Integrated analysis of mRNA and miRNA expression profiles in pancreatic ductal adenocarcinoma

  • Authors:
    • Hongwei Sun
    • Liang Zhao
    • Kehua Pan
    • Zhao Zhang
    • Mengtao Zhou
    • Guoquan Cao
  • View Affiliations

  • Published online on: March 24, 2017     https://doi.org/10.3892/or.2017.5526
  • Pages: 2779-2786
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the present study, to investigate the potential molecular mechanism of pancreatic ductal adenocarcinoma (PDAC), mRNA and miRNA expression profiles were integrated for systematic analysis. Results showed that a total of 76 common differentially expressed genes (DEGs) were identified from 2 mRNA expression profiles that contained 39 tumor and 15 normal samples. Notably, the tumor and normal samples were able to be clearly classified into 4 groups based on the DEGs. mRNA‑miRNA regulation network analysis indicated that 22 out of the 76 DEGs including MUC4, RRM2 and CCL2 are regulated by 5 reported miRNAs. Survival analysis using SurvExpress database demonstrated that the common DEGs were able to significantly differentiate low- and high-risk PDAC groups in 4 datasets. In summary, various biological processes are probably involved in the development and progression of PDAC. Firstly, activation of MUC4 induces nuclear translocation of β-catenin and promotes the process of angiogenesis that provides necessary nutrition or oxygen for cancer cells. Then, RRM2 induces the invasiveness of PDAC via NF-κB. Finally, the formation of an immunosuppressive tumor microenvironment by recruiting regulatory T cells with high expression of CCL2 further promotes cancer cell proliferation and vascularization. Identification of valuable biological processes and genes can be helpful for the understanding of the molecular mechanism of PDAC.

Introduction

Pancreatic cancer is the fourth most common cause of global cancer-related deaths (1) There were ~44,000 newly diagnosed cases of pancreatic cancer and more than 37,000 related deaths in 2012 in the US (2). In addition, the average 5-year survival rate is ~7.7% based on the data from 2006 to 2012 (3). Pancreatic ductal adenocarcinoma (PDAC), the most common histological subtype, comprises 90% of all pancreatic cancer cases (4). PDAC always displays local invasion and distant metastasis during early stages leading to poor prognosis with an overall 5-year survival rate of only 5% (5). World Health Organization (WHO) and Surveillance, Epidemiology and End Results (SEER) mortality data indicate that the occurrence of PDAC increases with age (~71 years) (6). In addition, researchers have shown that various risk factors can contribute to the development of PDAC. In Italy, PDAC risk was found to be 4.3-fold higher in heavy smokers (>20 cigarettes/day) compared with never smokers (7). In addition, alcohol intake was found to be associated with PDAC mortality based on data from the Cancer Prevention Study II. The results show evidence that alcohol consumption promotes PDAC mortality (8).

To explore the molecular mechanism of PDAC, numerous studies have been carried out using advanced microarray or next-generation sequencing technology. Previous studies based on microarrays have identified several genes that play an important role in PDAC. GNAI2, G protein subunit αi2, was found to be significantly upregulated in PDAC, and can mediate the functions of dopamine receptor D2 (DRD2) on cAMP signaling. Knockdown or inhibition of DRD2 was found to reduce the proliferation of PDAC cells (9). Teodorczyk at al revealed that CD95 is associated with stemness and epithelial-mesenchymal transition (EMT) in PDAC based on an in silico analysis of 36 RNA profiles (10). An in vitro experiment demonstrated that PDAC growth and metastasis can be significantly reduced by pharmacological inhibition of CD95 activity, and Sck is necessary for the CD95 induction of cell cycle progression (10). In addition, expression levels of microRNAs and lncRNAs were also explored in PDAC, and several markers have been identified including miR-10b, miR-155, miR-106b (11) and lncRNA AFAP1-AS1 (12). Recently, whole-exome sequencing of 109 micro-dissected PDAC samples identified multiple novel mutated genes in PDAC such as RBM10, KRAS, BRAF and high-frequency alterations in Wnt signaling, chromatin remodelling and cell cycle pathways (13).

Research has been carried out to explore the molecular mechanisms of PDAC based on microarray expression profiles or next-generation sequencing. However, studies with the integration of mRNA and miRNA expression profiles have not been widely applied in PDAC. In recent years, more and more microarray expression datasets have been submitted to the Gene Expression Omnibus (GEO) database, and re-analysis of the deposited datasets with various bioinformatics algorithms can be helpful (14). In the present study, we firstly identified the common differentially expressed genes (DEGs) in PDAC based on 2 mRNA expression profiles from 2 independent laboratories. Then functional annotation of the common DEGs based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases was carried out. Furthermore, an interaction network between the identified DEGs and documented miRNAs was constructed. Finally, the identified DEGs were virtually validated using SurvExpress online database.

Materials and methods

Acquisition of mRNA and miRNA expression profiles

In the present study, publicly available datasets from Expression Omnibus Database (GEO) (http://www.ncbi.nlm.nih.gov/geo/)were used. Firstly, we carefully searched the GEO database, and downloaded 2 mRNA expression profiles. GSE71989 submitted by Thomas Schmittgen in 2015 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE71989) consists of 14 PDAC and 8 normal pancreas tissues. The other dataset GSE32676 submitted by Tran in 2011 (15) consists of 25 PDAC and 7 normal pancreas tissues. Based on the instruction from manufacturer, RNAs were extracted and hybridized to Affymetrix Human Genome U133 Plus 2.0 array. Detailed sample information and experiment designs were documented in the previous studies.

Differentially expressed gene screening

Data analysis was carried out using in-house R script and publicly available annotation database. In brief, mRNA expression values were firstly subject to background correction, normalization and log2 transformation using GeneChip Robust Multi-array Analysis (GC-RMA) algorithm (16). Furthermore, uninformative control probe sets were filtered out. In addition, the average expression value was calculated for the genes with multiple probes. Finally, DEGs were screened using Linear Models for Microarray Data (Limma) package (17) within the bioconductor. The criteria were set to adjust p-value ≤0.05 and |log2 fold-change (FC)| ≥2. In addition, the common DEGs between the 2 datasets were identified based on a Venn diagram. The common DEGs were used to construct a heat map using Heatmap.2 method within ggplot package (18).

GO and KEGG pathway annotation

The functions of the identified DEGs were further annotated using GO and KEGG pathway databases using the online tools of Database for Annotation, Visualization and Integrated Discovery (DAVID) (19). The GO term consist of biological process (BP), cellular component (CC) and molecular function (MF). The criterion was set to p<0.05.

mRNA-miRNA interaction network

Numerous studies show that miRNAs play an important role in the regulation of carcinogenesis, malignant transformation and metastatic processes by preventing mRNA expression or via other processes (20). Cote et al (11) showed that 5 miRNAs, miR-10b-5p, miR-155-5p, miR-106b-5p, miR-30c-5p and miR-212-3p, have excellent performance to distinguish PDAC from normal samples. In addition, the sensitivity and specificity were 96 and 100%, respectively, in the training and validation cohorts. In the present study, we constructed the mRNA-miRNA interaction network based on the common DEGs and the 5 miRNAs. In brief, target genes of the 5 miRNAs were predicted based on the microCosm, mirTarbase and TargetScan databases. Then, the intersection between the common DEGs and the target genes were selected. Finally, the interaction network was constructed using CyTargetLinker (21) plugin in Cytoscape (22).

Virtual validation of the common DEGs

Clinical outcomes of the DEGs are critical for the diagnosis or treatment of PDAC. In the present study, virtual validation of the DEGs was carried out using SurvExpress online tool (23). This tool is based on a cancer-wide gene expression database with clinical outcomes. Four datasets were used for the virtual validation including GSE21501, GSE28735, TCGA PDAC and ICGC PDAC. Detailed information for the datasets can be found in previous studies. Parameter setting were carefully selected according to the developer's instructions.

Results

DEGs in PDAC

After background correction and normalization, the gene expression median values for different samples in the 2 datasets were almost at the same level (Fig. 1). Then, the datasets were subjected to DEG analysis. Results showed that a total of 364 and 816 DEGs were screened out for GSE32676 and GSE71989, respectively. For GSE32676, 292 genes (80.2%) were upregulated and 72 genes (19.8%) were downregulated. For GSE71989, 666 genes (81.6%) were upregulated and 150 genes (18.4%) were downregulated. Among those DEGs, 76 genes were differentially expressed in both GSE32676 and GSE71989 (Fig. 2). Fold-changes for the common DEGs are listed in Table I.

Table I.

The identified 76 common DEGs in GSE32676 and GSE71989.

Table I.

The identified 76 common DEGs in GSE32676 and GSE71989.

Fold-change Fold-change


GeneGSE32676GSE71989GeneGSE32676GSE71989
CCL2−3.712.77LAMA33.752.80
TMC53.332.93NR4A2−2.282.50
GJB24.542.58HOXB32.082.09
DPCR14.232.29EFNA52.122.16
MMP112.882.92ANLN2.392.08
CCL8−3.232.12CTSE5.123.49
LCN24.592.18ANO12.832.80
ZWINT2.302.12MTUS2−2.32−2.39
IFI272.103.37TSPAN14.412.18
MMP19−3.002.39 C15orf483.772.57
NQO13.093.41MMP283.062.26
SLC6A145.883.34 C19orf335.672.99
ADAMTS122.692.41LAMC22.252.37
RRM22.402.38VILL2.982.10
SFTA24.142.27 SERPINB55.622.29
PTGDS−2.792.17CAMK2N12.082.47
LAMB34.872.48 ST6GALNAC13.752.61
GPRC5A3.143.46ETV12.182.38
PHLDA23.043.40DCBLD12.262.40
PPP1R1A−2.42−2.03CST14.902.25
OAS12.512.10GCNT33.932.38
ECT22.403.04SOCS3−2.433.25
SDR16C54.783.21MUC44.302.44
LOC1005059844.963.34SDC12.682.36
MALL2.132.34EPPK13.542.31
THBS1−3.112.60AGR23.962.45
CEACAM13.472.08AGR33.763.11
CLDN233.212.21SFN2.643.75
KLF53.092.09CDK12.602.21
CEACAM56.813.96OGN−3.202.42
ITGA23.332.65AOC13.762.44
CEACAM65.854.22EMP1−2.633.38
KRT174.013.00S100P6.754.78
S100A63.112.92AHNAK23.762.55
FOSB−3.973.40MSLN6.443.48
TFF15.062.92CAPN85.282.99
TOP2A2.763.21KRT196.223.93
LY6E2.162.37CH25H−2.422.06

[i] DEGs, differentially expressed genes.

Furthermore, all samples from the 2 datasets were subjected to hierarchical clustering analysis based on the common DEGs. As shown in Fig. 3, tumor (red) and normal (blue) samples were abe to be clearly classified into different subgroups. Due to tumor heterogeneity and expression value variation, a few normal and tumor samples were erroneously classified.

GO and KEGG pathway annotation

In order to explore the biological functions of the common DEGs, functional annotation of the common DEGs was carried out using DAVID online tool based on GO and KEGG pathway databases. Results showed that the common DEGs can be significantly enriched into 4 KEGG pathways (Table II). Six genes participated in the ECM-receptor interaction pathway (p=0.000013), 5 genes were involved in the p53 signaling pathway (p=0.00012), 4 genes mapped to small cell lung cancer (p=0.0040) and 5 genes play a role in the pathway of focal adhesion (p=0.0069). In addition, the common DEGs were mainly related to the biological process of epidermis development (p=0.0001), ectoderm development (p=0.0002) and cell adhesion (p=0.0008) (Table III). In addition, the top 5 cellular components include extracellular matrix (p=0.0), extracellular region (p=0.0), extracellular region part (p=0.0), proteinaceous extracellular matrix (p=0.0002) and anchored to membrane (p=0.023) (Table III). In addition, 5 significant molecular function were enriched for the common DEGs including structural molecule (p=0.0035), enzyme inhibitor (p=0.0044), heparin binding (p=0.0081), metalloendopeptidase (p=0.0083) and protein kinase inhibitor activities (p=0.0091) (Table III).

Table II.

KEGG pathway enrichment result for the common DEGs.

Table II.

KEGG pathway enrichment result for the common DEGs.

TermGenesP-value
hsa04512:ECM-receptor interactionLAMB3, SDC1, LAMA3, ITGA2, LAMC2, THBS11.39E-05
hsa04115:p53 signaling pathwayCDK1, SERPINB5, RRM2, SFN, THBS11.21E-04
hsa05222:Small cell lung cancerLAMB3, LAMA3, ITGA2, LAMC20.0040471
hsa04510:Focal adhesionLAMB3, LAMA3, ITGA2, LAMC2, THBS10.0069739

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; DEGs, differentially expressed genes.

Table III.

Top 5 GO terms for the common DEGs.

Table III.

Top 5 GO terms for the common DEGs.

IDGO termP-valuesGenes
BP
  GO:0008544Epidermis development0.0001LAMB3, LAMA3, KRT17, AHNAK2, LAMC2, SFN, EMP1
  GO:0007398Ectoderm development0.0002LAMB3, LAMA3, KRT17, AHNAK2, LAMC2, SFN, EMP1
  GO:0007155Cell adhesion0.0008DCBLD1, LAMB3, LAMA3, CCL2, MSLN, ITGA2, LAMC2, THBS1, CEACAM1, CLDN23, MUC4
  GO:0022610Biological adhesion0.0008DCBLD1, LAMB3, LAMA3, CCL2, MSLN, ITGA2, LAMC2, THBS1, CEACAM1, CLDN23, MUC4
  GO:0048545Response to steroid hormone stimulus0.0015KRT19, SDC1, CCL2, SOCS3, TFF1, THBS1
CC
  GO:0031012Extracellular matrix0.0000OGN, LAMB3, LAMA3, MMP19, LAMC2, MMP28, ADAMTS12, THBS1, MMP11, MUC4
  GO:0005576Extracellular region0.0000CCL2, MMP19, CCL8, CST1, MMP28, SFN, MUC4, MMP11, LCN2, OGN, LAMB3, LAMA3, PTGDS, SERPINB5, MSLN, SFTA2, LAMC2, EFNA5, ADAMTS12, TFF1, AGR3, THBS1, AGR2, CEACAM1
  GO:0044421Extracellular region part0.0000CCL2, MMP19, CCL8, MMP28, SFN, MMP11, MUC4, OGN LAMB3, LAMA3, SERPINB5, LAMC2, EFNA5, TFF1, ADAMTS12, THBS1
  GO:0005578Proteinaceous extracellular matrix0.0002OGN, LAMB3, LAMA3, MMP19, LAMC2, MMP28, ADAMTS12, MMP11, MUC4
  GO:0031225Anchored to membrane0.0231LY6E, MSLN, CEACAM6, CEACAM5, EFNA5
MF
  GO:0005198Structural molecule activity0.0035KRT19, LAMB3, LAMA3, EPPK1, KRT17, THBS1, VILL, CLDN23, MUC4
  GO:0004857Enzyme inhibitor activity0.0044SERPINB5, SOCS3, PPP1R1A, CST1, SFN, CAMK2N1
  GO:0008201Heparin binding0.0081CCL2, CCL8, LAMC2, THBS1
  GO:0004222 Metalloendopeptidase activity0.0083MMP19, MMP28, ADAMTS12, MMP11
  GO:0004860Protein kinase inhibitor activity0.0091SOCS3, SFN, CAMK2N1

[i] GO, Gene Ontology; DEGs, differentially expressed genes; BP, biological process; CC, cellular component; MF, molecular function.

mRNA-miRNA network construction

To explore mRNA and miRNA regulation mechanism, the mRNA-miRNA pairings were constructed. The results indicated that the 5 miRNAs were able to target 3,760, 827 and 3,671 genes in the MicroCosm, mirTarbase and TargetScan database, respectively. Among those, 22 genes were also identified in the common DEGs. Then, the interaction network between the 22 genes and 5 miRNAs were constructed (Fig. 4). Based on the figure, we found that hsa-miR-212-3p can regulate 6 genes, hsa-miR-106p-5p can regulate 5 genes, hsa-miR-155-5p can regulate 5 genes, hsa-miR-10b-5p can regulate 5 genes and hsa-miR-30c-5p can regulate 6 genes.

Virtual validation

Biomarker validation is critical in the study of cancer molecular mechanism research. The prognostic performance of the common DEGs were validated using the SurvExpress online tool which provides survival analysis and risk assessment. Results are shown in Fig. 5 and are summarized in Table IV. The figure indicated that low- and high-risk PDAC groups can be significantly differentiated in the 4 datasets, and the p-values were 9.9e-15, 0.0, 6.0e-13 and 1.1e-06, respectively. In addition, the higher concordance index (CI) demonstrated that better prediction results can be achieved based on those DEGs (Table IV).

Table IV.

Virtual validation results of the common DEGs using 4 PDAC datasets.

Table IV.

Virtual validation results of the common DEGs using 4 PDAC datasets.

DEGs between risk groups

DatasetSamplesGenes foundRisk groups p-valueCINo.DEGs
TCGA176759.90E-1579.649TMC5, GJB2, DPCR1, MMP11, ZWINT
ICGC189750.00E+0082.117CCL8, ZWINT, NQO1, OAS1, ECT2
GSE21501132756.00E-1395.411SLC6A14, CEACAM5, FOSB, LAMA3, ANO1
GSE28735  90731.10E-0699.9  4RRM2, PTGDS, ITGA2, C15orf48

[i] DEGs, differentially expressed genes; PDAC, pancreatic ductal adenocarcinoma; CI, concordance index.

Discussion

With the development of microarray and next generation sequencing technologies, understanding of the molecular mechanism of pancreatic ductal adenocarcinoma (PDAC) has been significantly advanced. In the present study, we aimed to unveil the complex mechanism of PDAC by integrating different types of omics data. A total of 76 genes were simultaneously differentially expressed in 39 tumor tissues. Notably, a heat map showed that tumor and normal samples were able to be clearly distinguished based on the DEGs. The erroneous assignment of 2 samples was probably due to tumor heterogeneity.

KEGG pathway enrichment revealed that the DEGs were involved in the ECM-receptor interaction pathway. Studies have shown that PDAC is characterized by prominent desmoplasia (24). Extracellular matrix (ECM) proteins, ECM metabolizing enzymes and growth factors are main components of desmoplasia, and the components can promote the growth of cancer cells (25). In addition, ECM proteins and desmoplastic secreted growth factors can activate intracellular signals including reactive oxygen species that prevent the death of PDAC cancer cells (25). In addition, several ECM components such as collagens I/III/IV, decorin and versican may be of clinical prognostic significance in PDAC (26). Another significantly enriched pathway is focal adhesion. Researches have demonstrated that interaction between integrin and focal adhesion kinase can regulate cancer cell adhesion and invasion (27,28). Sawai et al showed that phosphorylation of focal adhesion kinase is involved in the aggressive capability of PDAC via the Ras/ERK signaling pathway (29). In addition, MUC16, a heavily glycosylated type-I transmembrane mucin, can facilitate PDAC growth and metastasis via focal adhesion signaling (30).

The mRNA and miRNA interaction network analysis further unveiled the complex mechanism of PDAC. The interaction network showed that hsa-miR-212-3p can regulate the DEG MUC4. This gene encodes highly glycosylated integral membrane glycoprotein in the cell surface (31). In addition, immunohistochemical analyses based on 135 PDAC tissues demonstrated that MUC4 was significantly highly expressed in patients with poor prognosis (p=0.0043) (32). In addition, hypomethylation of the MUC4 promoter probably participated in the carcinogenesis and malignant development of PDAC based on DNA methylation-specific PCR analysis of 116 microdissected foci (33). Jonckheere et al showed that MUC4 can interact with the ErbB2 oncogenic receptor via EGF domains, and inhibition of MUC4 expression can affect the downstream JNK pathway (34).

Another important gene is RRM2 which encodes one of 2 non-identical subunits for ribonucleotide reductase. Ribonucleotide reductase has been demonstrated to be a determinant of gemcitabine chemoresistance in human cancers (35), and the level of RRM2 can regulate enzyme activity (36). Duxbury et al showed that high expression of RRM2 is related to gemcitabine chemoresistance in PDAC (37). The suppression of RRM2 by siRNA significantly inhibited tumor growth, metastasis and increased tumor apoptosis (37). Research has also shown that NF-κB is the key mediator by which RRM2 induces invasiveness in PDAC (38). RRM2 and its downstream intermediaries can become potential drug targets (38). The mRNA-miRNA interaction network demonstrated that hsa-miR-106p-5p and hsa-miR-155-5p can regulate RRM2.

In addition, the CCL2 gene, which is also refered to as monocyte chemoattractant protein 1 and is a small cytokine (39), has been widely reported to be related to PDAC progression. In PDAC, the expression of CCL2 was found to be significantly elevated (40). In addition, Kalbasi et al demonstrated that highly expressed CCL2 can recruit Ly6C+CCR2+ inflammatory monocytes or macrophages to the regions surrounding the tumor and promote tumor proliferation and vascularization (41). In addition, PDAC cancer cells can construct an immunosuppressive tumor microenvironment by recruiting regulatory T cells with the high-expression of CCL5 (42). The mRNA-miRNA interaction network showed that hsa-miR-155-5p can regulate CCL2.

Apart from the above-mentioned 3 critical genes in PDAC, some rarely reported or novel genes such as ECT2, SDC1, SOCS3, TMC5 and NR4A2, also play a role in the development of PDAC. The ECT2 gene can promote Rho activity during cytokinesis, and RT-PCR results showed that it was highly expressed in PDAC (43). Statistical analysis showed that epithelial expression of SDC1 was positively correlated with survival time in PDAC patients (p=0.029) (44). Lesina et al showed that homozygous deletion of Socs3 can aberrantly activate Stat3 and promote PDAC development (45).

In summary, the development and progression of PDAC were probably induced via various processes. Firstly, activation of MUC4 induces nuclear translocation of β-catenin and promotes the process of angiogenesis that can provide necessary nutrition or oxygen for cancer cells. Then, RRM2 can induce the invasiveness of PDAC via NF-κB. Finally, the formation of an immunosuppressive tumor microenvironment by recruiting regulatory T cells with high expression of CCL2 further promotes cancer cell proliferation and vascularization.

Acknowledgements

The present study was funded by the Natural Science Foundation of Zhejiang Province (nos. LY15H030016 and LY15H160056) and Wenzhou Science & Technology Bureau (no. Y20140696).

References

1 

Hariharan D, Saied A and Kocher HM: Analysis of mortality rates for pancreatic cancer across the world. HPB. 10:58–62. 2008. View Article : Google Scholar : PubMed/NCBI

2 

American Cancer Society: Cancer Facts & Figures 2012. American Cancer Society; Atlanta, GA, USA: 2012

3 

Stat Fact Sheets SEER: Pancreas Cancer. National Cancer Institute; 2013

4 

Ishiwata T: Pancreatic ductal adenocarcinoma: Basic and clinical challenges for better prognosis. J Carcinog Mutagen. S9:2157–2518. 2013.http://dx.doi.org/4172/2157-2518.S9-005

5 

Siegel R, Ward E, Brawley O and Jemal A: Cancer statistics, 2011: The impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin. 61:212–236. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Partensky C: Toward a better understanding of pancreatic ductal adenocarcinoma: Glimmers of hope? Pancreas. 42:729–739. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Talamini R, Polesel J, Gallus S, Dal Maso L, Zucchetto A, Negri E, Bosetti C, Lucenteforte E, Boz G, Franceschi S, et al: Tobacco smoking, alcohol consumption and pancreatic cancer risk: A case-control study in Italy. Eur J Cancer. 46:370–376. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Gapstur SM, Jacobs EJ, Deka A, McCullough ML, Patel AV and Thun MJ: Association of alcohol intake with pancreatic cancer mortality in never smokers. Arch Intern Med. 171:444–451. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Jandaghi P, Najafabadi HS, Bauer AS, Papadakis AI, Fassan M, Hall A, Monast A, von Knebel Doeberitz M, Neoptolemos JP, Costello E, et al: Expression of DRD2 is increased in human pancreatic ductal adenocarcinoma and inhibitors slow tumor growth in mice. Gastroenterology. 151:1218–1231. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Teodorczyk M, Kleber S, Wollny D, Sefrin JP, Aykut B, Mateos A, Herhaus P, Sancho-Martinez I, Hill O, Gieffers C, et al: CD95 promotes metastatic spread via Sck in pancreatic ductal adenocarcinoma. Cell Death Differ. 22:1192–1202. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Cote GA, Gore AJ, McElyea SD, Heathers LE, Xu H, Sherman S and Korc M: A pilot study to develop a diagnostic test for pancreatic ductal adenocarcinoma based on differential expression of select miRNA in plasma and bile. Am J Gastroenterol. 109:1942–1952. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Ye Y, Chen J, Zhou Y, Fu Z, Zhou Q, Wang Y, Gao W, Zheng S, Zhao X, Chen T, et al: High expression of AFAP1-AS1 is associated with poor survival and short-term recurrence in pancreatic ductal adenocarcinoma. J Transl Med. 13:1372015. View Article : Google Scholar : PubMed/NCBI

13 

Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, et al: Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 6:67442015. View Article : Google Scholar : PubMed/NCBI

14 

Berger B, Peng J and Singh M: Computational solutions for omics data. Nat Rev Genet. 14:333–346. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Donahue TR, Tran LM, Hill R, Li Y, Kovochich A, Calvopina JH, Patel SG, Wu N, Hindoyan A, Farrell JJ, et al: Integrative survival-based molecular profiling of human pancreatic cancer. Clin Cancer Res. 18:1352–1363. 2012. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Kerr MK: Linear models for microarray data analysis: Hidden similarities and differences. J Comput Biol. 10:891–901. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Hadley W: ggplot2: Elegant Graphics for Data Analysis. Springer; Switzerland: 2016

19 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 4:32003. View Article : Google Scholar

20 

Acunzo M and Croce CM: MicroRNA in cancer and cachexia - A mini-review. J Infect Dis. 212:(Suppl 1). S74–S77. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Kutmon M, Kelder T, Mandaviya P, Evelo CT and Coort SL: CyTargetLinker: A cytoscape app to integrate regulatory interactions in network analysis. PLoS One. 8:e821602013. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Aguirre-Gamboa R, Gomez-Rueda H, Martínez-Ledesma E, Martínez-Torteya A, Chacolla-Huaringa R, Rodriguez-Barrientos A, Tamez-Peña JG and Treviño V: SurvExpress: An online biomarker validation tool and database for cancer gene expression data using survival analysis. PLoS One. 8:e742502013. View Article : Google Scholar : PubMed/NCBI

24 

Mollenhauer J, Roether I and Kern HF: Distribution of extracellular matrix proteins in pancreatic ductal adenocarcinoma and its influence on tumor cell proliferation in vitro. Pancreas. 2:14–24. 1987. View Article : Google Scholar : PubMed/NCBI

25 

Pandol S, Edderkaoui M, Gukovsky I, Lugea A and Gukovskaya A: Desmoplasia of pancreatic ductal adenocarcinoma. Clin Gastroenterol Hepatol. 7:(Suppl 11). S44–47. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Mahadevan D and Von Hoff DD: Tumor-stroma interactions in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 6:1186–1197. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Crowe DL and Ohannessian A: Recruitment of focal adhesion kinase and paxillin to beta1 integrin promotes cancer cell migration via mitogen activated protein kinase activation. BMC Cancer. 4:182004. View Article : Google Scholar : PubMed/NCBI

28 

Schmitz KJ, Grabellus F, Callies R, Otterbach F, Wohlschlaeger J, Levkau B, Kimmig R, Schmid KW and Baba HA: High expression of focal adhesion kinase (p125FAK) in node-negative breast cancer is related to overexpression of HER-2/neu and activated Akt kinase but does not predict outcome. Breast Cancer Res. 7:R194–R203. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Sawai H, Okada Y, Funahashi H, Matsuo Y, Takahashi H, Takeyama H and Manabe T: Activation of focal adhesion kinase enhances the adhesion and invasion of pancreatic cancer cells via extracellular signal-regulated kinase-1/2 signaling pathway activation. Mol Cancer. 4:372005. View Article : Google Scholar : PubMed/NCBI

30 

Muniyan S, Haridas D, Chugh S, Rachagani S, Lakshmanan I, Gupta S, Seshacharyulu P, Smith LM, Ponnusamy MP and Batra SK: MUC16 contributes to the metastasis of pancreatic ductal adenocarcinoma through focal adhesion mediated signaling mechanism. Genes Cancer. 7:110–124. 2016.PubMed/NCBI

31 

Mall AS: Analysis of mucins: Role in laboratory diagnosis. J Clin Pathol. 61:1018–1024. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Saitou M, Goto M, Horinouchi M, Tamada S, Nagata K, Hamada T, Osako M, Takao S, Batra SK, Aikou T, et al: MUC4 expression is a novel prognostic factor in patients with invasive ductal carcinoma of the pancreas. J Clin Pathol. 58:845–852. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Zhu Y, Zhang JJ, Zhu R, Zhu Y, Liang WB, Gao WT, Yu JB, Xu ZK and Miao Y: The increase in the expression and hypomethylation of MUC4 gene with the progression of pancreatic ductal adenocarcinoma. Med Oncol. 28:(Suppl 1). S175–S184. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Jonckheere N, Skrypek N, Merlin J, Dessein AF, Dumont P, Leteurtre E, Harris A, Desseyn JL, Susini C, Frénois F, et al: The mucin MUC4 and its membrane partner ErbB2 regulate biological properties of human CAPAN-2 pancreatic cancer cells via different signalling pathways. PLoS One. 7:e322322012. View Article : Google Scholar : PubMed/NCBI

35 

Goan YG, Zhou B, Hu E, Mi S and Yen Y: Overexpression of ribonucleotide reductase as a mechanism of resistance to 2,2-difluorodeoxycytidine in the human KB cancer cell line. Cancer Res. 59:4204–4207. 1999.PubMed/NCBI

36 

Eriksson S and Martin DW Jr: Ribonucleotide reductase in cultured mouse lymphoma cells. Cell cycle-dependent variation in the activity of subunit protein M2. J Biol Chem. 256:9436–9440. 1981.PubMed/NCBI

37 

Duxbury MS, Ito H, Zinner MJ, Ashley SW and Whang EE: RNA interference targeting the M2 subunit of ribonucleotide reductase enhances pancreatic adenocarcinoma chemosensitivity to gemcitabine. Oncogene. 23:1539–1548. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Duxbury MS and Whang EE: RRM2 induces NF-kappaB-dependent MMP-9 activation and enhances cellular invasiveness. Biochem Biophys Res Commun. 354:190–196. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Carr MW, Roth SJ, Luther E, Rose SS and Springer TA: Monocyte chemoattractant protein 1 acts as a T-lymphocyte chemoattractant. Proc Natl Acad Sci USA. 91:3652–3656. 1994. View Article : Google Scholar : PubMed/NCBI

40 

Monti P, Leone BE, Marchesi F, Balzano G, Zerbi A, Scaltrini F, Pasquali C, Calori G, Pessi F, Sperti C, et al: The CC chemokine MCP-1/CCL2 in pancreatic cancer progression: Regulation of expression and potential mechanisms of antimalignant activity. Cancer Res. 63:7451–7461. 2003.PubMed/NCBI

41 

Kalbasi A, Komar CA, Tooker GM, Liu M, Lee JW, Gladney WL, Ben-Josef E and Beatty GL: Tumor-derived CCL2 mediates resistance to radiotherapy in pancreatic ductal adenocarcinoma. Clin Cancer Res. 23:137–148. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Tan MC, Goedegebuure PS, Belt BA, Flaherty B, Sankpal N, Gillanders WE, Eberlein TJ, Hsieh CS and Linehan DC: Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer. J Immunol. 182:1746–1755. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Zhang ML, Lu S, Zhou L and Zheng SS: Correlation between ECT2 gene expression and methylation change of ECT2 promoter region in pancreatic cancer. Hepatobiliary Pancreat Dis Int. 7:533–538. 2008.PubMed/NCBI

44 

Hrabar D, Aralica G, Gomercic M, Ljubicic N, Kruslin B and Tomas D: Epithelial and stromal expression of syndecan-2 in pancreatic carcinoma. Anticancer Res. 30:2749–2753. 2010.PubMed/NCBI

45 

Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Klöppel G, Yoshimura A, Reindl W, Sipos B, Akira S, et al: Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell. 19:456–469. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 37 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun H, Zhao L, Pan K, Zhang Z, Zhou M and Cao G: Integrated analysis of mRNA and miRNA expression profiles in pancreatic ductal adenocarcinoma. Oncol Rep 37: 2779-2786, 2017
APA
Sun, H., Zhao, L., Pan, K., Zhang, Z., Zhou, M., & Cao, G. (2017). Integrated analysis of mRNA and miRNA expression profiles in pancreatic ductal adenocarcinoma. Oncology Reports, 37, 2779-2786. https://doi.org/10.3892/or.2017.5526
MLA
Sun, H., Zhao, L., Pan, K., Zhang, Z., Zhou, M., Cao, G."Integrated analysis of mRNA and miRNA expression profiles in pancreatic ductal adenocarcinoma". Oncology Reports 37.5 (2017): 2779-2786.
Chicago
Sun, H., Zhao, L., Pan, K., Zhang, Z., Zhou, M., Cao, G."Integrated analysis of mRNA and miRNA expression profiles in pancreatic ductal adenocarcinoma". Oncology Reports 37, no. 5 (2017): 2779-2786. https://doi.org/10.3892/or.2017.5526