complex involved in cell adhesion (CC, P=3.33×10−15) and the integrin-mediated signaling pathway (BP, P=1.34×10−14) (Table V). In module 2, DEGs were involved in the respiratory electron transport chain (BP, P=4.60×10−13) and the electron transport chain (BP, P=5.17×10−13) (Table VI).

Table V.

Top five enriched gene ontology terms in biological process, cellular component and molecular function categories for DEGs in module 1.

Table V.

Top five enriched gene ontology terms in biological process, cellular component and molecular function categories for DEGs in module 1.

A, Biological process

GO_IDTermCountP-valueDEG
GO:0007229Integrin-mediated signaling pathway7 1.34×10−14ITGB6, BCAR1, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0030198Extracellular matrix organization7 3.66×10−10ITGB6, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0043062Extracellular structure organization7 3.73×10−10ITGB6, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0007155Cell adhesion8 1.30×10−8ITGB6, BCAR1, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0022610Biological adhesion8 1.35×10−8ITGB6, BCAR1, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8

B, Cellular component

GO_IDTermCountP-valueDEG

GO:0008305Integrin complex6 3.33×10−15ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0098636Protein complex involved in cell adhesion6 3.33×10−15ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0043235Receptor complex6 2.87×10−9ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0030055Cell-substrate junction6 2.02×10−8BCAR1, VASP, ITGA6, ITGB4, ITGB5, ITGA3
GO:0009986Cell surface6 4.88×10−7ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8

C, Molecular function

GO_IDTermCountP-valueDEG

GO:0005178Integrin binding4 3.15×10−7ITGB6, ITGA6, ITGB5, ITGA3
GO:0050839Cell adhesion molecule binding4 2.35×10−6ITGB6, ITGA6, ITGB5, ITGA3
GO:0005102Receptor binding7 2.36×10−6ITGB6, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
GO:0043236Laminin binding2 1.47×10−4ITGA6, ITGA3
GO:0050840Extracellular matrix binding2 4.39×10−4ITGA6, ITGA3

[i] GO, gene ontology; DEGs, differentially expressed genes.

Table VI.

Top five enriched gene ontology terms in biological process, cellular component and molecular function categories for DEGs in module 2.

Table VI.

Top five enriched gene ontology terms in biological process, cellular component and molecular function categories for DEGs in module 2.

A, Biological process

GO_IDTermCountP-valueDEGs
GO:0022904Respiratory electron transport chain6 4.60×10−13CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0022900Electron transport chain6 5.17×10−13CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0045333Cellular respiration6 6.05×10−12CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0015980Energy derivation by oxidation of organic compounds6 5.80×10−10CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0006091Generation of precursor metabolites and energy6 2.32×10−9CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1

B, Cellular component

GO_IDTermCountP-valueDEGs

GO:0005743Mitochondrial inner membrane7 1.67×10−12ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0019866Organelle inner membrane7 3.57×10−12ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0070469Respiratory chain5 2.12×10−11CYC1, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0031966Mitochondrial membrane7 2.30×10−11ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
GO:0005740Mitochondrial envelope7 3.57×10−11ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1

C, Molecular function

GO_IDTermCountP-valueDEGs

GO:0015078Hydrogen ion transmembrane transporter activity4 4.15×10−8ATP5G3, COX8A, UQCRC1, UQCRFS1
GO:0008121 Ubiquinol-cytochrome-c reductase activity2 3.57×10−6UQCRC1, UQCRFS1
GO:0016681Oxidoreductase activity, acting on diphenols and related substances as donors, cytochrome as acceptor2 3.57×10−6UQCRC1, UQCRFS1
GO:0016679Oxidoreductase activity, acting on diphenols and related substances as donors2 4.76×10−6UQCRC1, UQCRFS1
GO:0015077Monovalent inorganic cation transmembrane transporter activity4 6.29×10−6ATP5G3, COX8A, UQCRC1, UQCRFS1

[i] DEG, differentially expressed genes; GO, gene ontology; BP, biological process; CC, cellular component; MF, molecular function.

The DEGs in module 1 were most significantly enriched in the focal adhesion pathway (P=5.20×10−14) and the extracellular matrix (ECM)-receptor interaction pathway (P=3.66×10−12) (Table VII). In addition, the DEGs in module 2 were enriched in Parkinson's disease (P=2.23×10−12), oxidative phosphorylation (P=2.49×10−12), Alzheimer's disease (P=1.33×10−11), Huntington's disease (P=2.56×10−11) and metabolic pathways (P=9.66×10−6) (Table VII).

Table VII.

The 13 and 6 enriched pathways for differentially expressed genes in modules 1 and 2, respectively.

Table VII.

The 13 and 6 enriched pathways for differentially expressed genes in modules 1 and 2, respectively.

PathwayCountP-valueGene symbol
A, Module 1

Focal adhesion9 5.20×10−14ITGB6, BCAR1, VASP, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
ECM-receptor interaction7s 3.66×10−12ITGB6, LAMA3, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
Arrhythmogenic right ventricular cardiomyopathy6 2.67×10−10ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
Hypertrophic cardiomyopathy6 5.41×10−10ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
Dilated cardiomyopathy6 8.90×10−10ITGB6, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
Regulation of actin cytoskeleton7 2.55×10−9ITGB6, BCAR1, ITGA6, ITGB4, ITGB5, ITGA3, ITGB8
Small cell lung cancer3 2.31×10−4LAMA3, ITGA6, ITGA3
Hematopoietic cell lineage2 7.47×10−3ITGA6, ITGA3
Pathways in cancer3 1.11×10−2LAMA3, ITGA6, ITGA3
Leukocyte transendothelial migration2 1.27×10−2BCAR1, VASP
Toxoplasmosis2 1.63×10−2LAMA3, ITGA6
Cell adhesion molecules2 1.65×10−2ITGA6, ITGB8

B, Module 2

Parkinson's disease7 2.23×10−12ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
Oxidative phosphorylation7 2.49×10−12ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
Alzheimer's disease7 1.33×10−11ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
Huntington's disease7 2.56×10−11ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1
Cardiac muscle contraction5 7.02×10−9CYC1, COX8A, UQCRC1, UQCRC2, UQCRFS1
Metabolic pathways7 9.66×10−6ATP5G3, CYC1, COX8A, UQCRC1, NDUFS3, UQCRC2, UQCRFS1

[i] ECM, extracellular matrix.

Co-expression analysis of DEGs and DE-lncRNAs

The pairs of co-expressed genes and lncRNAs were obtained and the enriched pathways for the DEGs co-expressed with each DE-lncRNAs are presented in Fig. 3. The DEGs co-expressed with lnc-SCD-1:13, lnc-LRR1-1:2, lnc-PTMS-1:3, lnc-S100P-3:1, lnc-AP000974.1-1:1 and lnc-RAB3IL1-2:1 were enriched in the pathways associated with cancer, such as basal cell carcinoma, pathways in cancer and ECM-receptor interaction (Table VIII). The DEGs co-expressed with lnc-SCD-1:13, lnc-LRR1-1:2 and lnc-S100P-3:1 were enriched in the Wnt signaling pathway (Table VIII). The DEGs co-expressed with lnc-SCD-1:13, lnc-LRR1-1:2, lnc-PTMS-1:3, lnc-S100P-3:1 and lnc-AP000974.1-1:1 were enriched in the Hedgehog signaling pathway (Table VIII).

Table VIII.

The DEGs co-expressed with differentially expressed lncRNAs associated with pathways in cancer.

Table VIII.

The DEGs co-expressed with differentially expressed lncRNAs associated with pathways in cancer.

lncRNA/pathwayDEG
lnc-SCD-1:13
  Wnt signaling pathwayDVL1, FZD7, NFAT5, WNT11, WNT7B
  Hedgehog signaling pathwayBMP4, WNT11, WNT7B
  Basal cell carcinomaBMP4, DVL1, FZD7, WNT11, WNT7B
  ECM-receptor interactionHSPG2, ITGA3, ITGB4, LAMA3, SDC1
  Glycolysis/gluconeogenesisALDH1A3, ALDOA, PKM
  Aldosterone-regulated sodium reabsorptionATP1A1, SFN, SGK1
  Glycerolipid metabolismAGPAT2, AKR1B1, LIPG
  Metabolism of xenobiotics by cytochrome P450AKR1C2, ALDH1A3, CYP1B1
  Steroid hormone biosynthesisAKR1C2, CYP1B1, SULT2B1
  Epithelial cell signaling in H. pylori infectionATP6AP1, CXCL8, MAP3K14
  Pathways in cancerBMP4, CXCL8, DVL1, FOS, FZD7, ITGA3, LAMA3, WNT11, WNT7B
  Arrhythmogenic right ventricular cardiomyopathyITGA3, ITGB4, PKP2
  MelanogenesisDVL1, FZD7, WNT11, WNT7B
lnc-LRR1-1:2
  Wnt signaling pathwayDVL1, NFAT5, WNT11, WNT7B
  Axon guidanceEFNA3, NFAT5, RHOD, UNC5B
  ECM-receptor interactionITGA3, LAMA3, SDC1
  Basal cell carcinomaBMP4, DVL1, WNT11, WNT7B
  Aldosterone-regulated sodium reabsorptionATP1A1, SGK1
  Hedgehog signaling pathwayBMP4, WNT11, WNT7B
  MalariaCXCL8, SDC1
  Glycerolipid metabolismAGPAT2, AKR1B1
  T cell receptor signaling pathwayFOS, MAP3K14, NFAT5
  Pathways in cancerBMP4, CXCL8, DVL1, FOS, ITGA3, LAMA3, SLC2A1, WNT11, WNT7B
  MelanogenesisDVL1, WNT11, WNT7B
  Protein digestion and absorptionATP1A1, KCNE3, SLC1A5
lnc-PTMS-1:3
  Basal cell carcinomaBMP4, DVL1, WNT11, WNT7B
  Aldosterone-regulated sodium reabsorptionSFN, SGK1
  Hedgehog signaling pathwayBMP4, WNT11, WNT7B
  ECM-receptor interactionITGA3, LAMA3, SDC1
  Pathways in cancerBMP4, DVL1, FOS, ITGA3, LAMA3, SLC2A1, WNT11, WNT7B
  MelanogenesisDVL1, WNT11, WNT7B
lnc-S100P-3:1
  ECM-receptor interactionITGA3, LAMA3, SDC1
  Basal cell carcinomaBMP4, DVL1, WNT11, WNT7B
  Glycolysis/gluconeogenesisACSS2, ALDH1A3, ALDOA, ENO2
  Aldosterone-regulated sodium reabsorptionATP1A1, SFN, SGK1
  Hedgehog signaling pathwayBMP4, WNT11, WNT7B
  Metabolism of xenobiotics by cytochrome P450AKR1C2, ALDH1A3, CYP1B1
  Steroid hormone biosynthesisAKR1C2, CYP1B1, SULT2B1
  Pathways in cancerBMP4, CXCL8, DVL1, FOS, ITGA3, LAMA3, SLC2A1, WNT11, WNT7B
  Fructose and mannose metabolismAKR1B1, ALDOA
  Protein digestion and absorptionATP1A1, KCNE3, SLC1A5
lnc-AP000974.1-1:1
  Wnt signaling pathwayDVL1, FZD7, NFAT5, WNT11, WNT7B
  Hedgehog signaling pathwayBMP4, WNT11, WNT7B
  Basal cell carcinomaBMP4, DVL1, FZD7, WNT11, WNT7B
  ECM-receptor interactionHSPG2, ITGA3, ITGB4, LAMA3, SDC1
  Glycolysis/gluconeogenesisALDH1A3, ALDOA, ENO2, PKM
  Aldosterone-regulated sodium reabsorptionATP1A1, SFN, SGK1
  Glycerolipid metabolismAGPAT2, AKR1B1, LIPG
  Metabolism of xenobiotics by cytochrome P450AKR1C2, ALDH1A3, CYP1B1
  Steroid hormone biosynthesisAKR1C2, CYP1B1, SULT2B1
  Pathways in cancerBMP4, CXCL8, DVL1, FOS, FZD7, ITGA3, LAMA3, WNT11, WNT7B
  Arrhythmogenic right ventricular cardiomyopathyITGA3, ITGB4, PKP2
  MelanogenesisDVL1, FZD7, WNT11, WNT7B
lnc-RAB3IL1-2:1
  Axon guidanceNFAT5, SEMA7A, UNC5B
  Basal cell carcinomaDVL1, FZD7
  Extracellular matrix-receptor interactionHSPG2, ITGA3, ITGA6
  Aldosterone-regulated sodium reabsorptionATP1A1, SGK1
  Folate biosynthesisALPP, ALPPL2
  Glycerolipid metabolismAGPAT2, LIPG
  N-Glycan biosynthesisMGAT3, ST6GAL1
  Regulation of actin cytoskeletonFGD3, ITGA3, ITGA6, PFN1
  Pathways in cancerCXCL8, DVL1, FZD7, ITGA3, ITGA6
  Arrhythmogenic right ventricular cardiomyopathyITGA3, ITGA6, PKP2

[i] lncRNA, long non-coding ribonucleic acid; DEGs, differentially expressed genes.

Discussion

In the present study, the RNA sequencing data between gastric cancer cells treated with celecoxib and those treated with DMSO was used to explore the mechanism of celecoxib treatment in gastric cancer cells. It has been previously demonstrated that altered patterns of DNA methylation associated with Helicobacter pylori infection of gastric epithelial cells may contribute to the risk of gastric cancer (29). Following Helicobacter pylori infection, the significant expression of CXCL5 and CXCL8 was observed in primary human gastric epithelial cells (30). Verbeke et al (31) also reported that CXC chemokines may contribute to the transition of chronic inflammation in esophageal and gastric cancer. In addition, CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7 and CXCL8) could promote the migration and proliferation of endothelial cells by interacting with CXCR2 (32). Furthermore, the overexpression of CXCL1 and CXCR2 may be involved in the tumor invasion in gastric cancer (33). The study by Park et al (34) demonstrated that the overexpression of CXCL5 may contribute to the pathogenesis of gastric cancer.

The results of the present study revealed that some DEGs (CXCL1 and CXCL8) were enriched in the epithelial cell signaling pathway in Helicobacter pylori infection whereas other DEGs (CXCL1, CXCL3, CXCL5 and CXCL8) were enriched in both the chemokine signaling and cytokine-cytokine receptor interaction pathways, which were consistent with the previous reports. Based on these results, CXCL1, CXCL3, CXCL5 and CXCL8 were suggested to contribute to the development of gastric cancer through multiple pathways.

ITGA3 is known to be involved in the development of gastric cancer (35). The MPS-1/ITGB4 signaling axis mediates cell migration and invasiveness, which may be used as targets during the therapy of gastric cancer (36). Song et al (35) revealed that the polymorphisms of microRNA-binding sites in the 3′UTR region of the integrin genes (ITGA3, ITGA6, ITGB3, ITGB4 and ITGB5) were associated with the susceptibility of gastric cancer. Pathway enrichment analysis revealed that integrin genes (ITGA3, ITGA6, ITGB4, ITGB5, ITGB6 and ITGB8) in module 1 were enriched in the integrin-mediated signaling pathway. Altogether, we could speculate that these integrin genes may participate in the celecoxib treatment of gastric cancer via the integrin-mediated signaling pathway.

Co-expression analysis revealed that the DEGs co-expressed with lnc-SCD-1:13, lnc-LRR1-1:2, lnc-PTMS-1:3, lnc-S100P-3:1, lnc-AP000974.1-1:1 or lnc-RAB3IL1-2:1 were enriched in a number of pathways, including ECM-receptor interaction, Wnt signaling and Hedgehog signaling pathways. A number of studies reported that lncRNAs are important in the pathogenesis of gastric cancer (3739). Chang et al (40) revealed that the genes in the ECM-receptor interaction pathway were involved in the metastasis and aggression of gastric cancer. In addition, Tang et al (41) demonstrated that miR-200b and miR-22 could synergistically inhibit the growth of gastric cancer through the Wnt-1 signaling pathway. Furthermore, Yan et al (42) reported that the activated Hedgehog signaling pathway was involved in the progression of gastric cancer. These results implied that lnc-SCD-1:13, lnc-LRR1-1:2, lnc-PTMS-1:3, lnc-S100P-3:1, lnc-AP000974.1-1:1 and lnc-RAB3IL1-2:1 may be important in the celecoxib treatment of gastric cancer via different pathways. However, the correlation between COX-2 and DEGs or DE-lncRNAs remains unclear, and needs to be confirmed by further experiments.

In conclusion, a total of 490 DEGs and 37 DE-lncRNAs were identified in the celecoxib group. Several DEGs (including CXCL1, CXCL3, CXCL5, CXCL8 and integrin genes) and DE-lncRNAs (including lnc-SCD-1:13, lnc-LRR1-1:2, lnc-PTMS-1:3, lnc-S100P-3:1, lnc-AP000974.1-1:1 and lnc-RAB3IL1-2:1) may affect celecoxib treatment of gastric cancer through different pathways. However, these results were obtained by bioinformatics analysis and require further validation.

Glossary

Abbreviations

Abbreviations:

lncRNAs

long non-coding RNAs

DEGs

differentially expressed genes

PPI

protein-protein interaction

ncRNAs

non-coding RNAs

miRNAs

microRNAs

DMSO

dimethylsulfoxide

QC

quality control

NGS

next generation sequencing

GO

gene ontology

BP

biological process

CC

cellular component

MF

molecular function

mPTP

mitochondrial permeability transition pore

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Akagi H, Higuchi H, Sumimoto H, Igarashi T, Kabashima A, Mizuguchi H, Izumiya M, Sakai G, Adachi M, Funakoshi S, et al: Suppression of myeloid cell leukemia-1 (Mcl-1) enhances chemotherapy-associated apoptosis in gastric cancer cells. Gastric Cancer. 16:100–110. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Hippo Y, Taniguchi H, Tsutsumi S, Machida N, Chong JM, Fukayama M, Kodama T and Aburatani H: Global gene expression analysis of gastric cancer by oligonucleotide microarrays. Cancer Res. 62:233–240. 2002.PubMed/NCBI

4 

Lee CH, Bang SH, Lee SK, Song KY and Lee IC: Gene expression profiling reveals sequential changes in gastric tubular adenoma and carcinoma in situ. World J Gastroenterol. 11:1937–1945. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Chia NY, Deng N, Das K, Huang D, Hu L, Zhu Y, Lim KH, Lee MH, Wu J, Sam XX, et al: Regulatory crosstalk between lineage-survival oncogenes KLF5, GATA4 and GATA6 cooperatively promotes gastric cancer development. Gut. 64:707–719. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Wadhwa R, Song S, Lee JS, Yao Y, Wei Q and Ajani JA: Gastric cancer-molecular and clinical dimensions. Nat Rev Clin Oncol. 10:643–655. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Wapinski O and Chang HY: Long noncoding RNAs and human disease. Trends Cell Biol. 21:354–361. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Du Z, Fei T, Verhaak RG, Su Z, Zhang Y, Brown M, Chen Y and Liu XS: Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat Struct Mol Biol. 20:908–913. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Passon DM, Lee M, Rackham O, Stanley WA, Sadowska A, Filipovska A, Fox AH and Bond CS: Structure of the heterodimer of human NONO and paraspeckle protein component 1 and analysis of its role in subnuclear body formation. Proc Natl Acad Sci USA. 109:4846–4850. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Song H, Sun W, Ye G, Ding X, Liu Z, Zhang S, Xia T, Xiao B, Xi Y and Guo J: Long non-coding RNA expression profile in human gastric cancer and its clinical significances. J Transl Med. 11:2252013. View Article : Google Scholar : PubMed/NCBI

11 

Hu Y, Wang J, Qian J, Kong X, Tang J, Wang Y, Chen H, Hong J, Zou W, Chen Y, et al: Long noncoding RNA GAPLINC regulates CD44-dependent cell invasiveness and associates with poor prognosis of gastric cancer. Cancer Res. 74:6890–6902. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Liu M, Li CM, Chen ZF, Ji R, Guo QH, Li Q, Zhang HL and Zhou YN: Celecoxib regulates apoptosis and autophagy via the PI3K/Akt signaling pathway in SGC-7901 gastric cancer cells. Int J Mol Med. 33:1451–1458. 2014.PubMed/NCBI

13 

Lan C, Yang L, Fan L, Zhang Y, Wang J, Guo GJ, Wan S, Yang S, Wang R and Fang D: Celecoxib inhibits helicobacter pylori-induced invasion of gastric cancer cells through an adenine nucleotide translocator-dependent mechanism. Anticancer Agents Med Chem. 13:1267–1272. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Yeh TS, Wu CW, Hsu KW, Liao WJ, Yang MC, Li AF, Wang AM, Kuo ML and Chi CW: The activated Notch1 signal pathway is associated with gastric cancer progression through cyclooxygenase-2. Cancer Res. 69:5039–5048. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Hu PJ, Yu J, Zeng ZR, Leung WK, Lin HL, Tang BD, Bai AH and Sung JJ: Chemoprevention of gastric cancer by celecoxib in rats. Gut. 53:195–200. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Thiel A, Mrena J and Ristimäki A: Cyclooxygenase-2 and gastric cancer. Cancer Metastasis Rev. 30:387–395. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Pang RP, Zhou JG, Zeng ZR, Li XY, Chen W, Chen MH and Hu PJ: Celecoxib induces apoptosis in COX-2 deficient human gastric cancer cells through Akt/GSK3β/NAG-1 pathway. Cancer Lett. 251:268–277. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Chen Z, Liu M, Liu X, Huang S, Li L, Song B, Li H, Ren Q, Hu Z, Zhou Y and Qiao L: COX-2 regulates E-cadherin expression through the NF-κB/Snail signaling pathway in gastric cancer. Int J Mol Med. 32:93–100. 2013.PubMed/NCBI

19 

Saito Y, Suzuki H, Imaeda H, Matsuzaki J, Hirata K, Tsugawa H, Hibino S, Kanai Y, Saito H and Hibi T: The tumor suppressor microRNA-29c is downregulated and restored by celecoxib in human gastric cancer cells. Int J Cancer. 132:1751–1760. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Patel RK and Jain M: NGS QC toolkit: A platform for quality control of next-generation sequencing data. Enc Metagenomics. 1–5. 2013. View Article : Google Scholar

21 

Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R and Salzberg SL: TopHat2: Accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14:R362013. View Article : Google Scholar : PubMed/NCBI

22 

Rosenbloom KR, Armstrong J, Barber GP, Casper J, Clawson H, Diekhans M, Dreszer TR, Fujita PA, Guruvadoo L, Haeussler M, et al: The UCSC genome browser database: 2015 update. Nucleic Acids Res. 43:D670–D681. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, Salzberg SL, Rinn JL and Pachter L: Differential gene and transcript expression analysis of RNA-seq experiments with tophat and cufflinks. Nat Protoc. 7:562–578. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Volders PJ, Helsens K, Wang X, Menten B, Martens L, Gevaert K, Vandesompele J and Mestdagh P: LNCipedia: A database for annotated human lncRNA transcript sequences and structures. Nucleic Acids Res. 41:(Database Issue). D246–D251. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al: Bioconductor: Open software development for computational biology and bioinformatics. Genome Biol. 5:R802004. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Nepusz T, Yu H and Paccanaro A: Detecting overlapping protein complexes in protein-protein interaction networks. Nat Methods. 9:471–472. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Niwa T, Tsukamoto T, Toyoda T, Mori A, Tanaka H, Maekita T, Ichinose M, Tatematsu M and Ushijima T: Inflammatory processes triggered by Helicobacter pylori infection cause aberrant DNA methylation in gastric epithelial cells. Cancer Res. 70:1430–1440. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Mustapha P, Paris I, Garcia M, Tran CT, Cremniter J, Garnier M, Faure JP, Barthes T, Boneca IG, Morel F, et al: Chemokines and antimicrobial peptides cag-dependent early response to helicobacter pylori infection in primary human gastric epithelial cells. Infect Immun. 82:2881–2889. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Verbeke H, Geboes K, Van Damme J and Struyf S: The role of CXC chemokines in the transition of chronic inflammation to esophageal and gastric cancer. Biochim Biophys Acta. 1825:117–129. 2012.PubMed/NCBI

32 

Mukaida N, Sasaki S and Baba T: Chemokines in cancer development and progression and their potential as targeting molecules for cancer treatment. Mediators Inflamm. 2014:1703812014. View Article : Google Scholar : PubMed/NCBI

33 

Cheng WL, Wang CS, Huang YH, Tsai MM, Liang Y and Lin KH: Overexpression of CXCL1 and its receptor CXCR2 promote tumor invasion in gastric cancer. Ann Oncol. 22:2267–2276. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Park JY, Park KH, Bang S, Kim MH, Lee JE, Gang J, Koh SS and Song SY: CXCL5 overexpression is associated with late stage gastric cancer. J Cancer Res Clin Oncol. 133:835–840. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Song X, Zhong H, Zhou J, Hu X, Zhou Y, Ye Y, Lu X, Wang J, Ying B and Wang L: Association between polymorphisms of microRNA-binding sites in integrin genes and gastric cancer in Chinese han population. Tumor Biol. 36:2785–2792. 2015. View Article : Google Scholar

36 

Yang ZY, Jiang H, Qu Y, Wei M, Yan M, Zhu ZG, Liu BY, Chen GQ, Wu YL and Gu QL: Metallopanstimulin-1 regulates invasion and migration of gastric cancer cells partially through integrin β4. Carcinogenesis. 34:2851–2860. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Lin XC, Zhu Y, Chen WB, Lin LW, Chen DH, Huang JR, Pan K, Lin Y, Wu BT, Dai Y and Tu ZG: Integrated analysis of long non-coding RNAs and mRNA expression profiles reveals the potential role of lncRNAs in gastric cancer pathogenesis. Int J Oncol. 45:619–628. 2014.PubMed/NCBI

38 

Chen S, Li P, Xiao B and Guo J: Long noncoding RNA HMlincRNA717 and AC130710 have been officially named as gastric cancer associated transcript 2 (GACAT2) and GACAT3, respectively. Tumor Biol. 35:8351–8352. 2014. View Article : Google Scholar

39 

Okugawa Y, Toiyama Y, Hur K, Toden S, Saigusa S, Tanaka K, Inoue Y, Mohri Y, Kusunoki M, Boland CR, et al: Metastasis-associated long non-coding RNA drives cancer development and promotes peritoneal metastasis. Carcinogenesis. 35:2731–2739. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Chang W, Ma L, Lin L, Gu L, Liu X, Cai H, Yu Y, Tan X, Zhai Y, Xu X, et al: Identification of novel hub genes associated with liver metastasis of gastric cancer. Int J Cancer. 125:2844–2853. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Tang H, Kong Y, Guo J, Tang Y and Xie X, Yang L, Su Q and Xie X: Diallyl disulfide suppresses proliferation and induces apoptosis in human gastric cancer through Wnt-1 signaling pathway by up-regulation of miR-200b and miR-22. Cancer Lett. 340:72–81. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Yan R, Peng X, Yuan X, Huang D, Chen J, Lu Q, Lv N and Luo S: Suppression of growth and migration by blocking the hedgehog signaling pathway in gastric cancer cells. Cell Oncol (Dordr). 36:421–435. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2016
Volume 12 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song B, Du J, Feng Y, Gao YJ and Zhao JS: Co-expressed differentially expressed genes and long non-coding RNAs involved in the celecoxib treatment of gastric cancer: An RNA sequencing analysis. Exp Ther Med 12: 2455-2468, 2016
APA
Song, B., Du, J., Feng, Y., Gao, Y., & Zhao, J. (2016). Co-expressed differentially expressed genes and long non-coding RNAs involved in the celecoxib treatment of gastric cancer: An RNA sequencing analysis. Experimental and Therapeutic Medicine, 12, 2455-2468. https://doi.org/10.3892/etm.2016.3648
MLA
Song, B., Du, J., Feng, Y., Gao, Y., Zhao, J."Co-expressed differentially expressed genes and long non-coding RNAs involved in the celecoxib treatment of gastric cancer: An RNA sequencing analysis". Experimental and Therapeutic Medicine 12.4 (2016): 2455-2468.
Chicago
Song, B., Du, J., Feng, Y., Gao, Y., Zhao, J."Co-expressed differentially expressed genes and long non-coding RNAs involved in the celecoxib treatment of gastric cancer: An RNA sequencing analysis". Experimental and Therapeutic Medicine 12, no. 4 (2016): 2455-2468. https://doi.org/10.3892/etm.2016.3648