Transcriptional regulatory networks in human lung adenocarcinoma

  • Authors:
    • Xiangrui Meng
    • Peng Lu
    • Hua Bai
    • Peng Xiao
    • Qingxia Fan
  • View Affiliations

  • Published online on: August 14, 2012     https://doi.org/10.3892/mmr.2012.1034
  • Pages: 961-966
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung adenocarcinoma (AC) is the most common histological subtype of lung cancer worldwide and its absolute incidence is increasing markedly. Transcriptional regulation is one of the most fundamental processes in lung AC development. However, high-throughput functional analyses of multiple transcription factors and their target genes in lung AC are rare. Thus, the objective of our study was to interpret the mechanisms of human AC through the regulatory network using the GSE2514 microarray data. Our results identified the genes peroxisome proliferator activated receptor-γ (PPARG), CCAAT/enhancer binding protein β (CEBPB), ets variant 4 (ETV4), Friend leukemia virus integration 1 (FLI1), T-cell acute lymphocytic leukemia 1 (TAL1) and nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1) as hub nodes in the transcriptome network. Among these genes, it appears that: PPARG promotes the PPAR signaling pathway via the upregulation of lipoprotein lipase (LPL) expression, but suppresses the cell cycle pathway via downregulation of growth arrest and DNA-damage-inducible, γ (GADD45G) expression; ETV4 stimulates matrix metallopeptidase 9 (MMP9) expression to induce the bladder cancer pathway; FLI upregulates transforming growth factor, β receptor II (TGFBR2) expression to activate TGF-β signaling and upregulates cyclin D3 (CCND3) expression to promote the cell cycle pathway; NFKB1 upregulates interleukin 1, β (IL-1B) expression and initiates the prostate cancer pathway; CEBPB upregulates IL-6 expression and promotes pathways in cancer; and TAL1 promotes kinase insert domain receptor (KDR) expression to promote the TGF-β signaling pathway. This transcriptional regulation analysis may provide an improved understanding of the molecular mechanisms and potential therapeutic targets in the treatment of lung AC.

Introduction

Lung carcinoma is the most common cancer in the world and the leading cause of cancer-related mortality, with over one million cases annually (1). Lung carcinomas are usually classified as small-cell lung carcinomas (SCLCs) or non-small-cell lung carcinomas (NSCLCs). NSCLCs are histopathologically and clinically distinct from SCLCs and are further subcategorized as adenocarcinomas (ACs), squamous cell carcinomas or large-cell carcinomas, of which ACs are the predominant form (2).

Several molecular changes are characteristic of lung AC. These include mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR), v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) (3,4), tumor protein 53 (TP53) (5), serine/threonine kinase 11 (STK11) (6) and cyclin-dependent kinase inhibitor 2A (CDKN2A) (7) genes. Among them, somatic-activating EGFR mutations, particularly deletions in exon 19 and L858R point mutations in exon 21, may activate the gp130/JAK/STAT3 pathway by means of interleukin 6 (IL-6) upregulation in primary human lung AC, thereby promoting cell cycle progression, cell growth and tumorigenesis (8). Further studies have revealed that thyroid transcription factor-1 (TTF-1) expression is positively associated with EGFR mutations in lung AC (9). These results indicate that transcriptional regulation is a fundamental process in lung AC development.

However, high-throughput functional analyses of multiple transcription factors (TFs) and their target genes in lung AC are rare. Therefore, the objective of this study was to identify potential transcription regulation correlations between TFs and differentially expressed genes (DEGs) in lung AC using microarray data and transcriptional network analysis. In addition, the underlying molecular mechanisms were explored by KEGG pathway enrichment.

Materials and methods

Affymetrix microarray data analysis

The transcription profiles of human AC GSE2514 (10) were obtained from a public functional genomics data repository GEO (http://www.ncbi.nlm.nih.gov/geo/) and are based on the Affymetrix GPL8300 platform data (Affymetrix Human Genome U95 Version 2 Array) (11). Only 20 AC chips and 19 control chips were useable. Each pair of samples (one derived from cancer cells and the other from normal cells) represented a single patient with AC.

All patients participating in this study were enrolled in a local Colorado Multiple Institutional Review Board (COMIRB)-approved protocol for use of remnant tissue with anonymization and analysis of specimens and clinical data. Informed consent was obtained from all the patients. All but one of the patients had a history of smoking. Patients ranged in age from 45 to 73 years of age. Tumors from five males and five females were used in the study. All specimens for microarray analysis were obtained at surgery with nine patients undergoing lobectomy and one wedge resection. Specimens were examined immediately following removal from the patient and grossly visible solid tumor tissue was snap-frozen for RNA extraction. The tumors were all invasive ACs, but five specimens exhibited evidence of bronchoalveolar differentiation at the edge of tumor nests. Most tumors were low to intermediate grade and low stage, although two stage III tumors were included in the analysis.

The limma method (12) was used to identify DEGs. The original expression datasets from all conditions were extracted into expression estimates using the robust multiarray average (RMA) method (13) with the default settings implemented in Bioconductor and the linear model was constructed. Only the DEGs with a fold-change >1.5 and p-value <0.05 were selected.

Regulatory network analysis for AC

The TRANSFAC database contains data on TFs, their experimentally demonstrated binding sites and regulated genes (14). The Transcriptional Regulatory Element Database (TRED) has been built in response to increasing requirements for an integrated repository for cis- and trans-regulatory elements in mammals (15). TRED has curated transcriptional regulation information, including TF binding motifs and experimental evidence. The curation is currently focused on the target genes of 36 cancer-related TF families. A total of 774 pairs of regulatory relationships between 219 TFs and 265 target genes were collected from TRANSFAC (http://www.gene-regulation.com/pub/databases.html). A total of 5722 pairs of regulatory relationships between 102 TFs and 2920 target genes were collected from TRED (http://rulai.cshl.edu/TRED/). By integrating the two regulation datasets, a total of 6328 regulatory relationships between 276 TFs and 3002 target genes were obtained. To demonstrate the potential regulatory relationships, the Pearson Correlation Coefficient (PCC) was calculated for all pair-wise comparisons of gene expression values between TFs and the DEGs. The regulatory relationships having an absolute PCC >0.7 were considered to be significant.

Pathway enrichment and TF-pathway regulatory network analysis

Kyoto Encyclopedia of Genes and Genomes (KEGG)is a collection of online databases dealing with genomes, enzymatic pathways and biological chemicals (16). The PATHWAY database records networks of molecular interactions in cells, and variants of them specific to particular organisms (http://www.genome.jp/kegg/). A total of 130 pathways, involving 2287 genes, were collected from KEGG (updated in July 2011).

DAVID (17), a high-throughput and integrated data-mining environment, analyzes gene lists derived from high-throughput genomic experiments. DAVID was used to identify over-represented KEGG pathways. Pathways with p<0.05 and a count >2 were considered to be significant.

To further investigate the regulatory relationships between TFs and significant pathways, we mapped target genes in the network to pathways and created a regulatory network comprising TFs and pathways.

Results

Microarray data and regulatory network analysis

Using the limma package, a total of 915 DEGs with p<0.05 and fold-change >1.5 were selected. Regulatory relationships with a PCC >0.7 were considered to be significant. A regulatory network for human AC comprising TFs and their target genes was constructed (Fig. 1). In this network, peroxisome proliferator activated receptor-γ (PPARG), CCAAT/enhancer binding protein [C/EBP], β (CEBPB), ets variant 4 (ETV4), Friend leukemia virus integration 1 (FLI1), T-cell acute lymphocytic leukemia 1 (TAL1), and nuclear factor of κ light polypeptide gene enhancer in B-cells 1 (NFκB1) with higher degrees of interaction formed local networks, suggesting that these TFs are significant in lung AC. Among them, it appears that: PPARG can upregulate lipoprotein lipase (LPL), but downregulate growth arrest and DNA-damage-inducible, γ (GADD45G) expression; FLI can upregulate transforming growth factor, β receptor II (TGFBR2) and cyclin D3 (CCND3) expression; ETV4 can downregulate TGFBR2 expression, but stimulate matrix metallopeptidase 9 (MMP9) expression; NFκB1 can upregulate IL1B expression; CEBPB can upregulate IL-6 expression; TAL1 could promote kinase insert domain receptor (KDR) expression.

Significant pathways and TF pathway regulatory network analysis

Using the KEGG pathways to describe the function of the regulatory network, several KEGG pathways among the pathways in the regulatory network were revealed to be enriched, including pathways in cancer (hsa05200), the PPAR signaling pathway (hsa03320), cell cycle (hsa04110) and the TGF-β signaling pathway (hsa04350). The 10 most enriched KEGG pathways are listed in Table I.

Table I

Pathway significance analysis.

Table I

Pathway significance analysis.

TermDescriptionCountp-valueFDR
hsa05200Pathways in cancer16 1.01e−9 1.05e−6
hsa05220Chronic myeloid leukemia7 9.72e−60.010121
hsa05216Thyroid cancer5 3.95e−50.041159
hsa05210Colorectal cancer6 2.40e−40.249483
hsa03320PPAR signaling pathway50.0011841.225733
hsa04110Cell cycle60.0014891.538652
hsa05222Small cell lung cancer50.0024572.527709
hsa04350TGF-β signaling pathway50.0027932.86916
hsa05219Bladder cancer40.0028542.931146
hsa05215Prostate cancer50.0030343.113205

[i] Term represents the pathway ID. Description is the pathway symbol. Count is the number of enrichment pathways. The p-value is the probability of obtaining a test statistic; the smaller the p-value, the more enriched the pathway. False discovery rate (FDR) control is a statistical method used in multiple hypothesis testing to correct for multiple comparisons; the smaller the FDR, the higher the correctness.

To further investigate the regulatory relationships between TFs and pathways, we mapped DEGs to significant pathways and obtained a regulatory network comprising TFs and pathways (Fig. 2). In the network, 9 TFs regulated 6 pathways. The network indicates that the PPAR pathway is upregulated by PPARG and CEBPB; the cell cycle is upregulated by FLI and STAT5B, but downregulated by PPARG; the TGF-β signaling pathway is upregulated by FLI and TAL1, but downregulated by ETV4; ETV4 promotes the bladder cancer pathway; NFκB1 promotes the prostate cancer pathway. Pathways in cancer may be upregulated by CEBPD and CEBPB.

Discussion

We investigated the comprehensive regulatory network of lung AC comprising TFs, target genes and their underlying molecular pathways. In our transcriptosome network, the genes PPARG, CEBPB, ETV4, FLI1, TAL1, and NFκB1 are hub nodes. PPARG may promote the PPAR signaling pathway via upregulation of LPL expression, but suppress the cell cycle pathway via downregulation of GADD45G expression; ETV4 can stimulate MMP9 expression to induce the bladder cancer pathway; FLI can upregulate TGFBR2 expression to activate TGF-β signaling, and upregulate CCND3 expression to promote the cell cycle pathway; NFκB1 can upregulate IL1B expression and initiate the prostate cancer pathway; CEBPB can upregulate IL-6 expression and promote pathways in cancer; TAL1 could upregulate KDR expression to promote the TGF-β signaling pathway.

PPARG is a ligand-activated TF, whose activation has been implicated in the pathology of numerous diseases, including lung AC. High PPARG expression levels have been detected in lung AC patients (18) and PPARG-positivity has been identified more frequently in well-differentiated AC cases than in moderately and poorly differentiated ones (19). The treatment of lung AC cells with PPARG ligands induces a dose-dependent inhibition of lung AC cell growth, that is, a cell cycle arrest at G0/G1 (18,20). GADD45 is a cell cycle-regulated nuclear protein that reaches maximal levels in the G1 phase of the cycle (21). Through its association with Cdc2, GADD45 disrupts the interactions of Cdc2 with cyclin B1 and, thus, may induce G2/M arrest (22). Previous studies indicate that the activation of PPARG may lead to apoptosis and growth arrest, at least in part, by inducing the Oct-1-mediated transcription of GADD45 (23). However, GADD45G methylation is significantly frequent in lung cancer patients and results in lung tumorigenesis (24). Similarly, we also found that GADD45G was downregulated by PPARG in lung AC. Moreover, PPARG is a transcriptional factor which mediates pleiotropic effects, including the regulation of genes involved in lipid metabolism, such as LPL, which is a component of the PPAR signaling pathway. PPARG and the 9-cis retinoic acid receptor (RXR) heterodimers bind to the promoter sequence (−169 TGCCCTTTCCCCC −157) of the LPL gene and thus promote the transcriptional activation of the LPL gene (25,26). Higher LPL levels accelerate the growth of cancer cells (27) and predict shorter NSCLC patient survival times (28).

ETV4 (also known as E1AF), which binds to the enhancer elements of the adenovirus type 5 E1A gene, is a TF of the ets oncogene family (29). ETV4 is expressed in NSCLC cells. Significantly, ETV4-transfected NSCLC cells show a 2-fold increase in cell motility and invasion compared with parental and vector-transfected control cells (30). ETV4 is able to upregulate multiple MMP genes that contribute to the malignant phenotype of cancer cells by inducing invasive and metastatic activities (31). A previous study has shown that the ERK-ETV4-MMP1 axis is upregulated in esophageal AC cells and is a potentially significant driver of the metastatic progression of esophageal ACs (32). In the current study, our results revealed that MMP9 expression was upregulated by ETV4, which may be involved in the development and invasion of lung AC.

FLI1 is a TF of the ETS family, defined by a highly conserved DNA-binding domain (33). Its clinical role is most evident in human Ewing’s sarcoma in which it is fused with EWS. It has been shown that transduction of the gene EWSR1-FLI1 transforms NIH3T3 cells and that mutants containing a deletion in either the EWS domain or the DNA-binding domain in FLI1 lose this ability (34,35). Significantly, EWS-FLI1 binds to the second positive regulatory element of the TGF-b RII promoter, a putative tumor suppressor gene in the TGF-β signaling pathway, and suppresses transcription of the TGF-b RII gene at the mRNA and protein levels (36). Antisense to EWSR1-FLI1 in ES cell lines positive for this gene fusion restores TGF-β RII expression (37) which blocks tumorigenicity. However, the expression of FLI1 in lung cancer cells is not well characterized. FLI1 expression is usually detected at lower levels in certain non-hematopoietic tissues, including the lung. Weak nuclear immunoreactivity is observed in lung AC (38). Therefore, TGF-β RII expression may be upregulated to block tumorigenicity. In addition, levels of CCND3 and FLI1 are also positively correlated, since FLI1 maintains high levels of CCND3 in erythroblasts, thereby promoting proliferation over differentiation (39). Our results also suggest that FLI1 upregulates CCND3 expression. Therefore, we also suggest that CCND3 expression is associated with the proliferation of lung AC cells.

NFκB1 is a subunit of the TF NFκB which is derived by proteolytic cleavage from the N-terminus of a 105-kDa precursor protein (40). Activated NFκB stimulates the expression of genes involved in a wide variety of biological functions; for example, NFκB target genes, including chemokine (C-C motif) ligand 19 (CCL19), CCL21, chemokine (C-X-C motif) ligand 12 (CXCL12), CXCL13 and B-cell-activating factor of the tumour-necrosis-factor family (BAFF), were markedly upregulated in pancreatic ductal AC cell lines (41). In addition, the −300 region of the IL-1B promoter contains a functional NFKB binding site composed of the decamer sequence 5′-GGGAAAATCC-3′ (42). Therefore, activated NFκB may also induce the expression of IL-1B, which is an important cytokine involved in inflammatory and immune diseases, including various cancers (43).

CEBPB is a bZIP TF which may bind as a homodimer to certain DNA regulatory regions or form heterodimers with the related proteins CEBP-α, CEBP-δ, and CEBP-γ. CEBPB protein is important in the regulation of genes involved in immune and inflammatory responses. It has been shown to activate the IL-6 promoter and induce elevated IL-6 expression levels (44), which are frequently observed in human lung ACs (45). A further study has revealed that the regulation of the IL-6 promoter by CEBPB is completely dependent upon co-operative functions with NFκB in autocrine human prostate cancer cells (46), suggesting a model in which the bZIP protein primarily functions to augment the activity of NF-κB.

The TAL1 (or SCL) gene encodes a basic helix-loop-helix (bHLH) TF that has been demonstrated to be significant in hematopoiesis and vasculogenesis (47). Previous studies have revealed that the expression of the SCL interrupting locus gene is increased in lung ACs and promotes metastatic spread, which may result from the controlling effect on its downstream gene, SCL (48). KDR (VERFR2) is one of the two VEGF receptors which is critical for mediating angiogenic endothelial cell responses via the VEGF pathway (49). Higher levels of soluble VEGFR2 have been observed in NSCLC patients compared with healthy controls (50). E-box protein E2-2 blocks endothelial cell activation via perturbation of VEGFR2 promoter activity (51). However, TAL1/SCL relieves the E2-2-mediated repression of VEGFR2 reporter activity in endothelial cells by interacting with certain DNA sequences of E2-2 (52).

A basic understanding of the mechanisms underlying AC is valuable. A deeper understanding of TFs and their regulated genes remains an area of intense study. Our present findings shed new light on the complex interacting mechanisms of TFs and their regulated genes in lung AC. These results may provide potential therapeutic targets for lung AC treatment.

References

1 

Kamangar F, Dores GM and Anderson WF: Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol. 24:2137–2150. 2006. View Article : Google Scholar

2 

Travis WD, Travis LB and Devesa SS: Lung cancer. Cancer. 75(Suppl 1): 191–202. 1995. View Article : Google Scholar : PubMed/NCBI

3 

Riely GJ, Kris MG, Rosenbaum D, et al: Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clin Cancer Res. 14:5731–5734. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Marks JL, Broderick S, Zhou Q, et al: Prognostic and therapeutic implications of EGFR and KRAS mutations in resected lung adenocarcinoma. J Thorac Oncol. 3:111–116. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Kosaka T, Yatabe Y, Onozato R, Kuwano H and Mitsudomi T: Prognostic implication of EGFR, KRAS, and TP53 gene mutations in a large cohort of Japanese patients with surgically treated lung adenocarcinoma. J Thorac Oncol. 4:22–29. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Gill RK, Yang SH, Meerzaman D, et al: Frequent homozygous deletion of the LKB1/STK11 gene in non-small cell lung cancer. Oncogene. 30:3784–3791. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Ding L, Getz G, Wheeler DA, et al: Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 455:1069–1075. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Gao SP, Mark KG, Leslie K, et al: Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. J Clin Invest. 117:3846–3856. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Hiramatsu M, Ninomiya H, Inamura K, et al: Activation status of receptor tyrosine kinase downstream pathways in primary lung adenocarcinoma with reference of KRAS and EGFR mutations. Lung Cancer. 70:94–102. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Stearman RS, Dwyer-Nield L, Zerbe L, et al: Analysis of orthologous gene expression between human pulmonary adenocarcinoma and a carcinogen-induced murine model. Am J Pathol. 167:1763–1775. 2005. View Article : Google Scholar : PubMed/NCBI

11 

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

12 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:Article 32004.

13 

Irizarry RA, Hobbs B, Collin F, et al: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar

14 

Wingender E: The TRANSFAC project as an example of framework technology that supports the analysis of genomic regulation. Brief Bioinform. 9:326–332. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Jiang C, Xuan Z, Zhao F and Zhang MQ: TRED: a transcriptional regulatory element database, new entries and other development. Nucleic Acids Res. 35:D137–D140. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Kanehisa M: The KEGG database (Review). Novartis Found Symp. 247:91–101. 2002. View Article : Google Scholar

17 

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.PubMed/NCBI

18 

Chang TH and Szabo E: Induction of differentiation and apoptosis by ligands of peroxisome proliferator-activated receptor γ in non-small cell lung cancer. Cancer Res. 60:1129–1138. 2000.

19 

Theocharis S, Kanelli H, Politi E, et al: Expression of peroxisome proliferator activated receptor-gamma in non-small cell lung carcinoma: correlation with histological type and grade. Lung Cancer. 36:249–255. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Keshamouni VG, Reddy RC, Arenberg DA, et al: Peroxisome proliferator-activated receptor-γ activation inhibits tumor progression in non-small-cell lung cancer. Oncogene. 23:100–108. 2004.

21 

Hall PA, Kearsey JM, Coates PJ, Norman DG, Warbrick E and Cox LS: Characterisation of the interaction between PCNA and Gadd45. Oncogene. 10:2427–2433. 1995.PubMed/NCBI

22 

Zhan Q, Antinore MJ, Wang XW, et al: Association with Cdc2 and inhibition of Cdc2/Cyclin B1 kinase activity by the p53-regulated protein Gadd45. Oncogene. 18:2892–2900. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Bruemmer D, Yin F, Liu J, et al: Regulation of the growth arrest and DNA damage-inducible gene 45 (GADD45) by peroxisome proliferator-activated receptor γ in vascular smooth muscle cells. Circ Res. 93:e38–e47. 2003.

24 

Na YK, Lee SM, Hong HS, Kim JB, Park JY and Kim DS: Hypermethylation of growth arrest DNA-damage-inducible gene 45 in non-small cell lung cancer and its relationship with clinicopathologic features. Mol Cells. 30:89–92. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, et al: PPARalpha and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. EMBO J. 15:5336–5348. 1996.PubMed/NCBI

26 

Li L, Beauchamp MC and Renier G: Peroxisome proliferator-activated receptor alpha and gamma agonists upregulate human macrophage lipoprotein lipase expression. Atherosclerosis. 165:101–110. 2002. View Article : Google Scholar

27 

Kuemmerle NB, Rysman E, Lombardo PS, et al: Lipoprotein lipase links dietary fat to solid tumor cell proliferation. Mol Cancer Ther. 10:427–436. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Cerne D, Zitnik IP and Sok M: Increased fatty acid synthase activity in non-small cell lung cancer tissue is a weaker predictor of shorter patient survival than increased lipoprotein lipase activity. Arch Med Res. 41:405–409. 2010. View Article : Google Scholar

29 

Shindoh M, Higashino F and Kohgo T: E1AF, an ets-oncogene family transcription factor (Review). Cancer Lett. 216:1–8. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Hiroumi H, Dosaka-Akita H, Yoshida K, et al: Expression of E1AF/PEA3, an Ets-related transcription factor in human non-small-cell lung cancers: Its relevance in cell motility and invasion. Int J Cancer. 93:786–791. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Higashino F, Yoshida K, Noumi T, Seiki M and Fujinaga K: Ets-related protein E1A-F can activate three different matrix metalloproteinase gene promoters. Oncogene. 10:1461–1463. 1995.PubMed/NCBI

32 

Keld R, Guo B, Downey P, Gulmann C, Ang YS and Sharrocks AD: The ERK MAP kinase-PEA3/ETV4-MMP-1 axis is operative in oesophageal adenocarcinoma. Mol Cancer. 9:3132010. View Article : Google Scholar : PubMed/NCBI

33 

Truong A and Ben-David Y: The role of Fli-1 in normal cell function and malignant transformation (Review). Oncogene. 19:6482–6489. 2000. View Article : Google Scholar : PubMed/NCBI

34 

May WA, Arvand A, Thompson AD, Braun BS, Wright M and Denny CT: EWS/FLI1-induced manic fringe renders NIH 3T3 cells tumorigenic. Nat Genet. 17:495–497. 1997. View Article : Google Scholar : PubMed/NCBI

35 

Thompson AD, Teitell MA, Arvand A and Denny CT: Divergent Ewing’s sarcoma EWS/ETS fusions confer a common tumorigenic phenotype on NIH3T3 cells. Oncogene. 18:5506–5513. 1999.

36 

Im YH, Kim HT, Lee C, et al: EWS-FLI1, EWS-ERG, and EWS-ETV1 oncoproteins of Ewing tumor family all suppress transcription of transforming growth factor β type II receptor gene. Cancer Res. 60:1536–1540. 2000.PubMed/NCBI

37 

Hahm KB, Cho K, Lee C, et al: Repression of the gene encoding the TGF-β type II receptor is a major target of the EWS-FLI1 oncoprotein. Nat Genet. 23:222–227. 1999.

38 

Rossi S, Orvieto E, Furlanetto A, Laurino L, Ninfo V and Dei Tos AP: Utility of the immunohistochemical detection of FLI-1 expression in round cell and vascular neoplasm using a monoclonal antibody. Mod Pathol. 17:547–552. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Pereira R, Quang CT, Lesault I, Dolznig H, Beug H and Ghysdael J: FLI-1 inhibits differentiation and induces proliferation of primary erythroblasts. Oncogene. 18:1597–1608. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Grumont RJ, Fecondo J and Gerondakis S: Alternate RNA splicing of murine nfkb1 generates a nuclear isoform of the p50 precursor NF-kappa B1 that can function as a transactivator of NF-kappa B-regulated transcription. Mol Cell Biol. 14:8460–8470. 1994.PubMed/NCBI

41 

Wharry CE, Haines KM, Carroll RG and May MJ: Constitutive non-canonical NFkappaB signaling in pancreatic cancer cells. Cancer Biol Ther. 8:1567–1576. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Hiscott J, Marois J, Garoufalis J, et al: Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: evidence for a positive autoregulatory loop. Mol Cell Biol. 13:6231–6240. 1993.PubMed/NCBI

43 

Zienolddiny S, Ryberg D, Maggini V, Skaug V, Canzian F and Haugen A: Polymorphisms of the interleukin-1 β gene are associated with increased risk of non-small cell lung cancer. Int J Cancer. 109:353–356. 2004.

44 

Hu HM, Tian Q, Baer M, et al: The C/EBP bZIP domain can mediate lipopolysaccharide induction of the proinflammatory cytokines interleukin-6 and monocyte chemoattractant protein-1. J Biol Chem. 275:16373–16381. 2000. View Article : Google Scholar

45 

Yamaji H, Iizasa T, Koh E, et al: Correlation between interleukin 6 production and tumor proliferation in non-small cell lung cancer. Cancer Immunol Immunother. 53:786–792. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Xiao W, Hodge DR, Wang L, Yang X, Zhang X and Farrar WL: Co-operative functions between nuclear factors NFkappaB and CCAT/enhancer-binding protein-beta (C/EBP-beta) regulate the IL-6 promoter in autocrine human prostate cancer cells. Prostate. 61:354–370. 2004. View Article : Google Scholar

47 

Tang T, Shi Y, Opalenik SR, et al: Expression of the TAL1/SCL transcription factor in physiological and pathological vascular processes. J Pathol. 210:121–129. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Erez A, Perelman M, Hewitt SM, et al: Sil overexpression in lung cancer characterizes tumors with increased mitotic activity. Oncogene. 23:5371–5377. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Glubb DM, Cerri E, Giese A, et al: Novel functional germline variants in the vascular endothelial growth factor receptor 2 gene (KDR) and their effect on gene expression and micro-vessel density in lung cancer. Clin Cancer Res. 17:5257–5267. 2011. View Article : Google Scholar

50 

Sanmartin E, Jantus Lewintre E, Sirera R, et al: Soluble vascular endothelial growth factor receptor 2 (VEGFR2): New biomarker in advanced non-small cell lung cancer (NSCLC)? J Clin Oncol (Suppl). 27:e221082009.

51 

Tanaka A, Itoh F, Nishiyama K, et al: Inhibition of endothelial cell activation by bHLH protein E2–2 and its impairment of angiogenesis. Blood. 115:4138–4147. 2010.

52 

Tanaka A, Itoh F, Itoh S and Kato M: TAL1/SCL relieves the E2-2-mediated repression of VEGFR2 promoter activity. J Biochem. 145:129–135. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November 2012
Volume 6 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
Meng X, Lu P, Bai H, Xiao P and Fan Q: Transcriptional regulatory networks in human lung adenocarcinoma. Mol Med Rep 6: 961-966, 2012
APA
Meng, X., Lu, P., Bai, H., Xiao, P., & Fan, Q. (2012). Transcriptional regulatory networks in human lung adenocarcinoma. Molecular Medicine Reports, 6, 961-966. https://doi.org/10.3892/mmr.2012.1034
MLA
Meng, X., Lu, P., Bai, H., Xiao, P., Fan, Q."Transcriptional regulatory networks in human lung adenocarcinoma". Molecular Medicine Reports 6.5 (2012): 961-966.
Chicago
Meng, X., Lu, P., Bai, H., Xiao, P., Fan, Q."Transcriptional regulatory networks in human lung adenocarcinoma". Molecular Medicine Reports 6, no. 5 (2012): 961-966. https://doi.org/10.3892/mmr.2012.1034