Open Access

Gene expression profile analysis of pancreatic cancer based on microarray data

  • Authors:
    • Jin Long
    • Zhe Liu
    • Xingda Wu
    • Yuanhong Xu
    • Chunlin Ge
  • View Affiliations

  • Published online on: March 21, 2016     https://doi.org/10.3892/mmr.2016.5021
  • Pages: 3913-3919
  • Copyright: © Long 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 present study identified differentially‑expressed genes (DEGs) between pancreatic cancer (PC) tissues and normal tissues, and assessed genetic factors associated with the pathogenesis of PC. The mRNA expression microarray dataset, GSE16515, containing 52 samples, including 16 paired tumor and normal tissue samples, and 20 tumor samples, was downloaded from Gene Expression Omnibus. Raw data were normalized and DEGs were identified. Subsequently, clustering was performed, protein‑protein interaction networks were drawn, and functional and pathway enrichment analyses of the DEGs were performed. Copy number variations of DEGs were also identified. A total of 1,765 DEGs between PC and normal tissues were identified, including 1,312 upregulated and 453 downregulated DEGs. Upregulated DEGs were associated with the regulation of nucleocytoplasmic and intracellular transport, whereas downregulated DEGs were associated with the response to organic substances and hormone stimulus. The pancreatic cancer pathway was connected to three DEGs, namely transforming growth factor β1 (TGFB1), TGFβ receptor 1 (TGFBR1) and epidermal growth factor (EGF), which had 2, 3 and 5 CNVs, respectively. These results indicated the important roles of TGFB1, TGFBR1 and EGF in the pathogenesis of PC. These genes may be potential therapeutic targets for the treatment of PC.

Introduction

Pancreatic cancer (PC; OMIM 260350) is a highly lethal disease, with an incidence rate that is constantly increasing (1). The 5-year survival rate of PC is <5% (2), and almost all patients with primary PC develop metastases.

Previous studies have indicated that PC has a complex genomic landscape with frequent copy number variations (CNVs) or copy number polymorphisms (CNPs) (3). Biankin et al (4) defined 16 significantly mutated genes [e.g., Kirsten rat sarcoma viral oncogene homolog (KRAS), tumor protein p53 (TP53), SMAD family member 4 (SMAD4) and transforming growth factor β receptor 2 (TGFBR2)] that were reaffirmed known mutations associated with PC. The commonly mutated genes, such as KRAS (74–100%), cyclin-dependent kinase inhibitor 2A (up to 98%), TP53 (43–76%), erb-b2 receptor tyrosine kinase 2 (ERBB2; ~65%) and fragile histidine triad (~70%) have been found in PC (59). Among these genes, KRAS and ERBB2 are proto-oncogenes, whereas the other genes are tumor suppressors (3). The progression of PC is correlated with the activation of oncogenes and the inactivation of tumor suppressor genes, as well as the deregulation of a number of signaling pathways, among which the epidermal growth factor receptor (EGFR), v-akt murine thymoma viral oncogene homolog 1 (v-AKT1) and nuclear factor of κ light polypeptide gene enhancer in B-cells 1 (NFκB1) pathways appear to be most relevant (10).

AKT1 is a central regulator of cell growth. AKT1 has been shown to inhibit apoptosis and promote cell survival, thus contributing to the pathogenesis of cancer (11,12). Pei et al (13) showed that FK506-binding protein 51 (FKBP51) acted as a scaffolding protein for Akt and that it promoted the activation of Akt. The expression of FKBP51 was downregulated in PC tissues. Decreased FKBP51 expression resulted in the hyper-phosphorylation of Akt, and then decreased the level of cell death in the PC tissues. Thus, Pei et al demonstrated FKBP51 to be a negative regulator of the Akt pathway (13). Pei et al also released the mRNA expression microarray dataset, GSE16515, consisting of 36 pancreatic tumor and 16 normal tissue samples. Numerous studies have since been performed using this dataset (1416). For example, using the GSE16515 dataset, Yang et al (14) screened the differentially-expressed genes (DEGs), such as TGF α (TGFA) and EGF, between PC tumor tissues and normal tissues, and selected the important single nucleotide polymorphisms (SNPs) of A/G and C/T in the DEGs. However, none of the studies based on the GSE16515 dataset performed an analysis of CNVs in the DEGs.

Using the GSE16515 dataset downloaded from Gene Expression Omnibus (GEO), the DEGs between PC tumor tissues and normal tissues were screened in the present study. Next, clustering analysis and construction of a protein-protein interaction (PPI) network of DEGs was performed. The underlying functions of these DEGs were investigated by functional and pathway enrichment analyses. Finally, the CNVs of these DEGs were also analyzed. This will be beneficial for developing therapeutic strategies for patients with PC.

Materials and methods

mRNA microarray data

The mRNA expression microarray data from the GSE16515 dataset (6) was downloaded from GEO (http://www.ncbi.nlm.nih.gov/geo/), based on the platform of GPL570 [HG-U133_Plus_2] Affymetrix Human Genome U133 Plus 2.0 Array (Affymetrix, Santa Clara, CA, USA). GSE16515 was composed of 52 samples (from 34 males and 18 females). In total, 32 samples consisted of tumor and normal expression data, whereas 20 samples consisted of only tumor data. These samples were obtained during clinically indicated surgical procedures and consent was obtained for experimental purposes. The raw data and the probe annotation files were downloaded for further analysis. The microarray data of the GSE16515 dataset was analyzed following the procedures presented in Fig. 1.

Data preprocessing and DEG identification

The Robust Multiarray Average in Affy package of R (http://www.biocon-ductor.org/packages/release/bioc/html/affy.html), provided by Bioconductor project (17), was applied to process the raw micro-array data. The processing included background correction, quantile normalization and probe summarization of expression values. The gene expression matrices were obtained for further analysis. Afterwards, the Linear Models for Microarray Data package was used to identify the gene signatures between the tumor and normal tissues, with significant differences indicated using a P-value of <0.05. Next, the Bonferroni correction (18) was applied to adjust the raw P-value for the false discovery rate (FDR) and to calculate the fold change (FC). In the present study, the cut-off criteria for the statistically significant DEGs were |log2FC| >1 and FDR <0.05.

Clustering analysis of DEGs

Based on the Euclidean distance between the expression profile of each DEG filtered from the samples, hierarchical clustering can be used to build a hierarchy of clusters of DEGs (19). The heatmap figure of the DEGs was drawn with the R package pheatmap (http://cran.r-project.org/web/packages/pheatmap/index.html) function. DEGs with the same signatures were clustered together, indicating the specificity of the DEGs.

Identification of PPIs of DEGs

Identification of protein complexes and functional modules from PPI networks is crucial to predict protein functions and to understand the principles of cellular organization (20). The Search Tool for the Retrieval of Interacting Genes (STRING; http://string-db.org/) database provides uniquely comprehensive coverage and ease of access for the prediction of interaction information (21). To better understand the interactions of the DEGs, the PPI network of their encoding products was predicted using the STRING database, with the reliability threshold of >0.9. Cytoscape software (http://cytoscape.org/), a standard tool for the integrated analysis and visualization of biological networks, was used to visualize the PPI network (22).

Functional enrichment analysis of DEGs

Gene Ontology (GO; http://www.geneontology.org/) analysis is an functional study method for large-scale transcriptomic or genomic data (23). In order to investigate the biofunctions of DEGs in tumor progression, the Database for Annotation, Visualization and Integrated Discovery (http://david.abcc.Ncifcrf.gov/), a high-throughput and integrated data-mining environment (24), was used to identify the enriched GO biological processes that the DEGs were associated with (FDR<0.05).

Pathway analysis of DEGs

The Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg/pathway.html) pathway database provides information on how molecules or genes function (25). Pathway analysis of all the DEGs was performed using the KEGG database. The KEGG maps of biological functions associated with DEGs were obtained (P<0.05).

CNV analysis of DEGs

The Database of Genomic Variants (DGV; http://dgv.tcag.ca/) (26) was used to identify the CNVs in the DEGs, including deletions, insertions, duplications, complex multi-site variants and SNPs.

Results

Data processing and identification of DEGs

After the normalization, the DEGs between the tumor and normal tissues of the 52 samples were identified, with the cut-off criteria of |log2FC| >1 and FDR <0.05. A total of 1,765 DEGs were identified between the PC and normal tissues, of which 1,312 were upregulated and 453 were downregulated.

Hierarchical clustering of DEGs

Hierarchical clustering of the 1,765 DEGs is presented in Fig. 2. The LogFC values of the DEGs ranged from 6-fold downregulated and 6-fold upregulated. The majority of the DEGs were upregulated in the PC tumors compared with the normal tissues. The tumor samples and the normal control samples could easily be distinguished from the characteristics of the DEGs.

PPIs analysis of DEGs

To identify the PPIs and predict protein functions, PPI network analysis was performed using the STRING database (threshold >0.9). The resulting PPI network of upregulated DEGs connected to 92 nodes (proteins) through 171 PPIs, whereas the PPI network of the downregulated DEGs connected to 82 nodes through 83 PPIs (Fig 3).

Functional enrichment analysis of DEGs

To obtain the enriched GO biological processes of the DEGs in the PPI networks, GO functional enrichment analysis was performed for the up- and downregulated DEGs, respectively (FDR <0.05). The upregulated DEGs (including TGFB1 and TGFBR1) were associated with significant biological processes, such as the regulation of nucleocytoplasmic transport, protein localization and intracellular transport (Table I), whereas the downregulated DEGs (e.g., EGF) were correlated with the biological processes of the positive regulation of catalytic activity, the response to organic substances and the response to hormone stimuli (Table I).

Table I

Enriched GO biological processes of the DEGs in the protein-protein interaction networks.

Table I

Enriched GO biological processes of the DEGs in the protein-protein interaction networks.

Term and functionCountGenesP-valueFDR
Upregulated DEGs
 GO:0046822 - Regulation of nucleocytoplasmic transport7CDKN2A, TGFBR1, SMAD3, CDH1, TACC3, FLNA, TGFB1 1.72×10−60.002848
 GO:0032880 - Regulation of protein localization9TGFBR1, SMAD3, CDH1, CDH2, CASP1, TACC3, FLNA, TGFB1, IL1A 2.60×10−60.004308
GO:0051222 - Positive regulation of protein transport7TGFBR1, SMAD3, CDH1, CASP1, FLNA, TGFB1, IL1A 4.05×10−60.006710
 GO:0032386 - Regulation of intracellular transport7CDKN2A, TGFBR1, SMAD3, CDH1, TACC3, FLNA, TGFB1 5.69×10−60.009436
Downregulated DEGs
 GO:0043085 - Positive regulation of catalytic activity13ADCY1, PTGER3, CCKBR, C6, C5, LPAR3, EDNRB, PRKAR2B, PLCE1, CLPS, GNAS, EGF, PSMD6 1.72×10−50.027131
 GO:0010033 - Response to organic substances15TF, ADCY1, PNLIPRP1, GATM, PDE3B, EPHX1, CFTR, PDE3A, NPY1R, PRKAR2B, ABAT, ANGPT1, GNAS, SST, GNG7 2.20×10−50.034801
 GO:0009725 - Response to hormone stimuli11PRKAR2B, PNLIPRP1, ADCY1, GATM, PDE3B, GNAS, ANGPT1, CFTR, NPY1R, SST, GNG7 2.29×10−50.036206
 GO:0006575 - Cellular amino acid derivative metabolic processes8GSTA1, P4HB, CTH, GATM, GPX3, ABAT, GAMT, GNMT 2.78×10−50.043955

[i] FDR, false discovery rate; DEG, differentially-expressed genes; GO, gene ontology.

Pathway analysis of DEGs

The KEGG maps of biological functions associated with DEGs in the PPI networks were obtained (P<0.05). The results showed that only the pancreatic cancer pathway was associated with DEGs in the PPI networks, including upregulated TGFB1 and TGFBR1, and downregulated EGF (Fig. 4).

CNV analysis of DEGs

The CNVs of the TGFB1, TGFBR1 and EGF genes were further identified using the DGV. The identification of CNVs of DEGs included deletions, insertions, duplications and complex multi-site variants. Finally, 2, 3 and 5 CNVs, were identified in the TGFB1, TGFBR1 and EGF genes, respectively (Table II) (2734). In total, 1 of the CNVs of TGFB1 was insertin; all 3 of the CNVs of TGFBR1 were insertins; and 1 of the 5 CNVs of EGF was insertin.

Table II

Copy number variations in TGFB1, TGFBR1 and EGF genes.

Table II

Copy number variations in TGFB1, TGFBR1 and EGF genes.

First author, yearGeneVariant IDSubtype(Ref.)
Xu et al, 2011TGFB1nsv911769Loss(27)
Shaikh et al, 2009nsv521311Insertion(28)
Xu et al, 2011TGFBR1nsv893619Insertion(27)
nsv893618Insertion
Wong et al, 2007nsv831666Insertion(29)
Abecasis et al, 2012EGFesv2672203Deletion(30)
McKernan et al, 2009esv2618042Insertion(31)
Conrad et al, 2010esv22936Loss(32)
Mills et al, 2006nsv290769Loss(33)
Kim et al, 2009nsv820232Loss(34)

Discussion

The early stages of PC are usually asymptomatic, and the majority of patients with PC are diagnosed at an advanced stage. The pathogenesis of PC is involved in a number of biological processes. Advanced studies of genetic factors have greatly improved our understanding of the pathogenesis of PC, which is associated with gene mutations, continuous changes to nuclei, loss of polarity and changes in cellular architecture (35).

In the present study, the DEGs between PC tumor tissues and normal tissues were systematically investigated. A total of 1,765 DEGs, including 1,312 upregulated and 453 down-regulated DEGs, were identified. The majority of DEGs were upregulated in the tumor tissues. The upregulated DEGs (including SMAD3, TGFB1 and TGFBR1) were associated with the regulation of nucleocytoplasmic and intracellular transport, and protein localization, whereas the downregulated DEGs (e.g., EGF) were associated with regulation of catalytic activity, and the responses to organic substances and hormone stimuli. All 4 of these DEGs were connected to the pancreatic cancer pathway. In addition, TGFB1, TGFBR1 and EGF exhibited 2, 3 and 5 CNVs, respectively. These results proposed an important role for these DEGs in PC development.

CNVs or CNPs, such as deletions, insertions, duplications and complex multi-site variants, have been found in all humans and in other mammals (36). CNVs can be simple in structures like tandem duplication, or may be complex in the genome, such as in gains or losses of homologous sequences at multiple sites (37). Yang et al (14) screened the DEGs between PC tissues and normal tissues using the GSE16515 dataset, and selected the important SNPs of A/G and C/T in DEGs such as TGFA and EGF. In the present study, 5 CNVs of EGF were identified, including 3 losses, 1 deletion and 1 insertion. These results demonstrate that the CNVs of EGF may be significant in the pathogenesis of PC.

EGFR is required for KRAS-induced PC (38). Accordingly, the overexpression of EGFR and TGFA has been reported as an important molecular abnormality in human PCs (39). EGF and TGFA are essential molecules for the VEGF signaling pathway (Fig. 4). The inhibition of the vascular endothelial growth factor (VEGF) signaling pathway is beneficial for the suppression of tumor metastasis and invasion, such as required in PC (40). In the present study, it was found that the expression of EGF was downregulated in the PC tissues. Taken together, these results suggest the significant role of EGF in PC and indicate that EGF may be a novel target for the therapy of PC.

TGFB1 and TGFBR1 were overexpressed in the PC tumors in the present study. The two genes are elements of the TGFB signaling pathway, which is a potent inhibitor of cell growth (41). There is growing evidence that members of the TGFB family are frequently mutated in cancer. CNVs, DNA segments that are ≥1 kb and present at variable copy number in comparison with a reference genome, affect the expression of genes, the variation and adaptation of phenotypes, and the pathogenesis of diseases, such as human immunodeficiency virus-1 infection, by disrupting genes and altering gene dosage in microdeletion or microduplication disorders (42,43). The present study identified 3 insertion CNVs in TGFBR1, and 1 insertion CNV and 1 loss CNV in TGFB1. Moreover, 3 of the CNVs in TGFBR1 and TGFB1 were located at the same segment as in a previous study (27). These indicate the vital roles of TGFBR1 and TGFB1 in PC development. The genes may be potential therapeutic targets for the treatment of PC.

In summary, 1,765 DEGs, including 1,312 upregulated (e.g., TGFB1 and TGFBR1) and 453 downregulated (e.g., EGF) DEGS, were identified in the PC tissues compared with the normal tissues in the present study. The upregulated DEGs were associated with the regulation of nucleocytoplasmic and intracellular transport, whereas the downregulated DEGs were associated with the regulation of catalytic activity, and the response to organic substances and hormone stimuli. A pancreatic cancer pathway was connected to the DEGs of TGFB1, TGFBR1 and EGF. In addition, TGFB1, TGFBR1 and EGF exhibited 2, 3 and 5 CNVs, respectively. These results suggested the significance of the DEGs in PC. TGFB1, TGFBR1 and EGF may be potential therapeutic targets for the treatment of PC. However, further clinical trials are required to validate these conclusions and hypotheses.

References

1 

Lowenfels AB and Maisonneuve P: Epidemiology and risk factors for pancreatic cancer. Best Pract Res Clin Gastroenterol. 20:197–209. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Thomas JK, Kim MS, Balakrishnan L, Nanjappa V, Raju R, Marimuthu A, Radhakrishnan A, Muthusamy B, Khan AA, Sakamuri S, et al: Pancreatic Cancer Database: An integrative resource for pancreatic cancer. Cancer Biol Ther. 15:963–967. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Strimpakos A, Saif MW and Syrigos KN: Pancreatic cancer: From molecular pathogenesis to targeted therapy. Cancer Metastasis Rev. 27:495–522. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, et al: Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 491:399–405. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA, et al: Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 4:437–450. 2003. View Article : Google Scholar

6 

Goldstein AM, Fraser MC, Struewing JP, Hussussian CJ, Ranade K, Zametkin DP, Fontaine LS, Organic SM, Dracopoli NC and Clark WH Jr: Increased risk of pancreatic cancer in melanoma-prone kindreds with p16INK4 mutations. N Engl J Med. 333:970–974. 1995. View Article : Google Scholar : PubMed/NCBI

7 

Pellegata NS, Sessa F, Renault B, Bonato M, Leone BE, Solcia E and Ranzani GN: K-ras and p53 gene mutations in pancreatic cancer: Ductal and nonductal tumors progress through different genetic lesions. Cancer Res. 54:1556–1560. 1994.PubMed/NCBI

8 

Grau AM, Zhang L, Wang W, Ruan S, Evans DB, Abbruzzese JL, Zhang W and Chiao PJ: Induction of p21waf1 expression and growth inhibition by transforming growth factor beta involve the tumor suppressor gene DPC4 in human pancreatic adenocarcinoma cells. Cancer Res. 57:3929–3934. 1997.PubMed/NCBI

9 

Sorio C, Baron A, Orlandini S, Zamboni G, Pederzoli P, Huebner K and Scarpa A: The FHIT gene is expressed in pancreatic ductular cells and is altered in pancreatic cancers. Cancer Res. 59:1308–1314. 1999.PubMed/NCBI

10 

Sarkar FH, Banerjee S and Li Y: Pancreatic cancer: Pathogenesis, prevention and treatment. Toxicol Appl Pharmacol. 224:326–336. 2007. View Article : Google Scholar

11 

Luo J, Manning BD and Cantley LC: Targeting the PI3K-Akt pathway in human cancer: Rationale and promise. Cancer Cell. 4:257–262. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Manning BD and Cantley LC: AKT/PKB signaling: Navigating downstream. Cell. 129:1261–1274. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Pei H, Li L, Fridley BL, Jenkins GD, Kalari KR, Lingle W, Petersen G, Lou Z and Wang L: FKBP51 affects cancer cell response to chemotherapy by negatively regulating Akt. Cancer Cell. 16:259–266. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Yang D, Zhu Z, Wang W, Shen P, Wei Z, Wang C and Cai Q: Expression profiles analysis of pancreatic cancer. Eur Rev Med Pharmacol Sci. 17:311–317. 2013.PubMed/NCBI

15 

Zhao LL, Zhang T, Zhuang LW, Yan BZ, Wang RF and Liu BR: Uncovering the pathogenesis and identifying novel targets of pancreatic cancer using bioinformatics approach. Mol Biol Rep. 41:4697–4704. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Söderquist F, Hellström PM and Cunningham JL: Human gastroenteropancreatic expression of melatonin and its receptors MT1 and MT2. PLoS One. 10:e01201952015. View Article : Google Scholar : PubMed/NCBI

17 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc B. 57:289–300. 1995.

19 

Deza MM and Deza E: Encyclopedia of distances. Springer; Heidelberg: 2009, View Article : Google Scholar

20 

Li M, Wu X, Wang J and Pan Y: Towards the identification of protein complexes and functional modules by integrating PPI network and gene expression data. BMC Bioinformatics. 13:1092012. View Article : Google Scholar : PubMed/NCBI

21 

Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, et al: The STRING database in 2011: Functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 39(Database issue): D561–D568. 2011. View Article : Google Scholar :

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 

Hulsegge I, Kommadath A and Smits MA: Globaltest and GOEAST: Two different approaches for Gene Ontology analysis. BMC Proc. 3(Suppl 4): S102009. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar

26 

MacDonald JR, Ziman R, Yuen RK, Feuk L and Scherer SW: The Database of Genomic Variants: A curated collection of structural variation in the human genome. Nucleic Acids Res. 42(Database issue): D986–D992. 2014. View Article : Google Scholar :

27 

Xu H, Poh WT, Sim X, Ong RT, Suo C, Tay WT, Khor CC, Seielstad M, Liu J, Aung T, et al: SgD-CNV, a database for common and rare copy number variants in three Asian populations. Hum Mutat. 32:1341–1349. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Shaikh TH, Gai X, Perin JC, Glessner JT, Xie H, Murphy K, O'Hara R, Casalunovo T, Conlin LK, D'Arcy M, et al: High-resolution mapping and analysis of copy number variations in the human genome: a data resource for clinical and research applications. Genome Res. 19:1682–1690. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Wong KK, deLeeuw RJ, Dosanjh NS, Kimm LR, Cheng Z, Horsman DE, MacAulay C, Ng RT, Brown CJ, Eichler EE and Lam WL: A comprehensive analysis of common copy-number variations in the human genome. Am J Hum Genet. 80:91–104. 2007. View Article : Google Scholar :

30 

Abecasis GR, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM, Korbel JO, Marchini JL, McCarthy S, McVean GA and Abecasis GR: 1000 Genomes Project Consortium: An integrated map of genetic variation from 1,092 human genomes. Nature. 491:56–65. 2012. View Article : Google Scholar : PubMed/NCBI

31 

McKernan KJ, Peckham HE, Costa GL, McLaughlin SF, Fu Y, Tsung EF, Clouser CR, Duncan C, Ichikawa JK, Lee CC, et al: Sequence and structural variation in a human genome uncovered by short-read, massively parallel ligation sequencing using two-base encoding. Genome Res. 19:1527–1541. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Conrad DF, Pinto D, Redon R, Feuk L, Gokcumen O, Zhang Y, Aerts J, Andrews TD, Barnes C, Campbell P, et al: Origins and functional impact of copy number variation in the human genome. Nature. 464:704–712. 2010. View Article : Google Scholar

33 

Mills RE, Luttig CT, Larkins CE, Beauchamp A, Tsui C, Pittard WS and Devine SE: An initial map of insertion and deletion (INDEL) variation in the human genome. Genome Res. 16:1182–1190. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Kim JI, Ju YS, Park H, Kim S, Lee S, Yi JH, Mudge J, Miller NA, Hong D, Bell CJ, et al: A highly annotated whole-genome sequence of a Korean individual. Nature. 460:1011–1015. 2009.PubMed/NCBI

35 

Hruban RH, Adsay NV, Albores-Saavedra J, Compton C, Garrett ES, Goodman SN, Kern SE, Klimstra DS, Klöppel G, Longnecker DS, et al: v Am J Surg Pathol. 25:579–586. 2001. View Article : Google Scholar

36 

Feuk L, Carson AR and Scherer SW: Structural variation in the human genome. Nat Rev Genet. 7:85–97. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Redon R, Ishikawa S, Fitch KR, Feuk L, Perry GH, Andrews TD, Fiegler H, Shapero MH, Carson AR, Chen W, et al: Global variation in copy number in the human genome. Nature. 444:444–454. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Ardito CM, Grüner BM, Takeuchi KK, Lubeseder-Martellato C, Teichmann N, Mazur PK, DelGiorno KE, Carpenter ES, Halbrook CJ, Hall JC, et al: EGF receptor is required for KRAS-induced pancreatic tumorigenesis. Cancer Cell. 22:304–317. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Barton CM, Hall PA, Hughes CM, Gullick WJ and Lemoine NR: Transforming growth factor alpha and epidermal growth factor in human pancreatic cancer. J Pathol. 163:111–116. 1991. View Article : Google Scholar : PubMed/NCBI

40 

Sennino B, Ishiguro-Oonuma T, Wei Y, Naylor RM, Williamson CW, Bhagwandin V, Tabruyn SP, You WK, Chapman HA, Christensen JG, et al: Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov. 2:270–287. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Pasche B, Pennison MJ, Jimenez H and Wang M: TGFBR1 and cancer susceptibility. Trans Am Clin Climatol Assoc. 125:300–312. 2014.PubMed/NCBI

42 

McCarroll SA, Hadnott TN, Perry GH, Sabeti PC, Zody MC, Barrett JC, Dallaire S, Gabriel SB, Lee C, Daly MJ, et al: Common deletion polymorphisms in the human genome. Nat Genet. 38:86–92. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Nguyen DQ, Webber C and Ponting CP: Bias of selection on human copy-number variants. PLoS Genet. 2:e202006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2016
Volume 13 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Long J, Liu Z, Wu X, Xu Y and Ge C: Gene expression profile analysis of pancreatic cancer based on microarray data. Mol Med Rep 13: 3913-3919, 2016
APA
Long, J., Liu, Z., Wu, X., Xu, Y., & Ge, C. (2016). Gene expression profile analysis of pancreatic cancer based on microarray data. Molecular Medicine Reports, 13, 3913-3919. https://doi.org/10.3892/mmr.2016.5021
MLA
Long, J., Liu, Z., Wu, X., Xu, Y., Ge, C."Gene expression profile analysis of pancreatic cancer based on microarray data". Molecular Medicine Reports 13.5 (2016): 3913-3919.
Chicago
Long, J., Liu, Z., Wu, X., Xu, Y., Ge, C."Gene expression profile analysis of pancreatic cancer based on microarray data". Molecular Medicine Reports 13, no. 5 (2016): 3913-3919. https://doi.org/10.3892/mmr.2016.5021