Open Access

Identification of potential therapeutic targets for lung cancer by bioinformatics analysis

  • Authors:
    • Li‑Quan Wang
    • Lan‑Hua Zhao
    • Yi‑Ze Qiao
  • View Affiliations

  • Published online on: December 31, 2015     https://doi.org/10.3892/mmr.2015.4752
  • Pages: 1975-1982
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to identify potential therapeutic targets for lung cancer and explore underlying molecular mechanisms of its development and progression. The gene expression profile datasets no. GSE3268 and GSE19804, which included five and 60 pairs of tumor and normal lung tissue specimens, respectively, were downloaded from Gene Expression Omnibus. Differentially expressed genes (DEGs) between lung cancer and normal tissues were identified, and gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis of the DEGs was performed. Furthermore, protein‑protein interaction (PPI) networks and a transcription factor (TF) regulatory network were constructed and key target genes were screened. A total of 466 DEGs were identified, and the PPI network indicated that IL‑6 and MMP9 had key roles in lung cancer. A PPI module containing 34 nodes and 547 edges was obtained, including PTTG1. The TF regulatory network indicated that TFs of FOSB and LMO2 had a key role. Furthermore, MMP9 was indicated to be the target of FOSB, while PTTG1 was the target of LMO2. In conclusion, the bioinformatics analysis of the present study indicated that IL‑6, MMP9 and PTTG1 may have key roles in the progression and development of lung cancer and may potentially be used as biomarkers or specific therapeutic targets for lung cancer.

Introduction

Lung cancer is one of the most common malignancies and has a significant socioeconomic impact on patients and their families (1). In western countries, the mortality rate of lung cancer is 15% and the worldwide mortality rate for patients with lung cancer is 86% (2). The high mortality of lung cancer is mainly attributable to the lack of effective therapeutic methods and the difficulty of obtaining an early diagnosis. Thus, the development of effective therapeutic targets is urgently required.

Differentially expressed genes (DEGs) have been reported to have important roles in lung cancer, and their identification may aid in the elucidation of its underlying molecular mechanisms as well as the discovery of novel biomarkers and treatments (3). Numerous genes, including p53 (3,4), EGFR (5,6), kRAS (7), PIK3CA (8) and EML4 (9), are known to be associated with lung cancer, while others have remained elusive. Futhermore, SEMA5A and -6A were identified as potential therapeutic targets for lung cancer (1012). Although tremendous efforts have been made to discover novel targets for lung cancer treatments, the current knowledge is insufficient and requires expansion.

In the present study, DEGs between lung cancer and normal lung tissues were identified. Protein-protein interaction (PPI) and transcription factor (TF) regulatory networks were constructed and key target genes were screened. Through the identification of key genes, the possible underlying molecular mechanisms as well as potential candidate biomarkers and treatment targets for lung cancer were explored.

Materials and methods

Affymetrix microarray data

The gene expression profile dataset no. GSE3268 deposited in the Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/) by Wachi et al (13) based on the GPL96 platform (HG-U133A; Affymetrix Human Genome U133A Array), was subjected to bioinformatics analysis in the present study. The dataset contained a total of 10 chips, including five squamous cell lung cancer tissues and five paired adjacent normal lung tissues obtained from patients with squamous cell lung cancer.

Furthermore, the gene expression profile dataset GSE19804 based on the platform GPL570 (HG-U133_Plus_2; Affymetrix Human Genome U133 Plus 2.0 Array), which was deposited in the GEO database by Lu et al (14), was used. The dataset contained 120 chips, including 60 samples of non-small cell lung cancer tissues and 60 samples of paired normal lung tissues from female Taiwanese patients.

Identification of DEGs

The raw data were pre-processed using the Affy package (15) in R language. DEGs of GSE3268 (DEG1) and GSE19804 (DEG2) between normal groups and disease groups were respectively analyzed using the limma package in R (16). Fold changes (FCs) in the expression of individual genes were calculated and DEGs with P<0.05 and |log FC| >1 were considered to be significant. DEG1 and DEG2 were then combined and the pooled dataset was referred to as the overlapping DEGs in the present study.

Gene ontology (GO) and pathway enrichment analysis of DEGs

GO analysis is a commonly used approach for functional studies of large-scale transcriptomic data (17). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database (18) contains information on networks of molecules or genes. The Database for Annotation, Visualization and Integrated Discovery (DAVID) (19) was used to systematically extract biological information from the large number of genes. GO functions and KEGG pathways of the overlapping DEGs were analyzed using DAVID 6.7 with P<0.05.

Construction of PPI network and screening of modules

The Search Tool for the Retrieval of Interacting Genes (STRING) (20) database was used to retrieve the predicted interactions for the DEGs; version 9.1 of STRING covers 1,133 completely sequenced species. All associations obtained in STRING are provided with a confidence score, which represents a rough estimate of the likelihood of a given association to describe a functional linkage between two proteins (21). The overlapping DEGs with a confidence score >0.4 were selected to construct the PPI network using Cytoscape software (version 3.0; http://cytoscape.org/) (22). Cytoscape allows for the visualization of complex networks and their integration to any type of attribute data. The MCODE (23) plugin in Cytoscape was used to divide the PPI into modules. GO functional analysis of genes in the modules was performed using the BinGo 2.44 plugin in Cytoscape (24) with a threshold of P<0.05 using the hypergeometric test.

Transcriptional regulatory network construction

The University of California at Santa Cruz (UCSC) database (http://genome.ucsc.edu) contains information on TF binding sites and the regulated genes (25). Using information collected from the UCSC database, DEGs were matched with their associated TFs. The TF regulatory network then was constructed using Cytoscape software (26).

Results

GO and pathway enrichment analysis of DEGs

From the GEO datasets, information on the expression of 8,172 genes was obtained. The normalized results showed that the expression median after normalization was in a straight line (Fig. 1). A total of 466 DEGs, including 156 upregulated and 310 downregulated genes, were selected.

Results of GO analysis showed that the upregulated DEGs were significantly enriched in biological processes, including collagen metabolic processes, multicellular organismal macromolecule metabolic processes and nuclear division (Table I); the downregulated DEGs were significantly enriched in biological processes, including response to wounding, immune response, defense response and inflammatory response (Table I).

Table I

GO and pathway analysis of the differentially expressed genes.

Table I

GO and pathway analysis of the differentially expressed genes.

ExpressionCategoryTerm/gene and functionCountP-value
UpregulatedKEGG_PATHWAYhsa04110 - Cell cycle12 6.94×10−7
KEGG_PATHWAYhsa04512 - ECM-receptor interaction10 1.50×10−6
KEGG_PATHWAYhsa04510 - Focal adhesion10 1.42×10−3
KEGG_PATHWAYhsa04115 - p53 signaling pathway6 2.14×10−3
KEGG_PATHWAYhsa00240 - Pyrimidine metabolism5 3.93×10−2
GOTERM_BP_FATGO:0032963 - Collagen metabolic process9 2.10×10−10
GOTERM_BP_FATGO:0044259 - Multicellular organismal macromolecule metabolic process9 5.19×10−10
GOTERM_BP_FATGO:0000280 - Nuclear division17 5.79×10−10
GOTERM_BP_FATGO:0007067 - Mitosis17 5.79×10−10
GOTERM_BP_FATGO:0000278 - Mitotic cell cycle21 7.04×10−10
GOTERM_BP_FATGO:0000087 - M phase of mitotic cell cycle17 7.55×10−10
GOTERM_CC_FATGO:0005576 - Extracellular region53 1.41×10−10
GOTERM_CC_FATGO:0005578 - Proteinaceous extracellular matrix19 7.80×10−9
GOTERM_CC_FATGO:0031012 - Extracellular matrix19 2.50×10−8
GOTERM_CC_FATGO:0044421 - Extracellular region part30 2.27×10−7
GOTERM_CC_FATGO:0005819 - Spindle12 4.55×10−7
GOTERM_MF_FATGO:0004222 - Metalloendopeptidase activity9 9.37×10−6
GOTERM_MF_FATGO:0048407 - Platelet-derived growth factor binding4 1.53×10−4
GOTERM_MF_FATGO:0004175 - Endopeptidase activity13 3.80×10−4
GOTERM_MF_FATGO:0004857 - Enzyme inhibitor activity11 3.81×10−4
DownregulatedKEGG_PATHWAYhsa04060 - Cytokine-cytokine receptor interaction20 6.99×10−5
KEGG_PATHWAYhsa04610 - Complement and coagulation cascades8 2.47×10−3
KEGG_PATHWAYhsa04062 - Chemokine signaling pathway13 4.53×10−3
KEGG_PATHWAYhsa04650 - Natural killer cell mediated cytotoxicity10 9.69×10−3
KEGG_PATHWAYhsa04614 - Renin-angiotensin system4 1.01×10−2
GOTERM_BP_FATGO:0009611 - Response to wounding48 2.23×10−17
GOTERM_BP_FATGO:0006952 - Defense response46 1.66×10−13
GOTERM_BP_FATGO:0006954 - Inflammatory response33 2.92×10−13
GOTERM_BP_FATGO:0006955 - Immune response43 4.20×10−10
GOTERM_BP_FATGO:0048545 - Response to steroid hormone stimulus21 3.81×10−9
GOTERM_CC_FATGO:0005615 - Extracellular space55 2.36×10−18
GOTERM_CC_FATGO:0044421 - Extracellular region part64 2.03×10−17
GOTERM_CC_FATGO:0005576 - Extracellular region93 3.37×10−15
GOTERM_CC_FATGO:0005886 - Plasma membrane131 2.25×10−12
GOTERM_CC_FATGO:0005887 - Integral to plasma membrane61 1.99×10−11
GOTERM_MF_FATGO:0019838 - Growth factor binding16 2.01×10−9
GOTERM_MF_FATGO:0030246 - Carbohydrate binding27 7.86×10−9
GOTERM_MF_FATGO:0019955 - Cytokine binding13 1.54×10−6
GOTERM_MF_FATGO:0005509 - Calcium ion binding39 1.04×10−5
GOTERM_MF_FATGO:0030247 - Polysaccharide binding14 1.11×10−5

[i] BP, biological process; CC, cellular component; MF, molecular function; Count, numbers of differentially expressed genes; ECM, extracellular matrix; GO, gene ontology; hsa, Homo sapiens; KEGG, Kyoto Encyclopedia of Genes and Genomes; FAT, functional annotation tool.

Pathway analysis showed that the upregulated DEGs were significantly enriched in cell cycle, extracellular matrix - receptor interaction and the p53 signaling pathway (Table I); the downregulated DEGs were significantly enriched in cytokine receptor interaction, complement and coagulation cascades as well as chemokine signaling pathways (Table I).

Construction of PPI network and screening of module

The PPI network was constructed based on the predicted interactions of the identified DEGs (Fig. 2). Genes of IL-6, FOSB, CDK1, MMP9 and ICAM1 were found to have a high degree of interaction in lung cancer. A sub-network containing 34 nodes and 547 edges was screened from the PPI network, such as PTTG1 (Fig. 3). The DEGs in the sub-net were significantly enriched in biological processes, such as the cell cycle, and pathway analysis showed that they were significantly enriched in cell cycle and oocyte meiosis (Table II).

Table II

GO and pathway analysis of genes in sub-network.

Table II

GO and pathway analysis of genes in sub-network.

CategoryTerm/gene and functionCountP-value
KEGG_PATHWAYhsa04110 - Cell cycle10 1.09×10−11
KEGG_PATHWAYhsa04114- Oocyte meiosis6 1.09×10−5
KEGG_PATHWAYhsa04914 - Progesterone-mediated oocyte maturation4 1.83×10−3
KEGG_PATHWAYhsa04115 - p53 signaling pathway3 1.65×10−3
KEGG_PATHWAYhsa00240 - Pyrimidine metabolism3 3.10×10−2
GOTERM_BP_FATGO:0000278 - Mitotic cell cycle19 7.13×10−21
GOTERM_BP_FATGO:0007049 - Cell cycle22 1.65×10−19
GOTERM_BP_FATGO:0000280 - Nuclear division16 2.14×10−19
GOTERM_BP_FATGO:0007067 - Mitosis16 2.14×10−19
GOTERM_BP_FATGO:0000087 - M phase of mitotic cell cycle16 2.82×10−19
GOTERM_CC_FATGO:0005819 - Spindle12 9.20×10−15
GOTERM_CC_FATGO:0000777 - Condensed chromosome kinetochore8 3.94×10−11
GOTERM_CC_FATGO:0015630 - Microtubule cytoskeleton14 5.31×10−11
GOTERM_CC_FATGO:0000779 - Condensed chromosome, centromeric region8 1.01×10−10
GOTERM_CC_FATGO:0000922 - Spindle pole7 1.01×10−10
GOTERM_MF_FATGO:0005524 - Adenosine triphosphate binding15 4.89×10−7
GOTERM_MF_FATGO:0032559 - Adenyl ribonucleotide binding15 5.78×10−7
GOTERM_MF_FATGO:0030554 - Adenyl nucleotide binding15 1.10×10−6
GOTERM_MF_FATGO:0001883 - Purine nucleoside binding15 1.32×10−6
GOTERM_MF_FATGO:0001882 - Nucleoside binding15 1.44×10−6

[i] BP, biological process; CC, cellular component; MF, molecular function; Count, numbers of DEGs; GO, gene ontology; hsa, Homo sapiens; KEGG, Kyoto Encyclopedia of Genes and Genomes; FAT, functional annotation tool.

TF-target gene regulatory network analysis

Associations between 44 TFs and their 47 target DEGs were collected from the TF regulatory network (Fig. 4). TFs of FOSB and LMO2, which exhibited a high degree of interaction, were selected from this network. Furthermore, the results also showed that MMP9 was the target of FOSB and PTTG1 was the target of LMO2.

Discussion

Lung cancer is the leading cause of cancer-associated mortality; however, the underlying molecular mechanisms of its development and progression have remained to be fully elucidated (1). The present study used a bioinformatics approach to predict the potential therapeutic targets and explore the possible molecular mechanisms for lung cancer. A total of 466 DEGs between tumorous and normal tissues was identified, among which 310 genes were downregulated and 156 were upregulated. By constructing a PPI network and a TF regulatory network, key genes, including IL6, MMP9 and PTTG1, were identified.

IL-6 is a multifunctional cytokine that was characterized as a regulator of immune and inflammatory responses (27,28). It is involved in the regulation of cell proliferation, survival and metabolism, and IL-6 signaling has an important role in tumorigenesis (29). Chung et al (30) found that IL-6 activated PI3K, which promoted apoptosis in human prostate cancer cell lines. Furthermore, studies have shown that IL-6 inhibited the growth of numerous types of cancer, including lung (31), breast (32) and prostate cancer (33). In the present study, IL-6 was shown to be downregulated in squamous cell and non-small cell lung cancer, and GO analysis showed that IL-6 was significantly enriched in biological processes, including defense response, inflammatory response, immune response and regulation of cell proliferation, which was consistent with a previous study (29). Combined with the above studies, it is indicated that IL-6 may be a diagnostic biomarker and therapeutic target in lung cancer.

MMP9 has a key role in cell migration, proliferation, differentiation, angiogenesis, apoptosis and host defense (34). Dysregulatoin of MMPs has been implicated in numerous diseases, including chronic ulcers and cancer (3537). Downregulation of MMPs has been shown to inhibit metastasis, while upregulation of MMPs led to enhanced cancer cell invasion (37). In the present study, MMP9 was overexpressed and regulated by FOSB in lung cancer tissues. Kim et al (38) found that FOSB was downregulated in pancreatic cancer and promoted tumor progression. Kataoka et al (39) found that FOSB gene expression in cancer stroma is a independent prognostic indicator for patients with epithelial ovarian cancer receiving standard therapy. Combined with the above studies, the present study indicated that MMP9 may have important roles in the progression of lung cancer, and that it may be utilized as a therapeutic target.

PTTG1 has tumorigenic activity and is highly expressed in various tumor types (40). Studies have shown that PTTG1 was overexpressed in esophageal cancer and associated with endocrine therapy resistance in breast cancer (41,42). Yoon et al (40) showed that the PTTG1 oncogene promoted tumor malignancy via epithelial-to-mesenchymal expansion of the cancer stem cell population. Hamid et al (43) found that PTTG1 promoted tumorigenesis in human embryonic kidney cells. A study by Li et al (44) indicated that PTTG1 promoted migration and invasion of human non-small cell lung cancer cells. Panguluri et al (45) showed that PTTG1 was an important target gene for ovarian cancer therapy. In the present study, PTTG1 was found to be overexpressed in lung cancer tissues and regulated by LMO2. LMO2 is an important regulator in determining cell fate and controlling cell growth and differentiation (46). Nakata et al (47) found that LMO2 was a novel predictive biomarker with the potential to enhance the accuracy of prognoses for pancreatic cancer. Yamada et al (48) showed that LMO2 is a key regulator of tumour angiogenesis. Combined with the above studies, the present study indicated that PTTG1 may have important roles in the progression of lung cancer and that it may represent a therapeutic target.

In conclusion, the bioinformatics analysis of the present study indicated that IL-6, MMP9 and PTTG1 may have key roles in the progression and development of lung cancer. They may be used as prognostic biomarkers as well as specific therapeutic targets for the treatment of lung cancer. However, molecular biology experiments are required to confirm these findings.

References

1 

Nugent M, Edney B, Hammerness PG, Dain BJ, Maurer LH and Rigas JR: Non-small cell lung cancer at the extremes of age: Impact on diagnosis and treatment. Ann Thorac Surg. 63:193–197. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Yang SP, Luh KT, Kuo SH and Lin CC: Chronological observation of epidemiological characteristics of lung cancer in Taiwan with etiological consideration-a 30-year consecutive study. Jpn J Clin Oncol. 14:7–19. 1984.PubMed/NCBI

3 

Andriani F, Roz E, Caserini R, Conte D, Pastorino U, Sozzi G and Roz L: Inactivation of both FHIT and p53 cooperate in deregulating proliferation-related pathways in lung cancer. J Thorac Oncol. 7:631–642. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Toyooka S, Tsuda T and Gazdar AF: The TP53 gene, tobacco exposure and lung cancer. Hum Mutat. 21:229–239. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Shigematsu H, Lin L, Takahashi T, Nomura M, Suzuki M, Wistuba II, Fong KM, Lee H, Toyooka S, Shimizu N, et al: Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 97:339–346. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Martin P, Kelly CM and Carney D: Epidermal growth factor receptor-targeted agents for lung cancer. Cancer Control. 13:129–140. 2006.PubMed/NCBI

7 

Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, Ince WL, Jänne PA, Januario T, Johnson DH, et al: Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. J Clin Oncol. 23:5900–5909. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Yamamoto H, Shigematsu H, Nomura M, Lockwood WW, Sato M, Okumura N, Soh J, Suzuki M, Wistuba II, Fong KM, et al: PIK3CA mutations and copy number gains in human lung cancers. Cancer Res. 68:6913–6921. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Wong DW, Leung EL, So KK, Tam IY, Sihoe AD, Cheng LC, Ho KK, Au JS, Chung LP, Pik Wong M, et al: The EML4-ALK fusion gene is involved in various histologic types of lung cancers from nonsmokers with wild-type EGFR and KRAS. Cancer. 115:1723–1733. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Castro-Rivera E, Ran S, Thorpe P and Minna JD: Semaphorin 3B (SEMA3B) induces apoptosis in lung and breast cancer, whereas VEGF165 antagonizes this effect. Proc Natl Acad Sci USA. 101:11432–11437. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Tomizawa Y, Sekido Y, Kondo M, Gao B, Yokota J, Roche J, Drabkin H, Lerman MI, Gazdar AF, Minna JD, et al: Inhibition of lung cancer cell growth and induction of apoptosis after reex-pression of 3p21. 3 candidate tumor suppressor gene SEMA3B. Proc Natl Acad Sci USA. 98:13954–13959. 2001. View Article : Google Scholar

12 

Brambilla E, Constantin B, Drabkin H and Roche J: Semaphorin SEMA3F localization in malignant human lung and cell lines: A suggested role in cell adhesion and cell migration. Am J Pathol. 156:939–950. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Wachi S, Yoneda K and Wu R: Interactome-transcriptome analysis reveals the high centrality of genes differentially expressed in lung cancer tissues. Bioinformatics. 21:4205–4208. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Lu TP, Tsai MH, Lee JM, Hsu CP, Chen PC, Lin CW, Shih JY, Yang PC, Hsiao CK, Lai LC, et al: Identification of a novel biomarker, SEMA5A, for non-small cell lung carcinoma in nonsmoking women. Cancer Epidemiol Biomarkers Prev. 19:2590–2597. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:2004.

17 

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

18 

Ogata H, Goto S, Sato K, Fujibuchi W, Bono H and Kanehisa M: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 27:29–34. 1999. View Article : Google Scholar

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:P32003. View Article : Google Scholar

20 

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

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:D561–D568. 2011. View Article : Google Scholar :

22 

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

23 

Bader GD and Hogue CW: An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 4:22003. View Article : Google Scholar : PubMed/NCBI

24 

Maere S, Heymans K and Kuiper M: BiNGO: A cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics. 21:3448–3449. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Wingender E, Dietze P, Karas H and Knüppel R: TRANSFAC: A database on transcription factors and their DNA binding sites. Nucleic Acids Res. 24:238–241. 1996. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Schafer ZT and Brugge JS: IL-6 involvement in epithelial cancers. J Clin Invest. 117:3660–3663. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Kishimoto T: Interleukin-6: From basic science to medicine-40 years in immunology. Annu Rev Immunol. 23:1–21. 2005. View Article : Google Scholar

29 

Hodge DR, Hurt EM and Farrar WL: The role of IL-6 and STAT3 in inflammation and cancer. Eur J Cancer. 41:2502–2512. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Chung TD, Yu JJ, Kong TA, Spiotto MT and Lin JM: Interleukin-6 activates phosphatidylinositol-3 kinase, which inhibits apoptosis in human prostate cancer cell lines. Prostate. 42:1–7. 2000. View Article : Google Scholar

31 

Takizawa H, Ohtoshi T, Ohta K, Yamashita N, Hirohata S, Hirai K, Hiramatsu K and Ito K: Growth inhibition of human lung cancer cell lines by interleukin 6 in vitro: A possible role in tumor growth via an autocrine mechanism. Cancer Res. 53:4175–4181. 1993.PubMed/NCBI

32 

Knüpfer H and Preiss R: Significance of interleukin-6 (IL-6) in breast cancer (review). Breast Cancer Res Treat. 102:129–135. 2007. View Article : Google Scholar

33 

Giri D, Ozen M and Ittmann M: Interleukin-6 is an autocrine growth factor in human prostate cancer. Am J Pathol. 159:2159–2165. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Sica A, Allavena P and Mantovani A: Cancer related inflammation: The macrophage connection. Cancer Lett. 267:204–215. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Benveniste EN: Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis. J Mol Med (Berl). 75:165–173. 1997. View Article : Google Scholar

36 

Firestein GS: Evolving concepts of rheumatoid arthritis. Nature. 423:356–361. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Coussens LM, Fingleton B and Matrisian LM: Matrix metal-loproteinase inhibitors and cancer: Trials and tribulations. Science. 295:2387–2392. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Kim JH, Lee JY, Lee KT, Lee JK, Lee KH, Jang KT, Heo JS, Choi SH and Rhee JC: RGS16 and FosB underexpressed in pancreatic cancer with lymph node metastasis promote tumor progression. Tumor Biol. 31:541–548. 2010. View Article : Google Scholar

39 

Kataoka F, Tsuda H, Arao T, Nishimura S, Tanaka H, Nomura H, Chiyoda T, Hirasawa A, Akahane T, Nishio H, et al: EGRI and FOSB gene expressions in cancer stroma are independent prognostic indicators for epithelial ovarian cancer receiving standard therapy. Gene Chromosome Cancer. 51:300–312. 2012. View Article : Google Scholar

40 

Yoon CH, Kim MJ, Lee H, Kim RK, Lim EJ, Yoo KC, Lee GH, Cui YH, Oh YS, Gye MC, et al: PTTG1 oncogene promotes tumor malignancy via epithelial to mesenchymal transition and expansion of cancer stem cell population. J Biol Chem. 287:19516–19527. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Shibata Y, Haruki N, Kuwabara Y, Nishiwaki T, Kato J, Shinoda N, Sato A, Kimura M, Koyama H, Toyama T, et al: Expression of PTTG (pituitary tumor transforming gene) in esophageal cancer. Jpn J Clin Oncol. 32:233–237. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Ghayad SE, Vendrell JA, Bieche I, Spyratos F, Dumontet C, Treilleux I, Lidereau R and Cohen PA: Identification of TACC1, NOV and PTTG1 as new candidate genes associated with endocrine therapy resistance in breast cancer. J Mol Endocrinol. 42:87–103. 2009. View Article : Google Scholar

43 

Hamid T, Malik MT and Kakar SS: Ectopic expression of PTTG1/securin promotes tumorigenesis in human embryonic kidney cells. Mol Cancer. 4:32005. View Article : Google Scholar : PubMed/NCBI

44 

Li H, Yin C, Zhang B, Sun Y, Shi L, Liu N, Liang S, Lu S, Liu Y, Zhang J, et al: PTTG1 promotes migration and invasion of human non-small cell lung cancer cells and is modulated by miR-186. Carcinogenesis. 34:2145–2155. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Panguluri SK, Yeakel C and Kakar SS: PTTG: An important target gene for ovarian cancer therapy. J Ovarian Res. 1:62008. View Article : Google Scholar : PubMed/NCBI

46 

Ma S, Guan XY, Beh PS, Wong KY, Chan YP, Yuen HF, Vielkind J and Chan KW: The significance of LMO2 expression in the progression of prostate cancer. J Pathol. 211:278–285. 2007. View Article : Google Scholar

47 

Nakata K, Ohuchida K, Nagai E, Hayashi A, Miyasaka Y, Kayashima T, Yu J, Aishima S, Oda Y, Mizumoto K, et al: LMO2 is a novel predictive marker for a better prognosis in pancreatic cancer. Neoplasia. 11:712–719. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Yamada Y, Pannell R, Forster A and Rabbitts TH: The LIM-domain protein Lmo2 is a key regulator of tumour angiogenesis: A new anti-angiogenesis drug target. Oncogene. 21:1309–1315. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 13 Issue 3

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
Wang LQ, Zhao LH and Qiao YZ: Identification of potential therapeutic targets for lung cancer by bioinformatics analysis. Mol Med Rep 13: 1975-1982, 2016
APA
Wang, L., Zhao, L., & Qiao, Y. (2016). Identification of potential therapeutic targets for lung cancer by bioinformatics analysis. Molecular Medicine Reports, 13, 1975-1982. https://doi.org/10.3892/mmr.2015.4752
MLA
Wang, L., Zhao, L., Qiao, Y."Identification of potential therapeutic targets for lung cancer by bioinformatics analysis". Molecular Medicine Reports 13.3 (2016): 1975-1982.
Chicago
Wang, L., Zhao, L., Qiao, Y."Identification of potential therapeutic targets for lung cancer by bioinformatics analysis". Molecular Medicine Reports 13, no. 3 (2016): 1975-1982. https://doi.org/10.3892/mmr.2015.4752