Open Access

Identification of key pathways and genes in lung carcinogenesis

  • Authors:
    • Xiang Jin
    • Xingang Liu
    • Zhen Zhang
    • Yinghui Guan
    • Ren Xv
    • Jun Li
  • View Affiliations

  • Published online on: July 24, 2018     https://doi.org/10.3892/ol.2018.9203
  • Pages: 4185-4192
  • Copyright: © Jin 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 aimed to identify key pathways and genes in the pathogenesis of lung cancer. The GSE10072 dataset was downloaded from the Gene Expression Omnibus database. Protein‑protein interaction data were collected from Human Protein Reference Database, and 201 pathways were downloaded from the Kyoto Encyclopedia of Genes and Genomes database. Signaling network impact analysis was performed to identify enriched pathways, followed by the construction of a pathway‑pathway crosstalk network. Benzopyrene was used to treat normal human lung cells at concentrations of 0.01, 0.1, 1 and 10 µM, and cell viability was measured. Furthermore, growth arrest and DNA damage inducible β (GADD45B), p53, cyclin B, Akt and nuclear factor (NF)‑κB protein levels were also measured via western blotting. Impact analysis identified 11 enriched lung cancer‑associated KEGG pathways, including ‘complement and coagulation cascades’, ‘ECM‑receptor interaction’, ‘P53 signaling pathway’, ‘cell adhesion molecules’ and ‘focal adhesion’. In addition, cell cycle, ‘drug metabolism‑cytochrome P450’, ‘metabolic pathways’, ‘pathways in cancer’, ‘focal adhesion’ and ‘antigen processing and presentation’ were central in the pathway‑pathway cross‑talk network. Furthermore, the upregulated gene GADD45B was associated with three of the pathways, including an activated pathway (‘MAPK signaling pathway’) and two repressed pathways (‘cell cycle’ and ‘P53 pathway’). Western blotting demonstrated that the expression of NF‑κB, Akt and GADD45B increased over time in lung cells treated with benzopyrene, whereas the expression levels of cyclin B and P53 decreased. In conclusion, GADD45B may contribute to lung carcinogenesis via affecting the MAPK, P53 signaling and cell cycle pathways.

Introduction

Lung cancer is one of the most common types of cancer (1). Lung cancer has two main histological types: Non-small cell lung cancer (NSCLC; 80.4%) and SCLC (16.8%) (2); however, the underlying mechanisms for the development of lung cancer are not yet completely characterized.

Accumulated genetic abnormalities are associated with cancer. Somatic mutations in a number of genes, including epidermal growth factor receptor (EGFR), P53, KRAS, BRAF, Erb-B2 receptor tyrosine kinase 2 (ERBB2), MET, serine/threonine kinase 11, PIK3CA and Parkin RBR E3 ubiquitin protein ligase, have been identified in patients with lung cancer (3,4). Gene amplifications, including of EGFR, ERBB2, MET, PIK3CA and NK2 homeobox 1, have also been detected in lung cancer (5). A number of single nucleotide polymorphisms (SNPs) are associated with lung cancer susceptibility, including in interleukin-1, cytochrome P450, a 5′SNP in the ERCC excision repair 6, chromatin remodeling factor gene and SNPs in the nicotinic acetylcholine receptor gene cluster on chromosome 15q25.1 (69). Genetic abnormalities have been identified in numerous pathways, including the Notch (10), EGFR (11), PI3K (12), phosphatase and tensin homolog/phospho-Akt/P53 (13), mitogen-activated protein kinase (MAPK) (14) and cell cycle pathways.

In the past decade, there has been a pervasive application of high-throughput molecular technologies, including microarrays, in lung cancer research (1517), which has greatly enriched the knowledge of the pathogenesis of the disease, and may potentially provide markers for the prognosis and targeted therapy of lung cancer. By enrolling 105 subjects in an Environment And Genetics in Lung cancer Etiology study (http://dceg.cancer.gov/eagle), Landi et al (18) produced a microarray dataset that included 107 expression values from tumor (n=58) and non-tumor tissues (n=49) from 74 subjects (non-smokers, n=20; former smokers, n=26; current smokers, n=28). Using this microarray analysis, 122 genes were identified that were differentially expressed between the tumor and non-tumor samples. In addition, several crucial smoking-associated genes and pathways were identified in the study, and a number of these, including Nima related kinase 2 and TTK protein kinase, were experimentally validated; however, the relationship between these genes and pathways were not considered in the original study.

In the present study, based on the microarray dataset produced by Landi et al (18), GSE10072, a novel pathway-pathway crosstalk approach was employed to identify pathways and genes that may have critical roles in the pathogenesis of lung cancer, and a number of the identified genes were experimentally validated.

Materials and methods

Source of pathway and microarray data

Protein-protein interaction data were downloaded from the Human Protein Reference Database (http://www.hprd.org/), and 201 lung cancer pathways were downloaded from the Kyoto Encyclopedia of Genes and Genomes (KEGG, http://www.kegg.jp/) database (19,20) using ‘lung cancer’ as the search term.

The raw data of the gene expression profile dataset GSE10072 in the ‘.CEL’ format were downloaded from the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/).

Identification of differentially expressed genes (DEGs)

The raw downloaded data were preprocessed and normalized using the R/Bioconductor package Affy with the Robust Multichip Average method for single-channel Affymetrix chips (21). A one-way analysis of variance was applied to each probe set to identify those that significantly changed expression level over time, as previously described (22). P<0.05 was considered to indicate a statistically significant result; the raw P-value was adjusted with the Bonferroni method (23).

Impact analysis

The pathway impact analysis as described by Draghici et al (24) was adopted, which considers the statistical significance of the enrichment of KEGG pathways, while also considering other crucial factors, including the magnitude of expression change for each gene, the topology of the signaling pathway, and the interactions between signaling pathways.

Construction of pathway-pathway crosstalk network

A hyper geometric distribution framework was applied to evaluate the significance of all non-empty intersections between two pathways, as previously described (25): Fisher's exact test computed the probability, p* using hyper geometric distribution with the parameters (S, NG, N).

p*=p(X=α|S,NG,N)=(Sα)(N-SNG-α)(NNG)

Where α was the number of DEGs in the pathway intersection; S, the number of DEGs in the pathway union; NG, the number of genes in the pathway intersection; and N, the number of genes in the pathway union.

The P-value to reject the null hypothesis with a probability of <p* was calculated using the sum of the probabilities with the same marginal totals, i.e.:

p=∑(Si)(N-sNG-i)(NNG)

This procedure gave a two-tailed probability for a Fisher's exact test. P<0.05 was considered to indicate a statistically significant result, indicative of the association of two pathways.

Analysis of cross-talk between pathways

The state of a pathway was initially determined. A pathway was considered to be activated when it met the following criteria: i) Number of DEGs in the pathway >10; ii) Q-value [(number of upregulated DEGs in the pathway-number of downregulated DEGs in the pathway)/total number of genes in the pathway] >0.5. A repressed pathway met the two criteria: i) Number of DEGs in this pathway >10; ii) Q-value <-0.5. Next, pairs of pathways sharing common DEGs were identified, and these DEGs were listed.

Cell culture and treatment

Normal lung K562 cells were purchased from the American Type Culture Collection (Manassas, VA, USA). The cells were grown in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% heat-inactivated fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 100 U/ml penicillin and streptomycin, in a humidified atmosphere with 5% CO2 at 37°C. To maintain drug resistance, adriamycin was supplemented at regular intervals for 2 weeks prior to any experiment. Benzopyrene was used to treat the cells at concentrations of 0.01, 0.1, 1 and 10 µM in the subsequent assays.

Measurement of cell viability

Cell proliferation was measured using an MTT assay. Cells (2×105 cells/ml) were seeded in 96-well plates with increasing concentrations of adriamycin, SNX-2112 and 17-AAG. After incubation at 37°C for 24, 48 and 72 h, 5 mg/ml MTT solution was added for incubation for 4 h. Then, 100 µl/well DMSO was added to solubilize the formazan crystals. Cell viability was assessed by measuring absorbance at 570 nm using a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Western blot analysis

Cells were first lysed in lysis buffer (Sangon Biotech Co., Ltd., Shanghai, China) and then total protein was extracted. Followed by protein concentration was measured by the bicinchoninic acid method. Equal aliquots (20 µg protein per lane) of protein lysate were separated by 8–12% SDS-PAGE and transferred to a polyvinylidene difluoride membrane. Subsequent to blocking with 5% skimmed milk at 37°C for 1 h, Western blots were probed with primary antibodies against nuclear factor (NF)-κB (1:1,000; cat no. ab32360; Abcam, Cambridge, MA, USA), Akt (1:1,000; cat no. ab126811; Abcam), cyclin B (1:1,000; cat no. ab18221; Abcam), P53 (1:1,000; cat no. ab21985; Abcam), growth arrest and DNA damage inducible β (GADD45B) (1:1,000; cat no. ab128920; Abcam) and β-actin (1:1,000; cat no. ab6276; Abcam) overnight at 4°C. Three consecutive washes were performed for 10 min in PBS-Tween, followed by incubation with the alkaline phosphatase-conjugated goat anti-rabbit IgG secondary antibody (1:5,000; cat no. ab6722; Abcam) diluted in 5% skimmed milk at room temperature for 1 h. The immunoblots were visualized with enhanced chemiluminescence (GE Healthcare, Chicago, IL, USA) and autoradiography. The experiments were repeated three times, and the results were detected using the Image Lab software (version 4.1; Bio-Rad Laboratories, Inc.) on ChemiDoc MP imaging system (Bio-Rad Laboratories, Inc.).

Statistical analysis

All data were presented as the mean ± standard deviation. Statistical analysis was performed by one-way analysis of variance with a Bonferroni post hoc test in SPSS software (version 13.0; SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of DEGs

Using a threshold of P<0.05, the genes with significantly differential expression were analyzed. A total of 1,763 DEGs were identified, of which 662 were associated with KEGG pathways.

Result of impact analysis

A total of 11 pathways were identified in the pathway impact analysis. Of these, ‘complement and coagulation cascades’, ‘ECM-receptor interaction’, ‘P53 signaling pathway’, ‘cell adhesion molecules (CAMs)’, ‘focal adhesion’ and ‘cell cycle’ were the top five pathways by impact factor value (Table I).

Table I.

Significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways in GSE10072.

Table I.

Significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways in GSE10072.

PathwayImpact factorP-value
Complement and coagulation cascades17.258 2.08×10−7
ECM-receptor interaction14.266 3.90×10−6
P53 signaling pathway9.017 3.41×10−4
Cell adhesion molecules70.734 4.42×10−4
Focal adhesion9.283 1.17×10−3
Cell cycle6.673 3.36×10−3
Renin-angiotensin system5.778 8.30×10−3
PPAR signaling pathway5.733 1.54×10−2
TGF-beta signaling pathway6.352 2.24×10−2
Leukocyte transendothelial migration114.039 2.34×10−2
migration Tight junction6.133 4.85×10−2
Analysis of cross-talk between pathways

It was determined that the ‘cell cycle’, ‘drug metabolism-cytochrome P450’, ‘metabolic pathways’, ‘pathways in cancer’, ‘focal adhesion’ and ‘antigen processing and presentation’ KEGG pathways were central in the pathway-pathway crosstalk network (Fig. 1).

Using the criteria defined for activated and repressed pathways, 2 pathways (‘cell cycle’ and ‘P53 signaling pathway’) were repressed, and 10 pathways were activated (Table II). The common DEGs between the pathways are listed in Table III. GADD45B was associated with three enriched pathways, including the activated ‘MAPK signaling pathway’ and the repressed ‘cell cycle’ and ‘P53 signaling pathway’ terms (Fig. 2).

Table II.

Status of the Kyoto Encyclopedia of Genes and Genomes pathways associated with the differentially expressed genes in lung cancer.

Table II.

Status of the Kyoto Encyclopedia of Genes and Genomes pathways associated with the differentially expressed genes in lung cancer.

DEGs

PathwayUpregulatedDownregulatedBothQ-value
Cell cycle21113−0.6923
P53 signaling pathway2810−0.6000
Cytokine-cytokine receptor interaction133160.6250
Tight junction92110.6364
Neuroactive ligand-receptor interaction153180.6667
Chemokine signaling pathway112130.6923
Complement and coagulation cascades142160.7500
Axon guidance91100.8000
Vascular smooth muscle contraction121130.8462
MAPK signaling pathway131140.8571
Regulation of actin cytoskeleton110111.0000
Endocytosis100101.0000

Table III.

Differentially expressed genes associated with multiple enriched Kyoto Encyclopedia of Genes and Genomes pathways.

Table III.

Differentially expressed genes associated with multiple enriched Kyoto Encyclopedia of Genes and Genomes pathways.

Pathway 1Pathway 2Gene IDs
Cytokine-cytokine receptor interactionChemokine signaling pathway6359, 1524, 2921, 2920, 6387
Neuroactive ligand-receptor interactionVascular smooth muscle contraction1906, 10203, 1909, 185
MAPK signaling pathwayRegulation of actin cytoskeleton2264, 6237, 2263
MAPK signaling pathwayEndocytosis2264, 7048, 2263
Regulation of actin cytoskeletonEndocytosis2264, 2263, 8395
Tight junctionRegulation of actin cytoskeleton10398, 6237, 4628
Cytokine-cytokine receptor interactionNeuroactive ligand-receptor interaction3953, 2690
Cytokine-cytokine receptor interactionChemokine signaling pathway10563, 9547
Tight junctionVascular smooth muscle contraction10398, 4629
Tight junctionMAPK signaling pathway6237, 10000
Vascular smooth muscle contractionRegulation of actin cytoskeleton10398, 4638
Cell cycleP53 signaling pathway4616
Cell cycleMAPK signaling pathway4616
Chemokine signaling pathwayAxon guidance6387
Chemokine signaling pathwayVascular smooth muscle contraction115
Chemokine signaling pathwayMAPK signaling pathway10000
Chemokine signaling pathwayRegulation of actin cytoskeleton5295
Chemokine signaling pathwayEndocytosis2869
Cytokine-cytokine receptor interactionAxon guidance6387
Cytokine-cytokine receptor interactionMAPK signaling pathway7048
Cytokine-cytokine receptor interactionEndocytosis7048
Neuroactive ligand-receptor interactionComplement and coagulation cascades728
Neuroactive ligand-receptor interactionEndocytosis154
P53 signaling pathwayMAPK signaling pathway4616
Tight junctionChemokine signaling pathway10000
Vascular smooth muscle contractionMAPK signaling pathway5319
Vascular smooth muscle contractionMAPK signaling pathway5321
Measurement of cell viability and western blot analysis

Benzopyrene treatment reduced the viability of lung cells at all concentrations from 72 h (Fig. 3). GADD45B, P53, cyclin, Akt and NF-κB protein levels were detected by Western blotting (Fig. 4). Western blotting demonstrated that the expression of NF-κB, Akt and GADD45B increased over time in lung cells treated with benzopyrene, whereas the expression of cyclin B and P53 was decreased (Fig. 5).

Discussion

In the present study, significantly enriched pathways were identified with impact analysis, and a pathway-pathway crosstalk network was constructed. GADD45B was identified as a connection between a number of the enriched pathways, and experimental validation of the expression of this gene and associated pathways was performed.

In comparison to classical pathway analysis, impact analysis also considers the over-representation of DEGs in a given pathway and the abnormal perturbation of that pathway, as measured by the expression changes across the pathway topology (26). Previously, the roles of the P53 signaling pathway, cell adhesion, focal adhesion and the cell cycle in the pathogenesis of lung cancer have been confirmed and studied extensively (27,28). In contrast, the complement and coagulation cascades in this disease have been less reported. Corrales et al (29), reported the activation of the complement system by detecting the anaphylatoxin C5a, a potent immune mediator generated subsequent to complement activation in lung cancer cell lines. Plasminogen activator inhibitor (PAI) variants PAI-1 A15T and PAI-2 S413C influence the prognosis of patients with lung cancer (30), and PAI-1 has been demonstrated to inhibit the activation of the coagulation system (31). Levels of the erythrocyte complement receptor 1 were significantly lower in patients with small cell lung cancer (32). A previous study revealed that activated coagulation factor X inhibited the migration of lung cancer cells and may serve a key role in cell migration (33). Thus, it was indicated that the complement and coagulation cascades may also serve an important role in the pathogenesis of lung cancer.

The pathway-pathway cross-talk network indicated the central roles of ‘cell cycle’, ‘drug metabolism-cytochrome P450’, ‘metabolic pathways’, ‘pathways in cancer’, ‘focal adhesion’ and ‘antigen processing and presentation’; metabolism-associated pathways may therefore be particularly important in the pathogenesis of lung cancer. Tumor cells sustain high rates of glycolysis even in aerobic conditions to maintain their rapid growth; alterations in in primary metabolites (34) and glycolysis-associated enzymes, including hexokinase II and glyceraldehyde-3-phosphate dehydrogenase (35,36), have been identified in lung cancer. Additionally, the alteration of metabolic pathways may affect the efficacy of anti-tumor drugs, e.g. the glutathione metabolic pathway, which is involved in the detoxification or inactivation of platinum drugs (37). The cytochromes P450 are a group of enzymes that catalyze the oxidative biotransformation of the majority of drugs and other lipophilic xenobiotics (38).

In the present study, GADD45B, an upregulated gene, was determined to be a common link between three KEGG pathways, including the upregulated ‘MAPK signaling pathway’ and the repressed ‘cell cycle’ and ‘P53 signaling pathway’. It was therefore considered to potentially have an important role in the pathogenesis of lung cancer. GADD45B, encoding MyD118, is part of a highly conserved Gadd45 gene family with GADD45A, encoding GADD45, and GADD45G, encoding CR6. Gadd45 proteins have been implicated for their involvement in tumorigenesis (39) and age-associated pathologies (40); however, MyD118, GADD45 and CR6 are considered to have similar but not identical functions via different apoptotic and growth suppressive pathways (41). The MAPK signaling pathway has been confirmed to have an important role in the pathogenesis of lung cancer (42). In the present study, this signaling pathway was activated in patients with lung cancer. Gupta et al (43) demonstrated that GADD45B promotes cell survival via activation of the GADD45a-P38-NF-κB pathway and inhibition of the MAPK kinase 4-c-Jun N-terminal kinases (JNK) pathway. P38 and JNK are members of the MAPK family (44). TGF-β can activate Smad by phosphorylation (45). One previous study demonstrates that following the induction by TGF-β, Smad transcription factors activate P38 via GADD45B (46). Another study demonstrates that SMAD3 and SMAD4 can activate GADD45B following the induction by TGF-β (47). GADD45B and GADD45G are cyclin-dependent kinase 2/cyclin B1 kinase inhibitors, and thus function in in G2/M cell cycle arrest (48). This is in accord with the observation in the present study that the cell cycle pathway was repressed.

It was also identified in the present study that the P53 signaling pathway was repressed. Previously, Mi et al (49) observed the upregulation of GADD45B and the downregulation of P53 in human prostate cancer cells exposed to silvestrol. This signaling pathway has been reported to correlate with the radioresponse of NSCLC (13). GADD45A is a target gene of P53 (50). Lambert et al (51) reported the upregulation of GADD45B mRNA in Saos-2-His273 cells exposed to proline rich membrane anchor 1, a P53-reactivating agent.

Benzopyrene is a carcinogen particularly associated with lung cancer (52); therefore, the normal lung cells were treated with benzopyrene in the present study to investigate the process of lung carcinogenesis. The harmful effects of benzopyrene against cell viability were observed by an MTT assay at various concentrations. Western blotting demonstrated that the expression of NF-κB, Akt and GADD45B were increased over time in lung cells treated with benzopyrene, whereas the expression of cyclin B and P53 were decreased. This corresponded with the data that the AKT-MAPK pathway was activated via NF-κB, while the P53 pathway and cell cycle pathway were repressed; thus, GADD45B may contribute to lung carcinogenesis via activating the MAPK signaling pathway and repressing the P53 signaling and cell cycle pathways.

Although a series of comprehensive bioinformatics analyses and validation experiments were performed, there were two major limitations in the present study: i) Only one lung cell line was used in the current study, which may not eliminate the heterogeneity of lung cancer; ii) knockdown of GADD45B was not performed, and the expressions of other proteins after GADD45B knockdown were not considered, which may weaken the regulatory associations between them.

In addition to ‘ECM-receptor interaction’, ‘P53 signaling pathway’, ‘cell adhesion molecules (CAMs)’, ‘focal adhesion’ and ‘cell cycle’, it was also identified that the ‘complement and coagulation cascades’ pathway may be associated with the pathogenesis of lung cancer; therefore, it was speculated that GADD45B may contribute to lung carcinogenesis via activating the MAPK signaling pathway, and repressing the P53 signaling and cell cycle pathways. Thus, the role of this gene in lung cancer should be studied further; GADD45B siRNA knockdown experiments will assist the further validation of this speculation.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and material

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

Authors' contributions

XJ and JL were involved in the conception and design of the research and drafting the manuscript. ZZ participated in the acquisition of data. RX performed the analysis and interpretation of data. YG was involved in the statistical analysis, XL participated in the design of the study and performed the statistical analysis. All authors read and approved the final 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 

McGuire S: World cancer report 2014. Geneva, switzerland: World health organization, international agency for research on cancer, WHO Press, 2015. Adv Nutr. 7:418–419. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Travis WD, Travis LB and Devesa SS: Lung cancer. Cancer. 75 Suppl 1:S191–S202. 1995. View Article : Google Scholar

3 

Ahrendt SA, Decker PA, Alawi EA, Zhu Yr YR, Sanchez-Cespedes M, Yang SC, Haasler GB, Kajdacsy-Balla A, Demeure MJ and Sidransky D: Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer. 92:1525–1530. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Naoki K, Chen TH, Richards WG, Sugarbaker DJ and Meyerson M: Missense mutations of the BRAF gene in human lung adenocarcinoma. Cancer Res. 62:7001–7003. 2002.PubMed/NCBI

5 

Weir BA, Woo MS, Getz G, Perner S, Ding L, Beroukhim R, Lin WM, Province MA, Kraja A, Johnson LA, et al: Characterizing the cancer genome in lung adenocarcinoma. Nature. 450:893–898. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Engels EA, Wu X, Gu J, Dong Q, Liu J and Spitz MR: Systematic evaluation of genetic variants in the inflammation pathway and risk of lung cancer. Cancer Res. 67:6520–6527. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Wenzlaff AS, Cote ML, Bock CH, Land SJ, Santer SK, Schwartz DR and Schwartz AG: CYP1A1 and CYP1B1 polymorphisms and risk of lung cancer among never smokers: A population-based study. Carcinogenesis. 26:2207–2212. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Lin Z, Zhang X, Tuo J, Guo Y, Green B, Chan CC, Tan W, Huang Y, Ling W, Kadlubar FF, et al: A variant of the Cockayne syndrome B gene ERCC6 confers risk of lung cancer. Hum Mutat. 29:113–122. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Amos CI, Wu X, Broderick P, Gorlov IP, Gu J, Eisen T, Dong Q, Zhang Q, Gu X, Vijayakrishnan J, et al: Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nat Genet. 40:616–622. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Westhoff B, Colaluca IN, D'Ario G, Donzelli M, Tosoni D, Volorio S, Pelosi G, Spaggiari L, Mazzarol G, Viale G, et al: Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci USA. 106:pp. 22293–22298. 2009; View Article : Google Scholar : PubMed/NCBI

11 

Leidner RS, Fu P, Clifford B, Hamdan A, Jin C, Eisenberg R, Boggon TJ, Skokan M, Franklin WA, Cappuzzo F, et al: Genetic abnormalities of the EGFR pathway in African American Patients with non-small-cell lung cancer. J Clin Oncol. 27:5620–5626. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Gustafson AM, Soldi R, Anderlind C, Scholand MB, Qian J, Zhang X, Cooper K, Walker D, McWilliams A, Liu G, et al: Airway PI3K pathway activation is an early and reversible event in lung cancer development. Sci Transl Med. 2:26ra252010. View Article : Google Scholar : PubMed/NCBI

13 

Jung IL, Kang HJ, Kim KC and Kim IG: PTEN/pAkt/p53 signaling pathway correlates with the radioresponse of non-small cell lung cancer. Int J Mol Med. 25:517–523. 2010.PubMed/NCBI

14 

Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, et al: Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 455:1069–1075. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Bremnes RM, Veve R, Gabrielson E, Hirsch FR, Baron A, Bemis L, Gemmill RM, Drabkin HA and Franklin WA: High-throughput tissue microarray analysis used to evaluate biology and prognostic significance of the E-cadherin pathway in non-small-cell lung cancer. J Clin Oncol. 20:2417–2428. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T, et al: Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 9:189–198. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Liu XG, Zhu WY, Huang YY, Ma LN, Zhou SQ, Wang YK, Zeng F, Zhou JH and Zhang YK: High expression of serum miR-21 and tumor miR-200c associated with poor prognosis in patients with lung cancer. Med Oncol. 29:618–626. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Landi MT, Dracheva T, Rotunno M, Figueroa JD, Liu H, Dasgupta A, Mann FE, Fukuoka J, Hames M, Bergen AW, et al: Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One. 3:e16512008. View Article : Google Scholar : PubMed/NCBI

19 

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 : PubMed/NCBI

20 

Joshi-Tope G, Gillespie M, Vastrik I, D'Eustachio P, Schmidt E, de Bono B, Jassal B, Gopinath GR, Wu GR, Matthews L, et al: Reactome: A knowledgebase of biological pathways. Nucleic Acids Res. 33:D428–D432. 2005. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Górecki T and Smaga Ł: A comparison of tests for the one-way ANOVA problem for functional data. Computational Statistics. 30:987–1010. 2015. View Article : Google Scholar

23 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: A practical and powerful approach to multiple testing. J Royal Statistical Society. Series B (Methodological). 1–300. 1995.

24 

Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, Georgescu C and Romero R: A systems biology approach for pathway level analysis. Genome Res. 17:1537–1545. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Francesconi M, Remondini D, Neretti N, Sedivy JM, Cooper LN, Verondini E, Milanesi L and Castellani G: Reconstructing networks of pathways via significance analysis of their intersections. BMC Bioinformatics. 9 Suppl 4:S92008. View Article : Google Scholar : PubMed/NCBI

26 

Tarca AL, Draghici S, Khatri P, Hassan SS, Mittal P, Kim JS, Kim CJ, Kusanovic JP and Romero R: A novel signaling pathway impact analysis. Bioinformatics. 25:75–82. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Zhong G, Chen X, Fang X, Wang D, Xie M and Chen Q: Fra-1 is upregulated in lung cancer tissues and inhibits the apoptosis of lung cancer cells by the P53 signaling pathway. Oncol Rep. 35:447–453. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Havel LS, Kline ER, Salgueiro AM and Marcus AI: Vimentin regulates lung cancer cell adhesion through a VAV2-Rac1 pathway to control focal adhesion kinase activity. Oncogene. 34:1979–1990. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Corrales L, Ajona D, Rafail S, Lasarte JJ, Riezu-Boj JI, Lambris JD, Rouzaut A, Pajares MJ, Montuenga LM and Pio R: Anaphylatoxin C5a creates a favorable microenvironment for lung cancer progression. J Immunol. 189:4674–4683. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Pappot H, Pedersen AN, Brünner N and Christensen IJ: The complex between urokinase (uPA) and its type-1 inhibitor (PAI-1) in pulmonary adenocarcinoma: Relation to prognosis. Lung Cancer. 51:193–200. 2006. View Article : Google Scholar : PubMed/NCBI

31 

McVey JH: Tissue factor pathway. Baillieres Best Pract Res Clin Haematol. 12:361–372. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Currie MS, Vala M, Pisetsky DS, Greenberg CS, Crawford J and Cohen HJ: Correlation between erythrocyte CR1 reduction and other blood proteinase markers in patients with malignant and inflammatory disorders. Blood. 75:1699–1704. 1990.PubMed/NCBI

33 

Borensztajn K, Peppelenbosch MP and Spek CA: Coagulation Factor Xa inhibits cancer cell migration via LIMK1-mediated cofilin inactivation. Thromb Res. 125:e323–e328. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Fan TW, Lane AN, Higashi RM, Farag MA, Gao H, Bousamra M and Miller DM: Altered regulation of metabolic pathways in human lung cancer discerned by (13)C stable isotope-resolved metabolomics (SIRM). Mol Cancer. 8:412009. View Article : Google Scholar : PubMed/NCBI

35 

Koukourakis MI, Giatromanolaki A and Sivridis E; Tumour and Angiogenesis Research Group, : Lactate dehydrogenase isoenzymes 1 and 5: Differential expression by neoplastic and stromal cells in non-small cell lung cancer and other epithelial malignant tumors. Tumor Biol. 24:199–202. 2003. View Article : Google Scholar

36 

Koukourakis MI, Giatromanolaki A, Sivridis E, Gatter KC and Harris AL; Tumor and Angiogenesis Research Group, : Pyruvate dehydrogenase and pyruvate dehydrogenase kinase expression in non small cell lung cancer and tumor-associated stroma. Neoplasia. 7:1–6. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Yang P, Ebbert JO, Sun Z and Weinshilboum RM: Role of the glutathione metabolic pathway in lung cancer treatment and prognosis: A review. J Clin Oncol. 24:1761–1769. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Zanger UM, Turpeinen M, Klein K and Schwab M: Functional pharmacogenetics/genomics of human cytochromes P450 involved in drug biotransformation. Anal Bioanal Chem. 392:1093–1108. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Tamura RE, de Vasconcellos JF, Sarkar D, Libermann TA, Fisher PB and Zerbini LF: GADD45 proteins: Central players in tumorigenesis. Curr Mol Med. 12:634–651. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Moskalev AA, Smit-McBride Z, Shaposhnikov MV, Plyusnina EN, Zhavoronkov A, Budovsky A, Tacutu R and Fraifeld VE: Gadd45 proteins: Relevance to aging, longevity and age-related pathologies. Ageing Res Rev. 11:51–66. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Azam N, Vairapandi M, Zhang W, Hoffman B and Liebermann DA: Interaction of CR6 (GADD45gamma) with proliferating cell nuclear antigen impedes negative growth control. J Biol Chem. 276:2766–2774. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Hosokawa S, Toyooka S, Fujiwara Y, Tokumo M, Soh J, Takigawa N, Hotta K, Yoshino T, Date H, Tanimoto M and Kiura K: Comprehensive analysis of EGFR signaling pathways in Japanese patients with non-small cell lung cancer. Lung Cancer. 66:107–113. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Gupta M, Gupta SK, Hoffman B and Liebermann DA: Gadd45a and Gadd45b protect hematopoietic cells from UV-induced apoptosis via distinct signaling pathways, including p38 activation and JNK inhibition. J Biol Chem. 281:17552–17558. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Takekawa M, Tatebayashi K, Itoh F, Adachi M, Imai K and Saito H: Smad-dependent GADD45beta expression mediates delayed activation of p38 MAP kinase by TGF-beta. EMBO J. 21:6473–6482. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Macias MJ, Martin-Malpartida P and Massagué J: Structural determinants of Smad function in TGF-β signaling. Trends Biochem Sci. 40:296–308. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Howley BV, Hussey GS, Link LA and Howe PH: Translational regulation of Inhibin βA by TGFβ via the RNA-binding protein hnRNP E1 enhances the invasiveness of epithelial-to-mesenchymal transitioned cells. Oncogene. 35:1725–1735. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Major MB and Jones DA: Identification of a gadd45beta 3′enhancer that mediates SMAD3- and SMAD4-dependent transcriptional induction by transforming growth factor beta. J Biol Chem. 279:5278–5287. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Vairapandi M, Balliet AG, Hoffman B and Liebermann DA: GADD45b and GADD45g are cdc2/cyclinB1 kinase inhibitors with a role in S and G2/M cell cycle checkpoints induced by genotoxic stress. J Cell Physiol. 192:327–338. 2002. View Article : Google Scholar : PubMed/NCBI

49 

Mi Q, Kim S, Hwang BY, Su BN, Chai H, Arbieva ZH, Kinghorn AD and Swanson SM: Silvestrol regulates G2/M checkpoint genes independent of p53 activity. Anticancer Res. 26:3349–3356. 2006.PubMed/NCBI

50 

Zhan Q: Gadd45a, a p53-and BRCA1-regulated stress protein, in cellular response to DNA damage. Mutat Res. 569:133–143. 2005. View Article : Google Scholar : PubMed/NCBI

51 

Lambert JM, Moshfegh A, Hainaut P, Wiman KG and Bykov VJ: Mutant p53 reactivation by PRIMA-1MET induces multiple signaling pathways converging on apoptosis. Oncogene. 29:1329–1338. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Denissenko MF, Pao A, Tang M and Pfeifer GP: Preferential formation of benzo[a]pyrene adducts at lung cancer mutational hotspots in p53. Science. 274:430–432. 1996. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 16 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jin X, Liu X, Zhang Z, Guan Y, Xv R and Li J: Identification of key pathways and genes in lung carcinogenesis. Oncol Lett 16: 4185-4192, 2018
APA
Jin, X., Liu, X., Zhang, Z., Guan, Y., Xv, R., & Li, J. (2018). Identification of key pathways and genes in lung carcinogenesis. Oncology Letters, 16, 4185-4192. https://doi.org/10.3892/ol.2018.9203
MLA
Jin, X., Liu, X., Zhang, Z., Guan, Y., Xv, R., Li, J."Identification of key pathways and genes in lung carcinogenesis". Oncology Letters 16.4 (2018): 4185-4192.
Chicago
Jin, X., Liu, X., Zhang, Z., Guan, Y., Xv, R., Li, J."Identification of key pathways and genes in lung carcinogenesis". Oncology Letters 16, no. 4 (2018): 4185-4192. https://doi.org/10.3892/ol.2018.9203