Downregulation of matrix metalloproteinase 14 by the antitumor miRNA, miR-150-5p, inhibits the aggressiveness of lung squamous cell carcinoma cells

  • Authors:
    • Takayuki Suetsugu
    • Keiichi Koshizuka
    • Naohiko Seki
    • Keiko Mizuno
    • Atsushi Okato
    • Takayuki Arai
    • Shunsuke Misono
    • Akifumi Uchida
    • Tomohiro Kumamoto
    • Hiromasa Inoue
  • View Affiliations

  • Published online on: December 21, 2017     https://doi.org/10.3892/ijo.2017.4232
  • Pages: 913-924
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the present study, in order to elucidate the aggressive nature of lung squamous cell carcinoma (LUSQ), we investigated the oncogenic RNA networks regulated by antitumor microRNAs (miRNAs or miRs) in LUSQ cells. The analysis of our original miRNA expression signatures of human cancers revealed that microRNA‑150‑5p (miR‑150‑5p) was downregulated in various types of cancer, indicating that miR‑150‑5p acts as an antitumor miRNA by targeting several oncogenic genes. Thus, the aims of this study were to investigate the antitumor roles of miR‑150‑5p in LUSQ cells and to identify oncogenes regulated by miR‑150‑5p that are involved in the aggressive behavior of LUSQ. The downregulation of miR‑150‑5p was validated in clinical samples of LUSQ and cell lines (SK-MES‑1 and EBC‑1). The ectopic overexpression of miR‑150‑5p significantly suppressed cancer cell aggressiveness. Comprehensive gene expression analyses revealed that miR‑150‑5p regulated 9 genes in the LUSQ cells. Among these, matrix metalloproteinase 14 (MMP14) was found to be a direct target of miR‑150‑5p, as shown by luciferase reporter assay. The knockdown of MMP14 using siRNA against MMP14 (si-MMP14) significantly inhibited cancer cell migration and invasion. The overexpression of MMP14 was detected in clinical specimens of LUSQ by immunohistochemistry. On the whole, these findings suggest that the downregulation of miR‑150‑5p and the overexpression of MMP14 may be deeply involved in the pathogenesis of LUSQ.

Introduction

The improvement of the treatment efficacy of lung cancer is an important issue worldwide. In 2012, 1.8 million individuals were diagnosed with lung cancer worldwide, resulting in the death of 1.6 million individuals (1). The majority of lung cancers (approximately 80%) are classified as non-small cell lung cancer (NSCLC). NSCLC is subdivided into four histopathological subtypes as follows: adenocarcinoma (LUAD), squamous cell carcinoma (LUSQ), large cell carcinoma and neuroendocrine cancer (2). The overall survival rate of patients with NSCLC is poor as almost half of patients have metastatic disease at initial diagnosis (3). In patients with adenocarcinoma, the survival rates are markedly improved by treatment with epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs), inhibitor of anaplastic lymphoma kinase (ALK) and immune check point drugs (4). By contrast, recent targeted molecular therapies have little benefit to the management of patients with LUSQ (57). Therefore, in order to improve the prognosis of patients with LUSQ, it is important to analyze the molecular mechanisms of metastatic pathways using the latest genomic approaches.

The human genome sequencing project has shown that a large proportion of non-coding RNAs (ncRNAs) are transcribed (8). The functions of ncRNAs are varied and they can act as effective regulatory molecules in a wide range of biological progresses (9). ncRNAs are categorized into two classes based on their molecular sizes: long ncRNAs (lncRNAs) and small ncRNAs (9). MicroRNAs (miRNAs or miRs) are members of the small ncRNA family. They are typically 19 to 23 nucleotides length, and they regulate the expression of protein-coding RNAs or non-coding RNAs (10). miRNAs possess unique properties, including the ability of a single miRNA species to regulate a vast number of protein-coding or ncRNAs in human cells (10). Thus, aberrantly expressed miRNAs can disrupt systematically regulated RNA networks in cancer cells. In fact, dysregulated miRNAs are deeply involved in the pathogenesis of human cancers (11).

To elucidate the aggressive nature of LUSQ, we previously identified miRNAs that play regulatory roles in this disease (1216). For example, our previous studies have revealed that the clustered miRNAs, miR-1 and miR-133a, markedly inhibit cancer cell aggressiveness by regulating actin binding protein CORO1C (12). All family members of miR-29 (miR-29a, miR-29b and miR-29c) act as anti-metastatic miRNAs by targeting lysyl oxidase homolog 2 (LOXL2) (13,14). The over-expression of LOXL2 has been observed in several types of cancer and the knockdown of LOXL2 interferes with cancer cell aggressiveness (17,18). Moreover, miR-206 has been shown to inhibit cancer cell malignancies by targeting two pivotal tyrosine kinase receptors, MET and EGFR, in LUSQ (16). These findings provide new knowledge into the novel molecular mechanisms underlying the pathogenesis of LUSQ.

The analysis of miRNA expression signatures of head and neck squamous cell carcinoma (HNSCC) by RNA sequencing revealed that miR-150-5p was downregulated in cancer tissues (19). In addition, the downregulation of miR-150-5p was detected in cancer signatures derived from prostate cancer and bladder cancer (20,21). However, the functional significance of miR-150-5p in LUSQ remains unknown. Thus, in this study, we focused on miR-150-5p and investigated its functional significance and the regulatory RNA networks in LUSQ cells.

Materials and methods

Clinical samples and cell lines

In this study, a total of 33 LUSQs and 24 non-cancerous lung specimens apart from the tumors were obtained from patients who underwent lobectomy at Kagoshima University Hospital from 2010 to 2013. The clinicopathological data of the patients with LUSQ (33 LUSQs and 24 non-cancerous lung specimens) are summarized in Table I. Our study was approved by the Institutional Review Board for Clinical Research of Kagoshima University Hospital. Each patient provided written informed consent and approval prior to obtaining the samples.

Table I

Characteristics of lung cancer and non-cancerous cases.

Table I

Characteristics of lung cancer and non-cancerous cases.

A, Characteristics of the lung cancer cases

Lung cancer patientsn(%)
Total number33
Median age (range, years)70 (55–88)
Sex
 Male3193.9
 Female26.1
Pathological stage
 IA515.2
 IB927.3
 IIA412.1
 IIB618.2
 IIIA824.2
 IIIB13.0
B, Characteristics of the non-cancerous cases

Non-cancerous tissuesn
Total number24
Median age (range, years)69 (50–88)
Sex
 Male24
 Female0

Two human LUSQ cell lines (SK-MES-1and EBC-1) were used in this study as previously described (15,16). The cell lines, SK-MES-1 and EBC-1, were obtained from the Japanese Cancer Research Resources Bank (JCRB) and the American Type Culture Collection (Manassas, VA, USA), respectively.

Mature miRNA and small interfering RNA transfection into LUSQ cells

The following RNA species were used in this study: mature miRNAs, Pre-miR™ miRNA Precursors (has-miR-150-5p, assay ID: PM 10070; Applied Biosystems, Foster City, CA, USA), negative control miRNA (Applied Biosystems, assay ID: AM 17111), small interfering RNA (Stealth Select RNAi siRNA, Invitrogen, si-MMP14 P/N: HSS106639 and HSS106640). RNA species were incubated with Lipofectamine RNAiMax reagent (Invitrogen, Carlsbad, CA, USA) and Opti-MEM (Invitrogen) prior to plating. Subsequently, the complex was added to suspended 1×105 cells per-well plated in 6-well plates. Mock-transfected cells were transfected only with Lipofectamine RNAiMax reagent and Opti-MEM at plating. The transfection procedures were as previously described (15,16).

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was isolated using Isogen (Nippon Gene, Tokyo, Japan) according to the manufacturer's instructions. The integrity of the RNA was checked using an RNA 6000 Nano assay kit and a 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA).

The procedure for PCR quantification was as previously described (12,15,16). In brief, we first synthesized cDNA from total RNA of each sample using the TaqMan Reverse Transcription kit (P/N N8080234 and 4366596, Applied Biosystems). Subsequently, we evaluated the expression of the gene by TaqMan Real-Time PCR Assays. TaqMan probes and primers for MMP14 (P/N: Hs1037003_g1; Applied Biosystems) were assay-on-demand gene expression products. Stem-loop RT-PCR for miR-150-5p (assay ID: 000473; Applied Biosystems) was used. Human GUSB (P/N: Hs99999908_m1; Applied Biosystems) and RNU48 (assay ID: 001006; Applied Biosystems) were used as normalized controls. All reactions were performed in triplicate and the ΔΔCt method was employed to calculate the fold change.

Cell proliferation, migration and invasion assays

To investigate the functional significance of miR-150-5p or MMP14 silencing by siRNA knockdown, we performed cell proliferation, migration and invasion assays using the SK-MES-1 and EBC-1 cells.

The cells were transfected with 10 nM miRNA or siRNA by reverse transfection and plated in 96-well plates at 5×104 cells per well. After 72 h, cell proliferation was determined by XTT assay using Cell Proliferation kit (Biological Industries, Kibbutz Beit Haemek, Israel).

Cell migration activity was evaluated by wound healing assay. The cells were seeded in 6-well plates at 1×105 cells per well. At 48 h after transfection, the cell monolayer was scraped using a P-200 micropipette tip. The initial gap length and residual gap length 24 h after wounding were calculated from photomicrographs.

Cell invasion assay was carried out using modified Boyden chambers, consisting Transwell pre-coated Matrigel membrane filter inserts with 8 µm pores in 24-well tissue culture plates (BD Biosciences, Bedford, MA, USA). At 48 h after transfection, the cells were planted in the inserts at 1×105 per well. At 72 h after transfection, the cells invaded to the lower side were fixed and stained with Diff-Quick (Sysmex Corp., Kobe, Japan). The number of cells invaded to the lower surface was determined microscopically by counting 8 areas of constant size per well. All experiments were performed in triplicate. The procedures of these functional assays were as previously described (15,16).

Western blot analysis

At 72 h after transfection, protein lysates (50 µg) were separated on NuPAGE on 4–12% Bis-Tris gels (Invitrogen) and transferred onto polyvinylidene fluoride membranes (GE Healthcare Japan, Tokyo, Japan). Immunoblotting was performed with monoclonal MMP14 antibody (1:2,000 dilution) (ab51074; Abcam, Cambridge, UK). GAPDH antibody (1:10,000 dilution) (ab8245; Abcam) was used as an internal control. The membranes were washed and then incubated with an anti-rabbit-IgG, HRP-linked anti body (#7074; Cell Signaling Technology, Danvers, MA, USA). Complexes were visualized with Clarity Western ECL Substrate (Bio-Rad, Hercules, CA, USA). The procedures have been described in our previous studies (15,16).

Identification of miR-150-5p target oncogenic genes in LUSQ cells

Specific genes regulated by miR-150-5p were identified by a combination of in silico and comprehensive gene expression analyses. Our target search strategies were described as previously (15,16). First, we selected putative miR-150-5p target genes using the TargetScan 7.1 database (http://www.targetscan.org/vert_71/). We then performed comprehensive gene analysis of downregulated genes in miR-150-5p transfected EBC-1 cells and upregulated genes in NSCLC from GEO database. The microarray data were deposited into GEO (http://www.ncbi.nlm.nih.gov/geo/), with the Accession number: GSE82108. Upregulated genes in NSCLC clinical specimens were obtained from the GEO database (Accession number: GSE19188).

Plasmid construction and dual-luciferase reporter assays

The wild-type or deletion-type sequences of the 3′-untranslated region (UTR) of MMP14 in miR-150-5p target sites were inserted in the psiCHECK-2 vector (C8021; Promega, Madison, WI, USA). We used 3 sequences that were putative miR-150-5p target sites of MMP14 by the TargetScan database (position 567–573: UUGGGAG; position 804–810: UGGGAGA and position 1381–1388: UUGGGAGA). The procedure for dual luciferase reporter assays was as previously described (15,16). The synthesized DNA was cloned into the psiCHECK-2 vector. EBC-1 cells were transfected with the vector, miRNAs and Lipofectamine 2000 in Opti-MEM (both from Invitrogen). The activities of Firefly and Renilla luciferases in cell lysates were determined with a dual-luciferase assay system (E1910; Promega). Normalized data were calculated as the quotient of Renilla/Firefly luciferase activities.

Immunohistochemistry staining and scoring

A tissue microarray containing a total of 30 lung samples, 20 LUSQ specimens and 10 normal lung samples was obtained from US Biomax (Derwood, MD, USA; Cat. no. BC04002). The patient characteristics for the tissue microarray are shown in Table II. The TNM classification of cancer tissues was according to the 7th edition of the American Joint Committee on Cancer (22).

Table II

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

Table II

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

A, Immunohistochemical status and characteristics of the LUSQ cases

Patient no.GradeTNMPathological stageImmunohistochemical staining intensity
11310IIIA(+)
21300IIIA(++)
32210II(+++)
42300IIIA(++)
51200I(++)
61210II(+++)
71310IIIA(++)
81200I(++)
91210II(++)
101200I(+++)
112220IIIA(++)
122200I(+++)
132100I(+++)
142100I(+++)
152210II(++)
162310IIIA(+++)
172200I(++)
182210II(++)
192320IIIA(+++)
202200I(++)
B, Immunohistochemical status of non-cancerous cases

Patient no.Immunohistochemical staining intensity
91(+)
92(+)
93(+)
94(−)
95(+)
96(+)
97(−)
98(++)
99(+)
100(+)

We confirmed the expression status of MMP14 in the LUSQ clinical specimens using immunohistochemical staining. The procedure for immunohistochemistry was as previously described (16,23,24). The tissue sections were incubated with the primary rabbit monoclonal antibody against MMP14 (1:5,000 dilution; ab51074; Abcam). The slide was treated with biotinylated goat anti-rabbit antibodies. Diaminobenzidine hydrogen peroxidase was the chromogen and counterstaining was done with 0.5% hematoxylin. Each tissues sample was scored on the basis of the intensity and area of staining. The procedure for score for staining tissues was as previously described (25).

Statistical analysis

Relationships between two or three variables and numerical values were analyzed using Mann-Whitney U tests or Bonferroni-adjusted Mann-Whitney U tests. Expert StatView software (version 5.0; SAS Institute Inc., Cary, NC, USA) was used for these analyses. Statistical analysis was carried out as previously described (19,23).

Results

Expression levels of miR-150-5p in LUSQ clinical specimens and cell lines

The expression levels of miR-150-5p were significantly decreased in the cancer tissues compared with non-cancerous tissues (P<0.0001; Fig. 1A). In addition, the expression levels of miR-150-5p in the cancer cell lines, SK-MES-1 and EBC-1, were markedly downregulated (Fig. 1A).

Effects of the ectopic overexpression of miR-150-5p on cell proliferation, migration and invasion in LUSQ cell lines

To validate the antitumor effects of miR-150-5p, we carried out gain-of-function assays by transfecting miRNA into two LUSQ cell lines (SK-MES-1 and EBC-1). Cell proliferation was significantly inhibited in the cells transfected with the mature miR-150-5p in comparison with the mock (transfection reagent only)- or miR-control-transfected cells (Fig. 1B). Furthermore, cell migration and invasion activities were markedly reduced in the cells transfected with the miR-150-5p expression plasmid compared to the mock- or miR-control-transfected cells (Fig. 1C and D).

Identification of putative targets of miR-150-5p regulation in LUSQ cells

Our strategy for the selection of miR-150-5p target oncogenic genes is shown in Fig. 2. First, we selected putative miR-150-5p target genes using the TargetScan 7.1 database and identified 351 genes. We then performed comprehensive gene expression analysis using miR-150-5p transfectants of EBC-1 cells, with negative control miRNA transfectants serving as controls (Accession number: GSE 82108). In this assessment, 15 genes were commonly downregulated (log2 ratio <−1.0). The gene set was then analyzed with a publicly available gene expression data set in GEO (Accession number: GSE 19188) and genes upregulated in LUSQ were selected (fold change >0). A total of 9 genes were identified as candidate targets of miR-150-5p regulation (Table III).

Table III

Putative target genes regulated by miR-150-5p in LUSQ cells.

Table III

Putative target genes regulated by miR-150-5p in LUSQ cells.

Gene symbolGene nameConservedPoorly conservedGEO82108 log2 ratioGEO19188 Fold change
MMP14Matrix metallopeptidase 14 (membrane-inserted)12−1.111.872
CPDCarboxypeptidase D10−1.071.071
LRRC58Leucine rich repeat containing 5816−1.190.733
CDC73Cell division cycle 7312−2.160.605
ENSAEndosulfine alpha15−1.790.501
DSELDermatan sulfate epimerase-like10−1.170.487
CSNK1A1Casein kinase 1, alpha 111−1.130.292
CHD2Chromodomain helicase DNA binding protein 212−2.140.260
CAPZBCapping protein (actin filament) muscle Z-line, beta14−1.180.201

Among these candidate genes, we focused on MMP14 as the aberrant expression of the matrix metalloproteinase family enhances cancer cell migration and invasion (26). The ectopic overexpression of miR-150-5p significantly inhibited cancer cell migration and invasion in the LUSQ cells (Fig. 1C and D).

Direct regulation of MMP14 by miR-150-5p in LUSQ cells

We investigated whether transfection of the LUSQ cell lines with the miR-150-5p expression plasmid would reduce the expression of MMP14/MMP14. The mRNA expression levels of MMP14 were decreased by transfection with the miR-150-5p expression plasmid compared with the mock- or miR-control-transfected cells (Fig. 3A). Furthermore, the protein expression levels of MMP14 were also decreased by the overexpression of miR-150-5p compared with the mock- or miR-control-transfected cells (Fig. 3B).

We also performed luciferase reporter assays with a vector that included the 3′-UTR of MMP14 to confirm that miR-150-5p directly regulated MMP14 in a sequence-dependent manner. We used vectors encoding the partial wild-type or deletion-type sequences of the 3′-UTR of the MMP14 with miR-150-5p target sites. Three binding sites for miR-150-5p in the 3′-UTR of MMP14 (positions 567–573, 804–810 and 1381–1388) were predicted by the TargetScan Human database (Fig. 3C). We observed that the luminescence intensities of the proteins coded by vectors carrying the wild-type sequences (positions 804–810 and 1381–1388) were significantly reduced by co-transfection with miR-150-5p (Fig. 3C). By contrast, transfection with the deletion-type vector blocked the reduction of luminescence intensities (Fig. 3C). These findings indicated that miR-150-5p directly bound to specific two sites in the 3′-UTR of MMP14.

Effects of knockdown of MMP14 on the proliferation, migration and invasion of LUSQ cell lines

Loss-of-function assays using siRNA were performed to examine the function of MMP14 in LUSQ cell lines (SK-MES-1 and EBC-1). The mRNA and protein expression levels of MMP14 were decreased by transfection with si-MMP14 in the LUSQ cell lines (Fig. 4A and B).

We then investigated the effects of MMP14 knockdown on the proliferation, migration, and invasion of LUSQ cell lines. Cancer cell proliferation was significantly reduced in the si-MMP14-transfected cells (Fig. 4C). In addition, the migration and invasion activities were significantly attenuated in the si-MMP14-transfected cells (Fig. 4D and E).

Expression of MMP14 in LUSQ clinical specimens

We examined the protein expression levels of MMP14 in LUSQ clinical specimens by immunostaining. We confirmed the overexpression of MMP14 in LUSQ lesions compared with that in normal tissues (Fig. 5).

Identification of MMP14-regulated genes in LUSQ cells

To investigate downstream genes regulated by MMP14, we performed genome-wide gene expression analysis using si-MMP14 in EBC-1 cells. A total of 92 genes were identified as MMP14-regulated genes (Table IV).

Table IV

Downstream genes regulated by MMP14 in LUSQ cells.

Table IV

Downstream genes regulated by MMP14 in LUSQ cells.

Gene symbolGene nameExpression log2 ratio
si-MMP14-1 si-MMP14-2Average
MMP14Matrix metallopeptidase 14 (membrane-inserted)−4.19−3.72−3.96
LMNB1Lamin B1−3.70−2.97−3.34
TMEM192Transmembrane protein 192−2.61−3.48−3.05
MKI67Marker of proliferation Ki-67−2.17−3.85−3.01
ENPP1Ectonucleotide pyrophosphatase/phosphodiesterase 1−2.77−3.11−2.94
LPLLipoprotein lipase−2.81−3.05−2.93
SLC7A11-AS1SLC7A11 antisense RNA 1−3.90−1.90−2.90
CDRT1CMT1A duplicated region transcript 1−3.92−1.77−2.85
FAM129AFamily with sequence similarity 129, member A−3.68−1.97−2.83
BZW1Basic leucine zipper and W2 domains 1−3.08−2.58−2.83
LYRM1LYR motif containing 1−2.63−2.90−2.76
CHRNA5Cholinergic receptor, nicotinic, alpha 5 (neuronal)−3.31−2.20−2.75
MARCH5Membrane-associated ring finger (C3HC4) 5−2.23−3.23−2.73
DEPDC1DEP domain containing 1−3.34−2.06−2.70
CBX3Chromobox homolog 3−3.06−2.27−2.66
ANKRD22Ankyrin repeat domain 22−1.50−3.76−2.63
JKAMP JNK1/MAPK8-associated membrane protein−1.66−3.37−2.51
ANTXR1Anthrax toxin receptor 1−2.39−2.59−2.49
SMAD1-AS1SMAD1 antisense RNA 1−2.29−2.69−2.49
ATP5EATP synthase, H+ transporting, mitochondrial F1 complex, epsilon subunit−2.31−2.64−2.47
TOMM20Translocase of outer mitochondrial membrane 20 homolog (yeast)−2.40−2.41−2.40
DEFB4ADefensin, beta 4A−2.62−2.12−2.37
ALDH7A1Aldehyde dehydrogenase 7 family, member A1−2.48−2.25−2.37
OSBPL8Oxysterol binding protein-like 8−2.70−1.99−2.34
GLYCTKGlycerate kinase−2.58−2.08−2.33
SLC16A1Solute carrier family 16 (monocarboxylate transporter), member 1−2.82−1.82−2.32
C5Complement component 5−2.18−2.42−2.30
ASNSAsparagine synthetase (glutamine-hydrolyzing)−3.09−1.50−2.29
POLE2Polymerase (DNA directed), epsilon 2, accessory subunit−1.53−3.05−2.29
SASS6Spindle assembly 6 homolog (C. elegans)−1.94−2.62−2.28
SMARCA2SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2−1.81−2.68−2.24
FANCD2Fanconi anemia, complementation group D2−2.19−2.27−2.23
TMEM154Transmembrane protein 154−2.69−1.77−2.23
ARHGAP9Rho GTPase activating protein 9−2.73−1.73−2.23
SPATA5Spermatogenesis associated 5−2.64−1.68−2.16
ARHGAP11ARho GTPase activating protein 11A−1.71−2.59−2.15
OSBPL8Oxysterol binding protein-like 8−2.12−2.16−2.14
SUV39H2Suppressor of variegation 3–9 homolog 2 (Drosophila)−1.77−2.50−2.13
ACTL8Actin-like 8−2.36−1.85−2.10
ESCO2Establishment of sister chromatid cohesion N-acetyltransferase 2−2.52−1.68−2.10
RAB1ARAB1A, member RAS oncogene family−2.07−2.13−2.10
FBXO4F-box protein 4−1.76−2.42−2.09
MAPK6Mitogen-activated protein kinase 6−2.32−1.86−2.09
RNF180Ring finger protein 180−1.91−2.25−2.08
DKFZP434I0714Uncharacterized protein DKFZP434I0714−1.77−2.38−2.08
MTBPMDM2 binding protein−1.93−2.21−2.07
AIF1LAllograft inflammatory factor 1-like−2.44−1.68−2.06
MOB3BMOB kinase activator 3B−1.57−2.53−2.05
CGGBP1CGG triplet repeat binding protein 1−1.75−2.34−2.04
HIST1H2AIHistone cluster 1, H2ai−2.46−1.62−2.04
FKBP5FK506 binding protein 5−1.79−2.27−2.03
NUDT21Nudix (nucleoside diphosphate linked moiety X)-type motif 21−1.54−2.45−2.00
NEIL3Nei endonuclease VIII-like 3 (E. coli)−2.02−1.90 −1.96
ATG4CAutophagy-related 4C, cysteine peptidase−1.64−2.26−1.95
ECEL1Endothelin converting enzyme-like 1−1.89−1.95−1.92
KIF11Kinesin family member 11−1.55−2.25−1.90
TCF19Transcription factor 19−1.64−2.16−1.90
ZNF681Zinc finger protein 681−2.03−1.75−1.89
RRM2Ribonucleotide reductase M2−1.66−2.10−1.88
PLAC8L1PLAC8-like 1−1.55−2.20−1.88
IL1BInterleukin 1, beta−1.69−2.05−1.87
DIS3LDIS3 like exosome 3′-5′ exoribonuclease−1.81−1.90−1.85
CKAP2LCytoskeleton associated protein 2-like−1.66−2.03−1.84
RRN3RRN3 RNA polymerase I transcription factor homolog (S. cerevisiae)−1.54−2.15−1.84
TMEM180Transmembrane protein 180−2.12−1.56−1.84
HS2ST1Heparan sulfate 2-O-sulfotransferase 1−1.57−2.11−1.84
ZNF414Zinc finger protein 414−1.74−1.89−1.82
TMEM194BTransmembrane protein 194B−1.69−1.94−1.82
STXBP5LSyntaxin binding protein 5-like−1.92−1.71−1.82
CENPICentromere protein I−1.98−1.64−1.81
ERCC6Lexcision repair cross-complementation group 6-like−1.60−2.02−1.81
SPC24SPC24, NDC80 kinetochore complex component−1.79−1.83−1.81
KRTAP10-3Keratin associated protein 10-3−1.85−1.76−1.80
ID2-AS1ID2 antisense RNA 1 (head to head)−1.55−2.02−1.78
GATA2-AS1GATA2 antisense RNA 1−2.03−1.53−1.78
FBF1Fas (TNFRSF6) binding factor 1−1.55−2.01−1.78
MED1Mediator complex subunit 1−1.64−1.91−1.78
PLCL2Phospholipase C-like 2−2.01−1.52−1.77
CDKN3Cyclin-dependent kinase inhibitor 3−1.56−1.92−1.74
CSF3Colony stimulating factor 3 (granulocyte)−1.55−1.92−1.73
PLAAPhospholipase A2-activating protein−1.53−1.88−1.70
MCM6Minichromosome maintenance complex component 6−1.81−1.59−1.70
TRIM14Tripartite motif containing 14−1.64−1.75−1.69
TUBA3FPTubulin, alpha 3f, pseudogene−1.66−1.71−1.68
OVOS2Ovostatin 2−1.76−1.59−1.67
NUCKS1Nuclear casein kinase and cyclin-dependent kinase substrate 1−1.58−1.75−1.67
HOOK1Hook microtubule-tethering protein 1−1.62−1.66−1.64
ZBTB33Zinc finger and BTB domain containing 33−1.72−1.50−1.61
GALNT4Polypeptide N-acetylgalactosaminyltransferase 4−1.70−1.53−1.61
RRM1Ribonucleotide reductase M1−1.61−1.61−1.61
MDM1Mdm1 nuclear protein homolog (mouse)−1.69−1.53−1.61
FAM72DFamily with sequence similarity 72, member D−1.51−1.66−1.59

Discussion

Recently, several treatment options have been approved for post-first-line therapy for LUSQ, such as chemotherapy with or without an angiogenesis inhibitor (27), or immunotherapy (4). However, treatment outcomes do not appear to have improved. Therefore, clinicians need new treatment options in their approach to LUSQ. Presumably, they should be based on the latest molecular analyses of the pathology of this disease.

In this study, we demonstrated that the restoration of miR-150-5p significantly attenuated with cancer cell aggressiveness, suggesting that this miRNA possesses antitumor activity in LUSQ cells. It has been demonstrated that miR-150-5p has multiple functions, possessing antitumor activity or oncogenic functions (28). The overexpression of miR-150-5p has been reported in several types of cancer (29,30). In contrast to the overexpression of miR-150-5p in cancer cells, the antitumor roles of miR-150-5p have been reported in several types of cancer through its targeting of oncogenic genes (19,31,32). Previous studies showed that contradiction resulted as to the expression pattern of miR-150-5p in lung cancer (28). Our preliminary data demonstrated that the expression levels of miR-150-5p were reduced in lung adenocarcinoma clinical specimens (data not shown). One such study showed that miR-150-5p expression was significantly reduced in HNSCC tissues and had antitumor roles. A low expression of miR-150-5p has been shown to be significantly associated with a poor prognosis of patients with HNSCC (19). Moreover, integrin subunit alpha 3 (ITGA3), integrin subunit alpha 6 (ITGA6) and tenascin C (TNC) have been identified as oncogenes in HNSCC that were downregulated by miR-150-5p (19). Previous studies have shown that the aberrant expression of integrin family genes and the activation of integrin-mediated oncogenic signaling promotes cancer cell metastasis and the epithelial-mesenchymal transition (EMT) phenotype (33,34). Other studies have demonstrated that SPOCK1 is directly regulated by miR-150-5p in prostate cancer and esophageal squamous cell carcinoma (31,32). The aberrant expression of SPOCK1 has been observed in several types of cancer and has been shown to play pivotal roles in cancer cell progression, metastasis and drug resistance (35,36). Of note, the ectopic expression of SPOCK1 induces EMT in lung cancer (37). These findings suggest that the downregulation of miR-150-5p induces several cancer-promoting genes and that its expression is deeply involved in cancer pathogenesis.

In this study, to identify oncogenic genes targeted by miR-150-5p, we applied gene expression analysis and in silico database searches. A total of 9 genes (MMP14, CPD, LRRC58, CDC73, ENSA, DSEL, CSNK1A1, CHD2 and CAPZB) were identified as putative targets of miR-150-5p regulation in LUSQ cells. Among these, we focused on MMP14 as the matrix metalloproteinase family contributes to cancer cell migration and invasion. Our luciferase reporter assay revealed that MMP14 was directly regulated by miR-150-5p in LUSQ cells. The overexpression of MMP14 was observed in LUSQ clinical specimens and the knockdown of MMP14 by siRNA significantly interfered in vitro with cancer cell malignancies. These findings indicate that MMP14 acts as an oncogene regulated by the antitumor miR-150-5p in LUSQ cells.

MMP14 belongs to the membrane-type matrix metalloprotease (MT-MMP) family that includes pivotal regulators of cell invasion, growth and survival in normal cells (26). The upregulation of MMP14 has been observed in many types of cancer, and the overexpression of MMP14 has been shown to correlate with a poor prognosis in patients with NSCLC, renal cell carcinoma and breast cancer (3840). Activated MMP14 cleaves pro-MMP2 and pro-MMP13 and the activated form of MMP2 and MMP13 are involved in cancer pathogenesis (26). A major substrate of MMP14 is type I collagen in ECM components. MMP14 is localized to the leading edge of invadopodia in migrating cells, achieving spatially coordinated matrix degradation to support invasion (41). MMP14 influences cancer cells and cancer niches (ECM and fibroblast cells). MMP14 is mediated through membrane proteins, e.g., receptor tyrosine kinases (RTK) and integrins, and these events promote cancer cell aggressiveness (42,43). For instance, he interaction of MMP14 and CD44 induces the phosphorylation of the EGF receptor and enhances downstream activation of the MAPK and PI3K signaling pathways (44,45).

Proteolytic enzymes, MMPs or MT-MMPs are tightly controlled by tissue inhibitors of metalloproteases (TIMPs) (26). The expression and activation of MMPs and TIMPs are important to biological processes and essential for tissue homeostasis. Dysregulated MMPs and TIMPs have been implicated in cancer cell progression and metastasis (46). Recent studies have indicated that several downregulated miRNAs caused the aberrant expression of MMP14 in cancer cells (47,48). Our recent studies showed the antitumor functions of miR-375 and miR-139-5p/-3p that targeted MMP13 and MMP11, respectively (49,50). These MMPs were overexpressed in cancer specimens, and the restoration of these miRNAs or knockdown of MMP13 or MMP11 inhibited cancer cell migration and invasion (49,50). The identification of antitumor miRNAs that regulate novel LUSQ networks may lead to a better understanding of the aggressiveness of this disease.

Taken together, this study demonstrates miR-150-5p acts as an antitumor miRNA by targeting MMP14 in LUSQ cells. The overexpression of MMP14 may be enhanced in lung cancer oncogenesis. The exploration of antitumor miRNA-mediated regulatory networks may lead to the development of novel treatment strategies for this disease.

Acknowledgments

This study was supported by the Japan Society for the Promotion of Science (JSPS) Grants-in-Aid for Scientific Research (KAKENHI), 17K16893, 15K10801, 16K19458, and 17K09660.

Notes

[1] Competing interests

The authors declare that they have no competing interests.

References

1 

Cheng TY, Cramb SM, Baade PD, Youlden DR, Nwogu C and Reid ME: The international epidemiology of lung cancer: Latest trends, disparities, and tumor characteristics. J Thorac Oncol. 11:1653–1671. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Travis WD: Pathology of lung cancer. Clin Chest Med. 32:669–692. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Reck M, Heigener DF, Mok T, Soria JC and Rabe KF: Management of non-small-cell lung cancer: Recent developments. Lancet. 382:709–719. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, Gottfried M, Peled N, Tafreshi A, Cuffe S, et al KEYNOTE-024 Investigators: Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 375:1823–1833. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Sandler AB, Schiller JH, Gray R, Dimery I, Brahmer J, Samant M, Wang LI and Johnson DH: Retrospective evaluation of the clinical and radiographic risk factors associated with severe pulmonary hemorrhage in first-line advanced, unresectable non-small-cell lung cancer treated with Carboplatin and Paclitaxel plus bevacizumab. J Clin Oncol. 27:1405–1412. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Scagliotti GV, Parikh P, von Pawel J, Biesma B, Vansteenkiste J, Manegold C, Serwatowski P, Gatzemeier U, Digumarti R, Zukin M, et al: Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 26:3543–3551. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Paz-Ares L, Tan EH, O'Byrne K, Zhang L, Hirsh V, Boyer M, Yang JC, Mok T, Lee KH, Lu S, et al: Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: Overall survival data from the phase IIb LUX-Lung 7 trial. Ann Oncol. 28:270–277. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Beermann J, Piccoli MT, Viereck J and Thum T: Non-coding RNAs in development and disease: Background, mechanisms, and therapeutic approaches. Physiol Rev. 96:1297–1325. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Wiemer EA: The role of microRNAs in cancer: No small matter. Eur J Cancer. 43:1529–1544. 2007. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Kamikawaji K, Seki N, Watanabe M, Mataki H, Kumamoto T, Takagi K, Mizuno K and Inoue H: Regulation of LOXL2 and SERPINH1 by antitumor microRNA-29a in lung cancer with idiopathic pulmonary fibrosis. J Hum Genet. 61:985–993. 2016. View Article : Google Scholar : PubMed/NCBI

14 

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. View Article : Google Scholar :

15 

Mataki H, Seki N, Mizuno K, Nohata N, Kamikawaji K, Kumamoto T, Koshizuka K, Goto Y and Inoue H: Dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p) coordinately targeted MTDH in lung squamous cell carcinoma. Oncotarget. 7:72084–72098. 2016. View Article : Google Scholar : PubMed/NCBI

16 

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. View Article : Google Scholar

17 

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

18 

Kurozumi A, Kato M, Goto Y, Matsushita R, Nishikawa R, Okato A, Fukumoto I, Ichikawa T and Seki N: Regulation of the collagen cross-linking enzymes LOXL2 and PLOD2 by tumor-suppressive microRNA-26a/b in renal cell carcinoma. Int J Oncol. 48:1837–1846. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Koshizuka K, Nohata N, Hanazawa T, Kikkawa N, Arai T, Okato A, Fukumoto I, Katada K, Okamoto Y and Seki N: Deep sequencing-based microRNA expression signatures in head and neck squamous cell carcinoma: Dual strands of pre-miR-150 as antitumor miRNAs. Oncotarget. 8:30288–30304. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Goto Y, Kurozumi A, Arai T, Nohata N, Kojima S, Okato A, Kato M, Yamazaki K, Ishida Y, Naya Y, et al: Impact of novel miR-145-3p regulatory networks on survival in patients with castration-resistant prostate cancer. Br J Cancer. 117:409–420. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Itesako T, Seki N, Yoshino H, Chiyomaru T, Yamasaki T, Hidakaå 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

22 

Mirsadraee S, Oswal D, Alizadeh Y, Caulo A and van Beek E Jr: The 7th lung cancer TNM classification and staging system: Review of the changes and implications. World J Radiol. 4:128–134. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Koshizuka K, Hanazawa T, Fukumoto I, Kikkawa N, Matsushita R, Mataki H, Mizuno K, Okamoto Y and Seki N: Dual-receptor (EGFR and c-MET) inhibition by tumor-suppressive miR-1 and miR-206 in head and neck squamous cell carcinoma. J Hum Genet. 62:113–121. 2017. View Article : Google Scholar

24 

Okato A, Goto Y, Kurozumi A, Kato M, Kojima S, Matsushita R, Yonemori M, Miyamoto K, Ichikawa T and Seki N: Direct regulation of LAMP1 by tumor-suppressive microRNA-320a in prostate cancer. Int J Oncol. 49:111–122. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Kojima S, Chiyomaru T, Kawakami K, Yoshino H, Enokida H, Nohata N, Fuse M, Ichikawa T, Naya Y, Nakagawa M, et al: Tumour suppressors miR-1 and miR-133a target the oncogenic function of purine nucleoside phosphorylase (PNP) in prostate cancer. Br J Cancer. 106:405–413. 2012. View Article : Google Scholar :

26 

Turunen SP, Tatti-Bugaeva O and Lehti K: Membrane-type matrix metalloproteases as diverse effectors of cancer progression. Biochim Biophys Acta. 1864(11 Pt A): 1974–1988. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Maione P, Sgambato A, Casaluce F, Sacco PC, Santabarbara G, Rossi A and Gridelli C: The role of the antiangiogenetic ramucirumab in the treatment of advanced non small cell lung cancer. Curr Med Chem. 24:3–13. 2017. View Article : Google Scholar

28 

Wang F, Ren X and Zhang X: Role of microRNA-150 in solid tumors. Oncol Lett. 10:11–16. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Wu Q, Jin H, Yang Z, Luo G, Lu Y, Li K, Ren G, Su T, Pan Y, Feng B, et al: miR-150 promotes gastric cancer proliferation by negatively regulating the pro-apoptotic gene EGR2. Biochem Biophys Res Commun. 392:340–345. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Huang S, Chen Y, Wu W, Ouyang N, Chen J, Li H, Liu X, Su F, Lin L and Yao Y: miR-150 promotes human breast cancer growth and malignant behavior by targeting the pro-apoptotic purinergic P2X7 receptor. PLoS One. 8:e807072013. View Article : Google Scholar : PubMed/NCBI

31 

Okato A, Arai T, Kojima S, Koshizuka K, Osako Y, Idichi T, Kurozumi A, Goto Y, Kato M, Naya Y, et al: Dual strands of pre-miR-150 (miR-150-5p and miR-150-3p) act as antitumor miRNAs targeting SPOCK1 in naïve and castration-resistant prostate cancer. Int J Oncol. 51:245–256. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Osako Y, Seki N, Koshizuka K, Okato A, Idichi T, Arai T, Omoto I, Sasaki K, Uchikado Y, Kita Y, et al: Regulation of SPOCK1 by dual strands of pre-miR-150 inhibit cancer cell migration and invasion in esophageal squamous cell carcinoma. J Hum Genet. 62:935–944. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Desgrosellier JS and Cheresh DA: Integrins in cancer: Biological implications and therapeutic opportunities. Nat Rev Cancer. 10:9–22. 2010. View Article : Google Scholar

34 

Gilcrease MZ: Integrin signaling in epithelial cells. Cancer Lett. 247:1–25. 2007. View Article : Google Scholar

35 

Shu YJ, Weng H, Ye YY, Hu YP, Bao RF, Cao Y, Wang XA, Zhang F, Xiang SS, Li HF, et al: SPOCK1 as a potential cancer prognostic marker promotes the proliferation and metastasis of gallbladder cancer cells by activating the PI3K/AKT pathway. Mol Cancer. 14:122015. View Article : Google Scholar : PubMed/NCBI

36 

Kim HP, Han SW, Song SH, Jeong EG, Lee MY, Hwang D, Im SA, Bang YJ and Kim TY: Testican-1-mediated epithelial-mesenchymal transition signaling confers acquired resistance to lapatinib in HER2-positive gastric cancer. Oncogene. 33:3334–3341. 2014. View Article : Google Scholar

37 

Miao L, Wang Y, Xia H, Yao C, Cai H and Song Y: SPOCK1 is a novel transforming growth factor-β target gene that regulates lung cancer cell epithelial-mesenchymal transition. Biochem Biophys Res Commun. 440:792–797. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Wang YZ, Wu KP, Wu AB, Yang ZC, Li JM, Mo YL, Xu M, Wu B and Yang ZX: MMP-14 overexpression correlates with poor prognosis in non-small cell lung cancer. Tumour Biol. 35:9815–9821. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Lu H, Hu L, Yu L, Wang X, Urvalek AM, Li T, Shen C, Mukherjee D, Lahiri SK, Wason MS, et al: KLF8 and FAK cooperatively enrich the active MMP14 on the cell surface required for the metastatic progression of breast cancer. Oncogene. 33:2909–2917. 2014. View Article : Google Scholar :

40 

Hagemann T, Gunawan B, Schulz M, Füzesi L and Binder C: mRNA expression of matrix metalloproteases and their inhibitors differs in subtypes of renal cell carcinomas. Eur J Cancer. 37:1839–1846. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Gawden-Bone C, Zhou Z, King E, Prescott A, Watts C and Lucocq J: Dendritic cell podosomes are protrusive and invade the extracellular matrix using metalloproteinase MMP-14. J Cell Sci. 123:1427–1437. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Gálvez BG, Matías-Román S, Yáñez-Mó M, Sánchez-Madrid F and Arroyo AG: ECM regulates MT1-MMP localization with beta1 or alphavbeta3 integrins at distinct cell compartments modulating its internalization and activity on human endothelial cells. J Cell Biol. 159:509–521. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Munshi HG and Stack MS: Reciprocal interactions between adhesion receptor signaling and MMP regulation. Cancer Metastasis Rev. 25:45–56. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Cho SH, Park YS, Kim HJ, Kim CH, Lim SW, Huh JW, Lee JH and Kim HR: CD44 enhances the epithelial-mesenchymal transition in association with colon cancer invasion. Int J Oncol. 41:211–218. 2012.PubMed/NCBI

45 

Zarrabi K, Dufour A, Li J, Kuscu C, Pulkoski-Gross A, Zhi J, Hu Y, Sampson NS, Zucker S and Cao J: Inhibition of matrix metalloproteinase 14 (MMP-14)-mediated cancer cell migration. J Biol Chem. 286:33167–33177. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Hojilla CV, Mohammed FF and Khokha R: Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer. 89:1817–1821. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Li Y, Kuscu C, Banach A, Zhang Q, Pulkoski-Gross A, Kim D, Liu J, Roth E, Li E, Shroyer KR, et al: miR-181a-5p inhibits cancer cell migration and angiogenesis via downregulation of matrix metalloproteinase-14. Cancer Res. 75:2674–2685. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Zuo QF, Cao LY, Yu T, Gong L, Wang LN, Zhao YL, Xiao B and Zou QM: MicroRNA-22 inhibits tumor growth and metastasis in gastric cancer by directly targeting MMP14 and Snail. Cell Death Dis. 6:e20002015. View Article : Google Scholar : PubMed/NCBI

49 

Osako Y, Seki N, Kita Y, Yonemori K, Koshizuka K, Kurozumi A, Omoto I, Sasaki K, Uchikado Y, Kurahara H, et al: Regulation of MMP13 by antitumor microRNA-375 markedly inhibits cancer cell migration and invasion in esophageal squamous cell carcinoma. Int J Oncol. 49:2255–2264. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Yonemori M, Seki N, Yoshino H, Matsushita R, Miyamoto K, Nakagawa M and Enokida H: Dual tumor-suppressors miR-139-5p and miR-139-3p targeting matrix metalloprotease 11 in bladder cancer. Cancer Sci. 107:1233–1242. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2018
Volume 52 Issue 3

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
Suetsugu T, Koshizuka K, Seki N, Mizuno K, Okato A, Arai T, Misono S, Uchida A, Kumamoto T, Inoue H, Inoue H, et al: Downregulation of matrix metalloproteinase 14 by the antitumor miRNA, miR-150-5p, inhibits the aggressiveness of lung squamous cell carcinoma cells. Int J Oncol 52: 913-924, 2018
APA
Suetsugu, T., Koshizuka, K., Seki, N., Mizuno, K., Okato, A., Arai, T. ... Inoue, H. (2018). Downregulation of matrix metalloproteinase 14 by the antitumor miRNA, miR-150-5p, inhibits the aggressiveness of lung squamous cell carcinoma cells. International Journal of Oncology, 52, 913-924. https://doi.org/10.3892/ijo.2017.4232
MLA
Suetsugu, T., Koshizuka, K., Seki, N., Mizuno, K., Okato, A., Arai, T., Misono, S., Uchida, A., Kumamoto, T., Inoue, H."Downregulation of matrix metalloproteinase 14 by the antitumor miRNA, miR-150-5p, inhibits the aggressiveness of lung squamous cell carcinoma cells". International Journal of Oncology 52.3 (2018): 913-924.
Chicago
Suetsugu, T., Koshizuka, K., Seki, N., Mizuno, K., Okato, A., Arai, T., Misono, S., Uchida, A., Kumamoto, T., Inoue, H."Downregulation of matrix metalloproteinase 14 by the antitumor miRNA, miR-150-5p, inhibits the aggressiveness of lung squamous cell carcinoma cells". International Journal of Oncology 52, no. 3 (2018): 913-924. https://doi.org/10.3892/ijo.2017.4232