miR-218 suppresses tumor growth and enhances the chemosensitivity of esophageal squamous cell carcinoma to cisplatin

Retraction in: /10.3892/or.2022.8265

  • Authors:
    • Hang Tian
    • Lei Hou
    • Yu-Mei Xiong
    • Jun-Xiang Huang
    • Ying-Jun She
    • Xiao-Bao Bi
    • Xing-Rong Song
  • View Affiliations

  • Published online on: December 8, 2014     https://doi.org/10.3892/or.2014.3657
  • Pages: 981-989
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A growing body of evidence suggests that microRNA-218 (miR-218) acts as a tumor suppressor and is involved in tumor progression, development and metastasis and confers sensitivity to certain chemotherapeutic drugs in several types of cancer. However, our knowledge concerning the exact roles played by miR-218 in esophageal squamous cell carcinoma (ESCC) and the underlying molecular mechanisms remain relatively unclear. Thus, the aims of this study were to detect the expression of miR-218 in human ESCC tissues and explore its effects on the biological features and chemosensitivity to cisplatin (CDDP) in an ESCC cell line (Eca109), so as to provide new insights for ESCC treatment. Here, we found increased expression of miR-218 in the ESCC tissues compared with that in the matched non-tumor tissues, and its expression level was correlated with key pathological characteristics including clinical stage, tumor depth and metastasis. We also found that enforced expression of miR-218 significantly decreased cell proliferation, colony formation, migration and invasion, induced cell apoptosis and arrested the cell cycle in the G0/G1 phase, as well as suppressed tumor growth in a nude mouse model. In addition, our results showed that miR-218 mimics increased the sensitivity to the antitumor effect of CDDP in the human Eca109 cells. Importantly, this study also showed that miR-218 regulated the expression of phosphorylated PI3K, AKT and mTOR, which may contribute to suppressed tumor growth of ESCC and enhanced sensitivity of ESCC cells. These findings suggest that miR-218 is a potential therapeutic agent for the treatment of ESCC.

Introduction

Esophageal cancer represents the sixth leading cause of cancer-related deaths and the eighth most common type of cancer worldwide with a variable geographic distribution (1,2). According to its pathological characteristics, esophageal cancer has two main histological subtypes: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) (1,2). ESCC is the major histological type of esophageal cancer in developing countries, particularly in China where the incidence in high-risk northern and central China exceeds 100 cases per 100,000 individuals per year (3). Despite the use of multimodal treatments such as radical surgery, chemotherapy and radiotherapy or a combination of these options, the prognosis of ESCC patients remains very poor, with a 5-year survival rate of <20% after surgery in China (3,4). To improve the overall outcome for patients with ESCC, there is a great need to disclose the molecular mechanisms of the pathogenesis of esophageal cancer; thereby useful biomarkers and novel therapeutic targets can be discovered.

microRNAs (miRNAs) are a class of highly conserved short noncoding small RNAs, usually 18–25 nucleotides in length that negatively regulate gene expression by repressing translation or cleaving RNA transcripts in a sequence-specific manner (57). Up until 2009, more than 700 miRs have been identified and registered in humans, with each individual miR predicted to target multiple genes based on the seed sequence matches in their 3′-untranslated regions (UTRs) (8). Some highly expressed miRNAs act as oncogenes by repressing tumor suppressors; on the other hand, some low level miRNAs function as tumor suppressors by negatively regulating oncogenes (9). A growing body of evidence indicates that alterations in miRNA expression are associated with human malignancies and suggests a key role of miRs in tumor initiation and progression and metastasis, since they can function as oncogenes or tumor suppressors (10,11). Furthermore, accumulating evidence shows that the profiles of miRNA expression differ between normal and tumor tissues and that they are emerging as highly tissue-specific biomarkers with potential clinical applicability for defining cancer type and origin (12,13). Given the close relationship between miRNAs and the biological progression of multiple cancers, miRNAs are presently considered as diagnostic biomarkers of various types of cancer and potential novel targets for anticancer therapies (14).

microRNA-218 (miR-218) is a recently discovered miRNA and is downregulated in several types of cancer, such as gastric cancer (15,16), cervical squamous cell carcinoma (17), medulloblastoma (18), lung cancer (19) and glioblastoma (20). Accumulating evidence demonstrates that miR-218 acts as a tumor suppressor and confers an inhibitory effect on cancer cell proliferation, migration and invasion by targeting genes (1522). In particular, recently studies have demonstrated that overexpression of miR-218 increased chemosensitivity to cisplatin (CDDP) in cervical cancer cells (23). Although it has recently been demonstrated that miR-218 is consistently downregulated in ESCC tissues when compared to that in the normal adjacent non-tumor tissues using miRNA microarrays (24), our knowledge concerning the exact roles played by miR-218 in ESCC and the underlying molecular mechanisms remain relatively unclear.

This study was therefore undertaken to evaluate the expression of miR-218 and its clinical diagnostic significance in patients with ESCC and to analyze its functional role, especially with respect to chemosensitivity. We found the elevated expression of miR-218 in ESCC tissues when compared to that in adjacent non-tumor tissues, and its expression level was correlated with key pathological characteristics including clinical stage, tumor depth and metastasis. We also found that enforced expression of miR-218 significantly suppressed tumor growth of ESCC in vitro and in vivo. Furthermore, miR-218 enhanced the chemosensitivity of ESCC cells to cisplatin. All the experimental data indicated that miR-218 was associated with the development of human ESCC and it may be a potential therapeutic strategy for human ESCC.

Materials and methods

Clinical ESCC specimens

A total of 50 pairs of primary ESCC tissues and corresponding normal esophageal epithelia were obtained from patients who underwent curative surgical resection for ESCC at the Shanxi Cancer Hospital (Shanxi, China) from August 2012 to March 2014. Normal esophageal epithelial tissues adjacent to the tumor were extracted 5 cm away from the peripheral tumor cells, and the lack of tumor cell infiltration was verified by pathological examination. Written consent for tissue donation for research purposes was obtained from each patient. The study protocol was approved by the Ethics Committee of Guangzhou Women and Children’s Medical Center (Guangdong, China). The specimens were snap frozen in liquid nitrogen and stored at −80°C until RNA extraction.

Cell lines and cell culture

Primary culture of normal esophageal epithelial cells (NEEC) was established from fresh specimens of the adjacent noncancerous esophageal tissues, which were over 5 cm distant from the cancerous tissue, according to a previous study (25). Human ESCC cell lines (Eca109 and EC9706) used in the present study were purchased from the Cell Center of the Shanghai Institute of Life Science, Chinese Academy of Science (Shanghai, China). All cells were cultured in RPMI-1640 medium (Gibco, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS; HyClone, Logan, UT, USA) and antibiotics (100 U/ml penicillin and 100 mg/ml streptomycin) at 37°C in a humidified atmosphere containing 5% CO2.

Cell transfection

The miR-218 mimics and a non-specific miR control (NC) were synthesized and purified by Genepharma Biotech (Shanghai, China). Cells cultured in a 6-well cell culture plate were transfected with miR-218 mimics or NC using Lipofectamine 2000 (Invitrogen Life Technologies, Grand Island, NY, USA) at a final concentration of 50 nM, following the manufacturer’s protocol. Transfection efficiencies were evaluated in every experiment by qRT-PCR at 48 h post-transfection. Cells were subdivided into three groups as follows: the untransfected group (blank), the transfected negative control group (NC) and the miR-218 mimic transfection group (miR-218).

miRNA real-time RT-PCR analysis

Total RNA of the cells or tissues, including miRNAs, were extracted using the Qiagen miRNeasy Mini kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. The purity and concentration of RNA were determined using a dual-beam ultraviolet spectrophotometer (Eppendorf, Hamburg, Germany). Then, the RNA was reversely transcribed into cDNA using the Universal cDNA synthesis kit from Exiqon (Woburn, MA, USA) following the manufacturer’s instructions. All PCR reactions were detected by the ABI 7900 Fast system (Applied Biosystems, Foster City, CA, USA). The primers for qRT-PCR reaction were as follows: miR-218 sense, 5′-CGGGCTTGTGCTGATCTA-3′ and antisense, 5′-CTCGCTTCGGCAGCACA-3′; U6 sense, 5′-CTCGCTTCGGCAGCACATATACT-3′ and antisense, 5′-ACGCTTCACGAATTTGCGTGTC-3′. The expression levels of U6 were used as an internal control. The relative quantification of each miRNA was presented as the fold change after normalized to the U6 RNA for the equation 2−ΔΔCt in the Rotor-Gene 6000 Series Software 1.7 (Qiagen). The above experiment was repeated at least three times.

Cell proliferation and colony formation assays

Cell viability was assessed by the Cell Counting Kit-8 assay (CCK-8; Dojindo, Kumamoto, Japan). After being transfected with miR-218 mimics or negative control (NC), Eca109 cells were cultured in 96-well plates at a density of 5,000 cells/well. The viability of the cells was determined at the indicated time (24, 48 and 72 h) according to the CCK-8 kit manufacturer’s instructions. When the media changed from red to yellow, the absorbance value at a wavelength of 450 nm was detected by an enzyme-linked immunosorbent assay reader (Thermo Labsystems, Helsinki, Finland). The experiment was performed at least three times with similar results.

For the colony formation assay, after cells were transfected with miR-218 mimics or the corresponding negative control for 48 h, the cells were seeded in a 6-well plate at a low density (1,000 cells/well) and cultured for 10 days. Then cells were fixed with 4% paraformaldehyde for 10 min and counted after being staining with 1% crystal violet. The experiments were carried out at least three times.

Cell cycle and cell apoptosis assay

The effects of miR-218 on ESCC cell cycle and apoptosis were examined by flow cytometry. In briefly, Eca109 cells were transfected with either miR-218 mimics or negative control miRNA for 48 h, and then the cells were harvested and washed twice with PBS, fixed with 70% ethanol at −20°C for 30 min and then stored at 4°C overnight. The cells were then washed with PBS again, treated with 100 ml 100 mg/l RNase at 37°C for 30 min and stained with 100 ml 50 mg/l propidium iodide (PI; Sigma, St. Louis, MO, USA) at 4°C for 30 min in the dark. The multiplication cycle and apoptotic rate were measured by flow cytometry (BD Biosciences, Mansfield, MA, USA), and the data were analyzed using CellQuest software (BD Biosciences, San Jose, CA, USA). In addition, caspase-3 activity was detected as an additional indicator of apoptosis.

Caspase activity

The activity of caspase-3 was determined by the caspases colorimetric protease assay kits (Millipore Corp, Billerica, MA, USA) following the manufacturer’s instructions. In brief, the transfected cells were washed twice with ice-cold PBS and harvested by centrifugation. The cell pellets were then lysed in 150 μl buffer provided in the kit. Protein concentrations of the lysates were measured by the Lowry method. An aliquot of lysates was incubated with a 1 μl substrate of caspase-3 and was cultured at 37°C for 2 h. Samples were measured in a microplate reader at 405 nm. The relative caspase-3 activity of the control blank group was referred to as 100.

Transwell migration and invasion assays

The migration and invasion assays were performed using Transwell insert chambers (Corning Inc., Corning, NY, USA). For the migration assay, 1×105 transfected cells were plated into the upper chamber in serum-free medium in triplicate. Medium containing 20% FBS in the lower chamber served as a chemoattractant. After culturing for 24 h, the media were removed from the upper chamber by being wiped with a cotton swab, and the cells that migrated to the lower surface of the filter were fixed in 70% ethanol for 30 min and stained with 0.2% crystal violet for 10 min. Cell migration was determined by counting five random fields/filter under a light microscope.

For the invasion assay, 3×105 transfected cells were seeded into upper chambers precoated with Matrigel (BD Biosciences) in a serum-free medium in triplicate. Media with 20% FBS were added to the lower chamber to serve as a chemoattractant, and the subsequent steps were similar to the migration assay. The number of cells invading the Matrigel were counted in five randomly selected fields using an inverted microscope (Olympus, Tokyo, Japan).

In vitro assay of CDDP

Transfected cells (5×103) were seeded into 96-well plates for 24 h. CDDP (Sigma) was freshly prepared and added to the cells with a final concentration series from 0 to 100 μM. After incubation for 72 h, cell viability was determined by the CCK-8 kit. The IC50 values were calculated and presented as means ± standard deviation (SD) from three independent experiments performed in sextuple.

In vitro CDDP treatment

Eca109 cells were transfected with miR-218 mimic and the corresponding negative controls as described above and at 24 h after transfection, freshly prepared CDDP was added at a concentration 2× the IC50 value. The chemotherapy-treated cells were further cultured for cell proliferation assay at different time points or for cell apoptosis and caspase-3 activity assay for 72 h.

Western blot analysis

The cells were harvested at 48 h after transfection and lysed with RIPA buffer (Sigma) for 30 min at 4°C. Equal amounts of protein lysates (20 μg/lane) were separated by 8–15% SDS-PAGE and then electrotransferred to nitrocellulose membranes (Invitrogen). The membranes were blocked with TBST containing 5% non-fat dry milk for 2 h and incubated with primary antibodies as followed: anti-MMP-2 (1:500, Cell Signaling Technology); anti-MMP-9 (1:1,000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA); anti-mTOR (1:2,000); anti-phosphorylated mTOR (p-mTOR; S2448, 1:1000); anti-PI3K (1:2,000); anti-p-PI3K (Tyr458, 1:1,500); anti-AKT (1:2,000); and anti-p-Akt (S473; 1:500) (all from Cell Signaling Technology) and β-actin (1:10,000; Santa Cruz Biotechnology, Inc.) overnight at 4°C. After washing for 3 times with TBST, the membranes were incubated with horseradish peroxidase-conjugated anti-mouse secondary antibody (1:5,000; Santa Cruz Biotechnology, Inc.) for 2 h. Then, the membranes were washed 3 times with TBST again and the proteins were detected by enhanced chemiluminescence (ECL; Thermo Scientific, Rockford, IL, USA). β-actin was used as a loading control. The integrated density of the band was quantified by Quantity One software (Bio-Rad, Hercules, CA, USA).

In vivo tumorigenesis assay

Thirty female BALB mice (5–6 weeks) were purchased from Jilin Institute of Experimental Animals (Changchun, China). All animals were housed under a pathogen-free condition. All experimental procedures were conducted according to the guidelines provided by NIH (National Cancer Institute, USA). For xenografts, nude mice were divided into three groups (n=10 for each) and were subcutaneously (s.c.) inoculated into the right flanks with 2×106 of miR-218-transfected or negative control-transfected cells, respectively. Mice (n=10) injected with 2×106 of Eca109 cells were used as the positive control. Tumor sizes were measured every 7 days using a caliper. Tumor volumes (cm3) were calculated using the following standard formula: 0.5236 × width2 × length. Mice were sacrificed at 4 weeks post-injection, and the tumors were resected and weighed. A portion of each tumor was used to measure the miR-218 level according to qRT-PCR.

Statistical analysis

All data are shown as mean ± SD, and the experiments were repeated three times. Comparisons between the groups were analyzed with one-way ANOVA or two-tailed Student’s t-test from SPSS version 16.0 software (SPSS Inc., Chicago, IL, USA) and the GraphPad Prism version 5.01 (GraphPad Software, Inc., San Diego, CA, USA) for Windows®. The difference was deemed statistically significant at P<0.05.

Results

Downregulation of miR-218 expression in the ESCC tissues

To investigate the role of miR-218 in esophageal malignancies, we first evaluated the expression levels of miR-218 in clinical samples of ESCCs and matched corresponding adjacent non-tumor tissues by real-time RT-PCR. Expression of U6 small RNA was used as an internal standard. Compared with the normal esophageal epithelial tissues, a significant downregulation in miR-218 expression in ESCC was observed (P<0.05, Fig. 1A). Meanwhile, the expression levels of miR-218 were decreased (P<0.05) in the two ESCC cell lines (Eca-109 and EC9706) examined, compared with the control NEEC cell line (Fig. 1B).

In addition, we further evaluated the correlation between clinicopathological factors and miR-218 expression (Table I). We found that there was no correlation between the miR-218 expression level and age, gender and tumor location, while the relative miR-218 expression level was significantly negatively correlated with TNM stage, lymph node metastasis and tumor depth (P<0.05, Table I). These findings imply that miR-218 may play a key role in ESCC development.

Table I

Correlation between the relative level of miR-218 in ESCC tissues and the clinicopathological features of the patients with ESCC.

Table I

Correlation between the relative level of miR-218 in ESCC tissues and the clinicopathological features of the patients with ESCC.

FeaturenmiR-218 expression levelP-value
Age (years)>0.05
 <60240.4214±0.026
 ≥60260.4182±0.025
Gender>0.05
 Male300.4210±0.025
 Female200.4195±0.021
Tumor location>0.05
 Middle310.4184±0.018
 Lower190.4301±0.028
Metastasis<0.05
 Yes220.3613±0.019
 No280.4856±0.029
TNM stage<0.05
 0–I250.5131±0.026
 II–IV250.3334±0.014
Tumor depth<0.05
 Tis, T1230.5312±0.036
 T2, T3, T4270.3258±0.013

[i] ESCC, esophageal squamous cell carcinoma.

miR-218 inhibits cell proliferation and colony formation in ESCC cells

To validate whether miR-218 regulates ESCC cell growth, we performed a proliferation assay by transfecting miR-218 mimics or NC into Eca109 cells. After transfection, the miR-218 level in the miR-218 mimic group was highly increased (P<0.05) when compared to the NC group and control blank group (Fig. 2A). As expected, increased expression of miR-218 inhibited cell proliferation (P<0.05) in the Eca109 cells (Fig. 2B). Meanwhile, the effects of miR-218 on cell colony formation were also examined in the Eca109 cells. Our results showed that upregulation of miR-218 significantly suppressed (P<0.05) cell colony formation in the Eca109 cells (Fig. 2C).

miR-218 induces cell cycle arrest and apoptosis in ESCC cells

To determine the effects of miR-218 on the cell cycle and cell apoptosis, FACScan flow cytometry assays were performed. Flow cytometric analysis revealed that the G1 phase cell population was increased in the miR-218 group compared to the blank group and NC group (P<0.05, Fig. 3A and B). In addition, enforced expression of miR-218 resulted in a much lower percentage of cells in the S phase compared with those of the blank group and NC group (P<0.05, Fig. 3A and B).

Our flow cytometry results also indicated that the apoptosis level of the miR-218 group was significantly enhanced compared to the levels in cells in the blank group and NC groups (P<0.01; Fig. 3C). Similarly, results from the caspase activity assay showed that transfection with miR-218 led to a significant increase in caspase-3 activity compared to the NC and blank groups (P<0.01). From these results we conclude that upregulation of miR-218 induces cell cycle arrest at the G0/G1 phase and the apoptosis of ESCC cells.

miR-218 inhibits cell migration and invasion of ESCC cells

Since invasiveness is one of the pathophysiological features of human malignant ESCC, we aimed to ascertain whether miR-218 is associated with the invasiveness of ESCC. Transwell assay (without Matrigel) showed that the migratory speed of the Eca109 cells was markedly slower in the miR-218 group than that of the blank group and NC group (Fig. 4A). Furthermore, Transwell matrix penetration (coated with Matrigel) assay showed that the upregulation of miR-218 markedly reduced the invasiveness of the Eca109 cells (Fig. 4B).

Furthermore, we analyzed the effects of miR-218 on the expression of MMP-2 and MMP-9 by western blot analysis. It was found that MMP-2 and MMP-9 protein expression levels were significantly decreased in the miR-218 treatment group compared to the blank group and NC group (P<0.05, Fig. 4C and D). These findings suggest that the upregulation of miR-218 inhibits the invasive ability of ESCC cells in vitro by inhibiting MMP-2 and MMP-9 expression.

miR-218 increases the chemosensitivity of ESCC cells to cisplatin

To investigate whether miR-218 plays a role in the chemosensitivity of ESCC cells, the transfected cells were treated with different concentrations of CDDP ranging from 0 to 100 μM for 72 h. Cell viability was then measured using the CCK-8 method. As shown in Fig 5A, enforced expression of miR-218 increased the sensitivity of Eca109 cells to CDDP. Data analysis showed that the IC50 of the CDDP-treated cancer cells in blank control group, NC group and miR-218 group was 18.75±1.26, 18.26±1.27 and 7.48±0.64, respectively, which indicated that miR-218 significantly increased the chemosensitivity to CDDP. To investigate the role of miR-218 in the CDDP-treated cells, we detected the proliferation and apoptosis of miR-218-overexpressing cells exposed to CDDP. The three groups of cells were treated with 15 μM of CDDP (~2× IC50 of the miR-218 group) for 72 h. The CCK-8 assay showed that enforced expression of miR-218 markedly enhanced the growth inhibitory effect of CDDP in the Eca109 cells (P<0.05, Fig. 5B). We also found that miR-218 mimics combined with CDDP significantly induced cell apoptosis (P<0.05, Fig. 5C) and increase caspase-3 activity (P<0.05, Fig. 5D) compared to the NC combined with CDDP group and the single treatment CDDP group. These results suggest that miR-218 increased the sensitivity of ESCC cells to CDDP.

miR-218 regulates the PI3K/AKT/mTOR signaling pathway

We then aimed to ascertain the signaling pathways involved during the regulation of miR-218 in ESCC cells. We examined the effect of miR-218 on the AKT/PI3K/mTOR signaling pathway, which participates in the main intracellular signaling required for cell proliferation and drug-resistance in ESCC cells. Measurements of the phosphorylation/activation pattern of PI3K, AKT and mTOR were performed using western blot analysis. It was found that enforced expression of miR-218 resulted in a marked decrease in phosphorylated PI3K, AKT and mTOR compared to the cells transfected with NC or the blank control, without altering the total protein levels of PI3K, AKT and mTOR in each group (Fig. 6). These results imply that miR-218 inhibits tumor growth and enhances the sensitivity of ESCC cells to CDDP, to some extent, by regulation of the PI3K/AKT/mTOR signaling pathway.

miR-218 suppresses tumor growth in vivo

Finally, we investigated the effect of miR-218 on tumor growth in nude mice. Tumors were monitored every 7 days from the time that they were apparent. Compared with the control group and NC group, tumor growth in the miR-218 group was significantly reduced (Fig. 7A and B). Day 28 after implantation, animals were sacrificed and tumor weights were determined. Consistent with tumor volumes, the tumor weights were decreased in the miR-218 group (Fig. 7C). To clarify miR-218 transfection activity, we examined miR-218 expression in the xenograft tumor tissues. The results showed that miR-218 expression was upregulated in the xenograft tumors in the miR-218 treatment group. These data indicate that miR-218 acts as a tumor suppressor in ESCC and suppresses tumor growth in vivo.

Discussion

Recently, a growing amount of studies suggest that a number of miRNAs are involved in several aspects of the pathogenesis of ESCC including cell cycle control, cell apoptosis, invasion, migration, as well as resistance to chemotherapy and radiotherapy. It has been reported that miR-21, miR-106b, miR- 373, miR-192, miR-196a and miR-147 were upregulated in patients with ESCC (2630), suggesting that they may exert tumor oncogene functions, while miR-655, miR-375, miR-625, miR-518b, miR-133a, miR-133b, miR-143 and miR-145 were downregulated in patients with ESCC (26,29,3133), suggesting that they may exert tumor suppressive functions. Here, we sought to provide evidence that upregulation of miR-218 inhibits tumor growth of ESCC in vitro and in vivo. In the present study, we found that miR-218 was frequently downregulated in both ESCC cell lines and human ESCC tissues and its expression level was correlated with key pathological characteristics including TNM stage, tumor depth and metastasis. This result is in line with a previous study showing that miR-218 was weakly expressed in patients with ESCC (24). In the present study, we also demonstrated that upregulation of miR-218 in an ESCC cell line inhibited cell proliferation, colony formation, migration and invasion, induced cell cycle arrest at the G0/G1 phase, as well as suppressed tumor growth in a nude mouse model, thus providing the direct evidence of an important role of miR-218 in regulating ESCC development and progression.

It has been well known that miRNAs play critical roles in the regulation of cancer cell proliferation, cell cycle, migration and invasion by regulating multiple target genes (34,35). It has been shown that miR-218 expression is downregulated in many tumors including ESCC (1520) and is involved in cancer initiation and development (1522). To date, some genes have been identified as target genes of miR-218, such as HMGB1 (19), LEF1 (20), Rictor (23), IKK-β (36), CDK6 (18), SH3GL1 (37), SOT (38), Rob1 receptor (39) as well as LAMB3 (40), which are involved in several different signaling pathways, suggesting that miR-218 is a tumor suppressor miRNA and plays a crucial role in the initiation, development and metastasis of tumors. Therefore, we reasonably speculated that miR-218 suppresses the tumor growth of ESCC in vitro and in vivo by regulating multiple target genes.

Cisplatin (cis-diamminedichloroplatinum or cis-DDP/CDDP) is an anticancer drug widely used in the treatment of various cancers including ESCC. However, resistance to cisplatin limits the effectiveness of treatment for ESCC. Therefore, elucidation of the mechanisms that regulate cisplatin resistance in ESCC is urgently needed. It was well known that the PI3K/Akt/mTOR signaling pathway contributes to the formation of a majority of human malignancies and is frequently implicated in the development of resistance to existing anticancer therapies (41). It has been shown that activation of the PI3K/Akt/mTOR pathway confers cisplatin resistance to various cancer therapies including ESCC (42). Thus, targeting of this pathway is a promising strategy for therapy in ESCC. Recent studies have demonstrated that miR-218 targets the mTOR component Rictor and blocks AKT phosphorylation in oral cancer (43). Xiao et al (44) also showed that upregulated expression of miR-218 in HCC cells significantly decreased cell proliferation through the PTEN/AKT/PI3K pathway. Of note, Li et al (23) found that miR-218 inhibited the proliferation of the human cervical cancer cell line HeLa and increased chemosensitivity to cisplatin in vitro by blocking the AKT-mTOR signaling pathway targeting Rictor. Similar with these results, in the present study, we found that upregulation of miR-218 inhibited cell proliferation, colony formation, migration and invasion, induced cell cycle arrest and apoptosis, enhanced the sensitivity of ESCC cells to CDDP, as well as decreased mTOR, AKT and PI3K phosphorylation. Thus, we proposed that the molecular mechanism by which miR-218 inhibits tumor growth and enhances the sensitivity of ESCC cells to CDDP may be attributed to, at least in part, suppression of the PI3K/AKT/mTOR signaling pathway.

In conclusion, to our knowledge, this is the first full-scale report concerning the association of miR-218 with esophageal squamous cell carcinoma. The present study demonstrated that miR-218 expression was decreased in ESCC tissues and a cell line and its expression level was correlated with key pathological characteristics including TNM stage, tumor depth and metastasis and that upregulation of miR-218 suppressed tumor growth of ESCC in vitro and in vivo and enhanced the sensitivity of ESCC cells to cisplatin, to some extent, by suppressing the PI3K/AKT/mTOR signaling pathway. Based on the multiple functions of miR-218 on the tumor growth of ESCC, miR-218 may be considered as a potential anticancer therapeutic target for esophageal squamous cell carcinoma.

References

1 

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

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

McCann J: Esophageal cancers: changing character, increasing incidence. Natl Cancer Inst. 91:497–498. 1999. View Article : Google Scholar

4 

Demeester SR: Epidemiology and biology of esophageal cancer. Gastrointest Cancer Res. 3(Suppl 2): S2–S5. 2009.PubMed/NCBI

5 

Brennecke J and Cohen SM: Towards a complete description of the microRNA complement of animal genomes. Genome Biol. 4:2282003. View Article : Google Scholar : PubMed/NCBI

6 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Carthew RW and Sontheimer EJ: Origins and mechanisms of miRNAs and siRNAs. Cell. 136:642–655. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Esquela-Kerscher A and Slack FJ: Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Volinia S, Calin GA, Liu CG, et al: A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci USA. 103:2257–2261. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Croce CM: Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 10:704–714. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Rosenfeld N, Aharonov R, Meiri E, et al: microRNAs accurately identify cancer tissue origin. Nat Biotechnol. 26:462–469. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Liang Y, Ridzon D, Wong L and Chen C: Characterization of microRNA expression profiles in normal human tissues. BMC Genomics. 8:1662007. View Article : Google Scholar : PubMed/NCBI

14 

Cho WC: OncomiRs: the discovery and progress of microRNAs in cancers. Mol Cancer. 6:602007. View Article : Google Scholar : PubMed/NCBI

15 

Xin SY, Feng XS, Zhou LQ, Sun JJ, Gao XL and Yao GL: Reduced expression of circulating microRNA-218 in gastric cancer and correlation with tumor invasion and prognosis. World J Gastroenterol. 20:6906–6911. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Gao C, Zhang Z, Liu W, Xiao S, Gu W and Lu H: Reduced microRNA-218 expression is associated with high nuclear factor kappa B activation in gastric cancer. Cancer. 116:41–49. 2010.

17 

Yu J, Wang Y, Dong R, Huang X, Ding S and Qiu H: Circulating microRNA-218 was reduced in cervical cancer and correlated with tumor invasion. J Cancer Res Clin Oncol. 138:671–674. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Venkataraman S, Birks DK, Balakrishnan I, et al: microRNA 218 acts as a tumor suppressor by targeting multiple cancer phenotype-associated genes in medulloblastoma. J Biol Chem. 288:1918–1928. 2013. View Article : Google Scholar :

19 

Zhang C, Ge S, Hu C, Yang N and Zhang J: MiRNA-218, a new regulator of HMGB1, suppresses cell migration and invasion in non-small cell lung cancer. Acta Biochim Biophys Sin (Shanghai). 45:1055–1061. 2013. View Article : Google Scholar

20 

Liu Y, Yan W, Zhang W, et al: miR-218 reverses high invasiveness of glioblastoma cells by targeting the oncogenic transcription factor LEF1. Oncol Rep. 28:1013–1021. 2012.PubMed/NCBI

21 

Prudnikova TY, Mostovich LA, Kashuba VI, Ernberg I, Zabarovsky ER and Grigorieva EV: miRNA-218 contributes to the regulation of D-glucuronyl C5-epimerase expression in normal and tumor breast tissues. Epigenetics. 7:1109–1114. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Yang L, Li Q, Wang Q, Jiang Z and Zhang L: Silencing of miRNA-218 promotes migration and invasion of breast cancer via Slit2-Robo1 pathway. Biomed Pharmacother. 66:535–540. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Li J, Ping Z and Ning H: miR-218 impairs tumor growth and increases chemo-sensitivity to cisplatin in cervical cancer. Int J Mol Sci. 13:16053–16064. 2012. View Article : Google Scholar

24 

Yang M, Liu R, Sheng J, et al: Differential expression profiles of microRNAs as potential biomarkers for the early diagnosis of esophageal squamous cell carcinoma. Oncol Rep. 29:169–176. 2013.

25 

Andl CD, Mizushima T, Nakagawa H, et al: Epidermal growth factor receptor mediates increased cell proliferation, migration, and aggregation in esophageal keratinocytes in vitro and in vivo. J Biol Chem. 278:1824–1830. 2003. View Article : Google Scholar

26 

Sakai NS, Samia-Aly E, Barbera M and Fitzgerald RC: A review of the current understanding and clinical utility of miRNAs in esophageal cancer. Semin Cancer Biol. 23:512–521. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Luthra R, Singh RR, Luthra MG, et al: microRNA-196a targets annexin A1: a microRNA-mediated mechanism of annexin A1 downregulation in cancers. Oncogene. 27:6667–6678. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Kan T, Sato F, Ito T, et al: The miR-106b-25 polycistron, activated by genomic amplification, functions as an oncogene by suppressing p21 and Bim. Gastroenterology. 136:1689–1700. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Lee KH, Goan YG, Hsiao M, et al: microRNA-373 (miR-373) post-transcriptionally regulates large tumor suppressor, homolog 2 (LATS2) and stimulates proliferation in human esophageal cancer. Exp Cell Res. 315:2529–2538. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Fassan M, Volinia S, Palatini J, et al: microRNA expression profiling in human Barrett’s carcinogenesis. Int J Cancer. 129:1661–1670. 2011. View Article : Google Scholar

31 

Zhang M, Zhou S, Zhang L, et al: miR-518b is downregulated, and involved in cell proliferation and invasion by targeting Rap1b in esophageal squamous cell carcinoma. FEBS Lett. 586:3508–3521. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Wang Z, Qiao Q, Chen M, et al: miR-625 downregulation promotes proliferation and invasion in esophageal cancer by targeting Sox2. FEBS Lett. 588:915–921. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Wang Y, Zang W, Du Y, et al: mir-655 upregulation suppresses cell invasion by targeting pituitary tumor-transforming gene-1 in esophageal squamous cell carcinoma. J Transl Med. 11:3012013. View Article : Google Scholar

34 

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

35 

Manikandan J, Aarthi JJ, Kumar SD and Pushparaj PN: Oncomirs: the potential role of non-coding microRNAs in understanding cancer. Bioinformation. 2:330–334. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Song L, Huang Q, Chen K, et al: miR-218 inhibits the invasive ability of glioma cells by direct downregulation of IKK-β. Biochem Biophys Res Commun. 402:135–140. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Shi J, Yang L, Wang T, et al: miR-218 is downregulated and directly targets SH3GL1 in childhood medulloblastoma. Mol Med Rep. 8:1111–1117. 2013.PubMed/NCBI

38 

Hassan MQ, Maeda Y, Taipaleenmaki H, et al: miR-218 directs a Wnt signaling circuit to promote differentiation of osteoblasts and osteomimicry of metastatic cancer cells. J Biol Chem. 287:42084–42092. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Tie J, Pan Y, Zhao L, et al: miR-218 inhibits invasion and metastasis of gastric cancer by targeting the Robo1 receptor. PLoS Genet. 6:e10008792010. View Article : Google Scholar : PubMed/NCBI

40 

Yamamoto N, Kinoshita T, Nohata N, et al: Tumor suppressive microRNA-218 inhibits cancer cell migration and invasion by targeting focal adhesion pathways in cervical squamous cell carcinoma. Int J Oncol. 42:1523–1532. 2013.PubMed/NCBI

41 

Courtney KD, Corcoran RB and Engelman JA: The PI3K pathway as drug target in human cancer. J Clin Oncol. 28:1075–1083. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Huang Y, Xi Q, Chen Y, et al: A dual mTORC1 and mTORC2 inhibitor shows antitumor activity in esophageal squamous cell carcinoma cells and sensitizes them to cisplatin. Anticancer Drugs. 24:889–898. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Uesugi A, Kozaki K, Tsuruta T, et al: The tumor suppressive microRNA miR-218 targets the mTOR component Rictor and inhibits AKT phosphorylation in oral cancer. Cancer Res. 71:5765–5778. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Xiao ZD, Jiao CY, Huang HT, et al: miR-218 modulate hepatocellular carcinoma cell proliferation through PTEN/ AKT/PI3K pathway and HoxA10. Int J Clin Exp Pathol. 7:4039–4044. 2014.

Related Articles

Journal Cover

February-2015
Volume 33 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian H, Hou L, Xiong Y, Huang J, She Y, Bi X and Song X: miR-218 suppresses tumor growth and enhances the chemosensitivity of esophageal squamous cell carcinoma to cisplatin Retraction in /10.3892/or.2022.8265. Oncol Rep 33: 981-989, 2015
APA
Tian, H., Hou, L., Xiong, Y., Huang, J., She, Y., Bi, X., & Song, X. (2015). miR-218 suppresses tumor growth and enhances the chemosensitivity of esophageal squamous cell carcinoma to cisplatin Retraction in /10.3892/or.2022.8265. Oncology Reports, 33, 981-989. https://doi.org/10.3892/or.2014.3657
MLA
Tian, H., Hou, L., Xiong, Y., Huang, J., She, Y., Bi, X., Song, X."miR-218 suppresses tumor growth and enhances the chemosensitivity of esophageal squamous cell carcinoma to cisplatin Retraction in /10.3892/or.2022.8265". Oncology Reports 33.2 (2015): 981-989.
Chicago
Tian, H., Hou, L., Xiong, Y., Huang, J., She, Y., Bi, X., Song, X."miR-218 suppresses tumor growth and enhances the chemosensitivity of esophageal squamous cell carcinoma to cisplatin Retraction in /10.3892/or.2022.8265". Oncology Reports 33, no. 2 (2015): 981-989. https://doi.org/10.3892/or.2014.3657