Open Access

Differences in the early stage gene expression profiles of lung adenocarcinoma and lung squamous cell carcinoma

  • Authors:
    • Nitin Venugopal
    • Justin Yeh
    • Sai Karthik Kodeboyina
    • Tae Jin Lee
    • Shruti Sharma
    • Nikhil Patel
    • Ashok Sharma
  • View Affiliations

  • Published online on: October 21, 2019     https://doi.org/10.3892/ol.2019.11013
  • Pages: 6572-6582
  • Copyright: © Venugopal 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 discovery of lung carcinoma subtype‑specific gene expression changes has the potential to elucidate the molecular differences and provide personalized therapeutic targets for these pathologies. The aim of the present study was to characterize the genetic profiles of the early stages (IA/IB) of two non‑small cell lung cancer subtypes, adenocarcinoma (AD) and squamous cell carcinoma (SC). RNA‑Seq gene expression data from The Cancer Genome Atlas was analyzed to compare the gene expression differences between AD and SC. The gene sets specific to each subtype were further analyzed to identify the enriched Gene Ontology terms, Kyoto Encyclopedia of Genes and Genomes pathways and biological functions. The results demonstrated that a unique set of genes (145 upregulated and 27 downregulated) was altered in AD, but not in SC; another set of genes (146 upregulated and 103 downregulated) was significantly altered in SC, but not in AD. Genes highly upregulated specifically in AD included albumin (1,732‑fold), protein lin‑28 homolog A, which is a positive regulator of cyclin‑dependent kinase 2 (150‑fold) and gastric lipase (81‑fold). Genes highly upregulated specifically in SC included amelotin (618‑fold), alcohol dehydrogenase 7 (57‑fold), aclerosteosis (55‑fold) and claudin‑22 (54‑fold). Several cancer/testis antigen family genes were notably upregulated in SC, but not in AD, whereas mucins were upregulated only in AD. Functional pathway analysis demonstrated that the dysregulation of genes associated with retinoid X receptors was common in AD and SC, genes associated with ‘lipid metabolism’ and ‘drug metabolism’ were dysregulated only in SC, whereas genes associated with ‘molecular transport’ and ‘cellular growth and proliferation’ were significantly enriched in AD specifically. These results reveal fundamental differences in the gene expression profiles of early‑stage AD and SC. In addition, the present study identified molecular pathways that are uniquely associated with the pathogenesis of these subtypes.

Introduction

Non-small cell lung cancers (NSCLC), which are classified into adenocarcinomas (AD) and squamous cell carcinomas (SC), account for ~85% of primary lung cancer cases and are responsible for ~25% of cancer deaths in the United States (14). Previous studies have identified key differences between these histological subtypes at the molecular level, and have demonstrated the potential of these differences as diagnostic biomarkers and predictors of overall survival (57). For example, the mammary serine protease inhibitor maspin has been demonstrated to be highly expressed in SC, but not in AD (5). In addition, thyroid transcription factor 1 has been effectively used as an immunohistochemical marker to differentiate AD from SC (7). Several studies have examined gene expression profiles in lung cancer, including studies differentiating AD and SC (812). Shi et al (9) identified 2,961 microRNA sequences that may regulate differentially expressed genes (DEGs) in both NSCLC and small cell lung cancer across all clinical stages. Lu et al (10) studied DEGs in NSCLC subtypes across all stages, identifying a set of upregulated and downregulated genes in AD and SC but had a limited sample size. A total of 1,127 DEGs in NSCLC were identified by Grigoroiu et al (12), however they focused specially on stage IIIA disease and did not differentiate between AD and SC. Thus, the number of studies focusing on gene expression profiles specifically at the early stages (IA and IB) of NSCLC is low. Therefore, the present study aimed to provide a unique perspective by identifying gene expression changes specific to the early stages of AD and SC. Gene expression profiling of early-stage lung cancer may have great value in identifying potential molecular targets for the early detection and treatment of NSCLC.

The 5-year survival rate of patients with NSCLC who start treatment during stage IA of the disease is ~92%; however, the 5-year survival rate is 60% for stage IIA, 36% for stage IIIA and <10% for stage IVA (13). Thus, diagnosis and treatment at the early stages are crucial for improving the survival rates of patients with NSCLC. Genomic profiles of early-stage NSCLC may be particularly advantageous with the advent of next generation sequencing panels that allow rapid identification of personalized therapies for cancer by analyzing genetic variants in tissue biopsies (14). This technology has been demonstrated to provide clinical benefits in NSCLC and is routinely used to identify common mutations in lung cancer, such as KRAS and epidermal growth factor receptor (15,16). The identification of novel genes and pathways uniquely expressed in early stages of AD and SC may provide more specific elements for evolving personalized therapies, such as specific drug targets or as a component of a panel for a prognostic screening test.

The aim of the present study was to identify the unique signatures of SC and AD, by comparing the gene expression levels in each carcinoma to fully characterize the genetic profiles of each subtype. These unique gene sets may improve our understanding of the molecular basis of each NSCLC subtype and may provide more specific targets for personalized therapy.

Materials and methods

The cancer genome atlas (TCGA) datasets

TCGA (https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga) is a landmark dataset, which comprises the molecular characterization of over 20,000 samples spanning 33 different cancer types, publicly available to the research community. TCGA gene expression RNA-Seq data was downloaded from Xenabrowser (http://xenabrowser.net). Data for the early stages (IA and IB) (AJCC 7th Edition TNM Staging System; http://cancerstaging.org) of AD and SC, as well as those for adjacent normal tissues, were selected. Gene expression levels were compared between the cancer and normal lung tissue samples to identify DEGs in each subtype.

Statistical analysis

All statistical analyses were performed using the R language and environment for statistical computing (R version 3.2.2; R Foundation for Statistical Computing; www.r-project.org). The edgeR package (https://bioconductor.org/packages/release/bioc/html/edgeR.html) was used to perform differential expression analysis of all genes with count per million (CPM)>1 in at ≥2 samples, and two separate differential gene expression analyses were performed for each cancer type (AD and SC). To identify DEGs in each subtype, the gene expression data for the early stages of each carcinoma were compared with those for the adjacent normal tissues. The Benjamini and Hochberg's method (17) was used to control the false discovery rate. The level of change in gene expression was expressed as the mean fold-change (FC) between the cancerous and adjacent normal tissues. To identify highly significantly upregulated genes, a filter of |FC|≥ 4 and adjusted P<0.001 was used. The FCs of downregulated genes were transformed with a negative reciprocal, as the negative reciprocal FC of a downregulated gene has a negative sign, but retains the fold difference information, which is similar to logFC. For example, FC of 0.5 is the same as 2-fold downregulation (−2 fold-change). To identify the genes uniquely differentially expressed in either subtype, DEGs were assigned to the following categories: i) Genes upregulated in AD (FCAD>4; PAD<0.001), but not in SC; ii) genes upregulated in SC (FCSC>4; PSC<0.001), but not in AD (Table I); iii) genes downregulated in AD (FCAD<-4; PAD<0.001), but not in SC; and iv) genes downregulated in SC (FCSC<-4; PAD<0.001), but not in AD (Table II).

Table I.

Upregulated genes in AD and SC.

Table I.

Upregulated genes in AD and SC.

A, Genes upregulated specifically in AD

GeneGene nameAD FCAD P-valueSC FCSC P-value
ALBAlbumin1,732.04 8.37×10−21a−1.480.205
LIN28AProtein lin-28 homolog A150.39 1.18×10−8a1.250.482
LIPFLipase, gastric81.24 5.48×10−7a2.690.057
TM4SF4Transmembrane 4 l six family member 468.74 4.11×10−18a−2.73 <0.001a
AGXT2L1Alanine-glyoxylate aminotransferase 2-like 166.16 4.11×10−11a−1.890.087
ACMSD Aminocarboxymuconate semialdehyde decarboxylase50.76 1.05×10−17a1.600.314
PLUNCPalate, lung and nasal epithelium carcinoma associated38.01 4.59×10−13a1.570.299
CDHR2Cadherin-related family member 234.47 9.39×10−11a1.490.135
CNGA3Cyclic nucleotide gated channel α 333.38 3.92×10−21a−1.630.020
SLC14A2Solute carrier family 14 (urea transporter), member 232.14 8.58×10−14a−1.020.894
TMEM229ATransmembrane protein 229A26.86 1.86×10−12a−1.470.125
GLTPD2Glycolipid transfer protein domain containing 2.25.59 4.10×10−13a1.230.671
COL25A1Collagen, type xxv, α 124.22 2.69×10−15a1.080.797
GKN1Gastrokine 123.84 4.54×10−5a−2.85 <0.001a
NPTX1Neuronal pentraxin I21.88 3.19×10−18a1.170.448
FAM177BFamily with sequence similarity 177, member b20.74 1.13×10−21a1.090.797
LOC643763Hypothetical protein loc64376319.57 6.23×10−12a1.920.077
LOC145837Hypothetical protein loc14583719.20 1.96×10−20a1.470.128
ZP2Zona pellucida glycoprotein 2 (sperm receptor)18.62 8.39×10−6a−2.63 <0.001a
TFPI2Tissue factor pathway inhibitor 217.86 2.40×10−17a−1.010.930
LGALS4Lectin, galactoside-binding, soluble, 4 (galectin 4)17.76 6.23×10−12a−2.71 <0.001a
KNG1Kininogen 117.56 1.99×10−7a1.830.097
TTRTransthyretin (prealbumin, amyloidosis type I)17.14 3.31×10−7a2.860.054
PHGR1Proline, histidine, and glysine-rich protein 117.13 4.30×10−6a2.380.076
MUC21Mucin 21, cell surfaceassociated14.87 1.96×10−20a−2.79 <0.001a
MUC5BMucin 5b, oligomeric mucus/gel-forming14.01 4.66×10−14a−1.130.640
FGAFibrinogen α chain12.68 2.45×10−10a−1.920.020
UPK3AUroplakin 3a12.53 2.47×10−20a1.280.239
SLC1A7Solute carrier family 1 (glutamate transporter), member 712.38 1.11×10−14a−3.80 <0.001a
RGS7Regulator of g-protein signaling 712.38 6.30×10−12a1.560.208

B, Genes upregulated specifically in SC

GeneGene nameAD FCAD P-valueSC FCSC P-value

AMTNAmelotin1.700.137618.16 4.66×10−25a
ADH7Alcohol dehydrogenase 7 (class iv), mu or sigma polypeptide1.520.27757.12 3.30×10−23a
SOSTSclerostin−1.810.02754.92 3.90×10−18a
CLDN22Claudin 222.460.05954.24 5.77×10−6a
SOX10SRY (sex determining region y)-box 101.760.05039.80 1.92×10−10a
C12orf54Chromosome 12 open reading frame 541.720.05339.51 2.34×10−34a
GPR149G protein-coupled receptor 1492.420.11638.62 7.30×10−13a
SCGNSecretagogin, ef-hand calcium binding protein1.960.17237.66 2.92×10−5a
SLC35D3Solute carrier family 35, member d3−1.200.45637.43 3.85×10−13a
CT45A3Cancer/testis antigen family 45, member a33.010.15135.61 2.05×10−5a
ADAM23Adam metallopeptidase domain 231.011.00034.46 1.71×10−29a
ST8SIA3St8 α-n-acetyl-neuraminide α-2,8-sialyltransferase 33.170.05933.69 7.23×10−5a
HOXD10Homeobox d101.530.12032.56 3.08×10−36a
LMO1Lim domain only 1 (rhombotin 1)1.590.07231.08 1.71×10−24a
ODZ2Odz, odd oz/ten-m homolog 21.000.97230.69 2.36×10−30a
CLDN19Claudin 19−1.620.07030.28 1.95×10−10a
FOXN1Forkhead box n11.190.33929.93 6.61×10−35a
APOA1Apolipoprotein a-i1.810.06429.75 8.79×10−9a
HS3ST4Heparan sulfate (glucosamine) 3-o-sulfotransferase 41.740.09729.69 3.25×10−11a
PAX1Paired box 11.630.22528.10 7.10×10−11a
OLFM3Olfactomedin 32.730.05026.58 2.93×10−9a
FAM181BFamily with sequence similarity 181, member b1.110.66825.19 1.25×10−39a
CRNNCornulin2.060.12924.94 1.37×10−8a
TP53AIP1Tumor protein p53 regulated apoptosis inducing protein 1−1.020.91623.03 8.00×10−25a
TCHHL1Trichohyalin-like 12.340.24222.08 1.38×10−17a
SERPINB2Serpin peptidase inhibitor, clade b (ovalbumin), member 2−1.060.80221.55 8.28×10−21a
QRFPRPyroglutamylated rfamide peptide receptor−2.300.00321.09 1.94×10−10a
TGM3Transglutaminase 3−1.590.03920.25 2.65×10−13a
CT45A1Cancer/testis antigen family 45, member a11.740.30220.10 5.63×10−7a
CT45A4Cancer/testis antigen family 45, member a43.550.07919.29 1.5×10−4a

a P<0.001. AD, lung adenocarcinoma; SC, squamous cell carcinoma; FC, fold change.

Table II.

Downregulated genes in AD and SC.

Table II.

Downregulated genes in AD and SC.

A, Genes downregulated specifically in AD

GeneGene nameAD FCAD P-valueSC FCSC P-value
ADCY8Adenylate cyclase 8 (brain)−11.65 8.60×10−24a2.130.073
SOSTDC1Sclerostin domain containing 1−10.96 2.73×10−39a−1.010.937
SLCO1A2Solute carrier organic anion transporter family, member−10.09 1.71×10−26a−1.290.146
CHRNA2Cholinergic receptor, nicotinic, α 2 (neuronal)−10.08 1.20×10−43a−1.430.292
ODAMOdontogenic, ameloblast associated−9.92 2.10×10−33a3.220.009
KRT79Keratin 79−8.53 3.65×10−33a−1.060.788
SYN2Synapsin ii−7.58 5.50×10−37a−1.050.790
S100A12S100 calcium binding protein a12−6.92 5.97×10−45a−1.260.310
TGM1Transglutaminase 1−6.87 1.19×10−78a2.44 <0.001a
ANXA8L2Annexin a8-like 2−6.84 2.81×10−29a1.78 <0.001a
SLITRK2Slit and ntrk-like family, member 2−6.63 3.63×10−36a−1.090.730
DCCDeleted in colorectal carcinoma−6.26 1.78×10−40a1.100.773
FGFBP2Fibroblast growth factor binding protein 2−5.78 1.31×10−24a5.56 <0.001a
VITVitrin−5.56 3.27×10−28a5.59 <0.001a
LPPR5Lipid phosphate phosphatase-related protein type 5−5.55 9.97×10−19a−1.340.311
VWC2Von willebrand factor c domain containing 2−4.62 4.26×10−31a2.150.008
NOS1Nitric oxide synthase 1 (neuronal)−4.50 1.75×10−11a1.110.769
HSPB3Heat shock 27 kda protein 3−4.47 3.32×10−26a1.730.021
SEMA6DSemaphoring 6D−4.28 1.70×10−33a−1.140.404
NTRK2Neurotrophic tyrosine kinase, receptor, type 2−4.21 1.10×10−18a9.48 <0.001a
SLC27A6Solute carrier family 27 (fatty acid transporter), member 6−4.12 4.37×10−10a−1.340.297
CHRNA4Cholinergic receptor, nicotinic, α 4−4.11 5.82×10−16a2.210.013
EDN3Endothelin 3−4.09 3.04×10−9a3.960.014
NDRG4Ndrg family member 4−4.08 6.96×10−34a1.730.002
KRT4Keratin 4−4.03 2.93×10−11a2.550.006
FEZ1Fasciculation and elongation protein zeta 1 (zygin i)−4.02 3.18×10−77a−1.190.141
ANXA8Annexin a8−4.01 1.57×10−12a3.94 <0.001a

B, Genes downregulated specifically in SC

GeneGene nameAD FCAD P-valueSC FCSC P-value

MYH1Myosin, heavy chain 1, skeletal muscle, adult−1.330.336−16.35 8.37×10−21a
PGCProgastricsin (pepsinogen c)3.89 <0.001a−16.06 1.18×10−8a
CHIAChitinase, acidic1.250.420−15.74 5.48×10−7a
SFTA2Surfactant associated 21.150.361−14.51 4.11×10−18a
APOHApolipoprotein h (β-2-glycoprotein i)1.820.035−13.95 4.11×10−11a
CAPN9Calpain 9−1.220.316−12.42 1.05×10−17a
DPCR1Diffuse panbronchiolitis critical region 14.44 <0.001a−12.26 4.59×10−13a
FOLR1Folate receptor 1 (adult)−1.330.076−11.98 9.39×10−11a
SFTA3 Surfactant-associated 3−1.200.184−11.69 3.92×10−21a
C16orf89Chromosome 16 open reading frame 891.190.394−11.54 8.58×10−14a
HNF1BHnf1 homeobox b1.230.114−11.46 1.86×10−12a
SLC10A2Solute carrier family 10 member 21.950.158−10.75 4.10×10−13a
NAPSANapsin a aspartic peptidase−1.150.382−10.39 2.69×10−15a
CYP2B7P1Cytochrome p450, family 2, subfamily b, polypeptide 7 pseudogene 11.230.316−9.97 4.54×10−5a
MIA2Melanoma inhibitory activity 2−1.050.783−9.17 3.19×10−18a
CCL14.CCL15C-C motif chemokine 14−1.910.053−9.13 1.13×10−21a
C4BPAComplement component 4 binding protein, α−1.380.075−8.55 6.23×10−12a
TDRD10Tudor domain containing 10−1.010.931−8.50 1.96×10−20a
AQP7Aquaporin 72.16 <0.001a−8.20 8.39×10−6a
FMO5Flavin containing monooxygenase 51.540.008−7.99 2.40×10−17a
NKX2.1Homeobox protein Nkx-2.11.260.067−7.72 6.23×10−12a
SLC26A9Solute carrier family 26, member 91.270.249−7.64 1.99×10−7a
SCGB3A1Secretoglobin, family 3a, member 11.770.062−7.60 3.31×10−7a

a P<0.001. AD, lung adenocarcinoma; SC, squamous cell carcinoma; FC, fold-change.

Functional analysis of DEGs

An Ingenuity Pathway Analysis (IPA) software tool (https://www.qiagenbioinformatics.com/products/ingenuity-pathway-analysis) was used to determine the underlying mechanisms, functions, pathways and associations between the gene sets identified during DEG analysis. Molecular and cellular functions and canonical pathways were identified using IPA to distinguish the complex biology underlying the pathogenesis of the two lung cancer subtypes. Upstream regulator analysis using IPA was performed to discover the upstream transcriptional factors regulating changes in the expression of the identified genes. The analysis is based on the expected effects between transcriptional regulators and their targets stored in the Ingenuity database. The analysis provides a P-value and an activation z-score based on the number of known targets present in the DEGs for each transcriptional regulator. Overall, this analysis is part of IPA core analysis examining the mechanisms, functions and pathways associated with a given set of genes.

Results

Identification of DEGs

RNA-Seq gene expression data from TCGA were analyzed to compare the gene expression changes in the early stages of two NSCLC subtypes. A total of 145 genes were upregulated specifically in AD, and 146 genes were upregulated specifically in SC. Among the downregulated genes, 27 were downregulated specifically in AD, whereas 103 were downregulated specifically in SC. Venn diagrams representing the number of upregulated and downregulated genes in the two NSCLC subtypes are presented in Fig. 1A and B. A heat map was constructed to identify the expression patterns of these unique gene sets in each subtype (Fig. 1C). The patients were stratified by smoking status to determine whether the DEGs were associated with smoking; the results demonstrated that early-stage differences unique to each subtype were not associated with smoking (data not shown).

Genes differentially expressed in AD

A total of 145 genes were highly upregulated specifically in AD (FCAD>4; PAD<0.001) with no significant upregulation in SC. The genes with the highest FC uniquely upregulated in each subtype are presented in Table I. In addition, 27 genes were significantly downregulated specifically in AD (FCAD<-4; PAD<0.001) with no significant downregulation in SC (Table II). Highly upregulated genes specific to AD included albumin (ALB; AD, FC=1,732.04; SC, FC=−1.48), protein lin-28 homolog A (LIN28A; AD, FC=150.39; SC, FC=1.25), gastric lipase (LIPF; AD, FC=81.24; SC, FC=2.69), transmembrane 4 L six family member 4 (TM4SF4; AD, FC=68.74; SC, FC=−2.73) and alanine-glyoxylate aminotransferase 2-like 1 (AGXT2L1; AD, FC=66.16; SC, FC=−1.89). The LIN28A gene is a cell cycle regulator, the role of which has been identified in a number of human cancers (18,19), but not in NSCLC. AD also demonstrated ~15-fold upregulation in the mucin (MUC) family of genes, which may be associated with the secretory nature of the tumor. The top downregulated genes specific to AD included adenylate cyclase 8 (AD, FC=−11.65; SC, FC=2.13), sclerostin domain-containing 1 (SOSTDC1; AD, FC=−10.96; SC, FC=−1.01), solute carrier organic anion transporter family member 1A2 (AD, FC=−10.09; SC, FC=−1.29), cholinergic receptor nicotinic α2 subunit (AD, FC=−10.08; SC, FC=−1.43) and odontogenic ameloblast-associated (ODAM; AD, FC=−9.92; SC, FC=3.22).

Genes differentially expressed in SC

A total of 146 genes were highly upregulated in SC (FCSC>4; PSC<0.001) with no significant upregulation in AD (Table I). In addition, 103 genes were significantly downregulated (FCSC<-4; PSC<0.001) in SC with no downregulation in AD (Table II). The top upregulated genes unique to SC were amelotin (AMTN; SC, FC=618.16; AD, FC=1.70), alcohol dehydrogenase 7 (ADH7; SC, FC=57.12; AD, FC=1.52), sclerostin (SOST; SC, FC=54.92; AD, FC=−1.81), claudin 22 (CLDN22; SC, FC=54.24; AD, FC=2.46) and SRY-box 10 (SC, FC=39.80; AD, FC=1.76). In addition, early-stage SC exhibited unique upregulation of several members of the cancer/testis antigen (CTA) family of genes. The top downregulated genes specific to SC were myosin heavy chain 1 (SC, FC=−16.35; AD, FC=−1.33), progastricsin (SC, FC=−16.06; AD, FC=3.89), chitinase acidic (SC, FC-15.74; AD, FC=1.25), surfactant-associated 2 (SC, FC=−14.51; AD, FC=1.15) and apolipoprotein H (SC, FC=−13.95; AD, FC=1.82).

Analysis of molecular pathways in AD

The IPA tool was used to generate an interaction network for genes specifically differentially regulated in AD and SC, based on known interactions (Fig. 2). The genes uniquely dysregulated in AD were enriched in a number of molecular and cellular functions, including ‘molecular transport’, ‘cell-to-cell signaling and interaction’, ‘amino acid metabolism’ and ‘cellular growth and proliferation’. A number of the dysregulated genes specific to AD were also involved in the canonical farnesoid X receptor (NR1H4)/retinoid X receptor (RXR) activation and liver X receptor (LXR)/RXR activation pathways. The roles of these regulators in NSCLCs have not been previously reported. A number of upstream regulators of these genes were identified, including hepatocyte nuclear factor 4 α (HNF4A), which regulated 26 genes, HNF1A, which regulates 22 genes, transcription activator BRG1 (SMARCA4), which regulates 14 genes, and Forkhead Box A2 (FOXA2), which regulated 10 genes (Table III). The HNF family of genes and FOXA2 have been independently associated with AD as positive and negative regulators of growth, respectively (20,21).

Table III.

Functional annotation terms of genes enriched in lung adenocarcinoma.

Table III.

Functional annotation terms of genes enriched in lung adenocarcinoma.

Gene Ontology termCountP-value
Molecular and cellular functions
  Small molecule biochemistry50 3.0×10−5
  Molecular transport43 1.6×10−5
  Cell-to-cell signaling and interaction42 2.1×10−5
  Amino acid metabolism18 3.0×10−5
  Cellular growth and proliferation16 2.1×10−5
Canonical pathways
  FXR/RXR activation13 1.1×10−11
  LXR/RXR activation9 3.2×10−7
  Acute phase response signaling9 7.3×10−6
  eNOS signaling6 1.9×10−3
  Coagulation system3 2.2×10−3
Upstream regulators
  Hepatocyte nuclear factor 4-α26 7.7×10−3
  Hepatocyte nuclear factor 1-α22 2.6×10−12
  Transcription activator BRG114 2.0×10−4
  Forkhead Box protein A210 1.9×10−6
  Peroxisome proliferator-activated receptor α10 2.1×10−3
  LIM/homeobox protein Lhx18 1.2×10−6
  Forkhead Box protein A18 1.8×10−5
  Bile acid receptor7 1.6×10−4
  PR domain zinc finger protein 17 1.6×10−3
  Myoblast determination protein 16 2.6×10−3
  Mothers against decapentaplegic homolog 36 7.4×10−3
Analysis of molecular pathways in SC

The genes unique to early-stage SC were enriched in ‘xenobiotic metabolism’, ‘lipid metabolism’, ‘vitamin and mineral metabolism’, ‘drug metabolism’ and ‘free radical scavenging’. These results suggested that impaired lipid metabolism is specific to SC. The canonical pathways ‘LPS/IL-1 mediated inhibition of RXR function’, ‘xenobiotic metabolism signaling’ and ‘aryl hydrocarbon receptor signaling’ were among those specifically dysregulated in SC. Peroxisome proliferator-activated receptor-γ (PPARG), which regulates 15 genes, c-Jun, which regulates 15 genes, and RXR α (RXRA), which regulates 14 genes, were among the upstream regulators of the differentially regulated genes (Table IV).

Table IV.

Functional annotation terms enriched in lung squamous cell carcinoma.

Table IV.

Functional annotation terms enriched in lung squamous cell carcinoma.

Gene Ontology termCountP-value
Molecular and cellular functions
  Small molecule biochemistry58 7.9×10−8
  Lipid metabolism45 7.9×10−8
  Vitamin and mineral metabolism26 7.9×10−8
  Drug metabolism15 3.8×10−6
  Free radical scavenging Canonical pathways10 1.1×10−5
  LPS/IL-1 mediated inhibition of RXR function14 4.6×10−8
  Xenobiotic metabolism signaling12 3.5×10−5
  Aryl hydrocarbon receptor signaling10 1.6×10−6
  PXR/RXR activation  6 5.8×10−5
  Glutathione-mediated detoxification  5 5.0×10−5
Upstream regulators
  Peroxisome proliferator-activated receptor γ15 5.8×10−5
  Transcription factor AP-115 5.4×10−5
  Retinoic acid receptor RXR-α14 5.0×10−7
  Estrogen receptor β13 7.2×10−5
  Tumor protein 6313 1.4×10−4
  Retinoic acid receptor α12 8.9×10−5
  Histone deacetylase 111 5.0×10−5
  Homeobox protein Nkx-2.110 1.4×10−5
  Zinc finger protein GLI2  7 7.9×10−5
  LIM/homeobox protein Lhx1  7 9.0×10−5

Discussion

Lung carcinomas account for >25% of cancer-associated mortalities worldwide, and the majority of primary lung cancers are NSCLC histological subtypes, including AD and SC (3). Personalized treatment for these cancers requires a complete and detailed understanding of the distinct molecular mechanisms that contribute to tumorigenesis, especially in early stages when survival rates are >90%. In the present study, a total of 172 genes with differential expression (145 upregulated and 27 downregulated) specific to AD, and another 249 genes (146 upregulated and 103 downregulated) specific to SC were identified.

The present study demonstrated that early-stage AD exhibited a 150-fold upregulation of the oncogene LIN28A, which is involved in cell cycle progression through the regulation of cyclin-dependent kinase 2 in lung, breast, ovarian, colon, liver and pancreatic cancer (22). In addition, LIN28 has been demonstrated to confer resistance to radiotherapy in lung carcinoma cell lines (23) and has been explored for its potential role in breast cancer therapy (24). The results of the present study revealed that the activity of this oncogene was unique to the AD subtype of NSCLC, whereas no significant changes in LIN28 expression levels were observed in SC. These results suggested that LIN28A may be a novel therapeutic target for AD. The palate, lung and nasal epithelium carcinoma-associated gene, which has a documented association with respiratory tumors with a glandular phenotype (25), was upregulated 38-fold in AD in the present study. Members of the mucin family of genes, mucin 21 cell surface-associated and mucin 5b oligomeric mucus/gel-forming, were identified to be upregulated ~15-fold in AD, and are likely to be involved in the excessive secretion of mucus by neoplastic cells in AD (26). Mucin peptides incorporated into liposomal vaccines are associated with extended survival times in patients with lung cancer (27). Downregulation of several tumor suppressor genes including SOSTDC1, ODAM, deleted in colorectal carcinoma, fasciculation and elongation proteins ζ 1 and annexin A8 was observed in AD, but not in SC. These genes are associated with a variety of cancers, including lung, breast, colon and prostate cancer (2831).

In the present study, early-stage SC exhibited specific upregulation of CTA family 45 members A1-4. Auto-antibodies against the genes of this family have been demonstrated to serve as biomarkers for NSCLC with low sensitivity and high specificity (32), and a RNAseq catalog of 90 cancer testis antigens were established by Djureinovic et al (33). The results of the present study also demonstrated that the expression of another member of the CTA family, X antigen family member 2, was downregulated in SC; this gene has previously been identified as a tumor suppressor in metastatic melanoma and Ewing sarcoma (34). In addition, SC exhibited downregulation of several tumor suppressor genes, including melanoma inhibitory activity 2, which is involved in hepatocellular carcinoma (30), and secretoglobin family 3a member 1, which serves a role in testicular germ cell tumors (35,36).

In the present study, ‘cellular function’ and ‘lipid metabolism’ were associated with the genes dysregulated specifically in early-stage SC. Alterations in lipid metabolism have been previously implicated in human cancers, particularly oral squamous cell carcinoma, in which increased lipid metabolism is associated with invasiveness (37). Previous studies have reported that impaired lipid metabolism in NSCLC results in the loss of malignant potential (3840). The results of the present study suggested that abnormal lipid metabolism may be specific to SC. AD exhibited significant upregulation of LIPF; however, no evidence of dysregulated lipid metabolism in AD was observed at a functional level. By contrast, AD exhibited alterations in ‘molecular transport’, ‘cell-to-cell signaling and interaction’ and ‘amino acid metabolism’.

Only SC exhibited strong enrichment of the ‘drug metabolism’ cellular function in the present study. Previous studies have suggested that high activity levels of cytochrome P450 isotypes, particularly cytochrome P450 family 1 (CYP1) subfamily B member 1 (CYP1B1), serve a role in carcinogenesis and drug resistance in human cancers, including NSCLC, and may serve as therapeutic targets or prognostic indicators (41,42). In addition, 5,7-dimethoxyflavone and resveratrol have been used to inhibit CYP1 family protein expression Hep G2 human hepatoma and MCF-10a non-tumorigenic human mammary epithelial cell lines, respectively (43,44). The results of the present study revealed that the ‘PXR/RXR’, ‘xenobiotic metabolism signaling’, ‘aryl hydrocarbon receptor signaling’ and ‘glutathione-mediated detoxification’ canonical pathways were also altered in early-stage SC. Previous studies have demonstrated that PXR serves a role in xenobiotic metabolism in human malignancies, such as colon, breast and gynecologic cancers (45). The involvement of PXR in NSCLC has not been previously reported. The aryl hydrocarbon transcription factor is also involved in cytochrome metabolism and activates the CYP1B1, CYP1A1 and CYP1A2 isotypes (46). The results of the present study suggest that the dysregulation of genes associated with drug metabolism may be specific to SC, and that the role of these catabolic enzymes may be evident in early-stage cancer. These results also identify several potential mechanisms of chemotherapy resistance in SC.

The canonical pathways ‘FXR/RXR activation’ and ‘LXR/RXR activation’ were altered in early-stage AD, and the NR1H4 gene was identified as an upstream regulator of AD in the present study. Previous studies of FXR/RXR in human cancers have demonstrated that it is activated in breast and esophageal cancers, but can be downregulated in hepatobiliary cancers (4749). Loss of LXR/RXR is involved in the growth and progression of prostatic carcinomas, and LXR agonists have emerged as a novel therapy for prostate cancer (50). The canonical pathways ‘LPS/IL-1 mediated inhibition of RXR function’ and ‘PXR/RXR activation’ were altered in SC in the present study. PXR/RXR is involved in the metabolism of xenobiotics and has been demonstrated to be involved in multiple types of human cancer, including colon, breast and gynecological cancers (45). Previous studies have demonstrated the use of retinoid receptor expression as a prognostic indicator in stage I NSCLC, but the role of specific retinoid receptors has not been explored (51,52). The involvement of FXR/RXR, LXR/RXR and PXR/RXR in NSCLC subtypes is a novel finding of the present study.

The results of the present study demonstrated that HNF4A and HNF1A were upstream regulators of the genes specifically dysregulated in AD. A previous study has identified the use of HNF4A as a biomarker for AD, and another study identified HNF4G to be involved in the AKT signaling pathway in lung cancer (20,53). The present results suggest that HNF may be an upstream driver of tumorigenesis. In addition, SMARCA4 was identified as another upstream regulator in AD. Upregulation of this gene in AD is associated with poor prognosis and a poor response to platinum-based chemotherapy (54,55). Analysis of the upstream regulators in the present study also identified FOXA2 and FOXA1 as specific regulators of AD; the FOXA2 gene product has been demonstrated to prevent lung tumor growth and metastasis by preventing epithelial-mesenchymal transition (21).

In the present study PPARG, c-JUN and RXRA were the most significant upstream regulators of the genes specifically differentially regulated in early-stage SC. The role of PPARG in lung cancer is unclear, although PPARG has been studied in the context of pulmonary fibrosis, where it was demonstrated to repress myofibroblast differentiation (56). However, upregulation of PPARG repressed tumor growth in pancreatic and colorectal cancer (57,58), and PPARG inhibitors have been used to induce anti-estrogen susceptibility in mammary tumors (59). The role of the c-JUN regulator in NSCLC may be related to the dysregulation of retinoid signaling by the inhibition of RXRA, which is another upstream regulator of the genes altered in SC (60). Based on prior studies (61,62), c-JUN may be activated and RXRA may be consequently inhibited in SC. Anti-tumor activity has been achieved through c-JUN protein inhibition using a bisphenazine anticancer drug (63). Several other upstream regulators were identified in the present study, such as estrogen receptor 2 and tumor protein p63, which have been previously demonstrated to serve roles in NSCLC (64,65).

The major limitation of the present study was the lack of experimental validation of the findings using in vivo or in vitro experiments. However, to minimize false positives, a very stringent cut-off was used to select the DEGs. In addition, the large sample set provided high statistical power to discover the differences with high confidence.

In conclusion, the present study revealed early-stage differences in the gene expression profiles of AD and SC. Unique sets of genes altered in each subtype were identified; for example, ALB, LIN28A, LIPF, TM4SF4, AGXT2L1 and ACMSD genes were upregulated >50-fold in AD, but were not significantly upregulated in SD. Similarly, AMTN, ADH7, SOST and CLDN22 were upregulated >50-fold in SC, but not in AD. Several CTA family genes were highly upregulated in SC, but not in AD, whereas several mucins were upregulated only in AD. In addition, ‘lipid metabolism’ and ‘drug metabolism’ pathways were associated with genes dysregulated specifically in SC, whereas ‘molecular transport’ and ‘cellular growth and proliferation’ were significantly enriched only in AD. The results of the present study provided gene expression alterations specific to each subtype, which may help to identify the molecular mechanisms underlying the pathogenesis of these subtypes. These findings also provide targets for future studies investigating novel diagnostic methods and personalized therapeutic approaches for AD and SC.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Institutional Start-Up Package to AS from the Medical College of Georgia at Augusta University, Augusta, GA, USA.

Availability of data and materials

The datasets analyzed for this study are available from The Cancer Genome Atlas (https://cancergenome.nih.gov).

Authors contributions

NV, JY and AS wrote the manuscript and created the figures and tables. TJL and SKK performed data analysis. NV, JY, SS, AS and NP contributed to the data interpretation. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study utilizes a publicly available data set from TCGA (https://cancergenome.nih.gov/abouttcga) and was granted an exemption from requiring ethics approval by the Institutional Review Boards at Augusta University Augusta GA USA.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Torre LA, Siegel RL and Jemal A: Lung cancer statistics. Adv Exp Med Biol. 893:1–19. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Muralidharan-Chari V, Clancy JW, Sedgwick A and DSouza-Schorey C: Microvesicles: Mediators of extracellular communication during cancer progression. J Cell Sci. 123:1603–1611. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Choy B, Findeis-Hosey JJ, Li F, McMahon LA, Yang Q and Xu H: High frequency of coexpression of maspin with p63 and p53 in squamous cell carcinoma but not in adenocarcinoma of the lung. Int J Clin Exp Pathol. 6:2542–2547. 2013.PubMed/NCBI

6 

Fukui T, Taniguchi T, Kawaguchi K, Fukumoto K, Nakamura S, Sakao Y and Yokoi K: Comparisons of the clinicopathological features and survival outcomes between lung cancer patients with adenocarcinoma and squamous cell carcinoma. Gen Thorac Cardiovasc Surg. 63:507–513. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Argon A, Nart D and Veral A: The value of cytokeratin 5/6, p63 and thyroid transcription factor-1 in adenocarcinoma, squamous cell carcinoma and non-small-cell lung cancer of the lung. Turk Patoloji Derg. 31:81–88. 2015.PubMed/NCBI

8 

Singhal S, Miller D, Ramalingam S and Sun SY: Gene expression profiling of non-small cell lung cancer. Lung Cancer. 60:313–324. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Shi WY, Liu KD, Xu SG, Zhang JT, Yu LL, Xu KQ and Zhang TF: Gene expression analysis of lung cancer. Eur Rev Med Pharmacol Sci. 18:217–228. 2014.PubMed/NCBI

10 

Lu C, Chen H, Shan Z and Yang L: Identification of differentially expressed genes between lung adenocarcinoma and lung squamous cell carcinoma by gene expression profiling. Mol Med Rep. 14:1483–1490. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Lacroix L, Commo F and Soria JC: Gene expression profiling of non-small-cell lung cancer. Expert Rev Mol Diagn. 8:167–178. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Grigoroiu M, Tagett R, Draghici S, Dima S, Nastase A, Florea R, Sorop A, Ilie V, Bacalbasa N, Tica V, et al: Gene-expression profiling in non-small cell lung cancer with invasion of mediastinal lymph nodes for prognosis evaluation. Cancer Genomics Proteomics. 12:231–242. 2015.PubMed/NCBI

13 

Yousef GM, Scorilas A, Nakamura T, Ellatif MA, Ponzone R, Biglia N, Maggiorotto F, Roagna R, Sismondi P and Diamandis EP: The prognostic value of the human kallikrein gene 9 (KLK9) in breast cancer. Breast Cancer Res Treat. 78:149–158. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Bahassi el M and Stambrook PJ: Next-generation sequencing technologies: Breaking the sound barrier of human genetics. Mutagenesis. 29:303–310. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Hagemann IS, Devarakonda S, Lockwood CM, Spencer DH, Guebert K, Bredemeyer AJ, Al-Kateb H, Nguyen TT, Duncavage EJ, Cottrell CE, et al: Clinical next-generation sequencing in patients with non-small cell lung cancer. Cancer. 121:631–639. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Xu X, Yang Y, Li H, Chen Z, Jiang G and Fei K: Assessment of the clinical application of detecting EGFR, KRAS, PIK3CA and BRAF mutations in patients with non-small cell lung cancer using next-generation sequencing. Scand J Clin Lab Invest. 76:386–392. 2016. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Wang T, Han P, He Y, Zhao C, Wang G, Yang W, Shan M, Zhu Y, Yang C, Weng M, et al: Lin28A enhances chemosensitivity of colon cancer cells to 5-FU by promoting apoptosis in a let-7 independent manner. Tumour Biol. 37:7657–7665. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Shen H, Yang Y, Zhao L, Yuan J and Niu Y: Lin28A and androgen receptor expression in ER/Her2+ breast cancer. Breast Cancer Res Treat. 156:135–147. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Sugano M, Nagasaka T, Sasaki E, Murakami Y, Hosoda W, Hida T, Mitsudomi T and Yatabe Y: HNF4α as a marker for invasive mucinous adenocarcinoma of the lung. Am J Surg Pathol. 37:211–218. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Tang Y, Shu G, Yuan X, Jing N and Song J: FOXA2 functions as a suppressor of tumor metastasis by inhibition of epithelial-to-mesenchymal transition in human lung cancers. Cell Res. 21:316–326. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Li N, Zhong X, Lin X, Guo J, Zou L, Tanyi JL, Shao Z, Liang S, Wang LP, Hwang WT, et al: Lin-28 homologue A (LIN28A) promotes cell cycle progression via regulation of cyclin-dependent kinase 2 (CDK2), cyclin D1 (CCND1), and cell division cycle 25 homolog A (CDC25A) expression in cancer. J Biol Chem. 287:17386–17397. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Oh JS, Kim JJ, Byun JY and Kim IA: Lin28-let7 modulates radiosensitivity of human cancer cells with activation of K-Ras. Int J Radiat Oncol Biol Phys. 76:5–8. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Xiong H, Zhao W, Wang J, Seifer BJ, Ye C, Chen Y, Jia Y, Chen C, Shen J, Wang L, et al: Oncogenic mechanisms of Lin28 in breast cancer: New functions and therapeutic opportunities. Oncotarget. 8:25721–25735. 2017.PubMed/NCBI

25 

Bingle L, Cross SS, High AS, Wallace WA, Devine DA, Havard S, Campos MA and Bingle CD: SPLUNC1 (PLUNC) is expressed in glandular tissues of the respiratory tract and in lung tumours with a glandular phenotype. J Pathol. 205:491–497. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Corfield AP: Mucins: A biologically relevant glycan barrier in mucosal protection. Biochim Biophys Acta. 1850:236–252. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Palmer M, Parker J, Modi S, Butts C, Smylie M, Meikle A, Kehoe M, MacLean G and Longenecker M: Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin Lung Cancer. 3:49–58. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Liu L, Wu S, Yang Y, Cai J, Zhu X, Wu J, Li M and Guan H: SOSTDC1 is down-regulated in non-small cell lung cancer and contributes to cancer cell proliferation. Cell Biosci. 6:242016. View Article : Google Scholar : PubMed/NCBI

29 

Kestler DP, Foster JS, Bruker CT, Prenshaw JW, Kennel SJ, Wall JS, Weiss DT and Solomon A: ODAM expression inhibits human breast cancer tumorigenesis. Breast Cancer (Auckl). 5:73–85. 2011.PubMed/NCBI

30 

Ishii H, Vecchione A, Murakumo Y, Baldassarre G, Numata S, Trapasso F, Alder H, Baffa R and Croce CM: FEZ1/LZTS1 gene at 8p22 suppresses cancer cell growth and regulates mitosis. Proc Natl Acad Sci USA. 98:10374–10379. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Lee MJ, Yu GR, Yoo HJ, Kim JH, Yoon BI, Choi YK and Kim DG: ANXA8 down-regulation by EGF-FOXO4 signaling is involved in cell scattering and tumor metastasis of cholangiocarcinoma. Gastroenterology. 137:1138–1150, 1150.e1-e9. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Shan Q, Lou X, Xiao T, Zhang J, Sun H, Gao Y, Cheng S, Wu L, Xu N and Liu S: A cancer/testis antigen microarray to screen autoantibody biomarkers of non-small cell lung cancer. Cancer Lett. 328:160–167. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Djureinovic D, Hallstrom BM, Horie M, Mattsson JSM, La Fleur L, Fagerberg L, Brunnström H, Lindskog C, Madjar K, Rahnenführer J, et al: Profiling cancer testis antigens in non-small-cell lung cancer. JCI Insight. 1:e868372016. View Article : Google Scholar : PubMed/NCBI

34 

Zendman AJ, Van Kraats AA, Weidle UH, Ruiter DJ and Van Muijen GN: The XAGE family of cancer/testis-associated genes: Alignment and expression profile in normal tissues, melanoma lesions and Ewings sarcoma. Int J Cancer. 99:361–369. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Hellerbrand C, Amann T, Schlegel J, Wild P, Bataille F, Spruss T, Hartmann A and Bosserhoff AK: The novel gene MIA2 acts as a tumour suppressor in hepatocellular carcinoma. Gut. 57:243–251. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Lind GE, Skotheim RI, Fraga MF, Abeler VM, Esteller M and Lothe RA: Novel epigenetically deregulated genes in testicular cancer include homeobox genes and SCGB3A1 (HIN-1). J Pathol. 210:441–449. 2006. View Article : Google Scholar : PubMed/NCBI

37 

SantAnna-Silva ACB, Santos GC, Campos SPC, Oliveira Gomes AM, Perez-Valencia JA and Rumjanek FD: Metabolic profile of oral squamous carcinoma cell lines relies on a higher demand of lipid metabolism in metastatic cells. Front Oncol. 8:132018. View Article : Google Scholar : PubMed/NCBI

38 

Long J, Zhang CJ, Zhu N, Du K, Yin YF, Tan X, Liao DF and Qin L: Lipid metabolism and carcinogenesis, cancer development. Am J Cancer Res. 8:778–791. 2018.PubMed/NCBI

39 

Luo X, Cheng C, Tan Z, Li N, Tang M, Yang L and Cao Y: Emerging roles of lipid metabolism in cancer metastasis. Mol Cancer. 16:762017. View Article : Google Scholar : PubMed/NCBI

40 

Guo JY and White E: Autophagy is required for mitochondrial function, lipid metabolism, growth, and fate of KRAS (G12D)-driven lung tumors. Autophagy. 9:1636–1638. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Murray GI, Taylor MC, McFadyen MC, McKay JA, Greenlee WF, Burke MD and Melvin WT: Tumor-specific expression of cytochrome P450 CYP1B1. Cancer Res. 57:3026–3031. 1997.PubMed/NCBI

42 

Su JM, Lin P, Wang CK and Chang H: Overexpression of cytochrome P450 1B1 in advanced non-small cell lung cancer: A potential therapeutic target. Anticancer Res. 29:509–515. 2009.PubMed/NCBI

43 

Chen ZH, Hurh YJ, Na HK, Kim JH, Chun YJ, Kim DH, Kang KS, Cho MH and Surh YJ: Resveratrol inhibits TCDD-induced expression of CYP1A1 and CYP1B1 and catechol estrogen-mediated oxidative DNA damage in cultured human mammary epithelial cells. Carcinogenesis. 25:2005–2013. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Wen X, Walle UK and Walle T: 5,7-Dimethoxyflavone downregulates CYP1A1 expression and benzo[a]pyrene-induced DNA binding in Hep G2 cells. Carcinogenesis. 26:803–809. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Qiao E, Ji M, Wu J, Ma R, Zhang X, He Y, Zha Q, Song X, Zhu LW and Tang J: Expression of the PXR gene in various types of cancer and drug resistance. Oncol Lett. 5:1093–1100. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Xue P, Fu J and Zhou Y: The aryl hydrocarbon receptor and tumor immunity. Front Immunol. 9:2862018. View Article : Google Scholar : PubMed/NCBI

47 

Swales KE, Korbonits M, Carpenter R, Walsh DT, Warner TD and Bishop-Bailey D: The farnesoid X receptor is expressed in breast cancer and regulates apoptosis and aromatase expression. Cancer Res. 66:10120–10126. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Yang F, Huang X, Yi T, Yen Y, Moore DD and Huang W: Spontaneous development of liver tumors in the absence of the bile acid receptor farnesoid X receptor. Cancer Res. 67:863–867. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Guan B, Li H, Yang Z, Hoque A and Xu X: Inhibition of farnesoid X receptor controls esophageal cancer cell growth in vitro and in nude mouse xenografts. Cancer. 119:1321–1329. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Fukuchi J, Kokontis JM, Hiipakka RA, Chuu CP and Liao S: Antiproliferative effect of liver X receptor agonists on LNCaP human prostate cancer cells. Cancer Res. 64:7686–7689. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Khuri FR, Lotan R, Kemp BL, Lippman SM, Wu H, Feng L, Lee JJ, Cooksley CS, Parr B, Chang E, et al: Retinoic acid receptor-beta as a prognostic indicator in stage I non-small-cell lung cancer. J Clin Oncol. 18:2798–2804. 2000. View Article : Google Scholar : PubMed/NCBI

52 

Chang YS, Chung JH, Shin DH, Chung KY, Kim YS, Chang J and Kim SK and Kim SK: Retinoic acid receptor-beta expression in stage I non-small cell lung cancer and adjacent normal appearing bronchial epithelium. Yonsei Med J. 45:435–442. 2004. View Article : Google Scholar : PubMed/NCBI

53 

Wang J, Zhang J, Xu L, Zheng Y, Ling D and Yang Z: Expression of HNF4G and its potential functions in lung cancer. Oncotarget. 9:18018–18028. 2017.PubMed/NCBI

54 

Bell EH, Chakraborty AR, Mo X, Liu Z, Shilo K, Kirste S, Stegmaier P, McNulty M, Karachaliou N, Rosell R, et al: SMARCA4/BRG1 is a novel prognostic biomarker predictive of cisplatin-based chemotherapy outcomes in resected non-small cell lung cancer. Clin Cancer Res. 22:2396–2404. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Guerrero-Martinez JA and Reyes JC: High expression of SMARCA4 or SMARCA2 is frequently associated with an opposite prognosis in cancer. Sci Rep. 8:20432018. View Article : Google Scholar : PubMed/NCBI

56 

Kulkarni AA, Thatcher TH, Olsen KC, Maggirwar SB, Phipps RP and Sime PJ: PPAR-γ ligands repress TGFβ-induced myofibroblast differentiation by targeting the PI3K/Akt pathway: Implications for therapy of fibrosis. PLoS One. 6:e159092011. View Article : Google Scholar : PubMed/NCBI

57 

Elnemr A, Ohta T, Iwata K, Ninomia I, Fushida S, Nishimura G, Kitagawa H, Kayahara M, Yamamoto M, Terada T and Miwa K: PPARgamma ligand (thiazolidinedione) induces growth arrest and differentiation markers of human pancreatic cancer cells. Int J Oncol. 17:1157–1164. 2000.PubMed/NCBI

58 

Ogino S, Shima K, Baba Y, Nosho K, Irahara N, Kure S, Chen L, Toyoda S, Kirkner GJ, Wang YL, et al: Colorectal cancer expression of peroxisome proliferator-activated receptor gamma (PPARG, PPARgamma) is associated with good prognosis. Gastroenterology. 136:1242–1250. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Yuan H, Kopelovich L, Yin Y, Lu J and Glazer RI: Drug-targeted inhibition of peroxisome proliferator-activated receptor-gamma enhances the chemopreventive effect of anti-estrogen therapy. Oncotarget. 3:345–356. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Srinivas H, Juroske DM, Kalyankrishna S, Cody DD, Price RE, Xu XC, Narayanan R, Weigel NL and Kurie JM: c-Jun N-terminal kinase contributes to aberrant retinoid signaling in lung cancer cells by phosphorylating and inducing proteasomal degradation of retinoic acid receptor alpha. Mol Cell Biol. 25:1054–1069. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Szabo E, Riffe ME, Steinberg SM, Birrer MJ and Linnoila RI: Altered cJUN expression: An early event in human lung carcinogenesis. Cancer Res. 56:305–315. 1996.PubMed/NCBI

62 

Xu XC, Sozzi G, Lee JS, Lee JJ, Pastorino U, Pilotti S, Kurie JM, Hong WK and Lotan R: Suppression of retinoic acid receptor beta in non-small-cell lung cancer in vivo: Implications for lung cancer development. J Natl Cancer Inst. 89:624–629. 1997. View Article : Google Scholar : PubMed/NCBI

63 

Dai J, Punchihewa C, Mistry P, Ooi AT and Yang D: Novel DNA bis-intercalation by MLN944, a potent clinical bisphenazine anticancer drug. J Biol Chem. 279:46096–46103. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Marquez-Garban DC, Chen HW, Fishbein MC, Goodglick L and Pietras RJ: Estrogen receptor signaling pathways in human non-small cell lung cancer. Steroids. 72:135–143. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Borczuk AC, Gorenstein L, Walter KL, Assaad AA, Wang L and Powell CA: Non-small-cell lung cancer molecular signatures recapitulate lung developmental pathways. Am J Pathol. 163:1949–1960. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2019
Volume 18 Issue 6

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
Venugopal N, Yeh J, Kodeboyina SK, Lee TJ, Sharma S, Patel N and Sharma A: Differences in the early stage gene expression profiles of lung adenocarcinoma and lung squamous cell carcinoma. Oncol Lett 18: 6572-6582, 2019
APA
Venugopal, N., Yeh, J., Kodeboyina, S.K., Lee, T.J., Sharma, S., Patel, N., & Sharma, A. (2019). Differences in the early stage gene expression profiles of lung adenocarcinoma and lung squamous cell carcinoma. Oncology Letters, 18, 6572-6582. https://doi.org/10.3892/ol.2019.11013
MLA
Venugopal, N., Yeh, J., Kodeboyina, S. K., Lee, T. J., Sharma, S., Patel, N., Sharma, A."Differences in the early stage gene expression profiles of lung adenocarcinoma and lung squamous cell carcinoma". Oncology Letters 18.6 (2019): 6572-6582.
Chicago
Venugopal, N., Yeh, J., Kodeboyina, S. K., Lee, T. J., Sharma, S., Patel, N., Sharma, A."Differences in the early stage gene expression profiles of lung adenocarcinoma and lung squamous cell carcinoma". Oncology Letters 18, no. 6 (2019): 6572-6582. https://doi.org/10.3892/ol.2019.11013