Differential expression of microRNAs and their target genes in non-small-cell lung cancer

  • Authors:
    • Hui-Young Lee
    • Seon-Sook Han
    • Hwanseok Rhee
    • Jung Hoon Park
    • Jae Seung Lee
    • Yeon-Mok Oh
    • Sun Shim Choi
    • Seung-Ho Shin
    • Woo Jin Kim
  • View Affiliations

  • Published online on: November 6, 2014     https://doi.org/10.3892/mmr.2014.2890
  • Pages: 2034-2040
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are single‑stranded RNA species that constitute a class of non‑coding RNAs, and are emerging as key regulators of gene expression. Since each miRNA is capable of regulating multiple genes, miRNAs are attractive markers for studies of coordinated gene expression. In this study, we investigated miRNA expression profiling using a massively parallel sequencing technique to compare non‑small‑cell lung cancer (NSCLC) tissue and normal lung tissue. Lung cancer tissue and normal lung tissue were obtained from nine NSCLC patients. RNA isolated from these samples was processed using RNA sequencing (RNA Seq) and the HiSeq 2000 system. Differentially expressed miRNAs and mRNAs were analyzed using a t‑test. We selected target pairs that showed a negative correlation among significantly differentially expressed miRNAs and their putative target mRNAs using miRBase Targets. The differences in the expression levels of 222 miRNAs and 1,597 genes were statistically significant, as indicated by an absolute fold change ≥1.5 and P<0.05. miR‑577, miR‑301b, miR‑944, miR‑891a and miR‑615‑3p were generally upregulated, and miR‑338‑3p was generally downregulated. miRNA‑mRNA target pair analysis revealed that 49 miRNAs had 696 target mRNAs. There were significantly differentially expressed miRNAs and mRNAs between lung cancer and normal tissue. Further investigation of miRNAs and their target genes is warranted to better understand NSCLC.

Introduction

Lung cancer is the most common cause of cancer-related mortality worldwide. More than 90 million individuals globally are at risk of developing lung cancer, and the disease is suspected to remain a major health problem for many years. The 5-year survival rate is ~10% (1). More than 75% of lung cancer cases are diagnosed at late stages, as no practical method of screening the large number of people at risk is available. The late-stage diagnosis is the major contributing factor to the poor prognosis of non-small-cell lung cancer (NSCLC), which accounts for 85% of all types of lung cancer (2).

To date, cancer genomic studies have focused on the protein-coding genome. However, it is increasingly evident that the non-protein-coding genome also plays a significant role in tumorigenesis (3). The most widely studied class of non-coding RNAs are microRNAs (miRNAs), which are small non-coding RNAs of approximately 22 nucleotides that mediate post-transcriptional gene silencing by controlling the translocation of mRNA into protein. miRNAs are estimated to regulate the translation of more than 60% of protein-coding genes. Since each miRNA species is capable of regulating multiple genes, miRNAs are attractive markers for studies of coordinated gene expression. In human cancer, the miRNA expression profile differs between normal tissue and tumor tissue, and miRNAs act as oncogenes or tumor suppressor genes and have key functions in tumorigenesis (3). Previous studies have reported differentially regulated miRNAs in diverse solid tumors, including breast (4), lung (5), prostate (6), colon (7) and ovarian (8) cancer.

In solid tumors, miRNAs are deregulated, suggesting their involvement in crucial cellular pathways including cell-cell adhesion and signaling, cell cycle regulation and apoptosis, which play a significant role in the pathogenesis of cancer.

The majority of these studies were performed using microarrays; however, next-generation sequencing (NGS) of small RNAs provides an efficient platform for the investigation of the ubiquitous and differentially expressed behavior of miRNAs (9). Sequencing has the advantage that all RNAs in a sample, not only those on the chip, may be detected; thus, novel miRNAs that were not reported previously may be identified. Furthermore, we aimed to investigate miRNA regulatory molecules by obtaining information on differentially expressed miRNA-mRNA pairs.

In the present study, we adopted the massively parallel sequencing approach to profile miRNA and provide crucial information with regard to the role of miRNAs in regulating tumorigenesis in NSCLC.

Materials and methods

Study subjects

The present study used tissue specimens obtained from the Bio-Resource Center of Asan Medical Center (Seoul, Korea) that were donated by nine patients who underwent surgery for NSCLC between March 2008 and March 2011. All of the paired NSCLC and adjacent normal tissue specimens used in the current study were acquired from surgical specimens. Cancer and normal tissue specimens were grossly dissected and preserved in liquid nitrogen immediately after surgery. Appropriate informed consent was obtained from the participants, and the Institutional Review Board of the Asan Medical Center (Seoul, Korea) approved the study (IRB no. AMC IRB 2011-0711).

mRNA expression

Raw data were extracted as fragments per kilobase of exon per million fragments mapped (FPKM) values for each gene from each sample using TopHat and Cufflinks software (10). Data with zeroed FPKM values across all samples and samples with zero values across more than 50% of the genes were excluded. We added 1 to the FPKM value to facilitate log10 transformation. Filtered data were logarithm transformed and normalized using the quantile method.

Statistical significance of the expression data was determined using fold changes and Student’s t-test or the paired t-test, in which the null hypothesis was that no difference exists between two groups. The false discovery rate (FDR) was controlled by adjusting the P-value using the Benjamini-Hochberg algorithm. Hierarchical clustering was performed using complete linkage and Euclidean distance as a measure of similarity. All data analyses and visualization of differentially expressed genes were conducted using R 2.15.1 (www.r-project.org).

Biological functional annotation analysis of the differentially expressed gene (DEG) list was performed using the DAVID tool (http://david.abcc.ncifcrf.gov/). In the DAVID annotation system, the modified Fisher’s exact P-value (EASE score) was adopted to evaluate gene enrichment in annotation terms.

Additionally, a gene set enrichment analysis was conducted for quality control-filtered expression data using GSEA-P (http://www.broadinstitute.org/gsea/index.jsp), which combines information from previously defined sets of genes (MSigDB). Filtered genes were ranked according to the difference in the log10 expression signal between the case and control using the ‘Diff_of_Classes’ metric option. The extent of association was measured using a nonparametric, running-sum statistic termed the enrichment score (ES), and the maximum ES (MES) over all gene sets in the actual data set was rescored from the cases. The significance of the MES score was calculated as the fraction of the 1,000 random permutations for phenotypes. From the analysis, a normalized ES (NES), nominal P-value, FDR q-value adjusting for a gene set size, and correlations between gene sets and actual data were obtained.

miRNA expression

Raw data (the reads for each miRNA) were normalized to the total reads of each individual sample as the standardized to reads per million (RPM; miRNA counts / total count of each sample × 1 million). We excluded miRNAs with zero values across >90% of the samples and samples with zero values across >80% of the miRNAs. We added 1 to the RPM value to facilitate log10 transformation. The remaining data processing was as for mRNA analysis.

Integrative (mRNA-miRNA) analysis

We performed mRNA-miRNA data integration for matched samples in each data set.

First, genes and miRNAs whose expression differed significantly (absolute fold change ≥1.5 and P<0.05) in each data set were identified. Next, putative mRNA-miRNA target pairs that showed a regulatory association were extracted using mirBase Targets from the Wellcome Trust Sanger Institute (http://microrna.sanger.ac.uk/targets/v5). This approach assumes that the expression of a given miRNA is negatively correlated with the expression of its mRNA targets. We identified negative correlations between any putative miRNA-mRNA pair. For miRNAs with a negative correlation with mRNA, the hypergeometric test was performed to evaluate statistical significance at P<0.05.

Gene ontology (GO) analysis

GO term enrichment analysis for the 696 candidate genes whose regulatory miRNAs were identified was performed using the ClueGo application (ver. 2.0.6) (11) in Cytoscape (ver. 3.0.0) (12). Default parameters were used, and only GO terms with P<0.01 were selected.

Identification of negatively correlated miRNA-mRNA target pairs associated with the cell cycle

The CluePedia application (ver. 1.0.7) (13) based on the miRBase software (miRanda-miRNAs-v5-2012-07-19) (14) in Cytoscape was used to investigate the interaction between miRNAs and cell-cycle-related genes identified in the GO enrichment analysis. Among the cell-cycle-related genes, the 100 mRNAs with the highest number of miRNA targets were selected, and negative correlations between the miRNAs and their target mRNAs were investigated.

Results

Baseline characteristics

Nine patients with completely resected NSCLC were included in our study. All of the patients were male, and the median age was 59 years (range, 45–68 years). The histology consisted of five squamous cell carcinomas (55.6%) and four adenocarcinomas (42.5%). The characteristics of the patients are summarized in Table I.

Table I

Clinical characteristics of NSCLC patients.

Table I

Clinical characteristics of NSCLC patients.

Clinical characteristicsNSCLC patients (n=9)
Age, years (mean ± SD)59 (45–68)
Male, n (%)9 (100)
Smoking (mean ± SD)33±15.6
Histological cell type, n (%)
 Squamous cell carcinoma5 (55.6)
 Adenocarcinoma4 (44.4)
TNM stage, n (%)
 I4 (44.4)
 II5 (55.6)
 III0 (0)

[i] NSCLC, non-small-cell lung cancer.

Expressed miRNAs and mRNA profiling

Overall, 1,117 miRNAs and 21,099 genes were differentially expressed in NSCLC and non-cancerous lung tissue. Of these, the differences in the expression levels of 222 miRNAs and 1,597 genes were statistically significant, as indicated by an absolute fold change ≥1.5 and P<0.05. miR-205-5p and miR-196a-5p were generally upregulated, and miR-133a and miR-139-5p were generally downregulated. The miRNAs with the greatest degrees of upregulation or downregulation in cancer tissue are listed in Table II.

Table II

Top seven up- or downregulated microRNAs in non-small-cell lung cancer.

Table II

Top seven up- or downregulated microRNAs in non-small-cell lung cancer.

miRNAFold change (lung cancer/normal tissue)P-value
Upregulated
hsa-miR-205-5p18.85950.0306
hsa-miR-196a-5p11.09680.0102
hsa-miR-12469.21120.0111
hsa-miR-5778.41140.0006
hsa-miR-301b7.64260.0004
hsa-miR-182-5p7.43880.0015
hsa-miR-196b-5p7.21260.0280
Downregulated
hsa-miR-133a−7.82520.0004
hsa-miR-139-5p−7.11650.0002
hsa-miR-144-5p−6.59231.41E-06
hsa-miR-338-3p−6.57420.0002
hsa-miR-1−6.11930.0002
hsa-miR-490-3p−6.10720.0001
hsa-miR-30a-3p−5.58120.0001
miRNA-mRNA integrative genomic analysis

We searched for negative correlations between putative miRNA and mRNA pairs; 49 putative negative regulatory miRNA-mRNA pairs were identified. Of these, 19 miRNAs were upregulated and 425 mRNAs were downregulated. Conversely, 30 miRNAs were downregulated and 332 mRNAs were upregulated. Fig. 1 shows the data analysis process. Table III shows miRNAs that have negatively correlated target mRNA. Table IV shows differentially expressed miRNAs in NSCLC and their predicted target genes.

Table III

miRNAs with their putative target genes.

Table III

miRNAs with their putative target genes.

miRNAmiRNA fold change (lung cancer/normal tissue)miRNA P-valueP-value (hypergeometric test)
hsa-miR-5778.4110.00060.0461
hsa-miR-301b7.6430.00040.0360
hsa-miR-9445.7650.04220.0416
hsa-miR-891a5.6830.02290.0020
hsa-miR-615-3p5.2110.00640.0299
hsa-miR-671-5p2.6060.01193.83E-06
hsa-miR-4292.5990.04290.0007
hsa-miR-2102.2460.03349.27E-06
hsa-miR-1372.0020.04120.0143
hsa-let-7c−2.0440.01050.0046
hsa-miR-338-3p−6.5740.00020.0442

Table IV

Differentially expressed miRNAs in non-small-cell lung cancer and their predicted target genes.

Table IV

Differentially expressed miRNAs in non-small-cell lung cancer and their predicted target genes.

miRNAmiRNA fold changeTarget geneGene fold change
miR-5778.411GNG11−5.143
EFEMP1−4.578
SCEL−3.625
GKN2−3.47
ATG16L2−2.723
miR-301b7.643HOXA5−4.096
SPARCL1−4.028
HYAL1−3.869
SERPING1−3.825
ALOX5−3.33
miR-9445.765SYT15−3.305
SVEP1−2.314
OMD−2.645
GATA6−2.089
CYB5A−2.552
miR-891a5.683IL1R1−5.836
CLIC5−5.621
SLC27A3−4.292
HOXA5−4.096
ITIH5−3.498
miR-615-3p5.211SFTPC−43.343
PTGDS−7.008
C10orf116−6.789
ABI3BP−6.529
TAGLN−4.285
miR-338-3p−6.574HIST2H4B2.862
CEACAM12.705
SF3B42.652
ZNF2382.477
MCM42.399
Physical interaction analysis

Fig. 2 shows the enrichment analysis for 696 DEGs using the ClueGO application in Cytoscape. The most relevant GO terms were positive regulation of a developmental process, regulation of multicellular organismal development and cell cycle regulation. Of these, deregulation of the cell cycle underlies the uncontrolled cell proliferation that characterizes the malignant phenotype. We analyzed the interaction between miRNAs and cell-cycle-related genes. Seven miRNAs were identified to have significant negative correlations with their target mRNAs (Fig. 3), as follows: Five miRNAs from the confirmed negative correlations between differentially expressed miRNA-mRNA targets (hsa-miR-490-5p, hsa-miR-423-5p, hsa-miR-328, hsa-miR-1 and hsa-let-7); hsa-miR-153, which is expected to regulate forkhead box M1, a gene identified as a putative hub in the protein-protein interaction network; and hsa-miR-215, which interacts with cell-cycle-related genes in miRcode (15).

Discussion

In the present study, we identified 222 significantly differentially expressed miRNAs using the massively parallel sequencing approach in human lung cancer tissue compared with normal lung tissue. Furthermore, we identified putative negative regulatory miRNA-mRNA target pairs. Nineteen miRNAs were upregulated, and 425 matched mRNAs were downregulated. Additionally, 30 miRNAs were downregulated, and 332 matched mRNAs were upregulated. miR-577, miR-301b, miR-944, miR-891a, miR-615-3p and miR-338-3p were highly differentially expressed in the current study.

We searched for putative target genes associated with lung cancer. miR-577 was generally upregulated (fold change=8.411) and negatively correlated with epidermal growth factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1, also known as fibulin-3). EFEMP1 belongs to the fibulin family of widely expressed extracellular matrix proteins that regulate cell proliferation. This family mediates cell-to-cell and cell-to-matrix communication, as well as providing organization and stabilization to extracellular matrix structures during organogenesis and vasculogenesis. Several studies have demonstrated that fibulin-3 and another fibulin family member, fibulin-5, antagonize tumor angiogenesis in vivo (16), suggesting that concerted deregulation of a set of antiangiogenic factors, including fibulin-3 and tissue inhibitor of metalloproteinase 3, contributes to tumor progression (17).

miR-301b is negatively correlated with homeobox A5 (HOXA5), which is known to be a tumor suppressor gene whose gene product positively regulates the expression of the TP53 tumor suppressor gene (18). Liu et al suggested that microRNA-196a promotes NSCLC cell proliferation and invasion by targeting HOXA5 (19). However, in our study, HOXA5 was negatively regulated by both miR-301b and miR-891a. Additionally, miR-301b was shown to be negatively correlated with secreted protein acidic and rich in cysteine-like 1 (SPARCL1) and hyaluronoglucosaminidase 1 (HYAL1). SPARCL1, also known as hevin, belongs to the matricellular protein family. SPARCL1 is downregulated in NSCLC (20). Additionally, Yu et al (21) reported that SPARCL1 is likely to be a significant negative regulator in the progression or metastasis of colorectal cancer. HYAL1, which encodes a lysosomal hyaluronidase, was downregulated in NSCLC tissue. Anedchenko et al (22) reported that HYAL1 and HYAL2 genes are downregulated in NSCLC, a finding that is consistent with our data.

miR-944 was negatively correlated with GATA binding protein 6 (GATA6), which is a member of a small family of zinc finger transcription factors that play a significant role in the regulation of cellular differentiation and organogenesis. This protein is expressed during early embryogenesis and localizes to endo- and mesodermally derived cells during later embryogenesis, thereby playing a significant role in lung development. Cheung et al (23) reported that lung adenocarcinoma progression is regulated in part by the lineage transcription factor GATA6.

miR 338-3p was generally downregulated (fold change=−6.574), and was negatively correlated with the putative target genes carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) and minichromosome maintenance protein 4 (MCM4). CEACAM1, a single-pass transmembrane type I glycoprotein, belongs to the carcinoembryonic antigen family. In lung cancer, accumulated immunohistochemical evidence indicates that epithelial CEACAM1 expression is associated with tumor metastasis and progression (24). Zhou et al demonstrated that CEACAM1 mRNA levels in tumors were higher those in adjacent tumor-free tissues, although not significantly (24). Additionally, MCM4 is one of six MCM proteins comprising the prereplicative complex that binds to replication origins in the G1 phase of the cell cycle and is essential for the initiation of DNA replication. Kikuchi et al (25) reported that MCM4 expression was higher in lung cancer cells than in adjacent normal bronchial epithelial cells (P<0.001), and high MCM4 expression was correlated with poorer differentiation (P<0.001).

miR-210 is one of the most consistently reported upregulated miRNAs in human lung cancer miRNA expression profiling studies (1,26). In our study, miR-210 was significantly differentially expressed (fold change=2.246; P=0.03). miR-210 has a number of validated targets associated with the regulation of mitochondrial metabolism, angiogenesis, cell cycle regulation and X-chromosome inactivation (1), and miR-210 is upregulated by hypoxia inducible factor 1α (HIF-1α) in response to hypoxic conditions. Recently, miR-210 was reported to increase the radioresistance of hypoxic cancer cells (27).

Our results were partly consistent with those of previous studies of miRNA in lung cancer, and novel miRNAs that may play an active role in cancer development were identified. Our study used the NGS approach to profile miRNA expression in human lung cancer tissue compared with matched normal lung tissue, although only a small number of samples were used. As the technology develops, the cost of sequencing is expected to decline, making it possible for NGS to be widely used in the future. The major advantages of NGS are its high-throughput capability, and that it is precise, accurate and repeatable. Its application includes new miRNA exploration, detection of miRNA, miRNA editing and isomiR and target mRNA detection. NGS of small RNAs facilitates the investigation of the ubiquitous and differentially expressed behavior of miRNAs, and will therefore promote miRNA research.

The current study had several limitations. Few samples were included, and no validation study was performed. Additionally, our study compared tumor and non-cancerous tissue only. The identification of markers associated with patient survival, disease prognosis or response to a specific anticancer drug is warranted. However, our results include overlapping miRNAs that were reported previously.

In conclusion, significantly differentially expressed miRNAs and mRNAs between lung cancer and normal tissue were identified by means of massively parallel sequencing. These miRNAs and their target genes may play a significant role in lung cancer. Further studies are warranted to fully understand the pathogenesis of NSCLC.

Acknowledgements

This study was supported by a grant from the National R&D Program for Cancer Control (1020420) and the Ministry for Health and Welfare and Basic Research Program through the National Research Foundation of Korea (C1010458-01-01) funded by the Ministry of Education, Republic of Korea.

References

1 

Võsa U, Vooder T, Kolde R, et al: Meta-analysis of microRNA expression in lung cancer. Int J Cancer. 132:2884–2893. 2013. View Article : Google Scholar

2 

Jemal A, Bray F, Center MM, et al: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Esteller M: Non-coding RNAs in human disease. Nat Rev Genet. 12:861–874. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Iorio MV, Ferracin M, Liu GC, et al: MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 65:7065–7070. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Yanaihara N, Caplen N, Bowman E, et al: Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 9:189–198. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Porkka KP, Pfeiffer MJ, Waltering KK, et al: MicroRNA expression profiling in prostate cancer. Cancer Res. 67:6130–6135. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Akao Y, Nakagawa Y and Naoe T: MicroRNA-143 and -145 in colon cancer. DNA Cell Biol. 26:311–320. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Yang H, Kong W, He L, et al: MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res. 68:425–433. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Sablok G, Milev I, Minkov G, et al: isomiRex: web-based identification of microRNAs, isomiR variations and differential expression using next-generation sequencing datasets. FEBS Lett. 587:2629–2634. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Trapnell C, Roberts A, Goff L, et al: Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nature protocols. 7:562–578. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Bindea G, Mlecnik B, Hackl H, et al: ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 25:1091–1093. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Shannon P, Markiel A, Ozier O, et al: Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Re. 13:2498–2504. 2003. View Article : Google Scholar

13 

Bindea G, Galon J and Mlecnik B: CluePedia Cytoscape plugin: Pathway insights using integrated experimental and in silico data. Bioinformatics. 29:661–663. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Kozomara A and Griffiths-Jones S: miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res. 39(Database issue): D152–D157. 2011. View Article : Google Scholar :

15 

Xiao F, Zuo Z, Cai G, et al: miRecords: an integrated resource for microRNA-target interactions. Nucleic Acids Res. 37(Database issue): D105–D110. 2009. View Article : Google Scholar :

16 

Albig AR, Neil JR and Schiemann WP: Fibulins 3 and 5 antagonize tumor angiogenesis in vivo. Cancer Res. 66:2621–2629. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Yue W, Dacic S, Sun Q, et al: Frequent inactivation of RAMP2, EFEMP1 and Dutt1 in lung cancer by promoter hypermethylation. Clin Cancer Res. 13:4336–4344. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Raman V, Martensen SA, Reisman D, et al: Compromised HOXA5 function can limit p53 expression in human breast tumours. Nature. 405:974–978. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Liu X, Lu K, Wang K, et al: MicroRNA-196a promotes non-small cell lung cancer cell proliferation and invasion through targeting HOXA5. BMC Cancer. 12:348. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Schraml P, Shipman R, Colombi M and Ludwig CU: Identification of genes differentially expressed in normal lung and non-small cell lung carcinoma tissue. Cancer Res. 54:5236–5240. 1994.PubMed/NCBI

21 

Yu S, Yu J, Ge W, et al: SPARCL1, Shp2, MSH2, E-cadherin, p53, ADCY-2 and MAPK are prognosis-related in colorectal cancer. World J Gastroenterol. 17:2028–2036. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Anedchenko EA, Dmitriev AA, Krasnov GS, et al: Down-regulation of RBSP3/CTDSPL, NPRL2/G21, RASSF1A, ITGA9, HYAL1 and HYAL2 genes in non-small cell lung cancer. Mol Biol. 42:965–976. 2008.(in Russian). View Article : Google Scholar

23 

Cheung W, Zhao M, Liu Z, et al: Control of alveolar differentiation by the lineage transcription factors GATA6 and HOPX inhibits lung adenocarcinoma metastasis. Cancer Cell. 23:725–738. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Zhou M, Du Y, Liu Y, et al: Clinical and experimental studies regarding the expression and diagnostic value of carcinoembryonic antigen-related cell adhesion molecule 1 in non-small-cell-lung cancer. BMC Cancer. 13:3592013. View Article : Google Scholar

25 

Kikuchi J, Kinoshita I, Shimizu Y, et al: Minichromosome maintenance (MCM) protein 4 as a marker for proliferation and its clinical and clinicopathological significance in non-small cell lung cancer. Lung Cancer. 72:229–237. 2011. View Article : Google Scholar

26 

Guan P, Yin Z, Li X, et al: Meta-analysis of human lung cancer microRNA expression profiling studies comparing cancer tissues with normal tissues. J Exp Clin Cancer Res. 31:542012. View Article : Google Scholar : PubMed/NCBI

27 

Grosso S, Doyen J, Parks SK, et al: MiR-210 promotes a hypoxic phenotype and increases radioresistance in human lung cancer cell lines. Cell Death Dis. 4:e5442013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 11 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee H, Han S, Rhee H, Park JH, Lee JS, Oh Y, Choi SS, Shin S and Kim WJ: Differential expression of microRNAs and their target genes in non-small-cell lung cancer. Mol Med Rep 11: 2034-2040, 2015
APA
Lee, H., Han, S., Rhee, H., Park, J.H., Lee, J.S., Oh, Y. ... Kim, W.J. (2015). Differential expression of microRNAs and their target genes in non-small-cell lung cancer. Molecular Medicine Reports, 11, 2034-2040. https://doi.org/10.3892/mmr.2014.2890
MLA
Lee, H., Han, S., Rhee, H., Park, J. H., Lee, J. S., Oh, Y., Choi, S. S., Shin, S., Kim, W. J."Differential expression of microRNAs and their target genes in non-small-cell lung cancer". Molecular Medicine Reports 11.3 (2015): 2034-2040.
Chicago
Lee, H., Han, S., Rhee, H., Park, J. H., Lee, J. S., Oh, Y., Choi, S. S., Shin, S., Kim, W. J."Differential expression of microRNAs and their target genes in non-small-cell lung cancer". Molecular Medicine Reports 11, no. 3 (2015): 2034-2040. https://doi.org/10.3892/mmr.2014.2890