Open Access

Regulation of MMP13 by antitumor microRNA-375 markedly inhibits cancer cell migration and invasion in esophageal squamous cell carcinoma

  • Authors:
    • Yusaku Osako
    • Naohiko Seki
    • Yoshiaki Kita
    • Keiichi Yonemori
    • Keiichi Koshizuka
    • Akira Kurozumi
    • Itaru Omoto
    • Ken Sasaki
    • Yasuto Uchikado
    • Hiroshi Kurahara
    • Kosei Maemura
    • Shoji Natsugoe
  • View Affiliations

  • Published online on: October 21, 2016     https://doi.org/10.3892/ijo.2016.3745
  • Pages: 2255-2264
  • Copyright: © Osako 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

Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignancies. Recently developed molecular targeted therapies are not available for patients with ESCC. After curative surgical resection, patients frequently suffer distant metastasis and recurrence. Exploration of novel ESCC metastatic pathways may lead to the development of new treatment protocols for this disease. Accordingly, we have sequentially identified microRNA (miRNA)-mediated metastatic pathways in several cancers. Our past studies of miRNA expression signatures have shown that microRNA-375 (miR-375) is frequently reduced in several types of cancers, including ESCC. In the present study, we aimed to investigate novel miR-375-mediated metastatic pathways in ESCC cells. The expression of miR-375 was downregulated in ESCC tissues, and ectopic expression of this miRNA markedly inhibited cancer cell migration and invasion, suggesting that miR-375 acted as an antimetastatic miRNA in ESCC cells. Our strategies for miRNA target searching demonstrated that matrix metalloproteinase 13 (MMP13) was directly regulated by miR-375 in ESCC cells. Overexpression of MMP13 was observed in ESCC clinical tissues, and the expression of MMP13 promoted cancer cell aggressiveness. Moreover, oncogenic genes, including CENPF, KIF14 and TOP2A, were shown to be regulated downstream of MMP13. Taken together, these findings demonstrated that the antitumor miR-375/oncogenic MMP13 axis had a pivotal role in ESCC aggressiveness. These results provide novel insights into the potential mechanisms of ESCC pathogenesis.

Introduction

Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive cancers and the major histological type of esophageal cancer in Japan and East Asia (13). ESCC cells frequently metastasize to the lymph nodes, liver, lungs and bone (24). Despite the use of multimodality therapies, the prognosis of patients with ESCC is still poor, with an overall 5-year survival rate of approximately 20–30% (2,4). Recently developed molecularly targeted therapeutics have not been shown to have beneficial effects in patients with ESCC (2). Additionally, the molecular pathogenesis of the aggressive phenotype in ESCC remains unclear. Thus, in order to improve disease outcomes in patients with ESCC, it is necessary to elucidate the molecular mechanisms of ESCC cell aggressiveness using advanced genomic approaches.

The discovery of microRNAs (miRNAs) has resulted in major advancements in cancer research (5,6). miRNAs are small non-coding RNAs that function to fine tune the expression of protein coding/non-coding RNAs by repressing translation or cleaving RNA transcripts in a sequence-depending manner (7). The unique characteristic function of miRNAs is to regulate RNA transcripts in human cells. Therefore, dysregulated expression of miRNAs can disrupt tightly regulated RNA networks in cancer cells. Currently, numerous studies have shown that miRNAs are aberrantly expressed in several cancers, including ESCC (6,8). Using miRNA expression signature analyses, we have sequentially identified tumor-suppressive miRNAs and shown that these miRNAs mediate novel cancer networks (913).

Our miRNA expression signatures revealed that microRNA-375 (miR-375) is frequently downregulated in several types of squamous cell carcinoma (10,13,14). Moreover, our previous studies demonstrated that ectopic expression of miR-375 suppressed cancer cell aggressiveness in several types of cancer cells (15). In ESCC cells, several studies have indicated that miR-375 has antitumor roles through targeting oncogenic genes (16,17). Moreover, miR-375-mediated cancer pathways are essential for cancer cell initiation, development and aggressiveness.

Accordingly, in the present study, we aimed to investigate the novel cancer networks regulated by miR-375 in ESCC cells. Our present data showed that matrix metalloproteinase 13 (MMP13) was directly regulated by miR-375 in ESCC cells. Overexpression of MMP13 was observed in ESCC clinical tissues, and knockdown of MMP13 expression markedly inhibited ESCC cell migration and invasion, indicating that MMP13 acted as a cancer-promoting gene in ESCC cells. Moreover, the oncogenic genes CENPF, KIF14 and TOP2 were found to function downstream of MMP13. Taken together, these results showed that the antitumor miR-375/oncogenic MMP13 axis had a pivotal role in ESCC aggressiveness.

Materials and methods

Clinical ESCC specimens and ESCC cell lines

Clinical specimens were collected from 25 patients with ESCC. All patients underwent primary surgical treatment and were pathologically proven to have ESCC at the Kagoshima University Hospital from 2010 to 2014. The present study was approved by the Bioethics Committee of Kagoshima University; written prior informed consent and approval were obtained from all patients. The clinicopathological characteristics of the patients are shown in Table I.

Table I

Clinical features of patients with ESCC.

Table I

Clinical features of patients with ESCC.

No.Age (years)Gender DifferentiationTNMStagelyvRecurrence
168MalePoor1b20IIIA13+
272MaleModerate1b00IA01
369MaleModerate1b00IIIA00
462MaleWell320IIIB11+
566MaleModerate300IIA11
674MaleModerate220IIIA31+
756MaleModerate200IB01
879MaleModerate210IIB11
968MaleModerate1b20IIIA11
1052MalePoor1b00IA11+
1167MaleWell320IIIB22+
1257MalePoor330IIIC11+
1370MaleModerate300IIA11+
1466MaleModerate300IIA11
1563MaleWell330IIIC21+
1655MaleModerate320IIIB11+
1760MaleWell1b10IIB11
1878MaleWell300IIA12
1971MaleWell300IIA12
2075MaleModerate320IIIB11+
2160MaleModerate210IIB12
2262MaleWell1a10IIB00
2371MaleModerate1b10IIB00
2469MaleModerate1b00IA10
2584MaleWell210IIB11

We used two ESCC cell lines: TE-8, which was moderately differentiated; and TE-9, which was poorly differentiated. Both of these cells lines were provided by Riken BioResourse Center (Tsukuba, Japan).

Extraction of total RNA from clinical specimens and cell lines was performed using ISOGEN (Nippon Gene, Tokyo, Japan) according to the manufacturer's protocol. The quality of RNA was checked using an Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA).

Quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR)

The procedure for PCR quantification was previously described (13,1820). The expression levels of miR-375 (assay ID: 000564; Applied Biosystems, Foster City, CA, USA) were analyzed by TaqMan qRT-PCR assays (TaqMan MicroRNA assays; Applied Biosystems) and RNU48 (assay ID: 001006) was used for normalization. TaqMan probes and primers for MMP-13 (assay ID: Hs00233992_m1; Applied Biosystems), CENPF (assay ID: Hs01118845_m1), KIF14 (assay ID: Hs00978236_m1) and GUSB (the internal control; assay ID: Hs00939627_ml; Applied Biosystems) were used for gene expression analysis.

Transfection with mature miRNAs and small interfering RNAs (siRNAs)

The following mature miRNA was used: Ambion Pre-miR miRNA precursor for hsa-miR-375 (product ID: PM10327; Applied Biosystems). The following siRNAs were used: Stealth Select RNAi siRNA, si-MMP13 (cat nos. HSS106637 and HSS106638; Invitrogen, Carlsbad, CA, USA), and negative control miRNA/siRNA (P/N: AM17111; Applied Biosystems). RNAs were incubated with Opti-MEM (Invitrogen) and Lipofectamine RNAiMax transfection reagent (Invitrogen), as previously described (13,1820).

Cell proliferation, migration and invasion assays

TE-8 and TE-9 cells were transfected with 10 nM miRNAs or siRNAs by reverse transfection. Cell proliferation, migration and invasion assays were performed as previously described (13,1820).

Screening of miR-375 target genes using in silico analysis and gene expression data

To identify miR-375 target genes, a combination of genome-wide gene expression and in silico analyses was conducted as previously described (13,1820). The microarray data were deposited into the GEO repository under accession number GSE77790. Next, we selected putative miRNA target genes using microRNA.org (August, 2010 release, http://www.microrna.org) databases. Our strategy for identification of miR-375 target genes is shown in Fig. 2.

Western blot analysis

Anti-human MMP-13 rabbit polyclonal IgG (1:1,000; sc30073; Santa Cruz Biotechnology, Santa Cruz, CA, USA) was used as a primary antibody. Anti-human GAPDH mouse monoclonal IgG (1:5,000; 010–25521; Wako Pure Chemical Industries, Osaka, Japan) was used as an internal loading control. The membrane was washed and incubated with a horseradish peroxidase-conjugated secondary antibody. Bands were visualized using Amersham ECL Prime Western Blotting detection reagent (GE Healthcare Life Sciences, Uppsala, Sweden).

Immunohistochemistry

Tumor samples were fixed with 10% formaldehyde in phosphate-buffered saline (PBS), embedded in paraffin and sectioned into 4-μm-thick slices. The sections were incubated with rabbit polyclonal anti-MMP-13 IgG (1:200; ab84594; Abcam, Cambridge, UK) at 4°C overnight. The procedure for immunohistochemistry was previously described (21).

Plasmid construction and Dual-luciferase reporter assays

Partial wild-type sequences of the 3′ untranslated region (UTR) of MMP13 containing the miR-375 target site (positions 100–113 of the MMP13 3′ UTR) or sequences with a deleted miR-375 target site were inserted between the XhoI and PmeI restriction sites in the 3′ UTR of the hRluc gene in the psiCHECK-2 vector (product ID: C8021; Promega, Madison, WI, USA). TE-8 and TE-9 cells were transfected with 50 ng of the vector and 10 nM miR-375 using Lipofectamine 2000 (Thermo Fisher Scientific) in Opti-MEM (Thermo Fisher Scientific). The activities of firefly and Renilla luciferases were determined in lysates of transfected cells using a Dual-luciferase reporter assay system according to the manufacturer's recommendations (product ID: E1960; Promega). Data were normalized to firefly luciferase activity (ratio of Renilla/firefly luciferase activities).

Identification of downstream genes mediated by MMP13 in ESCC cells

Gene expression analyses of si-MMP13-transfected TE-8 and TE-9 cells revealed molecular targets mediated by MMP13 in ESCC cells. This method is described in more detail in previous studies (13,1820). Microarray results were deposited in the GEO database (accession number GSE82108).

Statistical analysis

Relationships between two or three variables and numerical values were analyzed using the Mann-Whitney U test or the Bonferroni-adjusted Mann-Whitney test. Spearman's rank test was used to evaluate the correlations between the expression levels of miR-375 and MMP13. Expert StatView version 5.0 (SAS Institute, Inc., Cary, NC, USA) was used in these analyses.

Results

Expression levels of miR-375 in ESCC clinical specimens and cell lines

We evaluated the expression levels of miR-375 in ESCC tissues (n=25), normal esophageal specimens (n=13), and ESCC cell lines (TE-8 and TE-9). The patient backgrounds and clinicopathological characteristics are shown in Table I. The expression levels of miR-375 were significantly downregulated in cancer tissues and ESCC cell lines compared with those in normal tissues (P<0.0001; Fig. 1A). Additionally, there were no significant relationships between the expression level of miR-375 and any of the clinicopathological parameters examined in this study (recurrence, T stage, N stage, vascular invasion, or survival rate).

Effects of miR-375 restoration on cell proliferation, migration and invasion in ESCC cell lines

To investigate the antitumor functions of miR-375, we performed gain-of-function studies using mature miRNA transfection of TE-8 and TE-9 cells.

Cell proliferation was significantly suppressed by miR-375 transfection in TE-9 cells in comparison with that of mock or miR-control transfectants (Fig. 1B). However, no changes were detected in TE-8 cells (Fig. 1B).

Migration assays showed that cell migration activity was significantly inhibited by miR-375 transfection in TE-8 and TE-9 cells in comparison with that in mock or miR-control transfectants (Fig. 1C). Additionally, Matrigel invasion assays demonstrated that cell invasion activity was significantly inhibited by miR-375 transfection in TE-8 and TE-9 cells in comparison with that in mock or miR-control transfectants (Fig. 1D).

Identification of putative target genes regulated by miR-375 in ESCC cells

To gain additional insights into the molecular pathways regulated by antitumor miR-375 in ESCC cells, we used a combination of in silico and gene expression analyses. The strategy for identification of the miR-375-regulated genes in ESCC cells is shown in Fig. 2.

In gene expression analyses, 2,897 and 1,007 genes were downregulated (log2 ratio <-0.5) in TE-8 and TE-9 miR-375 transfectants, respectively, in comparison with that in control transfectants. Our present expression data were deposited in the Gene Expression Omnibus (GEO accession number GSE77790). Among these downregulated genes, we searched for genes having putative miR-375 binding sites in their 3′ UTRs using the microRNA.org database. A total of 55 genes were identified as putative target genes of miR-375, and nine genes were upregulated in ESCC clinical specimens, as determined using ESCC expression data (GEO accession number: GSE20347; Table II).

Table II

Highly expressed genes putatively regulated by miR-375 in ESCC.

Table II

Highly expressed genes putatively regulated by miR-375 in ESCC.

Entrez Gene IDGene symbolDescriptionmiR-375 target sitesExpression in miR-375 transfectants FC (Log2)GEO data (GSE20347) FC (Log2)

TE-8TE-9
4322MMP13Matrix metalloproteinase 131−2.24−1.765.12
6004RGS16Regulator of G-protein signaling 163−1.50−0.922.45
4920ROR2Receptor tyrosine kinase-like orphan receptor 21−0.80−0.592.14
10202DHRS2 Dehydrogenase/reductase (SDR family) member 23−3.07−0.832.02
1956EGFREpidermal growth factor receptor1−0.93−0.781.58
655BMP7Bone morphogenetic protein 71−0.85−0.741.54
23363OBSL1Obscurin-like 11−0.80−0.711.52
23035PHLPP2PH domain and leucine rich repeat protein phosphatase 21−0.69−0.641.15
1896EDAEctodysplasin A1−0.72−0.631.09

In this study, we focused on MMP13 because its expression was most upregulated in ESCC clinical specimens and most downregulated in miR-375 transfectants. Moreover, previous studies have shown that the activation of MMPs is associated with cancer cell aggressiveness (22).

Expression of MMP13 in ESCC clinical specimens

Next, we validated the upregulation of MMP13 in the ESCC clinical specimens at both the mRNA and the protein levels. The expression of MMP13 was significantly upregulated in 25 ESCC specimens and ESCC cell lines compared with that in 13 normal specimens (P<0.0001; Fig. 3A). The Spearman's rank tests showed negative correlations between the expression of miR-375 and that of MMP13 (r=−0.661, P<0.0001; Fig. 3B).

Immunohistochemistry showed that MMP13 tended to be strongly expressed in ESCC lesions, whereas low expression was observed in normal esophageal epithelium (Fig. 3C).

Direct regulation of MMP13 by miR-375 in ESCC cells

We performed qRT-PCR to validate miR-375-mediated repression of MMP13 expression in ESCC cell lines. Our results showed that MMP13 mRNA was significantly reduced in miR-375 transfectants in comparison with that in mock or miR-control transfectants (P<0.0001; Fig. 4A). MMP13 protein expression was also repressed in miR-375 transfectants (Fig. 4B).

Next, we performed luciferase reporter assays using TE-8 and TE-9 cells to determine whether MMP13 had an actual target site for miR-375 binding. The microRNA.org database predicted that there was one putative target site in the 3′ UTR of MMP13 (Fig. 4C). Compared with the miR-control, luminescence intensity was significantly reduced by transfection with miR-375 at the miR-375 target site, positions 100–113, in the 3′ UTR of MMP13 (Fig. 4D).

Effects of silencing MMP13 on proliferation, migration and invasion in ESCC cells

To investigate the functional roles of MMP13 in ESCC cell lines, we performed loss-of-function assays by transfection of si-MMP13 into TE-8 and TE-9 cells.

First, we evaluated the knockdown efficiency of si-MMP13 transfection in ESCC cell lines. In the present study, we used two siRNAs targeting MMP13 (si-MMP13-1 and si-MMP13-2). According to qRT-PCR and western blot analyses, both siRNAs effectively downregulated MMP13 expression in both cell lines (Fig. 5A and B).

Cell proliferation, migration and invasion assays demonstrated that cell proliferation, migration, and invasion were inhibited in si-MMP13-transfected cells compared with those in mock- or siRNA-control-transfected cells (Fig. 5C–E).

Identification of downstream genes regulated by MMP13 in ESCC cells

To determine which downstream genes were regulated by MMP13, genome-wide gene expression and in silico analyses were performed in TE-8 and TE-9 cells transfected with si-MMP13.

Our expression analysis showed that a total of 298 genes were commonly downregulated (log2 ratio <-2.0) in TE-8 and TE-9 cells following si-MMP13 transfection. Among these genes, 52 were upregulated in ESCC clinical specimens, as determined using ESCC expression data (GEO accession number: GSE20347; Fig. 6 and Table III).

Table III

Downregulated genes in si-MMP13-transfected ESCC cell lines.

Table III

Downregulated genes in si-MMP13-transfected ESCC cell lines.

Entrez gene IDGene symbolDescriptionExpression in si-MMP13 transfectants FC (log2)GEO data (GSE20347) FC (log2)

TE8TE9
4322MMP13Matrix metallopeptidase 13 (collagenase 3)−4.42−4.475.12
1063CENPFCentromere protein F, 350/400 kDa−2.96−5.182.31
9928KIF14Kinesin family member 14−2.28−4.662.14
2842GPR19G protein-coupled receptor 19−2.67−3.742.12
983CDK1Cyclin-dependent kinase 1−2.07−3.781.95
55165CEP55Centrosomal protein 55 kDa−3.33−4.791.94
1033CDKN3Cyclin-dependent kinase inhibitor 3−2.08−3.731.94
7153TOP2ATopoisomerase (DNA) II alpha 170 kDa−3.36−5.011.91
10403NDC80NDC80 kinetochore complex component−2.19−3.691.76
9787DLGAP5Discs, large (Drosophila) homolog-associated protein 5−2.27−3.321.72
55215FANCIFanconi anemia, complementation group I−2.27−3.971.70
23306 TMEM194ATransmembrane protein 194A−2.31−2.791.68
4751NEK2NIMA-related kinase 2−2.70−3.841.66
2735GLI1GLI family zinc finger 1−2.70−3.311.63
3161HMMRHyaluronan-mediated motility receptor (RHAMM)−4.06−5.291.60
259266ASPMAsp (abnormal spindle) homolog, microcephaly associated (Drosophila)−2.17−3.811.56
4998ORC1Origin recognition complex, subunit 1−2.23−3.081.53
57405SPC25SPC25, NDC80 kinetochore complex component−2.16−4.121.48
28951TRIB2Tribbles pseudokinase 2−2.28−2.351.44
9603NFE2L3Nuclear factor, erythroid 2-like 3−2.00−2.511.42
9638FEZ1Fasciculation and elongation protein zeta 1 (zygin I)−2.27−2.971.42
9918NCAPD2Non-SMC condensin I complex, subunit D2−2.12−2.791.38
7468WHSC1Wolf-Hirschhorn syndrome candidate 1−2.43−3.361.33
100288413 ERVMER34-1Endogenous retrovirus group MER34, member 1−2.76−3.781.32
1062CENPECentromere protein E, 312 kDa−2.60−3.911.29
55063ZCWPW1Zinc finger, CW type with PWWP domain 1−3.19−3.441.25
81624DIAPH3Diaphanous-related formin 3−2.22−3.541.25
6119RPA3Replication protein A3, 14 kDa−2.34−3.421.24
8318CDC45Cell division cycle 45−2.13−4.071.23
64151NCAPGNon-SMC condensin I complex, subunit G−3.25−3.921.22
7083TK1Thymidine kinase 1, soluble−2.11−3.861.22
55732 C1orf112Chromosome 1 open reading frame 112−2.06−2.621.22
1058CENPACentromere protein A−2.02−3.861.18
55635DEPDC1DEP domain containing 1−2.33−3.441.18
3925STMN1Stathmin 1−2.66−4.511.17
3092HIP1Huntingtin interacting protein 1−2.71−3.511.17
5427POLE2Polymerase (DNA directed), epsilon 2, accessory subunit−2.18−4.371.15
1719DHFRDihydrofolate reductase−2.46−3.631.14
54830NUP62CLNucleoporin 62 kDa C-terminal like−2.17−2.221.10
5062PAK2p21 protein (Cdc42/Rac)-activated kinase 2−2.37−2.601.09
100129361 LOC100129361Chromosome X open reading frame 69-like−2.57−2.461.09
5933RBL1Retinoblastoma-like 1−3.24−4.431.08
4288MKI67Marker of proliferation Ki-67−2.14−4.871.03
81691 LOC81691Exonuclease NEF-sp−2.62−3.611.03
675BRCA2Breast cancer 2, early onset−2.90−4.041.00

We then validated the upregulation of CENPF and KIF14 mRNAs in ESCC clinical specimens. The expression of CENPF and KIF14 mRNAs was significantly upregulated in 25 ESCC specimens and ESCC cell lines compared with that in 13 normal specimens (P<0.0001; Fig. 7A and C). The Spearman's rank tests showed correlations between the expression of MMP13 and that of CENPF or KIF14 (CENPF: r=0.554, P=0.0007, Fig. 7B; KIF14: r=0.729, P<0.0001, Fig. 7D).

Discussion

Numerous studies of miRNA expression signatures in ESCC have shown that miR-375 is frequently downregulated in cancer tissues and functions as an antitumor miRNA (14,23). In the present study, we confirmed that miR-375 was markedly downregulated in cancer tissues and that ectopic expression of miR-375 significantly suppressed cancer cell migration and invasion. Thus, we found that loss of miR-375 expression enhanced cancer cell aggressiveness in ESCC. Many previous studies have shown that the expression of miR-375 is markedly decreased in several types of cancers and that miR-375 functions as an antitumor miRNA (15,24). In contrast to these antitumor activities, miR-375 is upregulated in pediatric acute myeloid leukemia (AML) and prostate cancer, suggesting that miR-375 acts as an oncogenic miRNA in these diseases (25,26). The dual function of miR-375 is very unique; thus, it is important to identify miR-375-regulated pathways in various cancer types.

It is also important to elucidate novel RNA networks regulated by antitumor miR-375 in ESCC cells. Previous studies have shown that insulin-like growth factor 1 receptor (IGF1R), lactate dehydrogenase B (LDHB), and astrocyte elevated gene-1/metadherin (AEG-1/MTDH) are directly regulated by miR-375 in ESCC cells (16,17). These target genes are upregulated in ESCC clinical specimens and functioned as oncogenes in this disease. Another unique characteristic of miRNAs is that a single miRNA can regulate a large number of RNA transcripts in human cells (27,28). Thus, the continuous identification of miR-375-regulated oncogenes in ESCC cells is important for elucidation of the molecular pathogenesis of ESCC.

In this study, we identified MMP13 as a direct target of antitumor miR-375 in ESCC cells. MMP13 (also known as collagenase 3) is a member of the collagenase subfamily of MMPs and functions to degrade a wide range of extracellular matrix components, including tenascin C, fibronectin and type I–IV collagen (29). Thus, MMP13 has a wide range of proteolytic functions, suggesting that MMP13 is involved in several physiological and pathological processes (30). High expression of MMP13 has been reported in rheumatoid arthritis, osteoarthritis and several types of cancers (22). Previous studies have also shown that high expression of MMP13 is associated with vascular invasion and lymph node metastasis in ESCC (31). Our present data demonstrated that knockdown of MMP13 markedly reduced cancer cell migration and invasion in ESCC cells.

The MMP13 gene has also been reported to be epigenetically regulated by several other miRNAs, including miR-125b and miR-143, in cancer cells (3234). Notably, our miRNA signatures have shown that miR-125b and miR-143 are down-regulated in ESCC and in oral and hypopharyngeal squamous cell carcinoma (1214). Moreover, functional assays have indicated that these miRNAs act as tumor suppressors in several cancers, including ESCC cells (3235). Loss of the expression of several antitumor miRNAs and activation of MMP13 may enhance cancer cell aggressiveness and metastasis. Thus, identification of miR-375/MMP13-mediated cancer pathways may facilitate the discovery of candidate therapeutic targets in ESCC.

Based on the above, we further investigated the downstream genes mediated by MMP13 in ESCC cells using genome-wide gene expression analysis. Our data showed that several centromere-associated proteins were regulated by MMP13-mediated genes, such as CENPF, CENPE, CENPA, CEP55, NDC80 and SPC25. Moreover, cell cycle-promoting genes, e.g., KIF14, CDK1, TOP2A, CDC45 and PAK2, were also downregulated by si-MMP13 in this study. Recent studies have reported that several genes encoding mitotic apparatus components are upregulated in cancer cells and contribute to cancer cell phenotypes (36,37). Therefore, overexpression of genes encoding mitotic apparatus components may represent a potential target for cancer drug development (38). Several compounds that inhibit centromere proteins and mitotic kinesins are being tested as potential cancer therapies in clinical trials (39).

Among these genes, we validated the overexpression of CENPF and KIF14 in ESCC clinical specimens. Previous studies have shown that CENPF is a master regulator of prostate cancer malignancy and that high expression of CEPNF is a prognostic indicator of poor survival and metastasis in patients with ESCC (40). KIF14 is a member of the kinesin superfamily of proteins and functions as a microtubule motor protein involved in cytokinesis and chromosome segregation (41). Overexpression of KIF14 has been reported in several cancers, and its expression is associated with cancer cell phenotypes (42,43). An in-depth functional analysis of these genes in ESCC cells is necessary to further characterize these pathways. Identification of the downstream genes regulated by the miR-375/MMP13 axis may lead to a better understanding of ESCC aggressiveness.

In conclusion, downregulation of miR-375 was frequently observed in ESCC clinical specimens, and miR-375 was shown to function as an antitumor miRNA in ESCC cells. To the best of our knowledge, this is the first report demonstrating that MMP13 is directly regulated by antitumor miR-375 and acts to regulate several cell cycle promoting genes. The identification of novel molecular pathways and targets regulated by the miR-375/MMP13 axis may lead to a better understanding of ESCC molecular pathogenesis.

Acknowledgements

We wish to thank the Joint Research Laboratory, Kagoshima University Graduate School of Medical and Dental Sciences, for the use of their facilities. The present study was supported by KAKENHI (C) grant 15K10801.

References

1 

Hongo M, Nagasaki Y and Shoji T: Epidemiology of esophageal cancer: Orient to Occident. Effects of chronology, geography and ethnicity. J Gastroenterol Hepatol. 24:729–735. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Pennathur A, Gibson MK, Jobe BA and Luketich JD: Oesophageal carcinoma. Lancet. 381:400–412. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Ohashi S, Miyamoto S, Kikuchi O, Goto T, Amanuma Y and Muto M: Recent advances from basic and clinical studies of esophageal squamous cell carcinoma. Gastroenterology. 149:1700–1715. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Enzinger PC and Mayer RJ: Esophageal cancer. N Engl J Med. 349:2241–2252. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Harada K, Baba Y, Ishimoto T, Shigaki H, Kosumi K, Yoshida N, Watanabe M and Baba H: The role of microRNA in esophageal squamous cell carcinoma. J Gastroenterol. 51:520–530. 2016. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Nohata N, Hanazawa T, Kikkawa N, Sakurai D, Fujimura L, Chiyomaru T, Kawakami K, Yoshino H, Enokida H, Nakagawa M, et al: Tumour suppressive microRNA-874 regulates novel cancer networks in maxillary sinus squamous cell carcinoma. Br J Cancer. 105:833–841. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Nohata N, Hanazawa T, Kinoshita T, Inamine A, Kikkawa N, Itesako T, Yoshino H, Enokida H, Nakagawa M, Okamoto Y, et al: Tumour-suppressive microRNA-874 contributes to cell proliferation through targeting of histone deacetylase 1 in head and neck squamous cell carcinoma. Br J Cancer. 108:1648–1658. 2013. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Fukumoto I, Hanazawa T, Kinoshita T, Kikkawa N, Koshizuka K, Goto Y, Nishikawa R, Chiyomaru T, Enokida H, Nakagawa M, et al: MicroRNA expression signature of oral squamous cell carcinoma: Functional role of microRNA-26a/b in the modulation of novel cancer pathways. Br J Cancer. 112:891–900. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Kano M, Seki N, Kikkawa N, Fujimura L, Hoshino I, Akutsu Y, Chiyomaru T, Enokida H, Nakagawa M and Matsubara H: miR-145, miR-133a and miR-133b: Tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma. Int J Cancer. 127:2804–2814. 2010. View Article : Google Scholar

15 

Kinoshita T, Hanazawa T, Nohata N, Okamoto Y and Seki N: The functional significance of microRNA-375 in human squamous cell carcinoma: Aberrant expression and effects on cancer pathways. J Hum Genet. 57:556–563. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Isozaki Y, Hoshino I, Nohata N, Kinoshita T, Akutsu Y, Hanari N, Mori M, Yoneyama Y, Akanuma N, Takeshita N, et al: Identification of novel molecular targets regulated by tumor suppressive miR-375 induced by histone acetylation in esophageal squamous cell carcinoma. Int J Oncol. 41:985–994. 2012.PubMed/NCBI

17 

Kong KL, Kwong DL, Chan TH, Law SY, Chen L, Li Y, Qin YR and Guan XY: MicroRNA-375 inhibits tumour growth and metastasis in oesophageal squamous cell carcinoma through repressing insulin-like growth factor 1 receptor. Gut. 61:33–42. 2012. View Article : Google Scholar

18 

Matsushita R, Yoshino H, Enokida H, Goto Y, Miyamoto K, Yonemori M, Inoguchi S, Nakagawa M and Seki N: Regulation of UHRF1 by dual-strand tumor-suppressor microRNA-145 (miR-145-5p and miR-145-3p): Inhibition of bladder cancer cell aggressiveness. Oncotarget. 7:28460–28487. 2016.PubMed/NCBI

19 

Goto Y, Kojima S, Nishikawa R, Kurozumi A, Kato M, Enokida H, Matsushita R, Yamazaki K, Ishida Y, Nakagawa M, et al: MicroRNA expression signature of castration-resistant prostate cancer: The microRNA-221/222 cluster functions as a tumour suppressor and disease progression marker. Br J Cancer. 113:1055–1065. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Goto Y, Kojima S, Nishikawa R, Enokida H, Chiyomaru T, Kinoshita T, Nakagawa M, Naya Y, Ichikawa T and Seki N: The microRNA-23b/27b/24-1 cluster is a disease progression marker and tumor suppressor in prostate cancer. Oncotarget. 5:7748–7759. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Kita Y, Nishizono Y, Okumura H, Uchikado Y, Sasaki K, Matsumoto M, Setoyama T, Tanoue K, Omoto I, Mori S, et al: Clinical and biological impact of cyclin-dependent kinase subunit 2 in esophageal squamous cell carcinoma. Oncol Rep. 31:1986–1992. 2014.PubMed/NCBI

22 

Brinckerhoff CE, Rutter JL and Benbow U: Interstitial collagenases as markers of tumor progression. Clin Cancer Res. 6:4823–4830. 2000.

23 

Yan JW, Lin JS and He XX: The emerging role of miR-375 in cancer. Int J Cancer. 135:1011–1018. 2014. View Article : Google Scholar

24 

Hui AB, Bruce JP, Alajez NM, Shi W, Yue S, Perez-Ordonez B, Xu W, O'Sullivan B, Waldron J, Cummings B, et al: Significance of dysregulated metadherin and microRNA-375 in head and neck cancer. Clin Cancer Res. 17:7539–7550. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Wang Z, Hong Z, Gao F and Feng W: Upregulation of microRNA-375 is associated with poor prognosis in pediatric acute myeloid leukemia. Mol Cell Biochem. 383:59–65. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Szczyrba J, Nolte E, Wach S, Kremmer E, Stöhr R, Hartmann A, Wieland W, Wullich B and Grässer FA: Downregulation of Sec23A protein by miRNA-375 in prostate carcinoma. Mol Cancer Res. 9:791–800. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Kinoshita T, Yip KW, Spence T and Liu FF: MicroRNAs in extracellular vesicles: Potential cancer biomarkers. J Hum Genet. Jul 7–2016.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

28 

Yonemori K, Kurahara H, Maemura K and Natsugoe S: MicroRNA in pancreatic cancer. J Hum Genet. Jun 2–2016.(Epub ahead of print). View Article : Google Scholar

29 

Knäuper V, López-Otin C, Smith B, Knight G and Murphy G: Biochemical characterization of human collagenase-3. J Biol Chem. 271:1544–1550. 1996. View Article : Google Scholar : PubMed/NCBI

30 

Vincenti MP and Brinckerhoff CE: Transcriptional regulation of collagenase (MMP-1, MMP-13) genes in arthritis: Integration of complex signaling pathways for the recruitment of gene-specific transcription factors. Arthritis Res. 4:157–164. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Etoh T, Inoue H, Yoshikawa Y, Barnard GF, Kitano S and Mori M: Increased expression of collagenase-3 (MMP-13) and MT1-MMP in oesophageal cancer is related to cancer aggressiveness. Gut. 47:50–56. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Xu N, Zhang L, Meisgen F, Harada M, Heilborn J, Homey B, Grandér D, Ståhle M, Sonkoly E and Pivarcsi A: MicroRNA-125b down-regulates matrix metallopeptidase 13 and inhibits cutaneous squamous cell carcinoma cell proliferation, migration, and invasion. J Biol Chem. 287:29899–29908. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Wu D, Ding J, Wang L, Pan H, Zhou Z, Zhou J and Qu P: microRNA-125b inhibits cell migration and invasion by targeting matrix metallopeptidase 13 in bladder cancer. Oncol Lett. 5:829–834. 2013.PubMed/NCBI

34 

Osaki M, Takeshita F, Sugimoto Y, Kosaka N, Yamamoto Y, Yoshioka Y, Kobayashi E, Yamada T, Kawai A, Inoue T, et al: MicroRNA-143 regulates human osteosarcoma metastasis by regulating matrix metalloprotease-13 expression. Mol Ther. 19:1123–1130. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Liu J, Mao Y, Zhang D, Hao S, Zhang Z, Li Z and Li B: MiR-143 inhibits tumor cell proliferation and invasion by targeting STAT3 in esophageal squamous cell carcinoma. Cancer Lett. 373:97–108. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Yuen KW, Montpetit B and Hieter P: The kinetochore and cancer: What's the connection? Curr Opin Cell Biol. 17:576–582. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Sagona AP and Stenmark H: Cytokinesis and cancer. FEBS Lett. 584:2652–2661. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Rath O and Kozielski F: Kinesins and cancer. Nat Rev Cancer. 12:527–539. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Huszar D, Theoclitou ME, Skolnik J and Herbst R: Kinesin motor proteins as targets for cancer therapy. Cancer Metastasis Rev. 28:197–208. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Aytes A, Mitrofanova A, Lefebvre C, Alvarez MJ, Castillo-Martin M, Zheng T, Eastham JA, Gopalan A, Pienta KJ, Shen MM, et al: Cross-species regulatory network analysis identifies a synergistic interaction between FOXM1 and CENPF that drives prostate cancer malignancy. Cancer Cell. 25:638–651. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Zhu C, Zhao J, Bibikova M, Leverson JD, Bossy-Wetzel E, Fan JB, Abraham RT and Jiang W: Functional analysis of human microtubule-based motor proteins, the kinesins and dyneins, in mitosis/cytokinesis using RNA interference. Mol Biol Cell. 16:3187–3199. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Corson TW, Zhu CQ, Lau SK, Shepherd FA, Tsao MS and Gallie BL: KIF14 messenger RNA expression is independently prognostic for outcome in lung cancer. Clin Cancer Res. 13:3229–3234. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Thériault BL, Pajovic S, Bernardini MQ, Shaw PA and Gallie BL: Kinesin family member 14: An independent prognostic marker and potential therapeutic target for ovarian cancer. Int J Cancer. 130:1844–1854. 2012. View Article : Google Scholar

Related Articles

Journal Cover

December-2016
Volume 49 Issue 6

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
Osako Y, Seki N, Kita Y, Yonemori K, Koshizuka K, Kurozumi A, Omoto I, Sasaki K, Uchikado Y, Kurahara H, 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.
APA
Osako, Y., Seki, N., Kita, Y., Yonemori, K., Koshizuka, K., Kurozumi, A. ... Natsugoe, S. (2016). Regulation of MMP13 by antitumor microRNA-375 markedly inhibits cancer cell migration and invasion in esophageal squamous cell carcinoma. International Journal of Oncology, 49, 2255-2264. https://doi.org/10.3892/ijo.2016.3745
MLA
Osako, Y., Seki, N., Kita, Y., Yonemori, K., Koshizuka, K., Kurozumi, A., Omoto, I., Sasaki, K., Uchikado, Y., Kurahara, H., Maemura, K., Natsugoe, S."Regulation of MMP13 by antitumor microRNA-375 markedly inhibits cancer cell migration and invasion in esophageal squamous cell carcinoma". International Journal of Oncology 49.6 (2016): 2255-2264.
Chicago
Osako, Y., Seki, N., Kita, Y., Yonemori, K., Koshizuka, K., Kurozumi, A., Omoto, I., Sasaki, K., Uchikado, Y., Kurahara, H., Maemura, K., Natsugoe, S."Regulation of MMP13 by antitumor microRNA-375 markedly inhibits cancer cell migration and invasion in esophageal squamous cell carcinoma". International Journal of Oncology 49, no. 6 (2016): 2255-2264. https://doi.org/10.3892/ijo.2016.3745