Open Access

Regulation of TPD52 by antitumor microRNA-218 suppresses cancer cell migration and invasion in lung squamous cell carcinoma

  • Authors:
    • Tomohiro Kumamoto
    • Naohiko Seki
    • Hiroko Mataki
    • Keiko Mizuno
    • Kazuto Kamikawaji
    • Takuya Samukawa
    • Keiichi Koshizuka
    • Yusuke Goto
    • Hiromasa Inoue
  • View Affiliations

  • Published online on: September 13, 2016     https://doi.org/10.3892/ijo.2016.3690
  • Pages: 1870-1880
  • Copyright: © Kumamoto 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 development of targeted molecular therapies has greatly benefited patients with lung adenocarcinomas. In contrast, these treatments have had little benefit in the management of lung squamous cell carcinoma (lung SCC). Therefore, new treatment options based on current genomic approaches are needed for lung SCC. Aberrant microRNA (miRNA) expression has been shown to promote lung cancer development and aggressiveness. Downregulation of microRNA-218 (miR-218) was frequently observed in our miRNA expression signatures of cancers, and previous studies have shown an antitumor function of miR-218 in several types of cancers. However, the impact of miR-218 on lung SCC is still ambiguous. The present study investigated the antitumor roles of miR-218 in lung SCC to identify the target genes regulated by this miRNA. Ectopic expression of miR-218 greatly inhibited cancer cell migration and invasion in the lung SCC cell lines EBC-1 and SK-MES-1. Through a combination of in silico analysis and gene expression data searching, tumor protein D52 (TPD52) was selected as a putative target of miR-218 regulation. Moreover, direct binding of miR-218 to the 3'-UTR of TPD52 was observed by dual luciferase reporter assay. Overexpression of TPD52 was observed in lung SCC clinical specimens, and knockdown of TPD52 significantly suppressed cancer cell migration and invasion in lung SCC cell lines. Furthermore, the downstream pathways mediated by TPD52 involved critical regulators of genomic stability and mitotic checkpoint genes. Taken together, our data showed that downregulation of miR-218 enhances overexpression of TPD52 in lung SCC cells, promoting cancer cell aggressiveness. Identification of tumor-suppressive miRNA-mediated RNA networks of lung SCC will provide new insights into the potential mechanisms of the molecular pathogenesis of the disease.

Introduction

Lung cancer remains the most frequent cause of cancer-related death in developed countries (1). Approximately 80% of lung cancers are categorized as non-small cell lung cancer (NSCLC), and NSCLC is divided into four major subtypes pathologically: adenocarcinoma, squamous cell carcinoma (SCC), large cell carcinoma and neuroendocrine cancer (2). Development of targeted molecular therapies has remarkably improved the overall survival of patients with adenocarcinoma (36). In contrast, targeted molecular therapies have offered few benefits in the management of lung SCC (7). Therefore, there is a need for effective treatment options based on current genomic approaches for lung SCC.

After the human genome sequencing era, the discovery of an extremely large number of non-coding RNAs conceptually transformed cancer research. MicroRNAs (miRNAs) are small non-coding RNAs involved in the repression or degradation of target RNA transcripts in a sequence-dependent manner (1,2). The ability of miRNAs is unique; a single miRNA may regulate a vast number of protein-coding or non-coding RNAs in human cells. Therefore, aberrantly expressed miRNAs may upset tightly regulated cellular RNA networks. This failing of the RNA network contributes to cancer development, aggressiveness and drug resistance (35). In fact, aberrantly expressed miRNAs have been reported in various types of human cancers, including lung SCC (68). Based on miRNA expression signatures, we identified tumor-suppressive miRNAs and the cancer RNA networks regulated by these miRNAs (911). We hypothesize that elucidation of tumor-suppressive miRNA-regulated oncogenic networks will provide new insights into the potential molecular mechanisms of lung SCC.

In our earlier studies determining miRNA expression signatures, we showed that miRNA-218 was significantly downregulated in several types of cancer tissues (1215). Our previous studies also demonstrated that downregulation of miR-218 enhanced overexpression of extracellular matrix (ECM) protein components or actin-related proteins, and this promoted cancer cell migration and invasion (1618). Tumor-suppressive roles of miR-218 were reported in several types of cancer. However, the impact of miR-218 on lung SCC remains ambiguous.

The aim of the present study was to investigate the functional significance of miR-218 in lung SCC and to identify molecular targets regulated by this miRNA. We found that restoration of miR-218 significantly suppressed cancer cell migration and invasion. Using luciferase reporter assay, tumor protein D52 (TPD52) was shown to be directly regulated by miR-218. Overexpression of TPD52 was observed in lung SCC clinical specimens and downregulation of the TPD52 gene significantly inhibited cancer cell aggressiveness. miR-218/ TPD52-regulated RNA networks may provide new insights into the potential mechanisms of lung SCC pathogenesis.

Materials and methods

Clinical specimens, cancer cell lines and RNA extraction

In all, 31 lung SCC specimens and 24 non-cancerous lung specimens were obtained from patients who underwent thoracic surgery at Kagoshima University Hospital from 2010 to 2013. The present study was approved by the Institutional Review Board for Clinical Research of the Kagoshima University School of Medicine. Prior written informed consent and approval were obtained from each patient. Table I describes the patient background and clinical characteristics of collected specimens. Samples were staged using the TNM scheme according to the International Association for the Study of Lung Cancer and were graded histologically (19). These archival formalin-fixed paraffin-embedded (FFPE) samples were subsequently studied by qRT-PCR analysis and immunohistochemistry as previously described (911).

Table I

Characteristics of the lung cancer and non-cancerous cases.

Table I

Characteristics of the lung cancer and non-cancerous cases.

A, Characteristics of the lung cancer cases

Lung cancer patientsn(%)
Total no31
Median age (range)70 (50–88)
Gender
 Male29(93.5)
 Female2(6.5)
Pathological stage
 IA4(12.9)
 IB9(29.0)
 IIA4(12.9)
 IIB5(16.1)
 IIIA8(25.8)
 IIIB1(3.2)

B, Characteristics of the non-cancerous cases

Non-cancerous tissuesn

Total number24
Median age (range)69 (50–88)
Gender
 Male24
 Female0

The human lung SCC cell lines EBC-1 and SK-MES-1 were acquired from the Japanese Cancer Research Resources Bank (Osaka, Japan) and the American Type Culture Collection ATCC; Manassas, VA, USA), respectively.

Total RNA was isolated using Isogen II (code no: 311-07361; Nippon Gene, Co., Ltd., Tokyo, Japan) as directed by the manufacturer.

Quantitative reverse transcription PCR (qRT-PCR)

PCR quantification was performed as previously described (911). The expression of miR-218 was determined using stem-loop RT-PCR as directed by the manufacturer (P/N: 000521; Applied Biosystems, Foster City, CA, USA). The TaqMan probe and TPD52 primers were from Assay-on-Demand™ Gene Expression products (P/N: Hs00893105_m1; Applied Biosystems). For quantification, miRNA and mRNA data were normalized against human RNU48 (P/N: 001006; Applied Biosystems) and GUSB (P/N: Hs99999908_m1; Applied Biosystems), respectively.

Transfection of mature miRNA and small interfering RNA (siRNA)

Pre-miR™ miRNA precursors for miR-218 (hsa-miR-218-5p, P/N: AM1 7100; Applied Biosystems) and negative control miRNA (P/N: AM 17111; Applied Biosystems), Stealth Select RNAi siRNA, si-TPD52 (P/N: HSS120730 and HSS120731; Invitrogen, Carlsbad, CA, USA), and negative control siRNA (P/N: 4390843; Invitrogen) were used in this study. EBC-1 and SK-MES-1 cells in Opti-MEM medium (cat. no. 31985070; Thermo Fisher Scientific, Waltham, MA, USA) were transfected with Lipofectamine RNAiMAX transfection reagent (P/N: 56532; Invitrogen) with 10 nM mature miRNA or siRNA.

Cell proliferation, migration and invasion assays

Cell proliferation was determined by XTT assay using Cell Proliferation kit (SKU: 20-300-1000; Biological Industries, Kibbutz Beit Haemek, Israel). Cell migration activity was analyzed by wound-healing assay, and cell invasion was analyzed using Corning BioCoat Matrigel Invasion chamber (cat. no. 354480; BD Biosciences, Bedford, MA, USA). The cell proliferation, migration, and invasion assays were carried out as previously described (911).

Identification of putative miR-218 target genes in lung SCC cells

Genome-wide gene expression analysis of miR-218-transfected EBC-1 cells was performed (accession number: GSE77790). Putative miR-218-regulated genes were identified by searching the TargetScan database (http://www.targetscan.org/). We then examined the expression levels of putative miR-218 targets in lung SCC clinical expression data from the GEO database (accession number: GSE19188). Oligo-microarray procedures and data mining methods were conducted as previously described (20,21).

Western blot analysis

Cells were harvested 96 h after transfection, and proteins were extracted from lysed cells. Protein lysates (20 μg) were separated on NuPAGE 4–12% Bis-Tris gels (cat. no. NP0323BOX; Invitrogen) before transfer of proteins to a polyvinylidene fluoride membrane. Immunoblotting was performed using diluted primary anti-TPD52 antibodies (1:250 dilution; Human Protein Atlas no. HPA028427; Atlas Antibodies, Stockholm, Sweden) and anti-GAPDH antibodies (1:10,000 dilution; cat. no. MAB374; Chemicon International, Inc., Temecula, CA, USA). These assays were carried out as previously described (911).

Plasmid construction and dual-luciferase reporter assay

The procedure for the dual-luciferase reporter assay was previously described (911). A partial sequence of the wild-type TPD52 3′-UTR containing the miR-218 target site or the TPD52 3′-UTR partial sequence lacking the miR-218 target site was cloned into the psiDHECK-2 vector between the XhoI-PmeI restriction sites in the 3′-UTR of the hRluc gene (cat. no. C8021; Promega, Madison, WI, USA).

Immunohistochemistry

The expression status of TPD52 in lung SCC clinical specimens (BC04002; US Biomax, Inc., Rockville, MD, USA) was confirmed via immunohistochemistry using an UltraVision Detection system (cat. no. TP-015-HD, Thermo Fisher Scientific) according to the manufacturer's protocol. Tissues were incubated with primary rabbit polyclonal anti-TPD52 antibodies (1:3,000 dilution; HPA028427) then treated with biotinylated goat anti-rabbit secondary antibodies. Antibodies were visualized using diaminobenzidine hydrogen peroxidase as the chromogen, and slides were counterstained with 0.5% hematoxylin.

Identification of downstream targets regulated by TPD52 in lung SCC

Gene expression analysis using si-TPD52-transfected EBC-1 cells identified molecular targets regulated by TPD52 in lung SCC cells. Microarray expression profiles of si-TPD52 transfectants were compiled and deposited into the GEO database (accession number: GSE82108).

Statistical analysis

RT-PCR results were analyzed using Mann-Whitney U tests to evaluate the relationships between the 2 groups, while Bonferroni-adjusted Mann-Whitney U tests were used to analyze the relationships among three or more variables. All analyses were performed using Expert StatView (version 5; SAS Institute Inc., Cary, NC, USA).

Results

Expression of miR-218 in lung SCC tissues and cell lines

To confirm the miRNA expression signatures of lung SCC cells, we evaluated the expression level of miR-218 in lung SCC tissues (n=31) and non-cancerous lung tissues (n=24). The patient clinicopathological features are summarized in Table I. The expression level of miR-218 was significantly downregulated in cancer tissues compared to non-cancerous tissues (P<0.0001; Fig. 1A). Expression of miR-218 in EBC-1 and SK-MES-1 cell lines was also lower than in non-cancerous lung tissues (Fig. 1A).

Effects of miR-218 restoration on the proliferation, migration and invasion of EBC-1 and SK-MES-1 cells

To investigate the antitumor activity of miR-218, we performed gain-of-function assays using miRNA transfection into EBC-1 and SK-MES-1 cells. Cell proliferation assays showed that cancer cell growth was slightly inhibited in miR-218 transfectants compared with mock- or miR control-transfected EBC-1 and SK-MES-1 cells (Fig. 1B). Cancer cell migration and invasion activities were significantly inhibited in miR-218 transfectants compared with mock- or miR-control-transfectants (P<0.0001; Fig. 1C and D).

Identification of putative targets regulated by miR-218 in EBC-1 cells

To identify putative targets of miR-218, we employed a combination of in silico analysis, genome-wide gene expression analysis and analysis of gene expression in clinical specimens using the GEO database. First, we identified 513 genes downregulated by miR-218 transfection into EBC-1 cells (log2 ratio <−1.0). Among these 513 genes, 163 genes have putative miR-218 binding sites in their 3′-UTR regions according to the TargetScan database. Next, we cross-referenced these genes with genes upregulated (fold change >1.5) in NSCLC clinical specimens from the GEO gene expression data set (accession number: GSE19188). A total of 36 candidate genes were identified as putative target genes of miR-218 regulation in lung SCC (Table II). Fig. 2 shows our strategy for selecting putative miR-218 target genes.

Table II

Downregulated genes in miR-218 tranfectant.

Table II

Downregulated genes in miR-218 tranfectant.

Entrez gene IDGene symbolDescriptionmiR-218 target siteEBC-1 miR-218 transfectant (log2 ratio)GSE: 19188 (fold change)

TotalConserved sitePoorly conserved site
7163TPD52Tumor protein D52422−1.332.66
5563PRKAA2Protein kinase, AMP-activated, α 2 catalytic subunit404−1.042.50
2673GFPT1 Glutamine-fructose-6-phosphate transaminase 1413−1.171.92
332BIRC5Baculoviral IAP repeat containing 5202−2.0512.42
56938ARNTL2Aryl hydrocarbon receptor nuclear translocator-like 2202−1.097.85
112399EGLN3Egl-9 family hypoxia-inducible factor 3211−1.455.97
84181CHD6chromodomain helicase DNA binding protein 6211−1.352.30
29927SEC61A1Sec61 α 1 subunit (S. cerevisiae)211−1.092.13
8776MTMR1Myotubularin related protein 1211−1.061.96
131566DCBLD2Discoidin, CUB and LCCL domain containing 2220−1.461.88
2820GPD2 Glycerol-3-phosphate dehydrogenase 2 (mitochondrial)202−1.021.83
399694SHC4SHC (Src homology 2 domain containing) family, member 4211−1.251.58
3486IGFBP3Insulin-like growth factor binding protein 3101−2.094.12
144406WDR66WD repeat domain 66101−1.112.98
25907TMEM158Transmembrane protein 158 (gene/pseudogene)101−1.122.96
10447FAM3CFamily with sequence similarity 3, member C110−2.082.90
4017LOXL2Lysyl oxidase-like 2101−1.962.56
9139CBFA2T2Core-binding factor, runt domain, α subunit 2; translocated to, 2101−1.112.42
3691ITGB4Integrin, β 4101−1.122.20
6745SSR1Signal sequence receptor, α110−1.471.92
1457CSNK2A1Casein kinase 2, α 1 polypeptide110−1.301.90
157638FAM84BFamily with sequence similarity 84, member B101−1.021.90
949SCARB1Scavenger receptor class B, member 1101−1.541.84
2011MARK2MAP/microtubule affinity-regulating kinase 2110−1.411.75
9289GPR56G protein-coupled receptor 56101−1.531.70
55959SULF2Sulfatase 2101−1.111.67
65985AACSAcetoacetyl-CoA synthetase101−1.101.63
160AP2A1Adaptor-related protein complex 2, α 1 subunit110−1.141.63
54928IMPAD1Inositol monophosphatase domain containing 1110−1.011.62
1889ECE1Endothelin converting enzyme 1101−1.551.61
55609ZNF280CZinc finger protein 280C110−1.101.61
6533SLC6A6Solute carrier family 6 (neurotransmitter transporter), member 6110−1.131.61
80000GREB1LGrowth regulation by estrogen in breast cancer-like101−1.151.59
10776ARPP19cAMP-regulated phosphoprotein, 19 kDa110−1.331.52
83637ZMIZ2Zinc finger, MIZ-type containing 2110−1.591.52
79139DERL1Derlin 1101−1.161.51

In the present study, we focused on the TPD52 gene, investigating TPD52 function and downregulated TPD52-mediated pathways.

TPD52 is directly targeted by miR-218 in lung SCC cells

First, we measured the expression of TPD52/TPD52 using quantitative RT-PCR and western blotting to determine whether restoration of miR-218 in EBC-1 and SK-MES-1 cells suppressed the expression of TPD52/TPD52. As shown in Fig. 3A and B, the expression levels of TPD52/TPD52 were markedly repressed by miR-218 transfection compared with mock- or miR-control-transfected cells.

Next, we used luciferase reporter assays to determine whether miR-218 directly binds to the 3′-UTR of TPD52 mRNA. We used vectors encoding either partial wild-type sequences of the 3′-UTR of TPD52 including the predicted miR-218 target sites (positions 52–58, 778–784, 940–946 of the TPD52 3′-UTR), or deletion vectors lacking these sites (Fig. 3C). The luminescence intensities were significantly reduced by transfection with miR-218 and the vector carrying the wild-type 3′-UTR of TPD52, whereas transfection with the deletion vector blocked the decrease of luminescence in EBC-1 cells (P<0.0001). Thus, miR-218 bound directly to three sites in the 3′-UTR of TPD52 (Fig. 3D–F).

Effects of TPD52 silencing on cell proliferation, migration and invasion in lung SCC cell lines

We performed loss-of-function studies using si-TPD52 to investigate the oncogenic function of TPD52 in lung SCC cells. First, we measured the knockdown efficiency of si-TPD52 transfection in EBC-1 and SK-MES-1 cells. RT-PCR and western blotting showed that two siRNAs (si-TPD52-1 and si-TPD52-2) could effectively reduce the expression of TPD52/TPD52 in EBC-1 and SK-MES-1 cells (Fig. 4A and B).

Next, we carried out functional assays using these two siRNAs. XTT assays demonstrated that cell proliferation was inhibited by si-TPD52 transfection only in SK-MES-1 cells (Fig. 4C). Cell migration and invasion activities were significantly inhibited by si-TPD52 transfection in both EBC-1 and SK-MES-1 cells in comparison with mock- or negative-control transfectants (Fig. 4D and E).

TPD52 was strongly expressed in clinical lung SCC specimens

To analyze whether TPD52 was upregulated in lung SCC clinical specimens, we carried out immunohistochemical staining of lung SCC and non-cancerous lung tissues. Thirty specimens were stained in this study (20 lung SCC specimens and 10 non-cancerous tissues); all lung SCC specimens stained moderately or strongly, whereas all non-cancerous specimens stained weakly or negatively for TPD52 (Table III and Fig. 5).

Table III

Immunohistochemistry status and characteristics of the lung cancer and non-cancerous cases.

Table III

Immunohistochemistry status and characteristics of the lung cancer and non-cancerous cases.

A, Immunohistochemistry status and characteristics of the lung squamous cell carcinoma cases

Patient no.GradeTNMPathological stage Immunohistochemistry
11310IIIa(++)
21300IIIa(++)
32210II(++)
42300IIIa(++)
51200I(++)
61210II(+)
71310IIIa(+++)
81200I(++)
91210II(++)
101200I(++)
112220IIIa(+++)
122200I(+)
132100I(++)
142100I(++)
152210II(+)
162310III(++)
172200I(++)
182210II(+)
192320IIIa(+++)
202200I(++)

B, Immunohistochemistry status of non-cancerous cases

Patient no. Immunohistochemistry

91(−)
92(+)
93(+)
94(−)
95(−)
96(−)
97(−)
98(+)
99(+)
100(+)
Identification of TPD52-mediated downstream pathways in lung SCC cells

To identify the downstream genes regulated by TPD52, genome-wide gene expression analysis and in silico analysis were performed in lung SCC cells transfected with si-TPD52. A total of 2,278 genes were identified as downregulated in si-TPD52-transfected EBC-1 cells compared with the control (log2 ratio <−0.5). Among them, 215 genes were upregulated in NSCLC specimens in the GEO database (accession number: GSE19188). We categorized the 215 genes according to KEGG pathways, and 7 pathways were identified as significantly enriched pathways (Table IV). Fig. 6 shows our strategy for selecting TPD52-mediated downstream pathways. Among these pathways, we focused on the ‘Cell cycle pathway’, ‘DNA replication pathways’ and ‘p53 signaling pathway’. Genes involved in these pathways are listed in Table V.

Table IV

Significantly enriched annotations regulated by si-TPD52-1 and si-TPD52-2 in lung SCC cells.

Table IV

Significantly enriched annotations regulated by si-TPD52-1 and si-TPD52-2 in lung SCC cells.

No. of genesP-valueAnnotations
122.06E-11(KEGG) 04110: Cell cycle
88.84E-07(KEGG) 04114: Oocyte meiosis
72.27E-06(KEGG) 05322: Systemic lupus erythematosus
55.41E-06(KEGG) 03030: DNA replication
62.30E-05(KEGG) 04914: Progesterone-mediated oocyte maturation
59.53E-05(KEGG) 04115: p53 signaling pathway
24.91E-02(KEGG) 03410: Base excision repair

Table V

Representative pathways identified as downstream of TPD52.

Table V

Representative pathways identified as downstream of TPD52.

Cell cycle pathway

Gene symbolDescriptionGSE:19188 (log2 ratio)
TTKTTK protein kinase3.23
BUB1Budding uninhibited by benzimidazoles 1 homolog (yeast)3.21
CCNB2Cyclin B23.20
MCM4Minichromosome maintenance complex component 43.13
CCNB1Cyclin B12.82
PLK1Polo-like kinase 12.81
CDC25CCell division cycle 25 homolog C (S. pombe)2.77
CDK1Cyclin-dependent kinase 12.40
CCNE2Cyclin E22.04
DBF4DBF4 homolog (S. cerevisiae)1.42
MCM6Minichromosome maintenance complex component 61.31
BUB3Budding uninhibited by benzimidazoles 3 homolog (yeast)1.07

DNA replication pathway

Gene symbolDescriptionGSE:19188 (log2 ratio)

MCM4Minichromosome maintenance complex component 43.13
FEN1Flap structure-specific endonuclease 11.66
MCM6Minichromosome maintenance complex component 61.31
PRIM1Primase, DNA, polypeptide 1 (49 kDa)1.25
POLA2Polymerase (DNA directed), α 2 (70 kD subunit)1.00

p53 signaling pathway

Gene symbolDescriptionGSE:19188 (log2 ratio)

CCNB2Cyclin B23.20
CCNB1Cyclin B12.82
GTSE1G-2 and S-phase expressed 12.79
CDK1Cyclin-dependent kinase 12.40
CCNE2Cyclin E22.04

Discussion

A single miRNA can regulate an extremely large number of protein-coding or non-coding RNAs. Thus, aberrant expression of miRNAs disrupts the RNA network in cancer cells. Identification of aberrantly expressed miRNAs and the novel cancer networks they regulate is a research trend of the post-genome-sequencing era. Recent evidence shows that disruption of normally-regulated RNA networks by aberrantly expressed miRNAs triggers cancer cell development, progression and metastasis (3,4). Our recent studies of miRNA expression signatures in various cancers indicated that miR-218 is frequently downregulated in cancers (911). Investigating miR-218-regulated RNA networks in lung SCC is the first step in construction of a new treatment strategy for the disease.

Our current data showed that expression of miR218 was significantly reduced in lung SCC specimens. The mature form of miR-218 is generated from two separate loci, miR-218-1 and miR-218-2, which are located on chromosomes 4p15.31 and 5q35.1 within the introns of SLIT2 and SLIT3, respectively (22). Several lines of evidence suggest that miR-218 is frequently downregulated in various cancers (17,18,23,24). In this study, ectopic expression of miR-218 significantly inhibited cancer cell migration and invasion, suggesting this miRNA suppresses metastasis-promoting genes. Our previous studies showed that antitumor effects of miR-218 explicitly contribute to migration and invasion in head and neck cancer, cervical cancer, renal cell carcinoma and prostate cancer (17,18,23,24). Interestingly, miR-218-regulated genes were involved in ECM-related components such as collagens, laminins and integrins (17,18,23,24). Accumulating evidence indicates that the interaction of cancer cells with their microenvironment influences the initiation, development and metastasis of cancer cells (25,26). Overexpression of ECM components and activation of ECM-integrin signaling were observed in several types of cancers and shown to activate cancer cell aggressiveness (27). The discovery of tumor-suppressive miR-218-regulated genes and pathways may provide important insights into the potential mechanisms of lung SCC metastasis.

To better understand lung SCC development and metastasis, we identified miR-218 target genes using a combination of in silico and genome-wide gene expression analyses. We have identified antitumor miRNA-regulated oncogenes and novel cancer networks using this strategy (911). In the present study, a total of 36 putative target genes of miR-218 were identified. Among these genes, we demonstrated that LOXL2 (lysyl oxidase homolog 2) was directly regulated by miR-218 in head and neck cancer and prostate cancer (28). Another group showed direct regulation of BIRC5 (baculoviral IAP repeat containing 5) by miR-218 in cervical cancer (29). These findings show the effectiveness of our strategy in identifying target genes of miR-218-regulation in cancer cells.

In the present study, we focused on TPD52 and investigated the functional significance of this gene in lung SCC. TPD52 was initially cloned through differential screening using a breast cancer cDNA library (30). This cDNA clone had novel sequences and was expressed in breast and basal cell carcinomas (30). Interestingly, this gene is located on the human chromosome 8q21.13, a frequently observed region of chromosomal amplification in several types of cancers (3133). In lung adenocarcinoma, chromosome 8q21.13, which includes TPD52, was one of the most notable amplified genomic regions (34). A vast number of studies showed that TPD52 is overexpressed both at the mRNA and protein levels in several cancers. Likewise, overexpression of TPD52 was reported in small cell lung cancer, lung adenocarcinoma and lung SCC (3537). The influence of smoking is important in lung cancer pathogenesis. Expression levels of TPD52 were enhanced in airway epithelial cells in smokers with lung cancer compared to smokers without cancer (38). The functional roles of TPD52 were investigated using overexpression or knockdown analytical methods. Ectopic expression of mouse TPD52 in 3T3 fibroblasts resulted in a transformed phenotype that progressed to metastasis (39). Several studies have shown that increased expression of TPD52 enhances the proliferation of prostate cancer cell lines under both normal and androgen-resistant conditions (40,41). Other studies showed that TPD52 knockdown increases apoptotic cell death in ERBB2-amplified breast cancer cell lines (33). These findings suggest that over-expression of TPD52 enhances cancer cell aggressiveness and contributes to several oncogenic pathways.

Recently, several studies demonstrated that TPD52 expression is regulated by several miRNAs in cancer cells. Downregulation of TPD52 expression was observed by miR-107 and miR-185 transfection into non-small cell lung cancer cells and miR-34a transfection into colorectal cancer cells (42,43). Our previous data showed that the tumor-suppressive miR-224 directly regulates oncogenic TPD52 in prostate cancer cells, and silencing TPD52 results in significant reductions in cancer cell migration and invasion in prostate cancer cells (44). More recently, miR-218 was shown to directly regulate TPD52 in prostate cancer cells (45). These data agree with our present report in lung SCC and support our conclusions. Control of TPD52 expression by miRNAs is a novel molecular mechanism of cancer cells, and further investigation of the miRNA-TPD52 axis is needed.

In the present study, we identified TPD52-mediated cancer pathways using genome-wide gene expression analysis of si-TPD52-transfected lung SCC cells. Our data showed that several pathways were identified downstream of TPD52 pathways, such as the ‘Cell cycle pathway’, ‘DNA replication pathway’ and ‘p53 signaling pathway’. The genes involved in these pathways were critical regulators of genomic stability and were mitotic checkpoint genes (46,47). The functional insights obtained in the current studies indicated that several cell cycle kinases, such as TTK, BUB1 and PLK1, were multi-functional enzymes and contributed to cancer cell migration, invasion and metastasis (4850). The protein kinase TTK promoted the cell proliferation and migration through activation of AKT-mTOR and MDM2-p53 signaling pathways in hepatocellular carcinoma cells (48). Using the siRNA screen of the human kinome revealed that a serine/threonine kinase BUB1 acted as an essential mediator of TGFβ-dependent signaling (49). This study demonstrated that BUB1 interacted with both, TGFBRI and TGFBRII and promoted the TGFβ-dependent epithelial mesenchymal transition (EMT), cell migration and invasion (49). PLK1 is a serine/threonine kinase and a pivotal player of cell cycle regulator (50). Overexpression of PLK1 was observed in prostate cancer and involved in enhancing EMT and stimulation of cell migration and invasion (50). In bladder cancer, CCNB2 was overexpressed in cancer cells and knockdown of CCNB2 inhibited invasion and metastatic abilities (51). These findings have supported our present data of knockdown of TPD52 in lung cancer cells. Exploration of novel TPD52-mediated pathways may lead to the development of new treatment protocols for this disease.

In conclusion, expression of miR-218 was frequently downregulated in lung SCC clinical specimens and appeared to function in anti-migration and anti-invasion roles through targeting of TPD52. Elucidation of miR-218-regulated cancer networks should provide new information on potential therapeutic targets in the treatment of lung SCC development and metastasis.

Acknowledgements

We thank Dr Masami Sato (Department of General Thoracic Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan) for kindly providing surgically resected samples. The present study was supported in part by the Grants-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (JSPS, KAKENHI grants 15K09179 and 16K19458).

References

1 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Filipowicz W, Bhattacharyya SN and Sonenberg N: Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat Rev Genet. 9:102–114. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Hobert O: Gene regulation by transcription factors and microRNAs. Science. 319:1785–1786. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Iorio MV and Croce CM: MicroRNAs in cancer: Small molecules with a huge impact. J Clin Oncol. 27:5848–5856. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Rolfo C, Fanale D, Hong DS, Tsimberidou AM, Piha-Paul SA, Pauwels P, Van Meerbeeck JP, Caruso S, Bazan V, Cicero G, et al: Impact of microRNAs in resistance to chemotherapy and novel targeted agents in non-small cell lung cancer. Curr Pharm Biotechnol. 15:475–485. 2014. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Raponi M, Dossey L, Jatkoe T, Wu X, Chen G, Fan H and Beer DG: MicroRNA classifiers for predicting prognosis of squamous cell lung cancer. Cancer Res. 69:5776–5783. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Moriya Y, Nohata N, Kinoshita T, Mutallip M, Okamoto T, Yoshida S, Suzuki M, Yoshino I and Seki N: Tumor suppressive microRNA-133a regulates novel molecular networks in lung squamous cell carcinoma. J Hum Genet. 57:38–45. 2012. View Article : Google Scholar

9 

Mataki H, Enokida H, Chiyomaru T, Mizuno K, Matsushita R, Goto Y, Nishikawa R, Higashimoto I, Samukawa T, Nakagawa M, et al: Downregulation of the microRNA-1/133a cluster enhances cancer cell migration and invasion in lung-squamous cell carcinoma via regulation of Coronin1C. J Hum Genet. 60:53–61. 2015. View Article : Google Scholar

10 

Mataki H, Seki N, Chiyomaru T, Enokida H, Goto Y, Kumamoto T, Machida K, Mizuno K, Nakagawa M and Inoue H: Tumor-suppressive microRNA-206 as a dual inhibitor of MET and EGFR oncogenic signaling in lung squamous cell carcinoma. Int J Oncol. 46:1039–1050. 2015.

11 

Mizuno K, Seki N, Mataki H, Matsushita R, Kamikawaji K, Kumamoto T, Takagi K, Goto Y, Nishikawa R, Kato M, et al: Tumor-suppressive microRNA-29 family inhibits cancer cell migration and invasion directly targeting LOXL2 in lung squamous cell carcinoma. Int J Oncol. 48:450–460. 2016.

12 

Kikkawa N, Hanazawa T, Fujimura L, Nohata N, Suzuki H, Chazono H, Sakurai D, Horiguchi S, Okamoto Y and Seki N: miR-489 is a tumour-suppressive miRNA target PTPN11 in hypopharyngeal squamous cell carcinoma (HSCC). Br J Cancer. 103:877–884. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Hidaka H, Seki N, Yoshino H, Yamasaki T, Yamada Y, Nohata N, Fuse M, Nakagawa M and Enokida H: Tumor suppressive microRNA-1285 regulates novel molecular targets: Aberrant expression and functional significance in renal cell carcinoma. Oncotarget. 3:44–57. 2012.PubMed/NCBI

14 

Itesako T, Seki N, Yoshino H, Chiyomaru T, Yamasaki T, Hidaka H, Yonezawa T, Nohata N, Kinoshita T, Nakagawa M, et al: The microRNA expression signature of bladder cancer by deep sequencing: The functional significance of the miR-195/497 cluster. PLoS One. 9:e843112014. View Article : Google Scholar : PubMed/NCBI

15 

Kojima S, Enokida H, Yoshino H, Itesako T, Chiyomaru T, Kinoshita T, Fuse M, Nishikawa R, Goto Y, Naya Y, et al: The tumor-suppressive microRNA-143/145 cluster inhibits cell migration and invasion by targeting GOLM1 in prostate cancer. J Hum Genet. 59:78–87. 2014. View Article : Google Scholar

16 

Tatarano S, Chiyomaru T, Kawakami K, Enokida H, Yoshino H, Hidaka H, Yamasaki T, Kawahara K, Nishiyama K, Seki N, et al: miR-218 on the genomic loss region of chromosome 4p15.31 functions as a tumor suppressor in bladder cancer. Int J Oncol. 39:13–21. 2011.PubMed/NCBI

17 

Kinoshita T, Hanazawa T, Nohata N, Kikkawa N, Enokida H, Yoshino H, Yamasaki T, Hidaka H, Nakagawa M, Okamoto Y, et al: Tumor suppressive microRNA-218 inhibits cancer cell migration and invasion through targeting laminin-332 in head and neck squamous cell carcinoma. Oncotarget. 3:1386–1400. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Yamamoto N, Kinoshita T, Nohata N, Itesako T, Yoshino H, Enokida H, Nakagawa M, Shozu M and Seki N: Tumor suppressive microRNA-218 inhibits cancer cell migration and invasion by targeting focal adhesion pathways in cervical squamous cell carcinoma. Int J Oncol. 42:1523–1532. 2013.PubMed/NCBI

19 

Goldstraw P, Crowley J, Chansky K, Giroux DJ, Groome PA, Rami-Porta R, Postmus PE, Rusch V and Sobin L; International Association for the Study of Lung Cancer International Staging Committee; Participating Institutions. The IASLC Lung Cancer Staging Project: Proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM Classification of malignant tumours. J Thorac Oncol. 2:706–714. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Fukumoto I, Kinoshita T, Hanazawa T, Kikkawa N, Chiyomaru T, Enokida H, Yamamoto N, Goto Y, Nishikawa R, Nakagawa M, et al: Identification of tumour suppressive microRNA-451a in hypopharyngeal squamous cell carcinoma based on microRNA expression signature. Br J Cancer. 111:386–394. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Matsushita R, Seki N, Chiyomaru T, Inoguchi S, Ishihara T, Goto Y, Nishikawa R, Mataki H, Tatarano S, Itesako T, et al: Tumour-suppressive microRNA-144-5p directly targets CCNE1/2 as potential prognostic markers in bladder cancer. Br J Cancer. 113:282–289. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Lu YF, Zhang L, Waye MM, Fu WM and Zhang JF: MiR-218 mediates tumorigenesis and metastasis: Perspectives and implications. Exp Cell Res. 334:173–182. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Yamasaki T, Seki N, Yoshino H, Itesako T, Hidaka H, Yamada Y, Tatarano S, Yonezawa T, Kinoshita T, Nakagawa M, et al: MicroRNA-218 inhibits cell migration and invasion in renal cell carcinoma through targeting caveolin-2 involved in focal adhesion pathway. J Urol. 190:1059–1068. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Nishikawa R, Goto Y, Sakamoto S, Chiyomaru T, Enokida H, Kojima S, Kinoshita T, Yamamoto N, Nakagawa M, Naya Y, et al: Tumor-suppressive microRNA-218 inhibits cancer cell migration and invasion via targeting of LASP1 in prostate cancer. Cancer Sci. 105:802–811. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Weaver VM, Petersen OW, Wang F, Larabell CA, Briand P, Damsky C and Bissell MJ: Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies. J Cell Biol. 137:231–245. 1997. View Article : Google Scholar : PubMed/NCBI

26 

Bissell MJ and Radisky D: Putting tumours in context. Nat Rev Cancer. 1:46–54. 2001. View Article : Google Scholar

27 

Hood JD and Cheresh DA: Role of integrins in cell invasion and migration. Nat Rev Cancer. 2:91–100. 2002. View Article : Google Scholar

28 

Fukumoto I, Kikkawa N, Matsushita R, Kato M, Kurozumi A, Nishikawa R, Goto Y, Koshizuka K, Hanazawa T, Enokida H, et al: Tumor-suppressive microRNAs (miR-26a/b, miR-29a/b/c and miR-218) concertedly suppressed metastasis-promoting LOXL2 in head and neck squamous cell carcinoma. J Hum Genet. 61:109–118. 2016. View Article : Google Scholar

29 

Kogo R, How C, Chaudary N, Bruce J, Shi W, Hill RP, Zahedi P, Yip KW and Liu FF: The microRNA-218~Survivin axis regulates migration, invasion, and lymph node metastasis in cervical cancer. Oncotarget. 6:1090–1100. 2015. View Article : Google Scholar :

30 

Byrne JA, Tomasetto C, Garnier JM, Rouyer N, Mattei MG, Bellocq JP, Rio MC and Basset P: A screening method to identify genes commonly overexpressed in carcinomas and the identification of a novel complementary DNA sequence. Cancer Res. 55:2896–2903. 1995.PubMed/NCBI

31 

Byrne JA, Balleine RL, Schoenberg Fejzo M, Mercieca J, Chiew YE, Livnat Y, St Heaps L, Peters GB, Byth K, Karlan BY, et al: Tumor protein D52 (TPD52) is overexpressed and a gene amplification target in ovarian cancer. Int J Cancer. 117:1049–1054. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Rubin MA, Varambally S, Beroukhim R, Tomlins SA, Rhodes DR, Paris PL, Hofer MD, Storz-Schweizer M, Kuefer R, Fletcher JA, et al: Overexpression, amplification, and androgen regulation of TPD52 in prostate cancer. Cancer Res. 64:3814–3822. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Roslan N, Bièche I, Bright RK, Lidereau R, Chen Y and Byrne JA: TPD52 represents a survival factor in ERBB2-amplified breast cancer cells. Mol Carcinog. 53:807–819. 2014. View Article : Google Scholar

34 

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

35 

Rohrbeck A, Neukirchen J, Rosskopf M, Pardillos GG, Geddert H, Schwalen A, Gabbert HE, von Haeseler A, Pitschke G, Schott M, et al: Gene expression profiling for molecular distinction and characterization of laser captured primary lung cancers. J Transl Med. 6:692008. View Article : Google Scholar : PubMed/NCBI

36 

Zhu H, Lam DC, Han KC, Tin VP, Suen WS, Wang E, Lam WK, Cai WW, Chung LP and Wong MP: High resolution analysis of genomic aberrations by metaphase and array comparative genomic hybridization identifies candidate tumour genes in lung cancer cell lines. Cancer Lett. 245:303–314. 2007. View Article : Google Scholar

37 

Hanada S, Kakehashi A, Nishiyama N, Wei M, Yamano S, Chung K, Komatsu H, Inoue H, Suehiro S and Wanibuchi H: Myristoylated alanine-rich C-kinase substrate as a prognostic biomarker in human primary lung squamous cell carcinoma. Cancer Biomark. 13:289–298. 2013.PubMed/NCBI

38 

Boelens MC, van den Berg A, Fehrmann RS, Geerlings M, de Jong WK, te Meerman GJ, Sietsma H, Timens W, Postma DS and Groen HJ: Current smoking-specific gene expression signature in normal bronchial epithelium is enhanced in squamous cell lung cancer. J Pathol. 218:182–191. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Lewis JD, Payton LA, Whitford JG, Byrne JA, Smith DI, Yang L and Bright RK: Induction of tumorigenesis and metastasis by the murine orthologue of tumor protein D52. Mol Cancer Res. 5:133–144. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Ummanni R, Teller S, Junker H, Zimmermann U, Venz S, Scharf C, Giebel J and Walther R: Altered expression of tumor protein D52 regulates apoptosis and migration of prostate cancer cells. FEBS J. 275:5703–5713. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Li L, Xie H, Liang L, Gao Y, Zhang D, Fang L, Lee SO, Luo J, Chen X, Wang X, et al: Increased PrLZ-mediated androgen receptor transactivation promotes prostate cancer growth at castration-resistant stage. Carcinogenesis. 34:257–267. 2013. View Article : Google Scholar :

42 

Takahashi Y, Forrest AR, Maeno E, Hashimoto T, Daub CO and Yasuda J: MiR-107 and MiR-185 can induce cell cycle arrest in human non small cell lung cancer cell lines. PLoS One. 4:e66772009. View Article : Google Scholar : PubMed/NCBI

43 

Kaller M, Liffers ST, Oeljeklaus S, Kuhlmann K, Röh S, Hoffmann R, Warscheid B and Hermeking H: Genome-wide characterization of miR-34a induced changes in protein and mRNA expression by a combined pulsed SILAC and microarray analysis. Mol Cell Proteomics. 10:M111 010462. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Goto Y, Nishikawa R, Kojima S, Chiyomaru T, Enokida H, Inoguchi S, Kinoshita T, Fuse M, Sakamoto S, Nakagawa M, et al: Tumour-suppressive microRNA-224 inhibits cancer cell migration and invasion via targeting oncogenic TPD52 in prostate cancer. FEBS Lett. 588:1973–1982. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Han G, Fan M and Zhang X: microRNA-218 inhibits prostate cancer cell growth and promotes apoptosis by repressing TPD52 expression. Biochem Biophys Res Commun. 456:804–809. 2015. View Article : Google Scholar

46 

Aarts M, Linardopoulos S and Turner NC: Tumour selective targeting of cell cycle kinases for cancer treatment. Curr Opin Pharmacol. 13:529–535. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Dominguez-Brauer C, Thu KL, Mason JM, Blaser H, Bray MR and Mak TW: Targeting mitosis in cancer: Emerging strategies. Mol Cell. 60:524–536. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Liu X, Liao W, Yuan Q, Ou Y and Huang J: TTK activates Akt and promotes proliferation and migration of hepatocellular carcinoma cells. Oncotarget. 6:34309–34320. 2015.PubMed/NCBI

49 

Nyati S, Schinske-Sebolt K, Pitchiaya S, Chekhovskiy K, Chator A, Chaudhry N, Dosch J, Van Dort ME, Varambally S, Kumar-Sinha C, et al: The kinase activity of the Ser/Thr kinase BUB1 promotes TGF-β signaling. Sci Signal. 8:ra12015. View Article : Google Scholar

50 

Wu J, Ivanov AI, Fisher PB and Fu Z: Polo-like kinase 1 induces epithelial-to-mesenchymal transition and promotes epithelial cell motility by activating CRAF/ERK signaling. eLife. 5:52016. View Article : Google Scholar

51 

Lei CY, Wang W, Zhu YT, Fang WY and Tan WL: The decrease of cyclin B2 expression inhibits invasion and metastasis of bladder cancer. Urol Oncol. 34:237.e1–237.e10. 2016. View Article : Google Scholar

Related Articles

Journal Cover

November-2016
Volume 49 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kumamoto T, Seki N, Mataki H, Mizuno K, Kamikawaji K, Samukawa T, Koshizuka K, Goto Y and Inoue H: Regulation of TPD52 by antitumor microRNA-218 suppresses cancer cell migration and invasion in lung squamous cell carcinoma. Int J Oncol 49: 1870-1880, 2016
APA
Kumamoto, T., Seki, N., Mataki, H., Mizuno, K., Kamikawaji, K., Samukawa, T. ... Inoue, H. (2016). Regulation of TPD52 by antitumor microRNA-218 suppresses cancer cell migration and invasion in lung squamous cell carcinoma. International Journal of Oncology, 49, 1870-1880. https://doi.org/10.3892/ijo.2016.3690
MLA
Kumamoto, T., Seki, N., Mataki, H., Mizuno, K., Kamikawaji, K., Samukawa, T., Koshizuka, K., Goto, Y., Inoue, H."Regulation of TPD52 by antitumor microRNA-218 suppresses cancer cell migration and invasion in lung squamous cell carcinoma". International Journal of Oncology 49.5 (2016): 1870-1880.
Chicago
Kumamoto, T., Seki, N., Mataki, H., Mizuno, K., Kamikawaji, K., Samukawa, T., Koshizuka, K., Goto, Y., Inoue, H."Regulation of TPD52 by antitumor microRNA-218 suppresses cancer cell migration and invasion in lung squamous cell carcinoma". International Journal of Oncology 49, no. 5 (2016): 1870-1880. https://doi.org/10.3892/ijo.2016.3690