Open Access

Identification of key candidate genes for pancreatic cancer by bioinformatics analysis

  • Authors:
    • Kui Lv
    • Jianying Yang
    • Junfeng Sun
    • Jianguo Guan
  • View Affiliations

  • Published online on: May 28, 2019     https://doi.org/10.3892/etm.2019.7619
  • Pages: 451-458
  • Copyright: © Lv 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

Although pancreatic cancer has the highest mortality rate among all neoplasms worldwide, its exact mechanism remains poorly understood. In the present study, three Gene Expression Omnibus (GEO) datasets were integrated to elucidate the potential genes and pathways that contribute to the development of pancreatic cancer. Initially, a total of 226 differentially expressed genes (DEGs) were identified in the three GEO datasets, containing 179 upregulated and 47 downregulated DEGs. Furthermore, function and pathway enrichment analyses were performed to explore the function and pathway of these genes, and the results indicated that the DEGs participated in extracellular matrix (ECM) processes. In addition, a protein‑protein interaction network was constructed and 163 genes of the 229 DEGs were filtered into the network, resulting in a network complex of 163 nodes and 438 edges. Finally, 24 hub genes were identified in the network, and the top 2 most significant modules were selected for function and pathway analysis. The hub genes were involved in several processes, including activation of matrix, degradation of ECM and ECM organization. Taken collectively, the data demonstrated potential key genes and pathways in pancreatic cancer, which may provide novel insights to the mechanism of pancreatic cancer. In addition, these hub genes and pathways may be considered as targets for the treatment of pancreatic cancer.

Introduction

Pancreatic cancer is one of the most prevalent and lethal malignancies worldwide (1). Although substantial progress has been made in adjuvant and neo-adjuvant chemotherapies during previous decades, pancreatectomy remains the most effective treatment, notably for early stage pancreatic cancer cases. Despite this, a previous study demonstrated that only 20% of patients present with localized and non-metastatic disease, and are therefore suitable for initial resection (2). Due to its specific tumor biology, pancreatic cancer is characterized with early recurrence and metastasis and resistance to chemotherapy and radiotherapy. The 5-year overall survival rate is <5% (3). Therefore, an improved understanding of the underlying mechanism of pancreatic cancer is required for the development of effective therapy and the improvement of patient survival.

Previously, the development of high throughout sequencing has resulted in the production of numerous gene expression profiles of neoplasms that are freely available via the Gene Expression Omnibus (GEO) database (4). Based on these data, the different aspects of the mechanism of pancreatic neoplasm development and the resistance to chemotherapy may be investigated. However, only a small part of these data has been used, and the majority of them have only been deposited. Using a bioinformatic analysis, these data may be re-analysed and used to provide valuable information for subsequent investigation. During the re-analysis process, differentially expressed genes (DEGs) are initially identified, and subsequently the functions and pathways of the genes involved are investigated. Several studies performed in pancreatic cancer have been performed previously (5,6). Although the majority of these studies only focused on the identification of the most significant genes, the tumor and normal tissues were not paired in those analyses. Therefore, in the present study, three GEO datasets were selected, which contained paired tumor tissues and corresponding normal tissues, and the microarray data was analysed. The analysis led to the identification of the DEGs, and Gene Ontology (GO) and pathway enrichment analysis were subsequently performed to explore the biological functions and pathways of these genes. Furthermore, a protein-protein interaction (PPI) network was constructed and a module analysis was performed to explore the hub genes in pancreatic cancer. The present study may provide novel insights into the understanding of the mechanism of pancreatic cancer formation and its corresponding hub genes, and the pathways involved may serve as potential targets for the treatment of this cancer type.

Materials and methods

Data source

The microarray data for the investigation of pancreatic cancer were downloaded from the GEO datasets (www.ncbi.nlm.nih.gov/geo) as follows: GSE15471, GSE16515 and GSE28735. The gene expression profiles of GSE15471 and GSE16515 were obtained using the GPL570 platform, and GSE28735 was obtained using the GPL6244 platform. GSE15471 and GSE28735 included 36 and 45 pairs of tumor and corresponding normal tissues, respectively. GSE16515 consisted of 36 tumor and 16 normal tissues. A total of 16 pairs of tissues were selected for subsequent analysis.

DEG identification

DEGs between tumor and normal tissue samples were identified using the online analysis tool GEO2R (www.ncbi.nlm.nih.gov/geo/geo2r), and the results were saved as a .txt format. The cut-off criteria for DEG identification were defined as adjusted P<0.05 and |log fold change (FC)|>1. Subsequently, the DEGs of the three GEO datasets were processed to generate a Venn diagram using an online resource (bioinformatics.psb.ugent.be/webtools/Venn).

Gene Ontology (GO) and pathway enrichment analysis

To explore the primary functions and pathways of the DEGs involved, the Database for Annotation, Visualization and Integrated Discovery (DAVID; version 6.8; david.ncifcrf.gov) and the Panther (www.pantherdb.org) databases were employed to perform GO analysis. The Kyoto Encyclopedia of Genes and Genomes (7), and the Reactome pathway enrichment analysis were used for the pathway enrichment analysis (810). P<0.05 and a DEG count ≥2 were set as criteria.

PPI network construction and module analysis

The Search Tool for the Retrieval of Interacting Genes (STRING; string-db.org) was used to build a PPI network, and subsequently the network was visualized using Cytoscape v3.6.1 software (11). The cut-off criterion for the PPI network was a combined score >0.4, and the hub genes were defined by a node degree >10. The modules of the PPI network were calculated using the Molecular Complex Detection (MCODE) plug-in of Cytoscape with default parameters. Subsequently, the hub genes in high-scored modules (degree cut-off=2, node score cut-off=0.2) were selected for additional GO and pathway enrichment analyses.

Results

Identification of DEGs in pancreatic cancer

Gene expression profiles from three pancreatic cancer GEO datasets were analysed. Based on the cut-off criteria, 1,666, 1,501 and 412 DEGs were extracted from the GSE15471, GSE16515 and GSE28735 datasets, respectively. Subsequently, 226 DEGs were identified (Fig. 1) by integrated bioinformatics analysis, including 179 upregulated and 47 downregulated DEGs (Table I).

Table I.

Identification of DEGs. A total of 226 DEGs were identified, including 179 upregulated genes and 47 downregulated genes.

Table I.

Identification of DEGs. A total of 226 DEGs were identified, including 179 upregulated genes and 47 downregulated genes.

DEGsGene names
UpregulatedABHD17C, ACSL5, ADAM28, ADAM9, ADAMTS12, ADGRF1, AEBP1, AGR2, AHNAK2, AK4, ANKRD22, ANLN, ANO1, ANTXR1, ANXA10, ANXA3, ANXA8, APOL1, AREG, ARNTL2, ASAP2, ASPM, BGN, CAPG, CCL18, CCL20, CD109, CDH11, CDH3, CEACAM1, CEACAM5, CEACAM6, CEMIP, CLDN18, COL10A1, COL11A1, COL12A1, COL1A1, COL1A2, COL3A1, COL5A2, COL8A1, COMP, CORIN, CP, CST1, CST2, CTHRC1, CTSE, CXCL5, DDX60, DGKH, DHRS9, DKK1, DPCR1, DPYSL3, ECT2, EDIL3, EDNRA, EFNA5, EFNB2, ENO2, EPHA4, EPYC, ERO1A, ESM1, ETV1, FAP, FBXO32, FERMT1, FGD6, FN1, FOXQ1, FXYD3, GABRP, GALNT5, GCNT3, GJB2, GPRC5A, GPX2, GPX8, GREM1, HEPH, HK2, IFI27, IFI44L, IGF2BP3, IGFBP5, IL1R2, INHBA, INPP4B, ITGA2, ITGA3, ITGB4, KCNN4, KRT19, KRT7, KYNU, LAMA3, LAMB3, LAMC2, LCN2, LEF1, LOXL2, LRRN1, MALL, MATN3, MBOAT2, MELK, MET, MICAL2, MLPH, MMP1, MMP11, MMP12, MMP14, MMP7, MMP9, MTMR11, MXRA5, MYOF, NMU, NOX4, NPR3, NQO1, NRP2, NT5E, NTM, OAS1, OAS2, OLR1, OSBPL3, PCDH7, PGM2L1, PKM, PLA2R1, PLAC8, PLAT, PLAU, PLPP4, PLS1, POSTN, RAI14, RHBDL2, RUNX2, S100A16, S100P, SCEL, SCNN1A, SDR16C5, SERPINB3, SERPINB5, SLC22A3, SLC2A1, SLC44A4, SLC6A14, SLC6A6, SLPI, SRPX2, ST6GALNAC1, STYK1, SULF1, SULF2, SULT1C2, SYTL2, TCN1, TFF1, TGFBI, THBS2, TMC5, TMEM45B, TMPRSS4, TNFAIP6, TOP2A, TRIM29, TSPAN1, TSPAN8, VCAN, VSIG1
DownregulatedABAT, ACADL, ADAMTS6, ALB, ANPEP, AOX1, BACE1, BNIP3, BTG2, C5, CHRM3, CTNND2, DPP10, EGF, EPB41L4B, EPHX2, ERO1B, F11, F8, FAM129A, FAM150B, FGL1, GATM, GNMT, GP2, GSTA1, HOMER2, IAPP, KIAA1324, LIFR, MCOLN3, MT1G, NR5A2, NUCB2, PAIP2B, PDK4, PNLIPRP1, RBPJL, RGN, SERPINI2, SLC16A10, SLC1A2, SLC39A5, SLC43A1, SLC4A4, TMED6, TRHDE

[i] DEGs, differentially expressed genes.

GO analysis of DEGs in pancreatic cancer

Candidate DEGs function enrichment analysis was conducted with DAVID and Panther software. The functions of the DEGs were classified into three groups as follows: ‘Cellular Component’; ‘Molecular Function’; and ‘Biological Process’ (Fig. 2). DEGs were primarily involved in binding and catalytic activity with regard to the ‘Molecular Function’ group, cellular and metabolic processes with regard to the ‘Biological Process’ group, and cell membrane with regard to the ‘Cellular Component’ group. Furthermore, the top 30 GO terms of these DEGs (Fig. 3), and the top 10 GO terms of the upregulated and downregulated DEGs were classified (Table II). The upregulated DEGs that were primarily enriched were associated with extracellular matrix (ECM) organisation, cell adhesion, collagen catabolic process and ECM disassembly in the ‘Biological Process’ group, whereas with regard to the ‘Cellular Component’ group ECM, extracellular space, extracellular region and extracellular exosome formation were the primary processes identified. The downregulated DEGs that were primarily enriched were associated with reactive oxygen species, metabolic processes, proteolysis and cellular response to starvation in the ‘Biological Process’ group. The enriched downregulated DEGs were also associated with extracellular exosome formation, extracellular space, integral component of plasma membrane and extracellular region with regard to the ‘Cellular Component’ group. These results indicated that the majority of DEGs were significantly enriched in processes, including extracellular exosome formation, ECM, ECM organisation, extracellular space and extracellular region.

Table II.

Enrichment analysis of DEGs in pancreatic cancer.

Table II.

Enrichment analysis of DEGs in pancreatic cancer.

DEGsTermDescriptionCategoryP-value
UpregulatedGO:0030198Extracellular matrix organizationBP 8.62×10−16
GO:0031012Extracellular matrixCC 1.09×10−14
GO:0007155Cell adhesionBP 2.15×10−14
GO:0030574Collagen catabolic processBP 6.69×10−14
GO:0005615Extracellular spaceCC 3.16×10−13
GO:0005576Extracellular regionCC 3.37×10−11
GO:0005578Proteinaceous extracellular matrixCC 1.11×10−10
GO:0035987Endodermal cell differentiationBP 1.85×10−10
GO:0070062Extracellular exosomeCC 1.37×10−9
GO:0022617Extracellular matrix disassemblyBP 4.93×10−9
DownregulatedGO:0070062Extracellular exosomeCC 5.19×10−6
GO:0072593Reactive oxygen species metabolic processBP 9.67×10−5
GO:0006508ProteolysisBP 1.62×10−3
GO:0005615Extracellular spaceCC 5.84×10−3
GO:0015171Amino acid transmembrane transporterMF 6.37×10−3
GO:0009267Cellular response to starvationBP 6.44×10−3
GO:0005887Integral component of plasma membraneCC 8.01×10−3
GO:0031093Platelet α granule lumenCC 8.50×10−3
GO:0016323Basolateral plasma membraneCC 1.05×10−2
GO:0005576Extracellular regionCC 1.77×10−2

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

Pathway enrichment analysis of DEGs in pancreatic cancer

The DEG signaling pathway enrichment was performed with DAVID. The 30 significantly enriched pathway terms are demonstrated in Fig. 4. The data indicated that these enriched genes were involved in matrix formation, activation of matrix metalloproteinase enzymes, ECM organisation, degradation of the ECM, ECM proteoglycan formation, non-integrin membrane-ECM interactions and ECM-receptor interaction. The data suggested that the DEGs primarily participated in the regulation of the ECM.

PPI network construction and module analysis

The PPI network of the DEGs was constructed according to the STRING database and subsequently visualized and analysed using Cytoscape. The results indicated that 163 genes, including 133 upregulated and 30 downregulated genes, of the 229 DEGs were filtered into the PPI network, and the PPI network complex contained 163 nodes and 438 edges (Fig. 5). Among the 163 nodes, 24 hub genes were identified with the following filter: Node degree >10. The top 10 most significant hub genes were albumin, epidermal growth factor, matrix metalloproteinase (MMP) 9, collagen type I α 2 chain (COL1A2), fibronectin 1, collagen type I α 1 chain (COL1A1), collagen type III α 1 chain (COL3A1), tissue inhibitors of metalloproteinases 1 (TIMP1), integrin subunit α 2 and MMP2. Subsequently, MCODE was used to determine the role of the hub genes, and the top 2 significant modules were selected for subsequent analysis. Module 1 consisted of 10 genes, whereas module 2 consisted of 7 genes. GO analysis indicated that module 1 was primarily associated with ECM disassembly and ECM formation, whereas module 2 was primarily associated with ECM organisation and ECM formation (Table III). Furthermore, pathway enrichment analysis demonstrated that module 1 was mainly enriched in the activation of matrix and the degradation of the ECM, whereas module 2 was mainly enriched in ECM proteoglycan formation, ECM-receptor interaction and ECM organisation (Table IV). These data indicated that the hub genes served key roles in the regulation of the ECM.

Table III.

Gene Ontology analysis of the top 2 modules genes.

Table III.

Gene Ontology analysis of the top 2 modules genes.

ModulesTermDescriptionCategoryP-value
Module 1GO:0004252Serine-type endopeptidase activityMF 7.34×10−6
GO:0022617Extracellular matrix disassemblyBP 3.13×10−5
GO:0005576Extracellular regionCC 5.57×10−5
GO:0070062Extracellular exosomeCC 8.66×10−5
GO:0009986Cell surfaceCC 8.69×10−5
GO:0006508ProteolysisBP 2.14×10−4
GO:0005615Extracellular spaceCC 3.18×10−4
GO:0031093Platelet α granule lumenCC 3.31×10−4
GO:0031012Extracellular matrixCC 5.06×10−4
GO:0030574Collagen catabolic processBP 7.34×10−6
Module 2GO:0001501Skeletal system developmentBP 2.22×10−10
GO:0030198Extracellular matrix organizationBP 1.22×10−9
GO:0031012Extracellular matrixCC 6.47×10−9
GO:0005615Extracellular spaceCC 1.61×10−7
GO:0005578Proteinaceous extracellular matrixCC 6.70×10−7
GO:0005201Extracellular matrix structuralMF 1.19×10−6
GO:0007155Cell adhesionBP 7.91×10−6
GO:0005576Extracellular regionCC 2.97×10−5
GO:0005518Collagen bindingMF 1.85×10−4
GO:0030574Collagen catabolic processBP 2.12×10−4

[i] CC, cellular component; MF, molecular function; BP, biological process.

Table IV.

Pathway enrichment analysis of the top 2 module genes.

Table IV.

Pathway enrichment analysis of the top 2 module genes.

ModulesPathwayNameP-value
Module 1R-HSA-1592389Activation of matrix 3.63×10−6
R-HSA-1442490Collagen degradation 1.71×10−3
R-HSA-1474228Degradation of the extracellular 2.52×10−3
R-HSA-114608Platelet degranulation 6.86×10−3
R-HSA-75205Dissolution of fibrin clot 1.28×10−2
hsa04510Focal adhesion 2.10×10−2
hsa04810Regulation of actin cytoskeleton 2.30×10−2
R-HSA-210991Basigin interactions 2.45×10−2
R-HSA-3000157Laminin interactions 2.94×10−2
R-HSA-2022090Assembly of collagen fibrils and other multimeric structures 4.47×10−2
Module 2R-HSA-3000178ECM proteoglycans 2.14×10−8
R-HSA-216083Integrin cell surface interactions 8.11×10−6
hsa04512ECM-receptor interaction 1.89×10−5
R-HSA-1474244Extracellular matrix organization 2.54×10−5
R-HSA-3000170Syndecan interactions 8.48×10−5
R-HSA-3000171Non-integrin membrane-ECM 1.88×10−4
R-HSA-2022090Assembly of collagen fibrils and other multimeric structures 2.49×10−4
hsa04510Focal adhesion 2.50×10−4
R-HSA-1442490Collagen degradation 4.83×10−4
hsa04151PI3K-Akt signaling pathway 1.14×10−3

[i] CC, cellular component; MF, molecular function; BP, biological process; ECM, extracellular matrix; hsa, Homo sapiens.

Discussion

Pancreatic cancer is usually asymptomatic and is diagnosed in the advanced stages of progression due to lack of specific and sensitive detection markers that may be used during the early stages of the disease (12). The cancer cells are able to readily invade blood vessels and lymph nodes and metastasize to distant organs (13). Furthermore, pancreatic cancer is often resistant to conventional treatment, and it is characterised as one of the most lethal neoplasms (14). Previously, numerous studies have been performed to explore the underlying mechanism of pancreatic cancer progression and its treatment resistance. However, the disease prognosis has not changed significantly (15). The majority of these studies have focused on a single gene to investigate its potential role in pancreatic cancer, although cancer is a complex disease, and is not determined by only one or a few genes.

Therefore, the present study integrated three pancreatic cancer microarray datasets from different studies, using bioinformatics analysis. Consequently, 226 DEGs were identified, which included 176 upregulated and 47 downregulated genes. The function and pathway enrichment analysis were conducted, and the results indicated that these genes primarily participated in the ECM process. Furthermore, a PPI network was constructed in order to determine the role of the hub genes, and MCODE was used to determine the interactions of these genes with several pathways. The results identified 24 hub genes among the 226 DEGs that were associated with ECM regulation.

The ECM is a 3-dimensional non-cellular structure, which serves important roles in all tissues and biological process and is primarily responsible for the maintenance of tissue homeostasis and the regulation of development (16). As a complex network, the ECM consists of extracellular macromolecules, including glycoproteins, collagen and enzymes, which provide structural support to the surrounding cells and segregate cells from one another. This sequesters a variety of cellular growth factors and regulates intercellular communication (17,18). Although the composition of the ECM varies between tissues and species, its common functions comprise cell differentiation, cell adhesion and cell to cell communication (19). Stem cells are able to actively detect the elasticity and rigidity of the surrounding ECM, and subsequently adjust gene expression, which determines the differentiation process (20). Cells bind to the ECM via intermediate actin filaments, which is regulated by specific cell adhesion molecules, including integrin, fibronectin and laminin (21). The ECM interacts biochemically with the surrounding cells by serving as a ligand to transmit signals, which mediate cell adhesion, differentiation, apoptosis, survival, proliferation and migration (22).

Although the ECM is responsible for regulating normal tissue development and homeostasis, its dysregulation also contributes to neoplastic progression. The development of cancer is significantly affected by the microenvironment and the ECM is considered the major component of tumor associated microenvironment (23). Although ECM remodelling is strictly regulated and mediated by the activities of specific enzymes, the dysregulation of these activities may account for the progression and development of certain disease conditions. This may lead to changes in the amount and composition of the ECM, which may significantly alter the biochemical properties of the ECM and promote the oncogenic effects of various pathways, and deregulate cell behaviours during malignant transformation (24). Several studies have suggested that the ECM is important for the maintenance of the polarity and architecture of tissues, and that deregulated ECM promotes epithelial-mesenchymal transition, eventually facilitating tumor invasion (25). Furthermore, the abnormal ECM function may promote tumor angiogenesis and lymphangiogenesis (26) and tumor-associated inflammation (27). In summary, abnormal ECM may promote tumor progression through a number of mechanisms.

The present study revealed that the specific DEGs identified primarily participated in ECM formation, as demonstrated by analysis of specific hub genes, including MMP2, MMP9, TIMP1, COL1A2, COL1A1 and COL3A1. MMPs are the primary enzymes that degrade the ECM and are synthesized by tumor cells (MMP7) or tumor stromal cells (MMP2 and MMP9). A previous study has demonstrated that MMPs serves important roles in cancer progression by increasing tumor cell migration, invasion, metastasis and angiogenesis (28). In pancreatic cancer, MMPs were correlated with prognosis, survival time, local invasion and distant metastasis (29). TIMPs are the endogenous inhibitors of MMPs, which bind to active and alternative sites of the activated MMP enzymes, prevent angiostatin and endostatin production, and serve a role in the promotion of tumor angiogenesis (30). The balance between MMPs and TIMPs may determine the ECM dynamics and affect tumor progression (31). COL1A1, COL1A2 and COL3A1 are the genes that encode collagen proteins, and the target genes of transforming growth factor-β (TGF-β)/SMAD3. Collagens mediate the tumor metastatic process via the interactions with MMPs and may also serve significant roles in the immune response to cancer (32). β-TGF/SMAD3 may promote the expression of ECM-associated proteins (33). The aforementioned hub genes participated in ECM formation and may serve as potential targets for the treatment of pancreatic cancer.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

JG designed the study and revised the manuscript. KL performed the GEO database analysis, analysed the data and wrote the manuscript. JY and JS performed bioinformatics analyses and assisted with analysis of other data. All authors have read and approved the manuscript.

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.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Gillen S, Schuster T, Meyer Zum Buschenfelde C, Friess H and Kleeff J: Preoperative/neoadjuvant therapy in pancreatic cancer: A systematic review and meta-analysis of response and resection percentages. PLoS Med. 7:e10002672010. View Article : Google Scholar : PubMed/NCBI

3 

Kamisawa T, Wood LD, Itoi T and Takaori K: Pancreatic cancer. Lancet. 388:73–85. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, Kim IF, Soboleva A, Tomashevsky M and Edgar R: NCBI GEO: Mining tens of millions of expression profiles-database and tools update. Nucleic Acids Res. 35:D760–D765. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Li J, Tan W, Peng L, Zhang J, Huang X, Cui Q, Zheng J, Tan W, Wu C and Lin D: Integrative analysis of gene expression profiles reveals specific signaling pathways associated with pancreatic duct adenocarcinoma. Cancer Commun (Lond). 38:132018. View Article : Google Scholar : PubMed/NCBI

6 

Wang H, Zhan M, Yang R, Shi Y, Liu Q and Wang J: Elevated expression of NFE2L3 predicts the poor prognosis of pancreatic cancer patients. Cell Cycle. 17:2164–2174. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Kanehisa M, Sato Y, Furumichi M, Morishima K and Tanabe M: New approach for understanding genome variations in KEGG. Nucleic Acids Res. 47:D590–D595. 2019. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

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

10 

Mi H, Huang X, Muruganujan A, Tang H, Mills C, Kang D and Thomas PD: PANTHER version 11: Expanded annotation data from Gene Ontology and Reactome pathways, and data analysis tool enhancements. Nucleic Acids Res. 45:D183–D189. 2017. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Ilic M and Ilic I: Epidemiology of pancreatic cancer. World J Gastroenterol. 22:9694–9705. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Khadka R, Tian W, Hao X and Koirala R: Risk factor, early diagnosis and overall survival on outcome of association between pancreatic cancer and diabetes mellitus: Changes and advances, a review. Int J Surg. 52:342–346. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH and Neoptolemos JP: Pancreatic cancer. Nat Rev Dis Primers. 2:160222016. View Article : Google Scholar : PubMed/NCBI

15 

Cancer Research UK, . Pancreatic cancer statistics 2015. https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/pancreatic-cancerSeptember 20–2018

16 

Mammoto T and Ingber DE: Mechanical control of tissue and organ development. Development. 137:1407–1420. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Geiger B, Spatz JP and Bershadsky AD: Environmental sensing through focal adhesions. Nat Rev Mol Cell Biol. 10:21–33. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Theocharis AD, Skandalis SS, Gialeli C and Karamanos NK: Extracellular matrix structure. Adv Drug Deliv Rev. 97:4–27. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Abedin M and King N: Diverse evolutionary paths to cell adhesion. Trends Cell Biol. 20:734–742. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Engler AJ, Sen S, Sweeney HL and Discher DE: Matrix elasticity directs stem cell lineage specification. Cell. 126:677–689. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Gumbiner BM: Cell adhesion: The molecular basis of tissue architecture and morphogenesis. Cell. 84:345–357. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Hynes RO: The extracellular matrix: Not just pretty fibrils. Science. 326:1216–1219. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Lu P, Weaver VM and Werb Z: The extracellular matrix: A dynamic niche in cancer progression. J Cell Biol. 196:395–406. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Pickup MW, Mouw JK and Weaver VM: The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 15:1243–1253. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Radisky ES and Radisky DC: Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer. J Mammary Gland Biol Neoplasia. 15:201–212. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Avraamides CJ, Garmy-Susini B and Varner JA: Integrins in angiogenesis and lymphangiogenesis. Nat Rev Cancer. 8:604–617. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Sorokin L: The impact of the extracellular matrix on inflammation. Nat Rev Immunol. 10:712–723. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Egeblad M and Werb Z: New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2:161–174. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Garcea G, Neal CP, Pattenden CJ, Steward WP and Berry DP: Molecular prognostic markers in pancreatic cancer: A systematic review. Eur J Cancer. 41:2213–2236. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Jiang Y, Goldberg ID and Shi YE: Complex roles of tissue inhibitors of metalloproteinases in cancer. Oncogene. 21:2245–2252. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Bonnans C, Chou J and Werb Z: Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol. 15:786–801. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Nerenberg PS, Salsas-Escat R and Stultz CM: Collagen - A necessary accomplice in the metastatic process. Cancer Genomics Proteomics. 4:319–327. 2007.PubMed/NCBI

33 

Verrecchia F, Chu ML and Mauviel A: Identification of novel TGF-beta/Smad gene targets in dermal fibroblasts using a combined cDNA microarray/promoter transactivation approach. J Biol Chem. 276:17058–17062. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2019
Volume 18 Issue 1

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
Lv K, Yang J, Sun J and Guan J: Identification of key candidate genes for pancreatic cancer by bioinformatics analysis. Exp Ther Med 18: 451-458, 2019
APA
Lv, K., Yang, J., Sun, J., & Guan, J. (2019). Identification of key candidate genes for pancreatic cancer by bioinformatics analysis. Experimental and Therapeutic Medicine, 18, 451-458. https://doi.org/10.3892/etm.2019.7619
MLA
Lv, K., Yang, J., Sun, J., Guan, J."Identification of key candidate genes for pancreatic cancer by bioinformatics analysis". Experimental and Therapeutic Medicine 18.1 (2019): 451-458.
Chicago
Lv, K., Yang, J., Sun, J., Guan, J."Identification of key candidate genes for pancreatic cancer by bioinformatics analysis". Experimental and Therapeutic Medicine 18, no. 1 (2019): 451-458. https://doi.org/10.3892/etm.2019.7619