Molecular pathogenesis of esophageal squamous cell carcinoma: Identification of the antitumor effects of miR‑145‑3p on gene regulation

  • Authors:
    • Masataka Shimonosono
    • Tetsuya Idichi
    • Naohiko Seki
    • Yasutaka Yamada
    • Takayuki Arai
    • Takaaki Arigami
    • Ken Sasaki
    • Itaru Omoto
    • Yasuto Uchikado
    • Yoshiaki Kita
    • Hiroshi Kurahara
    • Kosei Maemura
    • Shoji Natsugoe
  • View Affiliations

  • Published online on: December 6, 2018     https://doi.org/10.3892/ijo.2018.4657
  • Pages: 673-688
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although miR‑145‑5p (the guide strand of the miR‑145 duplex) is established as a tumor suppressive microRNA (miRNA or miR), the functional significance of miR‑145‑3p (the passenger strand of the miR‑145 duplex) in cancer cells and its targets remains obscure. In our continuing analysis of esophageal squamous cell carcinoma (ESCC) pathogenesis, the aim of the present study was to identify important oncogenes and proteins that are controlled by miR‑145‑3p. Overexpression of miR‑145‑3p significantly reduced cancer cell proliferation, migration and invasive abilities, and further increased apoptotic abilities. In ESCC cells, 30 possible oncogenic targets were identified that might be regulated by miR‑145‑3p. Among these targets, dehydrogenase/reductase member 2 (DHRS2) and myosin IB (MYO1B) were focused on to investigate their functional roles in ESCC cells. DHRS2 and MYO1B were directly regulated by miR‑145‑3p in ESCC cells by dual luciferase reporter assays. Aberrantly expressed DHRS2 and MYOIB were detected in ESCC clinical specimens, and their overexpression enhanced cancer cell aggressiveness. Genes regulated by antitumor miR‑145‑3p were closely associated with the molecular pathogenesis of ESCC. The approach based on antitumor miRNAs may contribute to the understanding of ESCC molecular pathogenesis.

Introduction

Esophageal squamous cell carcinoma (ESCC) is the most prevalent type of esophageal cancer and is the sixth leading cause of cancer mortality worldwide (1). Due to ESCC's aggressive nature, the prognosis of ESCC patients with local invasion and distant metastasis at diagnosis is poor (2,3). Surgical resection is recognized as the preferred treatment for patients with newly diagnosed ESCC. However, high rates of tumor recurrence are notable (4,5). Neoadjuvant chemotherapy or chemoradiotherapy have been demonstrated to prolong overall survival for patients with ESCC (6-8). However, treatment options for recurrent cases are limited, and recently approved targeted therapies have not observed effective therapeutic effects (9,10). Therefore, ESCC patients with recurrence and metastasis require novel and effective treatment strategies.

The latest genomic analyses of ESCC cells have exhibited epigenetic modifications, e.g., DNA methylation, histone deacetylation, chromatin remodeling and non-coding RNA regulation (11,12). In that regard, microRNAs (miRNAs or miRs) consist of a class of small, well-conserved, non-coding RNAs that regulate RNA transcripts in a sequence-dependent manner (13). They participate in physiological and pathological conditions, e.g., cell differentiation, proliferation, motility and metabolism (14). A single miRNA can control a vast number of RNA transcripts in normal and diseased cells (15). Therefore, aberrantly expressed miRNAs may break down regulated RNA networks and contribute to cancer cells' development, metastasis and drug resistance (16).

A large number of miRNAs exhibit differential expression in ESCC, and they contribute to ESCC pathogenesis through their activities as oncogenes or tumor suppressors (11). Analyses of our original miRNA expression signatures by RNA-sequencing revealed that both strands of the miR-145 duplex (miR-145-5p, the guide strand and miR-145-3p, the passenger strand) were significantly downregulated in several types of cancers (17-20). The traditional view of miRNA function has held that only one strand of the miRNA duplex is incorporated into the RNA-induced silencing complex (RISC), becoming the active strand (guide strand). In contrast, the other strand (the passenger strand or miRNA*) was thought to be degraded and to have no function (21,22). However, recent studies of miRNA biogenesis have demonstrated that certain miRNA passenger strands are functional in plant and human cells (20,23).

Our recent studies have demonstrated that both strands of the miR-145 duplex have antitumor roles in lung cancer, bladder cancer, prostate cancer and head and neck cancer (17-20). In ESCC cells, both strands of the miR-150 duplex (miR-150-5p, the guide strand, and miR-150-3p, the passenger strand) acted as antitumor miRNAs through their targeting of SPOCK1 (24). A number of studies demonstrated that miR-145-5p acted as a pivotal antitumor miRNA in human cancers (25), including ESCC (26). In contrast, the functional significance and the targets of miR-145-3p are still obscure.

The aim of the present study was to demonstrate that miR-145-3p possesses antitumor functions and to identify its molecular targets, thereby elucidating ESCC pathogenesis. Thus, in ESCC cells, it was demonstrated that ectopic expression of miR-145-3p significantly blocked cancer cell proliferation, migration and invasion, similar to the actions of miR-145-5p. Furthermore, it was demonstrated that two genes, dehydrogenase/reductase member 2 (DHRS2) and myosin IB (MYO1B), were directly regulated by antitumor miR-145-3p in ESCC cells. Involvement of miR-145-3p (the passenger strand) is a novel concept in ESCC oncogenesis. The present approach, based on the roles of antitumor miRNA and its targets, will contribute to improved understanding of the molecular pathogenesis of ESCC.

Materials and methods

Human ESCC clinical specimens and cell lines

The present study was approved by the Bioethics Committee of Kagoshima University (Kagoshima, Japan; approval no. 28-65). Written prior informed consent and approval were obtained from all of the patients. All subjects in the patient cohort (n=29) were diagnosed with ESCC based upon pathologic criteria. ESCC with curative resection was included, salvage surgery was excluded. From this group, 22 clinical specimens and 12 noncancerous esophageal tissues were obtained. All samples were collected at the Kagoshima University hospital from March 2010 to September 2014. Collection of resected tissues occurred prior to preoperative therapy. The clinicopathological features of the patients are presented in Tables I and II. ESCC is more prevalent among males, thus almost all cases recruited for the present study were male.

Table I

Clinicopathological features of esophageal squamous cell carcinoma patients.

Table I

Clinicopathological features of esophageal squamous cell carcinoma patients.

No.Age (years)Sex DifferentiationTNMStagelyvRecurrence
152MalePoor1b00IA11+
272MaleModerate1b00IA01
369MaleModerate1b00IA00
456MaleModerate200IB01
566MaleModerate300IIA11
670MaleModerate300IIA11+
766MaleModerate300IIA11
871MaleWell300IIA12
962MaleWell1a10IIB00
1068MaleModerate1b10IIB11
1160MaleModerate1b10IIB11
1271MaleModerate1b10IIB00
1384MaleWell210IIB11
1479MaleModerate210IIB11
1560MaleModerate210IIB12
1668MalePoor1b20IIIA13+
1767MaleWell320IIIB22+
1855MaleModerate320IIIB11+
1975MaleModerate320IIIB11+
2074MaleModerate230IIIC31+
2157MalePoor330IIIC11+
2263MaleWell330IIIC21+

[i] ly, lymphatic invasion; M, metastasis; N, nodes; T, tumor; v, venous invasion.

Table II

Features of patients in noncancerous esophageal tissues.

Table II

Features of patients in noncancerous esophageal tissues.

No.Age (years)Sex
166Male
255Male
352Male
478Male
575Male
660Male
771Male
864Male
979Female
1081Male
1169Male
1284Male

In addition, 2 ESCC cell lines were used: TE-8, moderately differentiated and TE-9, poorly differentiated (27) (RIKEN BioResource Center, Tsukuba, Japan). Cell culture, extraction of total RNA and extraction of protein were performed as described in previous reports (28,29).

Transfection of mimic and inhibitor miRNA, small interfering (si)RNA into ESCC cells

In the present study, the following mimic and inhibitor miRNAs or siRNAs were transfected: mimic miRNAs (Ambion Pre-miR miRNA precursor; miR-145-5p: 5'-GUCCAGUUUUCCCAGGAAUCCCU-3', ID: PM11480; hsa-miR-145-3p: 5'-GGAUUCCUGGAAAUACUGUUCU-3', ID: PM13036; Thermo Fisher Scientific, Inc., Waltham, MA, USA), inhibitor miRNAs (Anti-miR miRNA Inhibitor; has-miR-145-3p: 5'-GGAUUCCUGGAAAUACUG UUCU-3', ID: AM13036; Applied Biosystems; Thermo Fisher Scientific, Inc.) and siRNAs (Stealth Select RNAi siRNA; si-DHRS2, ID: HSS145497 and HSS173461; si-MYO1B, ID: HSS106714 and HSS106716; Invitrogen; Thermo Fisher Scientific, Inc.) and negative control miRNA/siRNA (product ID: AM17111; Thermo Fisher Scientific, Inc.). The transfection procedures were performed as previously described (28-30).

Incorporation of miR-145-3p into the RISC: Assessment by argonaute 2 (Ago2) immunoprecipitation

miRNAs were transfected into TE-8 cells and miRNAs were isolated using an microRNA Isolation Kit, Human Ago2 (Wako Pure Chemical Industries, Ltd., Osaka, Japan) as described previously (28-30). The expression levels of Ago2-conjugated miRNAs were assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). miR-21 (assay ID; 000397; Applied Biosystems) was used as the internal control.

RT-qPCR

Quantification of miRNAs and mRNAs was performed by StepOnePlus™ Real-Time PCR System (Thermo Fisher Scientific, Inc.). The procedure used for RT-qPCR has been described previously (28,29). The expression levels of miRNAs were analyzed using TaqMan RT-qPCR assays (miR-145-5p, assay ID: 002278; miR-145-3p assay ID: 002149; Applied Biosystems; Thermo Fisher Scientific, Inc.). Data were normalized to RNU48 (assay ID: 001006; Applied Biosystems). In addition, the expression levels of DHRS2 and MYO1B were assessed with the following TaqMan probes: DHRS2, assay ID: Hs01061575_g1; MYO1B, assay ID: Hs00362654_m1; Applied Biosystems; Thermo Fisher Scientific, Inc.), and normalized to glucuronidase β (assay ID: Hs00939627_ml; Applied Biosystems; Thermo Fisher Scientific, Inc.).

Cell proliferation, migration, invasion and apoptosis assays

Protocols for determining cell proliferation (XTT assays), migration and invasion were described previously (28,29). For apoptosis assays, double staining with fluorescein isothiocyanate (FITC)-Annexin V and propidium iodide was carried out using a FITC Annexin V Apoptosis Detection kit (BD Biosciences, Franklin Lakes, NJ, USA) according to the manufacturer's recommendations. Stains were analyzed within 1 h using a flow cytometer (CyAn ADP analyzer; Beckman Coulter, Inc., Brea, CA, USA). Cells were identified as viable cells, dead cells, early apoptotic cells and late apoptotic cells using Summit 4.3 software (Beckman Coulter, Inc.). The percentages of early apoptotic and late apoptotic cells from each experiment were then compared. As a positive control, 1 µM gemcitabine hydrochloride (Tokyo Chemical Industry Co., Ltd., Tokyo, Japan) was used.

Identification of putative target genes regulated by miR-145-3p in ESCC cells

The present strategy for identification of miR-145-3p target genes is outlined in Fig. 1. The microarray data were deposited in the Gene Expression Omnibus (GEO) repository (https://www.ncbi.nlm.nih.gov/geo/) under accession number GSE107008. Putative target genes with a binding site for miR-145-3p were detected by TargetScanHuman ver.7.1 (http://www.targetscan.org/vert_71/). The GEO database (GSE20347) was used for assessment of the association between target genes and ESCC.

Exploration of downstream targets regulated by si-DHRS2 and si-MYO1B in ESCC

Genome-wide microarray analysis was used for identification of DHRS2 and MYO1B downstream targets. Expression data were deposited in a GEO database (GSE118966). A GEO database (GSE20347) was used for assessment of the association between target genes and ESCC. Our strategy for identification of DHRS2 and MYO1B downstream targets is outlined in Fig. 2. Expression analysis was performed on microarray data using SurePrint G3 Human 8×60K v3 (Agilent Technologies, Inc., Santa Clara, CA, USA).

Western blot analysis

Anti-human DHRS2 rabbit polyclonal immunoglobulin (Ig)G (1:1,000; HPA069551; Sigma-Aldrich; Merck KGaA) and anti-human MYO1B rabbit polyclonal IgG (1:250; HPA013607; Sigma-Aldrich; Merck KGaA) were used as primary antibodies. Anti-human β-actin mouse monoclonal IgG (1:2,000; A1978; Sigma-Aldrich; Merck KGaA) was used as an internal control. The protocol for Western blot analysis was described previously (29,30).

Immunohistochemistry

Tumor specimens were fixed, embedded and sectioned as described previously (31). Anti-human DHRS2 rabbit polyclonal IgG (1:250; HPA069551; Sigma-Aldrich, St. Louis, MO, USA) and anti-human MYO1B rabbit polyclonal IgG (1:300; HPA013607; Sigma-Aldrich; Merck KGaA were used as primary antibodies. The protocol followed was described previously (32).

Luciferase reporter assays

The following sequences were inserted into the psiCHECk-2 vector (C8021; Promega Corporation, Madison, WI, USA): The wild-type sequences of the 3'-untranslated regions (UTRs) of DHRS2 and MYO1B, or the deletion-type, which lacks the miR-145-3p target sites from DHRS2 (position 270-276) or MYO1B (position 88-94 or position 1,117-1,123). The cloned vectors were co-transfected into ESCC cells with mature miR-145-3p. The procedures for transfection and dual-luciferase reporter assays have been reported previously (28,29).

Statistical analysis

Associations between groups were analyzed using the Mann-Whitney U test or Tukey's multiple comparisons test following one-way analysis of variance. The differences between survival rates were analyzed by Kaplan-Meier survival curves and log-rank statistics. Spearman's rank test was used to evaluate the correlations between the expression levels of miR-145-3p, miR-145-5p, DHRS2 and MYO1B. Data are presented as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference. Expert StatView version 5.0 (SAS Institute, Inc., Cary, NC, USA) and GraphPad Prism version 7.04 (GraphPad Software, Inc., La Jolla, CA, USA) were used in these analyses.

Results

Expression levels of miR-145-5p and miR-145-3p in ESCC clinical specimens

Expression levels of miR-145-5p and miR-145-3p were significantly downregulated in cancer tissues and ESCC cell lines relative to normal tissues (Fig. 3A). Spearman's rank test demonstrated a positive correlation between the expression levels of miR-145-5p and miR-145-3p (Fig. 3B).

It was demonstrated that 5-year survival was significantly higher in patients who had high miR-145-3p expression than in patients with low miR-145-3p expression (Fig. 3C). There was no significant association between the expression level of miR-145-5p and patient survivals (Fig. 3C).

Ectopic expression of miR-145-5p and miR-145-3p: Impact on ESCC cells

To assess the ectopic expression of miR-145-5p and miR-145-3p, the mimic miRNA was transfected to ESCC cell lines (Fig. 3D). XTT assays demonstrated significant inhibition of cell proliferation in miR-145-5p and miR-145-3p transfectants (Fig. 3E). Likewise, cell migration and invasion were significantly inhibited following miR-145-5p or miR-145-3p transfection (Fig. 3F and G). The numbers of early apoptotic and late apoptotic cells were significantly larger in miR-145-5p or miR-145-3p transfectants than in mock or negative control transfectants (Fig. 4).

Incorporation of miR-145-3p into the RISC in ESCC cells

It was anticipated that the passenger strand of miR-145-3p was incorporated into the RISC and served as a tumor suppressor in ESCC cells. To verify that hypothesis, Ago2 was immunoprecipitated in cells that had been transfected with either miR-145-5p or miR-145-3p (Fig. 5A). Ago2 is an essential component of the RISC (16). Isolated Ago2-bound miRNAs were analyzed by RT-qPCR to confirm whether miR-145-5p and miR-145-3p bound to Ago2. In TE-8 cells, miR-145-5p transfectants demonstrated higher expression levels of miR-145-5p than mock transfectants, miR-control or miR-145-3p transfectants. Similarly, following miR-145-3p transfection, miR-145-3p was detected by Ago2 immunoprecipitation (Fig. 5B). miR-145-5p and miR-145-3p were demonstrated to bind to Ago2 separately and were incorporated into RISC, thereby demonstrating miRNA function.

Effects of co-transfection of mimic and inhibitor miR-145-3p into ESCC cells

To confirm the antitumor effects of miR-145-3p, rescue experiments were performed using mimic and inhibitor miR-145-3p with TE-8 cells (Fig. 6A). The rescue experiments indicated that cancer cell proliferation, migration and invasion were rescued in miR-145-3p inhibitor transfectants compared with restored miR-145-3p mimic only (Fig. 6B-D).

Searching for putative targets regulated by miR-145-3p in ESCC cells

The strategy to identify miR-145-3p target genes is presented in Fig. 1. Gene expression analyses demonstrated that 1,374 genes were downregulated (log2 ratio <-1.0) in miR-145-3p transfected TE-8 cells compared with control transfectants. The present expression data were deposited in the GEO repository under accession no. GSE107008. Among these downregulated genes, genes that had putative miR-145-3p binding sites in their 3'-UTRs were selected using information in the TargetScan database. A total of 280 genes were identified. Then, 30 genes were selected by restricting the identified genes to those strongly upregulated in ESCC clinical specimens (log2 ratio >1.0; GEO accession no. GSE20347; Table III).

Table III

Putative targets of miR-145-3p regulation in ESCC cells.

Table III

Putative targets of miR-145-3p regulation in ESCC cells.

Entrez gene IDGene symbolGene nameTE-8 miR-145-3p transfectantESCC GSE20347 fold-changeTarget site countPrognosis P-value: TCGA OncoLnc data
ESCAESCC
10202DHRS2 Dehydrogenase/reductase (SDR family) member 2−2.692.0210.0470.708
1848DUSP6Dual specificity phosphatase 6−2.611.0010.4560.469
55157DARS2Aspartyl-tRNA synthetase 2, mitochondrial−2.091.1720.5040.706
6646SOAT1Sterol O-acyltransferase 1−2.061.8110.7320.667
4430MYO1BMyosin IB−1.981.6120.3720.856
2115ETV1Ets variant 1−1.841.1010.1420.119
983CDK1Cyclin-dependent kinase 1−1.631.9510.6210.136
1719DHFRDihydrofolate reductase−1.481.1410.1990.465
51053GMNNGeminin, DNA replication inhibitor−1.461.3710.2740.189
23321TRIM2Tripartite motif containing 2−1.351.451<0.001a0.037a
55697VAC14Vac14 homolog (S. cerevisiae)−1.341.5310.0520.095
79789CLMNCalmin (calponin-like, transmembrane)−1.331.7920.5190.352
5654HTRA1HtrA serine peptidase 1−1.301.4410.8630.252
54830NUP62CLNucleoporin 62 kDa C-terminal like−1.281.1010.3130.313
204AK2Adenylate kinase 2−1.241.2120.6910.972
126321MFSD12Major facilitator superfamily domain containing 12−1.231.0510.8380.098
9532BAG2BCL2-associated athanogene 2−1.201.8020.1090.563
51029DESI2Desumoylating isopeptidase 2−1.181.2520.2610.19
6711SPTBN1Spectrin, β, non-erythrocytic 1−1.181.2110.5150.504
1163CKS1BCDC28 protein kinase regulatory subunit 1B−1.142.0210.6580.658
8534CHST1Carbohydrate (keratan sulfate Gal-6) Sulfotransferase 1−1.141.5210.9430.983
5174PDZK1PDZ domain containing 1−1.141.6510.5650.462
875CBS Cystathionine-β-synthase−1.143.5520.1990.160
23516SLC39A14Solute carrier family 39 (zinc transporter), member 14−1.122.6810.3220.101
6790AURKAAurora kinase A−1.072.1710.3220.051
79718TBL1XR1Transducin (β)-like 1 X-linked receptor 1−1.061.3010.5790.377
23141ANKLE2Ankyrin repeat and LEM domain containing 2−1.051.1610.4180.585
23649POLA2Polymerase (DNA directed), α 2, accessory subunit−1.041.3510.6420.842
64151NCAPGNon-SMC condensin I complex, subunit G−1.031.2210.1980.056
22848AAK1AP2 associated kinase 1−1.031.2730.6340.346

a Poor prognosis with low expression. miR, microRNA; ESCC, esophageal squamous cell carcinoma; ESCA, esophageal carcinoma.

In this fashion, DHRS2 was focused on, as its expression was the most downregulated in miR-145-3p transfectants and the most upregulated in ESCC clinical specimens. Additionally, MYO1B was examined, as it was more highly downregulated in miR-145-3p transfectants and was more upregulated in ESCC clinical specimens. In addition, our previous study had demonstrated that the activation of MYO1B was associated with cancer cell aggressiveness (20).

Direct regulation of DHRS2 and MYO1B by miR-145-3p in ESCC cells

The finding that DHRS2 and MYO1B were downregulated by expression of miR-145-3p was further investigated (Figs. 7 and 8). ESCC cells (TE-8) that had been transfected with miR-145-3p were examined. Using RT-qPCR, it was demonstrated that DHRS2 and MYO1B mRNA levels were significantly reduced by miR-145-3p transfection (Figs. 7A and 8A). Furthermore, western blot analysis was performed to measure the expression levels of DHRS2 and MYO1B proteins in the transfectants. Results demonstrated that the proteins were also reduced by miR-145-3p transfection (Figs. 7B and 8B).

Whether miR-145-3p directly regulated DHRS2 and MYO1B genes in a sequence-dependent manner was then evaluated.

The Human TargetScan database predicted that DHRS2 had 1 binding site (positions 270-276) for miR-145-3p in the 3'-UTR (Fig. 7C). Accordingly, luciferase reporter assays were carried out with vectors that included either the wild-type or deletion-type 3'-UTR of DHRS2. Co-transfection with miR-145-3p and vectors including the wild-type sequence significantly reduced luciferase activity compared with those in mock and miR-control transfectants in position 270-276 of the DHRS2 3'-UTR (Fig. 7D).

MYO1B had 2 binding sites (positions 88-94 and 1,117-1,123) for miR-145-3p in the 3'-UTR (Fig. 8C). Luciferase activities were significantly reduced in position 1,117-1,123 of the MYO1B 3'-UTR (Fig. 8D).

Effects of silencing DHRS2 and MYO1B in ESCC cells

Subsequently, siRNAs were transfected into TE-8 cells to examine the function of DHRS2 and MYO1B in ESCC cells (Figs. 9 and 10). The mRNA and protein expression levels of DHRS2 and MYO1B were decreased by si-DHRS2 and si-MYO1B, respectively (Figs. 9A and B, and 10A and B). Subsequently, the effects of DHRS2 or MYO1B knockdown on ESCC cell proliferation, migration and invasion were investigated.

Cancer cell proliferation was significantly suppressed in si-DHRS2 or si-MYO1B transfectants compared with mock and si-RNA-control transfectants. Additionally, migration and invasion activities were significantly inhibited in si-DHRS2 or si-MYO1B transfectants (Figs. 9C-E and 10C-E). In the apoptosis assays, si-DHRS2_1/si-DHRS2_2 and si-MYO1B_1/si-MYO1B_2 transfections significantly increased apoptotic TE-8 cells (Figs. 9F and 10F).

Expression of DHRS2 and MYO1B in ESCC clinical specimens

Based upon the findings above, it was of great interest to use RT-qPCR to determine the expression levels of DHRS2 and MYO1B in clinical specimens. DHRS2 and MYO1B expression levels were significantly upregulated in ESCC tumor tissues (Fig. 11A). Additionally, the 5-year survival rates of ESSC patients were significantly shorter in those with elevated DHRS2 expression compared with those with low expression (Fig. 11B). There was no significant association between the expression levels of MYO1B and the survival rate (Fig. 11B).

Spearman's rank test demonstrated a negative correlation between the expression of DHRS2 and miR-145-3p, and MYO1B and miR-145-3p (Fig. 11C). Furthermore, the protein expression levels of DHRS2 and MYO1B were examined in ESCC clinical specimens by immunostaining. Both DHRS2 and MYO1B were strongly expressed in cancer tissues, but not in noncancerous epithelia (Fig. 12).

Exploration of downstream targets regulated by si-DHRS2 and si-MYO1B in ESCC

The present strategy for selecting downstream genes regulated by DHRS2 and MYO1B is demonstrated in Fig. 2. A total of 756 genes were commonly downregulated (log2 ratio <-1.5) in si-DHRS2-transfected TE-8 cells. The upregulated genes in ESCC tissues were also assessed by GEO database analyses (GEO accession no. GSE20347). With that approach, 85 candidate genes downstream from DHRS2 were identified (Table IV). Furthermore, a total of 334 genes were commonly downregulated (log2 ratio <−1.5) in si-MYO1B-transfected TE-8 cells. In a similar approach, 32 candidate genes downstream from MYO1B were identified (Table V).

Table IV

Putative targets of si-DHRS2 regulation in ESCC cells.

Table IV

Putative targets of si-DHRS2 regulation in ESCC cells.

Entrez gene IDGene symbolGene nameESCC GSE20347 fold-changeTE-8 si-DHRS2 transfectant
1592CYP26A1Cytochrome P450, family 26, subfamily A, polypeptide 11.86−3.78
10112KIF20AKinesin family member 20A1.50−3.28
259266ASPMAsp (abnormal spindle) homolog, microcephaly associated (Drosophila)1.56−3.08
27074LAMP3 Lysosomal-associated membrane protein 31.56−3.03
11098PRSS23Protease, serine, 231.86−3.02
10202DHRS2 Dehydrogenase/reductase (SDR family) member 22.02−2.98
4322MMP13Matrix metallopeptidase 13 (collagenase 3)5.12−2.93
79075DSCC1DNA replication and sister chromatid cohesion 11.95−2.80
983CDK1Cyclin-dependent kinase 11.95−2.79
332BIRC5Baculoviral IAP repeat containing 51.59−2.72
995CDC25CCell division cycle 25C1.51−2.71
11065UBE2C Ubiquitin-conjugating enzyme E2C1.68−2.70
4751NEK2NIMA-related kinase 21.66−2.70
1033CDKN3Cyclin-dependent kinase inhibitor 31.94−2.70
11339OIP5Opa interacting protein 51.63−2.68
2842GPR19G protein-coupled receptor 192.12−2.64
10615SPAG5Sperm associated antigen 51.59−2.60
9055PRC1Protein regulator of cytokinesis 11.58−2.60
699BUB1BUB1 mitotic checkpoint serine/threonine kinase2.04−2.59
991CDC20Cell division cycle 201.54−2.58
55355HJURPHolliday junction recognition protein1.79−2.51
7153TOP2ATopoisomerase (DNA) II alpha 170 kDa1.91−2.46
10403NDC80NDC80 kinetochore complex component1.76−2.45
55388MCM10Minichromosome maintenance complex component 101.90−2.45
2263FGFR2Fibroblast growth factor receptor 21.65−2.44
3161HMMRHyaluronan-mediated motility receptor (RHAMM)1.60−2.42
1063CENPFCentromere protein F, 350/400 kDa2.31−2.40
7272TTKTTK protein kinase1.58−2.36
9401RECQL4RecQ protein-like 41.92−2.35
9355LHX2LIM homeobox 22.63−2.33
22836RHOBTB3Rho-related BTB domain containing 31.98−2.33
54478FAM64AFamily with sequence similarity 64, member A1.60−2.33
9133CCNB2Cyclin B21.68−2.27
9787DLGAP5Discs, large (Drosophila) homolog-associated protein 51.72−2.26
9156EXO1Exonuclease 11.89−2.26
3833KIFC1Kinesin family member C12.15−2.19
347733TUBB2BTubulin, beta 2B class IIb1.86−2.18
220134SKA1Spindle and kinetochore associated complex subunit 11.73−2.18
4291MLF1Myeloid leukemia factor 11.90−2.15
8438RAD54LRAD54-like (S. cerevisiae)2.31−2.14
79019CENPMCentromere protein M2.10−2.14
51514DTLDenticleless E3 ubiquitin protein ligase homolog (Drosophila)1.62−2.12
55872PBKPDZ binding kinase1.70−2.11
3790KCNS3Potassium voltage-gated channel, modifier subfamily S, member 32.32−2.11
51512GTSE1G-2 and S-phase expressed 12.02−2.10
9493KIF23Kinesin family member 231.96−2.10
5983RFC3Replication factor C (activator 1) 3, 38 kDa1.74−2.09
81611ANP32EAcidic (leucine-rich) nuclear phosphoprotein 32 family, member E1.52−2.07
83461CDCA3Cell division cycle associated 32.14−2.05
23350U2SURPU2 snRNP-associated SURP domain containing1.62−2.01
6790AURKAAurora kinase A2.17−2.00
55165CEP55Centrosomal protein 55 kDa1.94−1.99
80178C16orf59Chromosome 16 open reading frame 591.61−1.98
2305FOXM1Forkhead box M12.16−1.96
24137KIF4AKinesin family member 4A1.95−1.94
22974TPX2TPX2, microtubule-associated1.65−1.94
55215FANCIFanconi anemia, complementation group I1.70−1.91
10635RAD51AP1RAD51 associated protein 12.20−1.88
993CDC25ACell division cycle 25A1.88−1.88
2175FANCAFanconi anemia, complementation group A1.93−1.85
4171MCM2Minichromosome maintenance complex component 22.55−1.82
2491CENPICentromere protein I1.81−1.81
655BMP7Bone morphogenetic protein 71.54−1.77
4998ORC1Origin recognition complex, subunit 11.53−1.76
10036CHAF1AChromatin assembly factor 1, subunit A (p150)1.75−1.76
4085MAD2L1MAD2 mitotic arrest deficient-like 1 (yeast)1.67−1.76
3149HMGB3High mobility group box 32.01−1.74
29028ATAD2ATPase family, AAA domain containing 21.96−1.73
9837GINS1GINS complex subunit 1 (Psf1 homolog)1.64−1.73
51659GINS2GINS complex subunit 2 (Psf2 homolog)1.86−1.72
5984RFC4Replication factor C (activator 1) 4, 37 kDa2.08−1.69
55839CENPNCentromere protein N1.72−1.66
7078TIMP3TIMP metallopeptidase inhibitor 32.23−1.66
27346TMEM97Transmembrane protein 971.67−1.65
9928KIF14Kinesin family member 142.14−1.64
3625INHBBInhibin, β B1.75−1.63
10721POLQPolymerase (DNA directed), θ1.51 -1.62
1663DDX11DEAD/H (Asp-Glu-Ala-Asp/His) box helicase 112.08−1.60
9319TRIP13Thyroid hormone receptor interactor 132.02−1.58
4605MYBL2V-myb avian myeloblastosis viral oncogene3.08−1.56
homolog-like 2
51762RAB8BRAB8B, member RAS oncogene family1.76−1.54
91860CALML4Calmodulin-like 41.90−1.54
10293TRAIPTRAF interacting protein1.50−1.52
2237FEN1Flap structure-specific endonuclease 11.54−1.51
55753OGDHLOxoglutarate dehydrogenase-like2.37−1.51

[i] DHRS2, dehydrogenase/reductase member 2; ESCC, esophageal squamous cell carcinoma; si, small interfering RNA.

Table V

Putative targets of si-MYO1B regulation in ESCC cells.

Table V

Putative targets of si-MYO1B regulation in ESCC cells.

Entrez gene IDGene symbolGene nameESCC GSE20347 fold-changeTE-8 si-MYO1B transfectant
4430MYO1BMyosin IB1.61−3.60
347733TUBB2BTubulin, β 2B class IIb1.86−2.89
9837GINS1GINS complex subunit 1 (Psf1 homolog)1.64−2.72
4322MMP13Matrix metallopeptidase 13 (collagenase 3)5.12−2.66
79075DSCC1DNA replication and sister chromatid cohesion 11.95−2.55
4312MMP1Matrix metallopeptidase 1 (interstitial collagenase)6.54−2.40
51514DTLDenticleless E3 ubiquitin protein ligase homolog (Drosophila)1.62−2.38
4319MMP10Matrix metallopeptidase 10 (stromelysin 2)4.51−2.28
23657SLC7A11Solute carrier family 7 (anionic amino acid transporter light chain, xc- system), member 111.97−2.20
5983RFC3Replication factor C (activator 1) 3, 38 kDa1.74−2.11
10202DHRS2 Dehydrogenase/reductase (SDR family) member 22.02−2.04
2491CENPICentromere protein I1.81−2.01
4085MAD2L1MAD2 mitotic arrest deficient-like 1 (yeast)1.67−1.99
6574SLC20A1Solute carrier family 20 (phosphate transporter), member 11.52−1.91
4998ORC1Origin recognition complex, subunit 11.53−1.84
81611ANP32EAcidic (leucine-rich) nuclear1.52−1.83
phosphoprotein 32 family, member E
10669CGREF1Cell growth regulator with EF-hand domain 11.62−1.81
55388MCM10Minichromosome maintenance complex component 101.90−1.79
2119ETV5Ets variant 52.05−1.79
11199ANXA10Annexin A102.06−1.76
655BMP7Bone morphogenetic protein 71.54−1.73
983CDK1Cyclin-dependent kinase 11.95−1.72
55872PBKPDZ binding kinase1.70−1.70
4072EPCAMEpithelial cell adhesion molecule2.56−1.68
8914TIMELESSTimeless circadian clock1.54−1.65
9518GDF15Growth differentiation factor 151.58−1.65
55215FANCIFanconi anemia, complementation group I1.70−1.62
11339OIP5Opa interacting protein 51.63−1.61
10635RAD51AP1RAD51 associated protein 12.20−1.59
332BIRC5Baculoviral IAP repeat containing 51.59−1.58
995CDC25CCell division cycle 25C1.51−1.54
8438RAD54LRAD54-like (S. cerevisiae)2.31−1.52

[i] MYO1B, myosin IB; ESCC, esophageal squamous cell carcinoma; si, small interfering RNA.

Discussion

Downregulation of miR-145-5p has frequently been observed in a wide range of cancers, including ESCC (32). A number of previous studies have demonstrated that ectopic expression of miR-145-5p suppressed cancer cell proliferation, migration, invasion and drug resistance both in vitro and in vivo (25,26,33). Notably, the promoter region of pre-miR-145 has a p53 response element and its expression is controlled by activation of p53 under various conditions (34). Therefore, both strands of the miR-145 duplex are pivotal tumor suppressor miRNAs controlled by p53.

Analyses of the miRNA expression signatures demonstrated that certain miRNA passenger strands were downregulated and acted as antitumor miRNAs in several cancers, e.g., miR-145-3p, miR-150-3p, miR-148a-5p and miR-99a-3p (20,24,35,36). Our previous studies demonstrated that antitumor miR-145-3p directly targeted oncogenes, e.g., MTDH in lung adenocarcinoma, UHRF1 in bladder cancer, MYO1B in head and neck cancer and MELK, NCAPG, BUB1 and CDK1 in prostate cancer (17-20). Another group demonstrated that miR-145-3p inhibited cell growth, motility and chemotaxis in non-small cell lung cancer by targeting pyruvate dehydrogenase kinase 1 through suppressing the mechanistic target of rapamycin pathway (37). These findings indicated that antitumor miR-145-3p is associated with cancer pathogenesis.

Exploring the RNA network controlled by antitumor miR-145-3p expands the understanding of the novel molecular pathogenesis of ESCC. In the present study, 30 genes were identified as putative oncogenes based on miR-145-3p regulation in ESCC cells. Among these genes, the following 3 were reported to be cancer-promoting genes in ESCC: cyclin dependent kinase 1 (CDK1), aurora kinase A (AURKA) and transducin β like 1 X-linked receptor 1 (TBL1XR1) (38-43). Identification of the target genes controlled by miR-145-3p is important for understanding the underlying molecular pathogenesis of ESCC.

The present study demonstrated that both DHRS2 and MYO1B were directly regulated by miR-145-3p in ESCC cells. Overexpression of DHRS2 and MYO1B was observed in ESCC clinical specimens, and overexpression was associated with cancer cell aggressiveness. DHRS2 was initially cloned from a HepG2 human hepatocarcinoma cDNA library and named HEP27 (44). DHRS2 is a member of the short-chain dehydrogenase/reductase (SDR) family that metabolizes many different compounds (45). HEP27 protein interacts with MDM2, which is a negative regulator of p53, resulting in p53 stabilization and induction of p53 transcriptional target genes (46). More recently, downregulation of DHRS2 was reported in ESCC tissues and its downregulation was associated with ESCC aggressiveness and clinical staging (47). Thus, that report arrived at the opposite conclusions from those in our study. Further investigation of DHRS2 function in cancer cells will be necessary.

MYO1B is a member of the membrane-associated class I myosin family and it bridges membrane and actin cytoskeleton in several cellular processes (48). It was recently demonstrated that overexpression of MYO1B is associated with head and neck cancer pathogenesis (20). Importantly, antitumor miR-145-3p directly regulated expression of MYO1B in head and neck cells (20). A previous in vivo study demonstrated that downregulation of MYO1B inhibited cervical lymph node metastasis in head and neck cancer cells (49). These findings indicate that aberrantly expressed MYO1B is associated with cancer cell aggressiveness and metastasis. MYO1B may be a novel diagnostic and therapeutic target for patients with ESCC.

Downstream genes modulated by DHRS2 or MYO1B in ESCC cells were investigated. Previous studies have demonstrated that several aberrantly expressed oncogenes (CDK1, BIRC5, BUB1, TOP2A, CENPF, FOXM1 and AURKA) enhanced cancer cell aggressiveness (38,50-54). Notably, MMP13 may be controlled by DHRS2 and MYO1B in ESCC cells. Our recent study demonstrated that overexpression of MMP13 occurred in ESCC clinical specimens and the expression of MMP13 promoted cancer cell proliferation, migration and invasion (28). Identification of the downstream genes regulated by the miR-145-3p/DHRS2 or miR-145-3p/MYO1B axis may improve the understanding of ESCC aggressiveness.

In conclusion, genes controlled by the antitumor activity of miR-145-3p were closely associated with ESCC pathogenesis. Association of the passenger strand of miRNA is a novel concept of cancer research. DHRS2 and MYO1B were directly regulated by miR-145-3p in ESCC cells. Aberrantly expressed DHRS2 and MYO1B enhanced ESCC cell aggressiveness. Elucidation of antitumor miRNAs controlling RNA networks may provide novel prognostic markers and therapeutic targets for this disease.

Funding

The present study was supported by KAKENHI grants nos. 15K10801, 18K16322, 16K10508, 17K10706, 16K10510, 18K08687, 18K08626 and 17H04285.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

MS and TI performed the majority of the study and wrote the manuscript. NS and SN designed the study and wrote the manuscript. YY, TAra, YK and HK performed the experiments and data interpretation. TAri, KS, IO, YU and KM provided sample collection and clinical support. All authors reviewed, edited and approved the final version of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Bioethics Committee of Kagoshima University (Kagoshima, Japan; approval no. 28-65). Written prior informed consent and approval were obtained from all patients.

Patient consent for publication

All patients had provided written informed consent prior to surgery.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar

2 

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

3 

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

4 

Mariette C, Piessen G and Triboulet JP: Therapeutic strategies in oesophageal carcinoma: Role of surgery and other modalities. Lancet Oncol. 8:545–553. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Cai XW, Yu WW, Yu W, Zhang Q, Feng W, Liu MN, Sun MH, Xiang JQ, Zhang YW and Fu XL: Tissue-based quantitative proteomics to screen and identify the potential biomarkers for early recurrence/metastasis of esophageal squamous cell carcinoma. Cancer Med. 7:2504–2517. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Cooper JS, Guo MD, Herskovic A, Macdonald JS, Martenson JA Jr, Al-Sarraf M, Byhardt R, Russell AH, Beitler JJ, Spencer S, et al Radiation Therapy Oncology Group: Chemoradiotherapy of locally advanced esophageal cancer: Long-term follow-up of a prospective randomized trial (RTOG 85-01). JAMA. 281:1623–1627. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Natsugoe S, Ikeda M, Baba M, Churei H, Hiraki Y, Nakajo M and Aikou T; Kyushu Study Group for Adjuvant Therapy of Esophageal Cancer: Long-term survivors of advanced esophageal cancer without surgical treatment: A multicenter questionnaire survey in Kyushu, Japan. Dis Esophagus. 16:239–242. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Ando N, Kato H, Igaki H, Shinoda M, Ozawa S, Shimizu H, Nakamura T, Yabusaki H, Aoyama N, Kurita A, et al: A randomized trial comparing postoperative adjuvant chemotherapy with cisplatin and 5-fluorouracil versus preoperative chemotherapy for localized advanced squamous cell carcinoma of the thoracic esophagus (JCOG9907). Ann Surg Oncol. 19:68–74. 2012. View Article : Google Scholar

9 

Dutton SJ, Ferry DR, Blazeby JM, Abbas H, Dahle-Smith A, Mansoor W, Thompson J, Harrison M, Chatterjee A, Falk S, et al: Gefitinib for oesophageal cancer progressing after chemotherapy (COG): A phase 3, multicentre, double-blind, placebo-controlled randomised trial. Lancet Oncol. 15:894–904. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Suntharalingam M, Winter K, Ilson D, Dicker AP, Kachnic L, Konski A, Chakravarthy AB, Anker CJ, Thakrar H, Horiba N, et al: Effect of the addition of cetuximab to paclitaxel, cisplatin, and radiation therapy for patients with esophageal cancer: The NRG Oncology RTOG 0436 phase 3 randomized clinical trial. JAMA Oncol. 3:1520–1528. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Hemmatzadeh M, Mohammadi H, Karimi M, Musavishenas MH and Baradaran B: Differential role of microRNAs in the pathogenesis and treatment of Esophageal cancer. Biomed Pharmacother. 82:509–519. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Lin DC, Wang MR and Koeffler HP: Genomic and epigenomic aberrations in esophageal squamous cell carcinoma and implications for patients. Gastroenterology. 154:374–389. 2018. View Article : Google Scholar :

13 

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

14 

Chen CZ: MicroRNAs as oncogenes and tumor suppressors. N Engl J Med. 353:1768–1771. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Friedman RC, Farh KK, Burge CB and Bartel DP: Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 19:92–105. 2009. View Article : Google Scholar :

16 

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

17 

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

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. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Yamada Y, Koshizuka K, Hanazawa T, Kikkawa N, Okato A, Idichi T, Arai T, Sugawara S, Katada K, Okamoto Y, et al: Passenger strand of miR-145-3p acts as a tumor-suppressor by targeting MYO1B in head and neck squamous cell carcinoma. Int J Oncol. 52:166–178. 2018.

21 

Schwarz DS, Hutvágner G, Du T, Xu Z, Aronin N and Zamore PD: Asymmetry in the assembly of the RNAi enzyme complex. Cell. 115:199–208. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Mah SM, Buske C, Humphries RK and Kuchenbauer F: miRNA*: A passenger stranded in RNA-induced silencing complex? Crit Rev Eukaryot Gene Expr. 20:141–148. 2010. View Article : Google Scholar

23 

Liu WW, Meng J, Cui J and Luan YS: Characterization and Function of MicroRNA*s in Plants. Front Plant Sci. 8:22002017. View Article : Google Scholar

24 

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

25 

Sachdeva M and Mo YY: MicroRNA-145 suppresses cell invasion and metastasis by directly targeting mucin 1. Cancer Res. 70:378–387. 2010. View Article : Google Scholar :

26 

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

27 

Nishihira T, Hashimoto Y, Katayama M, Mori S and Kuroki T: Molecular and cellular features of esophageal cancer cells. J Cancer Res Clin Oncol. 119:441–449. 1993. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Yonemori K, Seki N, Kurahara H, Osako Y, Idichi T, Arai T, Koshizuka K, Kita Y, Maemura K and Natsugoe S: ZFP36L2 promotes cancer cell aggressiveness and is regulated by antitumor microRNA-375 in pancreatic ductal adenocarcinoma. Cancer Sci. 108:124–135. 2017. View Article : Google Scholar :

30 

Yoshino H, Chiyomaru T, Enokida H, Kawakami K, Tatarano S, Nishiyama K, Nohata N, Seki N and Nakagawa M: The tumour-suppressive function of miR-1 and miR-133a targeting TAGLN2 in bladder cancer. Br J Cancer. 104:808–818. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Harada K, Baba Y, Ishimoto T, Kosumi K, Tokunaga R, Izumi D, Ohuchi M, Nakamura K, Kiyozumi Y, Kurashige J, et al: Suppressor microRNA-145 is epigenetically regulated by promoter hypermethylation in esophageal squamous cell carcinoma. Anticancer Res. 35:4617–4624. 2015.PubMed/NCBI

32 

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. View Article : Google Scholar : PubMed/NCBI

33 

Zeng JF, Ma XQ, Wang LP and Wang W: MicroRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer. World J Gastroenterol. 23:2337–2345. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Sachdeva M, Zhu S, Wu F, Wu H, Walia V, Kumar S, Elble R, Watabe K and Mo YY: p53 represses c-Myc through induction of the tumor suppressor miR-145. Proc Natl Acad Sci USA. 106:3207–3212. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Arai T, Okato A, Yamada Y, Sugawara S, Kurozumi A, Kojima S, Yamazaki K, Naya Y, Ichikawa T and Seki N: Regulation of NCAPG by miR-99a-3p (passenger strand) inhibits cancer cell aggressiveness and is involved in CRPC. Cancer Med. 7:1988–2002. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Idichi T, Seki N, Kurahara H, Fukuhisa H, Toda H, Shimonosono M, Okato A, Arai T, Kita Y, Mataki Y, et al: Molecular pathogenesis of pancreatic ductal adenocarcinoma: Impact of passenger strand of pre-miR-148a on gene regulation. Cancer Sci. 109:2013–2026. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Chen GM, Zheng AJ, Cai J, Han P, Ji HB and Wang LL: microRNA-145-3p inhibits non-small cell lung cancer cell migration and invasion by targeting PDK1 via the mTOR signaling pathway. J Cell Biochem. 119:885–895. 2018. View Article : Google Scholar

38 

Dutertre S, Descamps S and Prigent C: On the role of aurora-A in centrosome function. Oncogene. 21:6175–6183. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Fung TK, Yam CH and Poon RY: The N-terminal regulatory domain of cyclin A contains redundant ubiquitination targeting sequences and acceptor sites. Cell Cycle. 4:1411–1420. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Li JY, Daniels G, Wang J and Zhang X: TBL1XR1 in physiological and pathological states. Am J Clin Exp Urol. 3:13–23. 2015.PubMed/NCBI

41 

Liu L, Lin C, Liang W, Wu S, Liu A, Wu J, Zhang X, Ren P, Li M and Song L: TBL1XR1 promotes lymphangiogenesis and lymphatic metastasis in esophageal squamous cell carcinoma. Gut. 64:26–36. 2015. View Article : Google Scholar

42 

Tamotsu K, Okumura H, Uchikado Y, Kita Y, Sasaki K, Omoto I, Owaki T, Arigami T, Uenosono Y, Nakajo A, et al: Correlation of Aurora-A expression with the effect of chemoradiation therapy on esophageal squamous cell carcinoma. BMC Cancer. 15:3232015. View Article : Google Scholar : PubMed/NCBI

43 

Zhang HF, Alshareef A, Wu C, Jiao JW, Sorensen PH, Lai R, Xu LY and Li EM: miR-200b induces cell cycle arrest and represses cell growth in esophageal squamous cell carcinoma. Carcinogenesis. 37:858–869. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Gabrielli F, Donadel G, Bensi G, Heguy A and Melli M: A nuclear protein, synthesized in growth-arrested human hepa-toblastoma cells, is a novel member of the short-chain alcohol dehydrogenase family. Eur J Biochem. 232:473–477. 1995. View Article : Google Scholar : PubMed/NCBI

45 

Gabrielli F and Tofanelli S: Molecular and functional evolution of human DHRS2 and DHRS4 duplicated genes. Gene. 511:461–469. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Deisenroth C, Thorner AR, Enomoto T, Perou CM and Zhang Y: Mitochondrial Hep27 is a c-Myb target gene that inhibits Mdm2 and stabilizes p53. Mol Cell Biol. 30:3981–3993. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Zhou Y, Wang L, Ban X, Zeng T, Zhu Y, Li M, Guan XY and Li Y: DHRS2 inhibits cell growth and motility in esophageal squamous cell carcinoma. Oncogene. 37:1086–1094. 2018. View Article : Google Scholar :

48 

Wessels D, Murray J, Jung G, Hammer JA III and Soll DR: Myosin IB null mutants of Dictyostelium exhibit abnormalities in motility. Cell Motil Cytoskeleton. 20:301–315. 1991. View Article : Google Scholar : PubMed/NCBI

49 

Ohmura G, Tsujikawa T, Yaguchi T, Kawamura N, Mikami S, Sugiyama J, Nakamura K, Kobayashi A, Iwata T, Nakano H, et al: Aberrant Myosin 1b Expression Promotes Cell Migration and Lymph Node Metastasis of HNSCC. Mol Cancer Res. 13:721–731. 2015. View Article : Google Scholar

50 

Elowe S: Bub1 and BubR1: At the interface between chromosome attachment and the spindle checkpoint. Mol Cell Biol. 31:3085–3093. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Athanasoula KC, Gogas H, Polonifi K, Vaiopoulos AG, Polyzos A and Mantzourani M: Survivin beyond physiology: Orchestration of multistep carcinogenesis and therapeutic potentials. Cancer Lett. 347:175–182. 2014. View Article : Google Scholar : PubMed/NCBI

52 

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

53 

Asghar U, Witkiewicz AK, Turner NC and Knudsen ES: The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov. 14:130–146. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Chen T, Sun Y, Ji P, Kopetz S and Zhang W: Topoisomerase IIα in chromosome instability and personalized cancer therapy. Oncogene. 34:4019–4031. 2015. View Article : Google Scholar

Related Articles

Journal Cover

February-2019
Volume 54 Issue 2

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
Shimonosono M, Idichi T, Seki N, Yamada Y, Arai T, Arigami T, Sasaki K, Omoto I, Uchikado Y, Kita Y, Kita Y, et al: Molecular pathogenesis of esophageal squamous cell carcinoma: Identification of the antitumor effects of miR‑145‑3p on gene regulation. Int J Oncol 54: 673-688, 2019
APA
Shimonosono, M., Idichi, T., Seki, N., Yamada, Y., Arai, T., Arigami, T. ... Natsugoe, S. (2019). Molecular pathogenesis of esophageal squamous cell carcinoma: Identification of the antitumor effects of miR‑145‑3p on gene regulation. International Journal of Oncology, 54, 673-688. https://doi.org/10.3892/ijo.2018.4657
MLA
Shimonosono, M., Idichi, T., Seki, N., Yamada, Y., Arai, T., Arigami, T., Sasaki, K., Omoto, I., Uchikado, Y., Kita, Y., Kurahara, H., Maemura, K., Natsugoe, S."Molecular pathogenesis of esophageal squamous cell carcinoma: Identification of the antitumor effects of miR‑145‑3p on gene regulation". International Journal of Oncology 54.2 (2019): 673-688.
Chicago
Shimonosono, M., Idichi, T., Seki, N., Yamada, Y., Arai, T., Arigami, T., Sasaki, K., Omoto, I., Uchikado, Y., Kita, Y., Kurahara, H., Maemura, K., Natsugoe, S."Molecular pathogenesis of esophageal squamous cell carcinoma: Identification of the antitumor effects of miR‑145‑3p on gene regulation". International Journal of Oncology 54, no. 2 (2019): 673-688. https://doi.org/10.3892/ijo.2018.4657