Open Access

MicroRNA‑200a promotes esophageal squamous cell carcinoma cell proliferation, migration and invasion through extensive target genes

  • Authors:
    • Bian Yang
    • Yumeng Liu
    • Lipeng Li
    • Hailong Deng
    • Lei Xian
  • View Affiliations

  • Published online on: February 26, 2020     https://doi.org/10.3892/mmr.2020.11002
  • Pages: 2073-2084
  • Copyright: © Yang 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

Despite investigations into microRNA (miRNA) expression in esophageal cancer (EC) tissue, miRNAs that participate in EC pathogenesis and their subsequent mechanisms of action remain to be determined. The present study aimed to identify important miRNAs that contribute to EC development, and to assess miRNA biomarkers that could be used in EC diagnosis, prognosis and therapy. Bioinformatics analysis was performed to reanalyze EC tissue miRNA expression microarray dataset GSE113776, which was followed by in vitro verification of miRNA functions using reverse transcription‑quantitative PCR, western blot analysis and a dual‑luciferase reporter assay. Out of 93 miRNAs extracted, only miR‑200a was significantly increased in EC tissues. Transfection of KYSE150 esophageal squamous cell carcinoma (ESCC) cells with miR‑200a mimics significantly increased their proliferative, migratory and invasive ability, whereas the opposite cell behaviors were observed in ESCC cells transfected with a miR‑200a inhibitor. A total of six miR‑200a target genes [catenin β1 (CTNNB1), cadherin‑1 (CDH1), PTEN, adenomatous polyposis coli (APC), catenin α1 (CTNNA1) and superoxide dismutase 2 (SOD2)] were selected for further analysis based on Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes pathway analysis, protein‑protein interaction network map data and protein expression in esophageal tissue. These target genes were downregulated under miR‑200a expression and upregulated in the presence of the miR‑200a inhibitor. The association between miR‑200a and the 3'‑untranslated region of target genes in ESCC cells was confirmed using a dual‑luciferase reporter assay. In conclusion, the present study demonstrated that miR‑200a may participate in the promotion of ESCC cell proliferation, migration and invasion, and provided novel evidence for the direct interaction between miR‑200a and CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2, which may contribute to the observed altered cell behavior.

Introduction

Esophageal cancer (EC) is one of the most common types of malignant tumor worldwide and it is associated with a high incidence (5.9 per 100,000) and patient mortality (5 per 100,000) (1). China exhibits one of the highest incidence rates of EC (12.5 per 100,000) and a high patient mortality (~9.1 per 100,000), with a total of ~50% new EC cases identified in 2012 (2,3). Esophageal squamous cell carcinoma (ESCC) is the major histological type of EC, accounting for > 90% of new diagnoses (4,5). However, clinical practices, including the diagnosis and treatment of EC remain challenging worldwide, thus there is an urgent requirement to identify novel biomarkers that can be used to improve the diagnosis and treatment of EC.

MicroRNAs (miRNAs) are widely reported to participate in the pathogenesis of tumor development, and are often used as biomarkers to monitor disease progression (4). miRNAs are small, single-stranded non-coding RNA molecules of ~22 nucleotides in length that can inhibit the expression of target mRNAs through binding to complementarity sites in the 3′-untranslated region (UTR) to induce RNA silencing and post-transcriptional regulation (5,6). Each miRNA can target multiple genes, thus functioning in complex regulatory networks involved in a variety of biological processes, including cell proliferation, migration and invasion, especially within tumors (79). In EC, it was reported that miRNA (miR)-100 significantly inhibited EC cell proliferation, migration and invasion through targeting the C-X-C chemokine receptor type 7 (10). miR-373 is highly expressed in EC tissues and was demonstrated to increase cell proliferation, migration and invasion through directly targeting metalloproteinase inhibitor 3, with opposite results occurring following miR-373 silencing (11). In a cohort of 102 patients, miR-451a, miR-144-3p and miR-144-5p expression levels in EC tissues were reported to be significantly lower compared with normal tissues (12). Furthermore, low expression levels of miR-144-3p and miR-144-5p is an independent risk factor for the occurrence of EC (12). Multiple other miRNAs, including miR-133a, miR-138, miR-375 and miR-593 serve as tumor suppressors, whereas miR-16, miR-21, miR-31, miR-34b, miR-208, miR-223, miR-373 and miR-423 exhibit oncogenic abilities (1318). It has been reported that miR-106b, miR-204, miR-371-3p, miR-574-3p, miR-886-3p, miR-1203, miR-1303 and miR-1909 were differentially expressed between patients with and without tumor relapse following surgery (13). miR-21 and miR-375 have previously been used as diagnostic and prognostic biomarkers of EC (14) and miR-100 has been suggested as a promising treatment owing to its tumor suppressor role in EC (10). These studies provide reasoning to identify additional miRNAs that may contribute to EC, alongside determining their mechanism of action in EC pathogenesis. The present study used bioinformatics analysis to reanalyze the miRNA expression microarray dataset of EC tissues (GSE113776), with the aim of identifying novel miRNAs to use as biomarkers for this disease. Since miRNAs demonstrated potential to be used for treatment and as biomarkers of EC according to the research cited above, the current study aimed to further investigate their functions in vitro through identifying key miRNAs that contribute to the development of EC, and to identify possible miRNA biomarkers for use in EC diagnosis, prognosis and therapy.

Materials and methods

Bioinformatics analysis

The miRNA profile dataset GSE113776 was downloaded from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo/). Considering the identical tissue variation and potential mutual effects or interactions in ESCC and neuroendocrine carcinoma (NEC), the GSE113776 dataset contained profiled miRNAs expressed in paired NEC and normal tissues based on an Agilent-041686 Unrestricted Human miRNA Microarray platform was used. Since only one sample was collected for sequencing in each group, no related research was cited in the dataset summary. R version 3.6 software (RStudio, Inc.) was used to analyze and visualize the dataset. To select miRNAs, the cut-off value of absolute fold change (|FC|) was set to 2. In addition, differentially expressed miRNAs (DEMs) that are associated with EC development were extracted from The Cancer Genome Atlas (TCGA; http://portal.gdc.cancer.gov) to intersect with the selected miRNAs. Verification of DEM expression was based on the miRNA profile dataset GSE112840 of ESCC (19).

miRNA target gene extraction and associated pathway enrichment

Using the miRNA databases TargetScanHuman (http://www.targetscan.org), miRTarBase (http://mirtarbase.mbc.nctu.edu.tw) and mirRDB (http://mirdb.org), intersections were filtered as the ready-for-test genes. Representative immunohistochemistry images of these genes in normal esophageal tissues were identified using the Human Protein Atlas (http://proteinatlas.org) to determine their protein expression. The Database for Annotation, Visualization and Integrated Discovery (DAVID; http://david.ncifcrf.gov) was used for pathway enrichment of the filtered genes and ClueGO and Search Tool for Recurring Instances of Neighbouring Genes (STRING; http://string-db.org) were used for visualization, which was performed using Cytoscape version 3.7.0 (https://cytoscape.org/) software.

Cell culture and transfection

The human ESCC cell line KYSE150 was purchased from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. KYSE150 cells were incubated in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.), supplemented with 10% FBS (HyClone; GE Healthcare Life Sciences) and 1% penicillin-streptomycin (Thermo Fisher Scientific, Inc.), and maintained in a humidified incubator at 37°C with 5% CO2. A total of 5×104 KYSE150 cells/well were seeded in a 6-well plate and were transfected with 50 nM miR-200a mimic (5′-UAACACUGUCUGGUAACGAUGU-3′), miR-200a inhibitor (5′-UAACCUCAUGGUGUACGAAUGU-3′) or scramble control sequences (5′-UUGUACUACACAAAAGUACUG-3′) (Shanghai GenePharma Co., Ltd.) separately for 24 h at 37°C using 10 nM Lipofectamine® 3000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturers' protocol. Cell proliferation and Transwell assays were subsequently performed and transfected cells were harvested at 48 h for reverse transcription-quantitative PCR (RT-qPCR) or western blot analysis.

Cell proliferation assay

Following transfection, a total of 1×104 KYSE150 cells/well were seeded into 96-well plates to assess cell viability. Cells were cultured for 2 days post-transfection in RPMI-1640 medium prior to the addition of 10 µl Cell Counting kit-8 (CCK-8) solution (WST-8, Dojindo Molecular Technologies, Inc.) to each well. Following continuous incubation for 2 h at 37°C, cell viability was determined by measuring the absorbance at 450 nm using an ELISA reader (Tecan Group, Ltd.) at 0, 12, 24 and 48 h.

Migration and invasion assays

Migratory and invasive abilities of transfected KYSE150 cells were examined using Transwell permeable supports (Corning, Inc.). Following cell transfection for 24 h at 37°C, cells were subsequently cultured in 200 µl serum-free RPMI-1640 medium before being transferred into 24-well plates separated into upper and lower chambers. A total of 1×105 KYSE150 cells/well were plated in the upper chambers of Transwell plates with serum-free RPMI-1640 medium, of which membranes were or were not precoated with Matrigel (BD Biosciences) for the invasion and migration assay, respectively. A total of 800 µl RPMI-1640 medium supplemented with 10% FBS was plated in the lower chambers. After 24 h incubation in wells without Matrigel or 48 h with Matrigel for the migration and invasion assay, respectively, cells in the lower chambers were fixed for 20 min with absolute methanol and subsequently stained with 0.1% crystal violet solution (Sigma-Aldrich; Merck KGaA) for 10 min at room temperature. Stained cells were counted in three randomly selected fields using an inverted microscope (magnification, ×200). ImageJ version 1.49 software (National Institutes of Health) was used for image analysis and quantification. Experiments were performed in triplicate.

RT-qPCR

Total RNA was extracted from KYSE150 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Total RNA was reverse transcribed into cDNA using PrimeScript RT Reagent kit purchased from Takara Biotechnology Co., Ltd., and the concentration of RNA was determined as described in a previous study (20). qPCR was subsequently performed in 96-well reaction plates using the ABI StepOnePlus Real-Time PCR system (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Briefly, in each well, a total of 1 µl cDNA template, 0.2 µl primer (0.1 µl forward and 0.1 µl reverse primer), 3.6 µl diethyl pyrocarbonate-H2O and 5 µl SYBR® Green dye were mixed. The primers (Sangon Biotech Co., Ltd.) used for the qPCR are presented in Table SI. The following thermocycling conditions were used for the qPCR: Initial denaturation at 95°C for 30 sec; followed by 40 cycles of denaturation at 95°C for 5 sec, annealing at 60°C for 10 sec and extension at 72°C for 30 sec. Expression levels were quantified using the 2−ΔΔCq method and normalized to the internal reference gene, U6 for miR-200a expression or GAPDH for miR-200a target genes (21).

Western blotting

Total protein was extracted from KYSE150 cells using RIPA lysis buffer (Beyotime Institute of Biotechnology) and protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA). Total protein was quantified using a bicinchoninic acid assay kit (Thermo Fisher Scientific, Inc.) and western blot analysis was performed according to previous protocols (22). A total of 30 µg protein/lane was separated by 6 or 12% SDS-PAGE. The separated proteins were subsequently transferred onto a PVDF membrane (Merck KGaA) and blocked for 1 h at room temperature using 5% non-fat milk. The membranes were incubated overnight at 4°C with the following primary antibodies diluted in 5% milk: Anti-catenin β1 (CTNNB1; 1:1,000; cat. no. sc-59737; Santa Cruz Biotechnology, Inc.), anti-cadherin-1 (CDH1; 1:500; cat. no. sc-71009; Santa Cruz Biotechnology, Inc.), anti-PTEN (1:100; cat. no. sc-73420; Santa Cruz Biotechnology, Inc.), anti-adenomatous polyposis coli (APC; 1:100; cat. no. sc-393704; Santa Cruz Biotechnology, Inc.), anti-catenin α1 (CTNNA1; 1:500; cat. no. sc-47753; Santa Cruz Biotechnology, Inc.), anti-superoxide dismutase 2 (SOD2; 1:200; cat. no. sc-130345; Santa Cruz Biotechnology, Inc.) and anti-GAPDH (1:3,000; cat. no. sc-47724; Santa Cruz Biotechnology, Inc.). Following the primary antibody incubation, membranes were incubated with horseradish peroxidase-conjugated goat anti-mouse secondary antibodies (1:5,000; cat. no. A0216; Beyotime Institute of Biotechnology) for 45 min at room temperature. Protein bands were visualized using the ECL luminol reagent (PerkinElmer Inc.) and a ChemiDoc image analyzer (Bio-Rad Laboratories, Inc.). Protein expression was quantified using ImageJ software (version 1.49v; National Institutes of Health) and normalized to the internal reference gene GAPDH.

Dual-luciferase reporter assay

The luciferase reporter assay was performed as previously described (22). The 3′-UTR sequence of miR-200a target genes (CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2) was separately amplified and inserted into the luciferase reporter vector pGL3-enhancer (Promega Corporation). The primers of the 3′-UTR of miR-200a target genes were designed and are presented in Table SII. A total of 1×104 KYSE150 cells/well were seeded into 24-well plates and incubated for 24 h at 37°C. Wild-type or mutant miR-200a target gene 3′-UTR vectors, combined with the miR-200a mimic, were subsequently co-transfected into KYSE150 cells using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. The construction of the miR-200a target genes' 3′-UTR wild-type or mutant reporter genes were performed using the miRNA databases (Fig. S1). Following incubation for 48 h at 37°C, KYSE150 cells were lysed and firefly and Renilla luciferase activity was detected using a Luciferase assay system (Promega Corporation), according to the manufacturer's protocol. Firefly luciferase activity was normalized to Renilla luciferase activity.

Statistical analysis

Data were analyzed using GraphPad Prism version 6.0 software (GraphPad Software, Inc.). Statistical differences between two groups were compared using a Student's t-test, whereas three groups were compared using a one-way ANOVA with Bonferroni correction post hoc test. Data were presented as the mean ± SEM from 3 independent experiments. P<0.05 was considered to indicate a statistically significant difference.

Results

Heat map of miRNA expression and identification of DEMs in a paired NEC sample

ESCC and neuroendocrine carcinoma (NEC) are from the identical sites in esophageal tissues, indicating a potentially similar mechanism. A heat map was constructed to identify the expression of miRNAs in esophageal tissues using a Heat map package in R. For the miRNA y dataset GSE113776, 494 miRNAs were detected (Fig. 1A). A total of 28 miRNAs were selected, of which the absolute expression value threshold was |FC|>2 (Fig. 1B). In addition, 93 DEMs that were associated with the development of EC were identified from TCGA (Table I). One miRNA was observed to overlap between the selected miRNAs and DEMs from TCGA, which was miR-200a (Fig. 1C). In addition, under the cross-verification with the GSE112840 dataset, miR-200a was confirmed to be upregulated in ESCC compared with the control (Fig. 1D), which suggests the critical role of miR-200a in ESCC.

Table I.

miRNAs identified from the tissue microarray and TCGA.

Table I.

miRNAs identified from the tissue microarray and TCGA.

OriginCountElements
miRNAs profiling28hsa-miR-136 hsa-miR-1469 hsa-miR-16 hsa-miR-181c hsa-miR-181d hsa-miR-18a hsa-miR-200a hsa-miR-23b hsa-miR-2861 hsa-miR-29b hsa-miR-362 hsa-miR-3663 hsa-miR-374c hsa-miR-377 hsa-miR-424 hsa-miR-4486 hsa-miR-4522 hsa-miR-4732 hsa-miR-500a hsa-miR-590 hsa-miR-601 hsa-miR-605 hsa-miR-625 hsa-miR-7-1 hsa-miR-718 hsa-miR-744 hsa-miR-887 hsa-miR-99b
TCGA93microRNA 663b microRNA 770 microRNA 520b microRNA 485 microRNA 1270 AC116165.1 microRNA 1302-3 microRNA 498 AC005631.1 microRNA 3156-3 AC100757.1 microRNA 516a-1 microRNA 920 AC233702.1 microRNA 519d microRNA 5787 microRNA 515-2 microRNA 6511a-3 microRNA 181b-1 microRNA 4697 AL391261.1 microRNA 6511a-1 AC005071.4 microRNA 1208 AC106782.1 AF254983.1 AJ271736.1 AC010203.1 microRNA 600 microRNA 6511a-2 AL354833.1 microRNA 4300 microRNA 1468 AL078621.1 microRNA 513c microRNA 3142 AC011453.4 microRNA 412 microRNA let-7f-1 microRNA 221 AC245033.1 microRNA 6859-3 AL354820.1 microRNA 300 microRNA 519b microRNA 182 AC106788.1 microRNA 514a-2 microRNA 622 microRNA 507 AC126544.1 microRNA 205 microRNA 15a Z83819.1 microRNA 409 microRNA 6859-4 AC090825.1 microRNA 2682 microRNA 410 microRNA 154 microRNA 411 microRNA 548× microRNA 519a-1 microRNA 6859-1 microRNA 548i-4 microRNA 1197 microRNA 495 AL161651.1 microRNA 892a microRNA 3680-2 AC133555.1 AC092375.1 microRNA 509-2 AC105339.2 AC023310.1 AC024937.1 microRNA 570 microRNA 517b AC092017 microRNA 6080 microRNA 509-3 microRNA 3147 AC215219.2 microRNA 466 microRNA 129-2 microRNA 548i-1 AC011467.1 microRNA 1-1 microRNA 520d AC011453.3 microRNA 518f microRNA 758
Effects of miR-200a on the proliferation, migration and invasion of ESCC cells

KYSE150 ESCC cells were transiently transfected with miR-200a mimics or inhibitors and the effects of miR-200a on ESCC cell proliferation were assessed using a CCK-8 assay. RT-qPCR was used to confirm the transfection efficiency of the mimic and inhibitor in KYSE150 ESCC cells; the miR-200a mimic significantly increased miR-200a expression levels compared with the scramble control, whereas the miR-200a inhibitor significantly decreased miR-200a expression levels compared with the scramble controls in ESCC cells (Fig. 2A). The viability of ESCC cells was significantly increased in miR-200a mimic-transfected cells compared with the scramble control at 24 and 48 h, whereas the inhibition of miR-200a expression significantly decreased ESCC cell viability compared with the scramble control at all of the time points (Fig. 2B). To evaluate the effects of miR-200a on cell migration and invasion, a Transwell assay was performed in ESCC cells. miR-200a mimics transfection significantly increased the migratory and invasive ability of ESCC cells compared with the scramble control, whereas the miR-200a inhibitor-transfected cells exhibited significantly reduced migratory and invasive capacity compared with the scramble control (Fig. 2C).

Identification of target genes and pathway enrichment analysis

As miR-200a was the only miRNA selected from the two groups, the current study aimed to identify target genes that miR-200a may regulate in ESCC. The miRNA databases TargetScanHuman, miRTarBase and mirRDB were searched and the intersections were filtered using 54 genes. DAVID was used for pathway enrichment and ClueGO and STRING were used for visualization. The results of the GO analysis indicated that the main biological processes affected by miR-200a were embryonic heart tube development and transcriptional regulation, and the main cellular components regulated were transcription factor complexes and cell-cell adherens junctions (Table II). The most significant molecular functions associated with miR-200a were DNA binding, transcription factor binding and cell-cell adhesion. The results of the KEGG pathway analysis reported that basal cell carcinoma pathways in cancer and carbon metabolism were the most significantly enriched pathways (P<0.05), whereas the Wnt and the Hippo signaling pathways were classed as critical pathways despite not being significant (P=0.05 and P=0.06, respectively; Table II). Subsequently, miR-200a target genes were assigned to ClueGO to assess the association between miR-200a target genes and GO biological process, molecular function and cellular component terms, which yielded the terms ‘regulation of transcription’, ‘positive regulation of core promoter region’ and others (Fig. 3A) and GO terms distribution associated with the number of genes and percentage (Fig. 3B). According to the relationships, the protein-protein interaction map of target genes was created using STRING, and the network visualization was performed using Cytoscape (Fig. 3C). In the network, CTNNB1, CDH1, TP53, PTEN, CDK1, APC, CTNNA1, FYN and SOD2 were the most prominent, as these genes exhibited multiple associations with other genes.

Table II.

GO analysis and KEGG pathway enrichment analyses of miR-200a target genes.

Table II.

GO analysis and KEGG pathway enrichment analyses of miR-200a target genes.

CategoryTermCountP-value
GO term-biologicalEmbryonic heart tube development3<0.001
processesNegative regulation of transcription from RNA polymerase II promoter90.001
Positive regulation of transcription from RNA polymerase II promoter100.002
Transcription from RNA polymerase II promoter70.004
Response to zinc ion30.005
Regulation of transcription, DNA-templated110.01
Response to drug50.01
Cell proliferation50.02
Prostate gland growth20.02
Canonical Wnt signaling pathway30.02
Embryonic foregut morphogenesis20.02
Positive regulation of skeletal muscle tissue development20.03
Negative regulation of cell proliferation50.03
Positive regulation of sequence-specific DNA binding transcription factor activity30.04
Epithelial tube branching involved in lung morphogenesis20.04
GO term-cellular componentsNucleus29<0.001
Transcription factor complex50.001
Nuclear transcription factor complex20.03
Cell-cell adherens junction40.05
Mitochondrial matrix40.05
GO term-molecular functionRNA polymerase II core promoter proximal region sequence-specific DNA binding10<0.0001
Nucleic acid binding120.0001
Transcription factor activity, sequence-specific DNA binding100.001
Transcription regulatory region DNA binding50.003
Transcriptional activator activity, RNA polymerase II core promoter proximal region sequence-specific binding50.005
Metal ion binding130.01
Transcription coactivator activity40.04
Transcription factor binding40.05
DNA binding100.05
Cadherin binding involved in cell-cell adhesion40.05
KEGG_PathwaySignaling pathways regulating pluripotency of stem cells40.006
Basal cell carcinoma30.009
Biosynthesis of amino acids30.01
Pathways in cancer50.02
Carbon metabolism30.04
Wnt signaling pathway30.05a
Hippo signaling pathway30.06a

a According to the literature review, these pathways are critical even though P>0.05. GO, gene ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Interactions of miR-200a and its target genes

As previously described, CTNNB1, CDH1, TP53, PTEN, CDK1, APC, CTNNA1, FYN and SOD2 were identified as the most significant genes targeted by miR-200a in the network. Representative immunohistochemistry images of these genes were obtained from the Human Protein Atlas and were used to determine the protein expression of each gene in normal esophageal tissues. These proteins were highly expressed in normal tissues according to the Human Protein Atlas (Fig. S2). Accordingly, the highly expressed genes (CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2) were selected for subsequent analysis. To detect the effects of miR-200a on the target genes, CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2 mRNA and protein expression levels were examined in KYSE150 ESCC cells transfected with a miR-200a mimic or inhibitor. RT-qPCR and western blot analysis demonstrated that target genes and their protein levels were significantly decreased in cells transfected with miR-200a mimic compared with the scramble-transfected cells, whereas expression levels were significantly increased in cells transfected with the miR-200a inhibitor compared with the control (Fig. 4A). A dual-luciferase reporter assay was performed to evaluate the direct interaction between miR-200a and the 3′-UTR of the target genes. The results indicated that KYSE150 ESCC cells transfected with the miR-200a mimic significantly suppressed the luciferase activity of the wild-type reporters containing the 3′-UTR of all target genes (CTNNB1, CDH1, APC, PTEN, CTNNA1 and SOD2) compared with the scramble control; this inhibition disappeared when the miR-200a target site was mutated (Fig. 4B).

Discussion

ESCC is the main histological type of EC, accounting for >90% of new cases (23); the majority of new ESCC cases occur in Asia, especially in China (2). Clinical treatment for ESCC remains challenging, and novel diagnosis and treatment strategies are urgently required. miRNAs have been widely reported to be associated with tumor development, and are often regarded as disease biomarkers; it has been demonstrated that miRNAs affect ESCC cell behaviors, including cell proliferation, migration and invasion (79). However, miRNAs function in a complicated regulatory network, which involves a number of different biological processes (24). The present study analyzed the miRNA expression microarray of NEC (with its incidence being rare but of similar effects with ESCC) tissues from dataset GSE113776 of the GEO database. A total of 28 miRNAs were selected according to the inclusion criteria. miR-200a was selected following the identification of an intersection between the 28 selected miRNAs and 93 DEMs using the TCGA. Subsequent in vitro experiments demonstrated that miR-200a may be involved in promoting ESCC cell proliferation, migration and invasion. Furthermore, the results provided evidences that miR-200a interacts with its target genes, CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2, which may contribute to the abnormal cell behaviors in EC.

A number of previous studies have performed high-throughput sequencing, while a total of 140 DEMs have been extracted, with 113 upregulated and 27 downregulated miRNAs being identified in ESCC tissue (25). In a previous study, only five miRNAs (miR-103-1, miR-18a, miR-324, miR-369 and miR-320b-2), which have previously been associated with survival rates (25), were studied. In another study, out of a total of 136 DEMs identified in EC, the top five DEMs were revealed to be miRNA-21, miRNA-93, miRNA-196a-1, miRNA-196a-2 and miRNA-4746 (26). In addition, within a cohort of 102 patients with EC, miR-451a, miR-144-3p and miR-144-5p were identified; other miRNAs including miR-133a miR-138, miR-375 and miR-593 were observed to serve as tumor suppressors, whereas miR-16, miR-21, miR-31, miR-34b, miR-208, miR-223, miR-373 and miR-423 exhibited oncogenic properties (1318). Meanwhile, researchers identified miR-28-5p, miR-34a-5p and miR-186-5p as the significant biomarkers of ESCC (27). Furthermore, previous studies explored the underlying miRNAs that can play key roles in ESCC development. For instance, miRNA-10b can promote ESCC cell proliferation, migration, invasion and colony formation, as well as metastasis; miRNA-548 and miRNA-576 enhance the migration and invasion of ESCC cells; miRNA-1 can suppress the proliferation, migration and invasion of ESCC cells (2830). Based on these studies, a number of miRNAs have been reported to serve a role in EC; however, to the best of our knowledge, no previous studies have predicted the potential role of miR-200a in EC pathogenesis using bioinformatics. In the present study, a total of 28 miRNAs were identified in the NEC microarray dataset GSE113776, including miR-200a. Additionally, a number of important miRNAs, including miR-181c-5p, hsa-miR-500a-3p, hsa-miR-601 and hsa-miR-605 were matched with identified DEMs in previous research (25,26).

To the best of our knowledge, no previous bioinformatics study has identified miR-200a as a crucial miRNA, and this may be due to the fact that a single sample of NEC tissue was used. The GSE113776 dataset may not have been investigated owing to the difficulty of using statistical analysis to assess it. However, this disadvantage permitted the identification of novel miRNAs because the high expression of miRNAs in the tissue may have excluded the significance of the less expressed, but critical miRNAs. In addition, previous studies may not have focused on the most important miRNAs, but instead on determining DEMs. Owing to the single sample comparison in the present study, miR-200a was identified and its was demonstrated to be upregulated in ESCC according to the miRNA profile GSE112840 (19). The importance of miR-200a was confirmed by matching with TGCA records and RT-qPCR verification. It is hypothesized that different types of cells exhibit mutual effects or interactions with each other (3134); NEC, squamous cell carcinoma and sarcoma exhibit metastatic and site-transfer-direction (32,33), and definitive chemoradiotherapy promoted the conversion of NEC to squamous cell carcinoma (34). In addition, NEC, squamous cell carcinoma and adenocarcinoma can coexist (31,35,36). Although miR-220a was singled out from NEC tissues, the present study demonstrated the effects of miR-200a in ESCC cells; however, the direct association between ESCC and NEC and adenocarcinoma was not assessed due to the lack of tissue.

The alterations of ESCC cell behaviors following miR-200a overexpression are not well determined. The role of miRNAs depends on the type of miRNAs. It is reported that miR-100 inhibits EC cell proliferation, migration and invasion, (10), while miR-373 enhances cell proliferation, migration and invasion (11). The present study demonstrated that the upregulation of miR-200a could promote ESCC cell proliferation, migration and invasion. This was a similar role to the effects of miR-373 in decreasing the proliferation, migration and invasion of ESCC cells. In addition to the effects of miR-200a in ESCC, miR-200a has been studied in a number of tumor types, including breast, ovarian, gastric, colorectal and pancreatic cancers (3741); miR-200a is also associated with cell proliferation, migration and invasion in these tumor types (3741).

The present study investigated the putative target genes of miR-200a, as miRNAs function in the development of ESCC via binding to the UTR region of target genes: miRNA-10b targeting FOXO3, micRNA-548 and miRNA-576 downregulating NRIP1 and miRNA-1 binding to Notch2 (2830). Bioinformatics analysis revealed that the main biological processes that miR-200a was involved in included embryonic development and transcription regulation, the main cellular components were transcription factor complexes and cell-cell adherence junctions and the main molecular functions were related to DNA binding, transcription factor binding and cell-cell adhesion, which showed the similar results with the findings of previous studies enriched from other miRNAs (25,26,42). The results of KEGG pathway enrichment, except for the suggestions of basic pathological pathways, also indicated that miR-200a may serve a role in the Wnt signaling pathway and the Hippo signaling pathway, which was consistent with previous findings (4347). According to target gene prediction and their expression in normal tissues from the Human Protein Atlas, miR-200a was identified to target CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2. CTNNB1 is a crucial downstream component of the Wnt signaling pathway, forming a complex to promote phosphorylation on N-terminal serine and threonine residues in the absence of Wnt, while acting as a coactivator for Wnt responsive genes in the presence of the Wnt ligand (48,49). Cadherins are calcium-dependent cell adhesion proteins that interact in a homophilic manner during cell communications and CDH1 is associated with cell-cell adhesions regulation, mobility and proliferation (50). The major function of CTNNB1 is to regulate cell adhesion by participating in the E-cadherin/catenin adhesion complex (51). CTNNA1, which is an isotype of the catenin protein, is associated with a number of cadherins, including E- and N-cadherins, and stabilizes E-cadherin/catenin adhesion complexes (52). APC, which is correlated with its phosphorylation state, participates in the Wnt signaling pathway as a negative regulator and promotes the rapid degradation of CTNNB1 (53,54). PTEN is a dual-specificity protein phosphatase, which mediates the phosphorylation of N-terminal tyrosine, serine and threonine residues (55). The dephosphorylation of tyrosine-phosphorylated focal adhesion kinase inhibits focal adhesion formation, cell migration and integrin-mediated cell spreading (56). SOD2 has been reported to delay tumor cell growth in a number of tumor types (57,58); its activity affects different stages of the cell cycle and its downregulation may stimulate cell cycle progression (5961). In addition, it has been demonstrated through immunohistochemical staining that the protein expression levels of CTNNB1, CDH1, TP53 and PTEN are decreased in ESCC tissues compared with normal tissues (6264), while the results of the present study showed the high expression of CTNNB1, CDH1, TP53 and PTEN in normal tissue with IHC staining (Fig. S2). Previous studies reported that miR-200a regulated CTNNB1, CDH1 and PTEN, but not APC, CTNNA1 and SOD2 in tumors. miR-200a was demonstrated to downregulate CTNNB1 and inhibit nasopharyngeal carcinoma cell growth, migration and invasion (65). miR-200a suppressed CDH1 and resulted in the induction of EMT, which has critical functions in tumor cell migration and invasion (66). miR-200a inhibited the expression of PTEN and promoted the invasion and migration of ovarian cancer cells (67). Thus, determining the regulation of miR-200a on APC, CTNNA1 and SOD2 during EC oncogenesis may be useful in the future. Since these target genes have been reported to be associated with the development of a number of types of cancer by influencing the cell cycle and cell adhesion, it is suggested that miR-200a may alter the cell behaviors of ESCC due to the role that the downstream genes have in cell proliferation, migration and invasion pathways.

In conclusion, the present study demonstrated that miR-200a participated in promoting ESCC cell proliferation, migration and invasion, and provided novel evidence for the direct interaction between miR-200a and CTNNB1, CDH1, PTEN, APC, CTNNA1 and SOD2, which may be responsible for this observed cell behavior.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

BY performed the experiments and wrote the manuscript. YL and LL performed the bioinformatics analysis. HD contributed to the interpretation of the data and helped write and revise the manuscript. LX conceived and designed the experiments. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Karamanou M, Markatos K, Papaioannou TG, Zografos G and Androutsos G: Hallmarks in history of esophageal carcinoma. J BUON. 22:1088–1091. 2017.PubMed/NCBI

2 

Klingelhöfer D, Zhu Y, Braun M, Brüggmann D, Schöffel N and Groneberg DA: A world map of esophagus cancer research: A critical accounting. J Transl Med. 17:1502019. View Article : Google Scholar : PubMed/NCBI

3 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Liu F, Wu K, Wu W, Chen Y, Wu H, Wang H and Zhang W: miR203 contributes to preeclampsia via inhibition of VEGFA expression. Mol Med Rep. 17:5627–5634. 2018.PubMed/NCBI

5 

Liu F, Wu W, Wu K, Chen Y, Wu H, Wang H and Zhang W: MiR-203 participates in human placental angiogenesis by inhibiting VEGFA and VEGFR2 Expression. Reprod Sci. 25:358–365. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Jopling CL, Schütz S and Sarnow P: Position-dependent function for a tandem microRNA miR-122-binding site located in the hepatitis C virus RNA genome. Cell Host Microbe. 4:77–85. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Shukla GC, Singh J and Barik S: MicroRNAs: Processing, maturation, target recognition and regulatory functions. Mol Cell Pharmacol. 3:83–92. 2011.PubMed/NCBI

8 

Tutar L, Özgür A and Tutar Y: Involvement of miRNAs and pseudogenes in cancer. Methods Mol Biol. 1699:45–66. 2018. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Zhou SM, Zhang F, Chen XB, Jun CM, Jing X, Wei DX, Xia Y, Zhou YB, Xiao XQ, Jia RQ, et al: miR-100 suppresses the proliferation and tumor growth of esophageal squamous cancer cells via targeting CXCR7. Oncol Rep. 35:3453–3459. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Xu DD, Zhou PJ, Wang Y, Zhang L, Fu WY, Ruan BB, Xu HP, Hu CZ, Tian L, Qin JH, et al: Reciprocal activation between STAT3 and miR-181b regulates the proliferation of esophageal cancer stem-like cells via the CYLD pathway. Mol Cancer. 15:402016. View Article : Google Scholar : PubMed/NCBI

12 

Gao Z, Liu R, Liao J, Yang M, Pan E, Yin L and Pu Y: Possible tumor suppressive role of the miR-144/451 cluster in esophageal carcinoma as determined by principal component regression analysis. Mol Med Rep. 14:3805–3813. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Okumura T, Kojima H, Miwa T, Sekine S, Hashimoto I, Hojo S, Nagata T and Shimada Y: The expression of microRNA 574-3p as a predictor of postoperative outcome in patients with esophageal squamous cell carcinoma. World J Surg Oncol. 14:2282016. View Article : Google Scholar : PubMed/NCBI

14 

Lv H, He Z, Wang H, Du T and Pang Z: Differential expression of miR-21 and miR-75 in esophageal carcinoma patients and its clinical implication. Am J Transl Res. 8:3288–3298. 2016.PubMed/NCBI

15 

Sun J, Song K, Feng X and Gao S: MicroRNA-367 is a potential diagnostic biomarker for patients with esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 473:363–369. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Sun L, Dong S, Dong C, Sun K, Meng W, Lv P, Yin H, Ming L and He F: Predictive value of plasma miRNA-718 for esophageal squamous cell carcinoma. Cancer Biomark. 16:265–273. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Matsuzaki J and Suzuki H: Role of MicroRNAs-221/222 in digestive systems. J Clin Med. 4:1566–1577. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Phatak P, Byrnes KA, Mansour D, Liu L, Cao S, Li R, Rao JN, Turner DJ, Wang JY and Donahue JM: Overexpression of miR-214-3p in esophageal squamous cancer cells enhances sensitivity to cisplatin by targeting survivin directly and indirectly through CUG-BP1. Oncogene. 35:2087–2097. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Zheng D, Ding Y, Ma Q, Zhao L, Guo X, Shen Y, He Y, Wei W and Liu F: Identification of serum MicroRNAs as novel biomarkers in esophageal squamous cell carcinoma using feature selection algorithms. Front Oncol. 8:6742018. View Article : Google Scholar : PubMed/NCBI

20 

Di Stefano V, Wang B, Parobchak N, Roche N and Rosen T: RelB/p52-mediated NF-κB signaling alters histone acetylation to increase the abundance of corticotropin-releasing hormone in human placenta. Sci Signal. 8:ra852015. View Article : Google Scholar : PubMed/NCBI

21 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Zhu X, Er K, Mao C, Yan Q, Xu H, Zhang Y, Zhu J, Cui F, Zhao W and Shi H: miR-203 suppresses tumor growth and angiogenesis by targeting VEGFA in cervical cancer. Cell Physiol Biochem. 32:64–73. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Lepage C, Rachet B, Jooste V, Faivre J and Coleman MP: Continuing rapid increase in esophageal adenocarcinoma in England and Wales. Am J Gastroenterol. 103:2694–2699. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Chekouo T, Stingo FC, Doecke JD and Do KA: miRNA-target gene regulatory networks: A Bayesian integrative approach to biomarker selection with application to kidney cancer. Biometrics. 71:428–438. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Zhao JY, Wang F, Li Y, Zhang XB, Yang L, Wang W, Xu H, Liu DZ and Zhang LY: Five miRNAs considered as molecular targets for predicting esophageal cancer. Med Sci Monit. 21:3222–3230. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Zeng JH, Xiong DD, Pang YY, Zhang Y, Tang RX, Luo DZ and Chen G: Identification of molecular targets for esophageal carcinoma diagnosis using miRNA-seq and RNA-seq data from The Cancer Genome Atlas: A study of 187 cases. Oncotarget. 8:35681–35699. 2017.PubMed/NCBI

27 

Chen L, Jin Y, Wang L, Sun F, Yang X, Shi M, Zhan C, Shi Y and Wang Q: Identification of reference genes and miRNAs for qRT-PCR in human esophageal squamous cell carcinoma. Med Oncol. 34:22017. View Article : Google Scholar : PubMed/NCBI

28 

Lu YF, Yu JR, Yang Z, Zhu GX, Gao P, Wang H, Chen SY, Zhang J, Liu MY, Niu Y, et al: Correction to: Promoter hypomethylation mediated upregulation of MicroRNA-10b-3p targets FOXO3 to promote the progression of esophageal squamous cell carcinoma (ESCC). J Exp Clin Cancer Res. 39:192020. View Article : Google Scholar : PubMed/NCBI

29 

Ni XF, Zhao LH, Li G, Hou M, Su M, Zou CL and Deng X: MicroRNA-548-3p and MicroRNA-576-5p enhance the migration and invasion of esophageal squamous cell carcinoma cells via NRIP1 down-regulation. Neoplasma. 65:881–887. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Liu W, Li M, Chen X, Zhu S, Shi H, Zhang D, Cheng C and Li B: MicroRNA-1 suppresses proliferation, migration and invasion by targeting Notch2 in esophageal squamous cell carcinoma. Sci Rep. 8:51832018. View Article : Google Scholar : PubMed/NCBI

31 

Kaneko Y, Saito S, Takahashi K, Kanamaru R, Hosoya Y, Yamaguchi H, Kitayama J, Niki T, Lefor AK and Sata N: Neuroendocrine carcinoma of the esophagus with an adenocarcinoma component. Clin J Gastroenterol. 12:534–538. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Tsuchihashi K, Arita S, Fujiwara M, Iwasaki K, Hirano A, Yoshihiro T, Nio K, Koga Y, Esaki M, Ariyama H, et al: Metastatic esophageal carcinosarcoma comprising neuroendocrine carcinoma, squamous cell carcinoma, and sarcoma: A case report. Medicine (Baltimore). 97:e127962018. View Article : Google Scholar : PubMed/NCBI

33 

Fujihara S, Kobayashi M, Nishi M, Yachida T, Yoshitake A, Deguchi A, Muraoka A, Kobara H and Masaki T: Composite neuroendocrine carcinoma and squamous cell carcinoma with regional lymph node metastasis: A case report. J Med Case Rep. 12:2272018. View Article : Google Scholar : PubMed/NCBI

34 

Morita M, Saeki H, Nakaji YU, Zaitsu Y, Hirahashi M, Ohguri T, Oki E, Toh Y, Oda Y and Maehara Y: Conversion to neuroendocrine carcinoma from squamous cell carcinoma of the esophagus after definitive chemoradiotherapy. Anticancer Res. 36:4045–4049. 2016.PubMed/NCBI

35 

Yazıcı O, Aksoy S, Özhamam EU and Zengin N: Squamous cell and neuroendocrine carcinoma of esophagus: Collision versus composite tumor: A case report and review of literature. Indian J Cancer. 52:603–604. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Yang L, Sun X, Zou Y and Meng X: Small cell type neuroendocrine carcinoma colliding with squamous cell carcinoma at esophagus. Int J Clin Exp Pathol. 7:1792–1795. 2014.PubMed/NCBI

37 

Yu SJ, Yang L, Hong Q, Kuang XY, Di GH and Shao ZM: MicroRNA-200a confers chemoresistance by antagonizing TP53INP1 and YAP1 in human breast cancer. BMC Cancer. 18:742018. View Article : Google Scholar : PubMed/NCBI

38 

Sun Q, Zou X, Zhang T, Shen J, Yin Y and Xiang J: The role of miR-200a in vasculogenic mimicry and its clinical significance in ovarian cancer. Gynecol Oncol. 132:730–738. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Liu X, Du P, Han L, Zhang A, Jiang K and Zhang Q: Effects of miR-200a and FH535 combined with taxol on proliferation and invasion of gastric cancer. Pathol Res Pract. 214:442–449. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Yang W, Ning N and Jin X: The lncRNA H19 promotes cell proliferation by competitively binding to miR-200a and Derepressing β-catenin expression in colorectal cancer. Biomed Res Int. 2017:27674842017.PubMed/NCBI

41 

Hu B, Qiu-Lan H, Lei RE, Shi C, Jiang HX and Qin SY: Interleukin-9 promotes pancreatic cancer cells proliferation and migration via the miR-200a/Beta-catenin axis. Biomed Res Int. 2017:28310562017. View Article : Google Scholar : PubMed/NCBI

42 

Chen F, Zhou H, Wu C and Yan H: Identification of miRNA profiling in prediction of tumor recurrence and progress and bioinformatics analysis for patients with primary esophageal cancer: Study based on TCGA database. Pathol Res Pract. 214:2081–2086. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Wang L, Zhang Z, Yu X, Huang X, Liu Z, Chai Y, Yang L, Wang Q, Li M, Zhao J, et al: Unbalanced YAP-SOX9 circuit drives stemness and malignant progression in esophageal squamous cell carcinoma. Oncogene. 38:2042–2055. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Qi B, Wang Y, Chen ZJ, Li XN, Qi Y, Yang Y, Cui GH, Guo HZ, Li WH and Zhao S: Down-regulation of miR-30a-3p/5p promotes esophageal squamous cell carcinoma cell proliferation by activating the Wnt signaling pathway. World J Gastroenterol. 23:7965–7977. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Gao Y, Yi J, Zhang K, Bai F, Feng B, Wang R, Chu X, Chen L and Song H: Downregulation of MiR-31 stimulates expression of LATS2 via the hippo pathway and promotes epithelial-mesenchymal transition in esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 36:1612017. View Article : Google Scholar : PubMed/NCBI

46 

Zang B, Huang G, Wang X and Zheng S: HPV-16 E6 promotes cell growth of esophageal cancer via downregulation of miR-125b and activation of Wnt/β-catenin signaling pathway. Int J Clin Exp Pathol. 8:13687–13694. 2015.PubMed/NCBI

47 

Ge C, Wu S, Wang W, Liu Z, Zhang J, Wang Z, Li R, Zhang Z, Li Z, Dong S, et al: miR-942 promotes cancer stem cell-like traits in esophageal squamous cell carcinoma through activation of Wnt/β-catenin signalling pathway. Oncotarget. 6:10964–10977. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Weiske J, Albring KF and Huber O: The tumor suppressor Fhit acts as a repressor of beta-catenin transcriptional activity. Proc Natl Acad Sci USA. 104:20344–20349. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Lillehoj EP, Lu W, Kiser T, Goldblum SE and Kim KC: MUC1 inhibits cell proliferation by a beta-catenin-dependent mechanism. Biochim Biophys Acta. 1773:1028–1038. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Meigs TE, Fedor-Chaiken M, Kaplan DD, Brackenbury R and Casey PJ: Galpha12 and Galpha13 negatively regulate the adhesive functions of cadherin. J Biol Chem. 277:24594–24600. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Yu Y, Wu J, Wang Y, Zhao T, Ma B, Liu Y, Fang W, Zhu WG and Zhang H: Kindlin 2 forms a transcriptional complex with β-catenin and TCF4 to enhance Wnt signalling. EMBO Rep. 13:750–758. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Escobar DJ, Desai R, Ishiyama N, Folmsbee SS, Novak MN, Flozak AS, Daugherty RL, Mo R, Nanavati D, Sarpal R, et al: α-Catenin phosphorylation promotes intercellular adhesion through a dual-kinase mechanism. J Cell Sci. 128:1150–1165. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Zaoui K, Benseddik K, Daou P, Salaün D and Badache A: ErbB2 receptor controls microtubule capture by recruiting ACF7 to the plasma membrane of migrating cells. Proc Natl Acad Sci USA. 107:18517–18522. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Sagara M, Kawasaki Y, Iemura SI, Natsume T, Takai Y and Akiyama T: Asef2 and Neurabin2 cooperatively regulate actin cytoskeletal organization and are involved in HGF-induced cell migration. Oncogene. 28:1357–1365. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Vazquez F, Ramaswamy S, Nakamura N and Sellers WR: Phosphorylation of the PTEN tail regulates protein stability and function. Mol Cell Biol. 20:5010–5018. 2000. View Article : Google Scholar : PubMed/NCBI

56 

Costa HA, Leitner MG, Sos ML, Mavrantoni A, Rychkova A, Johnson JR, Newton BW, Yee MC, De La Vega FM, Ford JM, et al: Discovery and functional characterization of a neomorphic PTEN mutation. Proc Natl Acad Sci USA. 112:13976–13981. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Weydert C, Roling B, Liu J, Hinkhouse MM, Ritchie JM, Oberley LW and Cullen JJ: Suppression of the malignant phenotype in human pancreatic cancer cells by the overexpression of manganese superoxide dismutase. Mol Cancer Ther. 2:361–369. 2003.PubMed/NCBI

58 

Zhong W, Oberley LW, Oberley TD and St Clair DK: Suppression of the malignant phenotype of human glioma cells by overexpression of manganese superoxide dismutase. Oncogene. 14:481–490. 1997. View Article : Google Scholar : PubMed/NCBI

59 

Sarsour EH, Kalen AL and Goswami PC: Manganese superoxide dismutase regulates a redox cycle within the cell cycle. Antioxid Redox Signal. 20:1618–1627. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Oberley LW, Oberley TD and Buettner GR: Cell division in normal and transformed cells: The possible role of superoxide and hydrogen peroxide. Med Hypotheses. 7:21–42. 1981. View Article : Google Scholar : PubMed/NCBI

61 

Oberley LW, Oberley TD and Buettner GR: Cell differentiation, aging and cancer: The possible roles of superoxide and superoxide dismutases. Med Hypotheses. 6:249–268. 1980. View Article : Google Scholar : PubMed/NCBI

62 

Taghavi N, Biramijamal F, Sotoudeh M, Khademi H, Malekzadeh R, Moaven O, Memar B, A'rabi A and Abbaszadegan MR: p16INK4a hypermethylation and p53, p16 and MDM2 protein expression in esophageal squamous cell carcinoma. BMC Cancer. 10:1382010. View Article : Google Scholar : PubMed/NCBI

63 

Lu J, Pan Y, Xia X, Gu Y and Lei Y: Prognostic significance of mTOR and PTEN in patients with esophageal squamous cell carcinoma. Biomed Res Int. 2015:4172102015. View Article : Google Scholar : PubMed/NCBI

64 

Ishiguro H, Wakasugi T, Terashita Y, Sakamoto N, Tanaka T, Mizoguchi K, Sagawa H, Okubo T and Takeyama H: Decreased expression of CDH1 or CTNNB1 affects poor prognosis of patients with esophageal cancer. World J Surg Oncol. 14:2402016. View Article : Google Scholar : PubMed/NCBI

65 

Xia H, Ng SS, Jiang S, Cheung WK, Sze J, Bian XW, Kung HF and Lin MC: miR-200a-mediated downregulation of ZEB2 and CTNNB1 differentially inhibits nasopharyngeal carcinoma cell growth, migration and invasion. Biochem Biophys Res Commun. 391:535–541. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Asakura T, Yamaguchi N, Ohkawa K and Yoshida K: Proteasome inhibitor-resistant cells cause EMT-induction via suppression of E-cadherin by miR-200 and ZEB1. Int J Oncol. 46:2251–2260. 2015. View Article : Google Scholar : PubMed/NCBI

67 

Suo HB, Zhang KC and Zhao J: MiR-200a promotes cell invasion and migration of ovarian carcinoma by targeting PTEN. Eur Rev Med Pharmacol Sci. 22:4080–4089. 2018.PubMed/NCBI

Related Articles

Journal Cover

May-2020
Volume 21 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang B, Liu Y, Li L, Deng H and Xian L: MicroRNA‑200a promotes esophageal squamous cell carcinoma cell proliferation, migration and invasion through extensive target genes. Mol Med Rep 21: 2073-2084, 2020
APA
Yang, B., Liu, Y., Li, L., Deng, H., & Xian, L. (2020). MicroRNA‑200a promotes esophageal squamous cell carcinoma cell proliferation, migration and invasion through extensive target genes. Molecular Medicine Reports, 21, 2073-2084. https://doi.org/10.3892/mmr.2020.11002
MLA
Yang, B., Liu, Y., Li, L., Deng, H., Xian, L."MicroRNA‑200a promotes esophageal squamous cell carcinoma cell proliferation, migration and invasion through extensive target genes". Molecular Medicine Reports 21.5 (2020): 2073-2084.
Chicago
Yang, B., Liu, Y., Li, L., Deng, H., Xian, L."MicroRNA‑200a promotes esophageal squamous cell carcinoma cell proliferation, migration and invasion through extensive target genes". Molecular Medicine Reports 21, no. 5 (2020): 2073-2084. https://doi.org/10.3892/mmr.2020.11002