MicroRNA‑655 suppresses cell proliferation and invasion in oral squamous cell carcinoma by directly targeting metadherin and regulating the PTEN/AKT pathway

Retraction in: /10.3892/mmr.2022.12897

  • Authors:
    • Qiang Wang
    • Longkun Lv
    • Yuan Li
    • Honghai Ji
  • View Affiliations

  • Published online on: July 16, 2018     https://doi.org/10.3892/mmr.2018.9292
  • Pages: 3106-3114
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRNAs) are important regulators of a variety of biological processes and their dysregulation is closely related to cancer formation and progression. Therefore, examination of aberrantly expressed miRNAs in oral squamous cell carcinoma (OSCC) may provide important clues for the diagnosis and treatment of patients with OSCC. The aim of the present study was to determine miRNA (miR)‑655‑3p expression in OSCC tissues and cell lines, and to investigate the biological roles and mechanisms of miR‑655‑3p associated with OSCC. Data from the present study indicated that miR‑655 expression was significantly downregulated in human OSCC tissues and cell lines. Overexpression of miR‑655 attenuated cell proliferation and invasion in OSCC in vitro. Metadherin (MTDH) mRNA was predicted as a potential target of miR‑655 by bioinformatics analysis, and this was confirmed by luciferase reporter assay, reverse transcription‑quantitative polymerase chain reaction and western blot analysis. In OSCC tissues, MTDH was highly expressed and inversely correlated with miR‑655 expression levels. MTDH overexpression reversed the inhibitory effects of miR‑655 mimics in OSCC cells. Notably, the upregulation of miR‑655 expression inhibited the activation of the phosphatase and tensin homolog (PTEN)/RAC‑α serine/threonine‑protein kinase (AKT) pathway in OSCC cells. Therefore, these results may provide the first evidence that miR‑655 targets MTDH to inhibit proliferation and invasion of OSCC by inhibiting PTEN/AKT signaling. Thus, the restoration of miR‑655 expression may be a novel therapeutic strategy for patients with OSCC.

Introduction

Oral cancer is the sixth most common malignant neoplasm worldwide (1); oral squamous cell carcinoma (OSCC) is the most common type of oral cancer and accounts for ~90% of all oral cancer cases, with an estimated 300,000 new cases diagnosed per year (2). Human papillomavirus infection, alcohol consumption and smoking were identified as major risk factors of OSCC (3). Currently, the main effective treatments for patients are radical operation in combination with radiotherapy, neoadjuvant chemotherapy and targeted therapy (4). Despite remarkable improvements in diagnostic strategies and surgery, the clinical outcome of patients diagnosed with OSCC remains unsatisfactory, with a 5-year survival rate of <50% (5). High local recurrence rates and metastasis are largely responsible for the poor prognosis of patients with OSCC (6). Therefore, further elucidation of the mechanisms underlying the pathogenesis of OSCC may aid the development of tumor-specific biomarkers and novel therapeutic methods for early diagnosis and therapy of patients with this malignancy.

microRNAs (miRNAs) are a class of endogenous, highly conserved, noncoding, short RNAs that serve a role in gene regulation (7). miRNAs interact directly with the 3′-untranslated region (UTR) of their target mRNAs in a base-pairing manner and induce mRNA degradation and/or inhibit transcription (8). A miRNA may regulate the expression of various mRNAs simultaneously; it has been estimated that the expression of ~67% of all human protein coding genes are modulated by miRNAs (9). Previous studies have reported that abnormally expressed miRNAs may serve a role in a number of human disorders, particularly cancers (10). For example, abnormal miRNA expression was reported in OSCC (11), bladder cancer (12), gastric cancer (13), thyroid cancer (14) and lung cancer (15). Dysregulated miRNAs may serve as oncogenes or tumor suppressors, and are involved in the regulation of numerous cellular biological processes, including cell proliferation, apoptosis, invasion, metastasis, angiogenesis and epithelial-mesenchymal transition (1618). Therefore, miRNAs may serve as novel, effective biomarkers for cancer diagnosis, therapy and prognosis.

miRNA-655-3p (miR-655), which is located on chromosome 14q32, was previously reported to be aberrantly expressed in multiple human cancers, including hepatocellular carcinoma (19,20), triple-negative breast cancer (21) and esophageal squamous cell carcinoma (22). However, the expression, role and molecular mechanisms of miR-655 in OSCC have not yet been elucidated. Therefore, the aim of the present study was to detect miR-655 expression in OSCC tissues and cell lines, and to investigate its biological roles in OSCC. The mechanisms underlying the involvement of miR-655 in OSCC were also investigated. Bioinformatics analyses were performed to determine potential targets of miR-655, and the results revealed that metadherin (MTDH) was a candidate target of miR-655. Subsequent experiments were performed to determine whether MTDH was a direct target gene of miR-655 in OSCC cells. MTDH has previously been reported to contribute to the regulation of the phosphatase and tensin homolog (PTEN)/protein kinase (AKT) signaling pathway (23,24); therefore, the present study also investigated whether miR-655 participated in the regulation of the PTEN/AKT signaling pathway in OSCC cells.

Materials and methods

Collection of OSCC tumoral tissues

A total of 26 pairs of OSCC tissues and adjacent non-tumoral oral tissues were collected from patients diagnosed with OSCC and treated with radical surgery at Yidu Central Hospital of Weifang (Weifang, China) between November 2014 and February 2017. All patients (15 males and 11 females; aged 49–72 years old) enrolled in the study received no radiotherapy, chemotherapy, targeted therapy or other treatments prior to surgery. Patients treated with radiotherapy, chemotherapy, targeted therapy or other treatments prior to surgical resection was excluded from the present study. All tissues were quickly frozen in liquid nitrogen following surgery and stored in liquid nitrogen until used in subsequent experiments. The present study was approved by the Ethics Committee of Yidu Central Hospital of Weifang, and written informed consent was provided by all patients prior to sample collection.

Cell culture and transfection assay

The OSCC cell lines Tca8113, CAL-27 and SCC-9 were purchased from the American Type Culture Collection (Manassas, VA, USA) and cultured in Dulbecco's modified Eagle's medium/Ham's F-12 (DMEM/F-12) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Primary normal human oral keratinocytes (NHOK) were acquired from ScienCell Research Laboratories, Inc. (San Diego, CA, USA) and grown in the oral keratinocyte medium (ScienCell Research Laboratories, Inc.). All cell lines were maintained at 37°C in a humidified chamber with 5% CO2.

miR-655 mimics and mimic negative-controls (miR-NC) were synthesized by Guangzhou Ruibo Biological Technology, Co., Ltd. (Guangzhou, China). The miR-655 mimic sequence was 5′-AUAAUACAUGGUUAACCUCUUU-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. MTDH overexpression vector (pCMV-MTDH) and empty pCMV vector were produced by Shanghai GenePharma Co., Ltd. (Shanghai, China). For transfection assays, cells (5×105 cells/well) were plated onto 6-well plates and transfected with miR-655 mimics (100 pmol), miR-NC (100 pmol), pCMV-MTDH (4 µg) or empty pCMV vectors (4 µg) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature, according to the manufacturer's protocol. Following transection for 24 h, CCK-8 assay was performed. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and cell invasion assays were performed a total of 48 h post-transfection. Western blot analysis was used to detect protein expression in transfected cells a total of 72 h post-incubation.

RNA isolation and RT-qPCR

RT-qPCR was performed to determine the expression levels of miR-655 and MTDH mRNA. Total RNA was extracted from tissue samples (100 mg) or cells (1×106) using the TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. To detect miR-655 expression, reverse transcription was carried out using a TaqMan miRNA Reverse Transcription kit followed by qPCR with a TaqMan miRNA assay kit (both from Applied Biosystems; Thermo Fisher Scientific, Inc.). The temperature protocol for RT was as follows: 16°C for 30 min, 42°C for 30 min and 85°C for 5 min. The thermocycling conditions used for qPCR were as follows: 50°C for 2 min and 95°C for 10 min; followed by 40 cycles of denaturation at 95°C for 15 sec; and subsequently annealing/extension at 60°C for 60 sec. For the quantification of MTDH mRNA levels, a PrimeScript RT Reagent kit was used to synthesize cDNA, and qPCR was performed using SYBR Premix Ex Taq II (both from Takara Biotechnology Co., Ltd., Dalian, China). The temperature protocol for RT was as follows: 37°C for 15 min and 85°C for 5 sec. The thermocycling conditions used for qPCR were as follows: 5 min at 95°C; followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. RT-qPCR was performed in an ABI PRISM 7000 Fluorescent Quantitative PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). U6 small nuclear RNA and GAPDH were used as the endogenous controls to normalize the levels of miR-655 and MTDH mRNA, respectively. The primers used were as follows: miR-655 forward, 5′-TCCGAACATGGTTAA-3′ and reverse, 5′-GTGCAGGGTCCGAGGT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; MTDH forward, 5′-TGTTGAAGTGGCTGAGGG-3′ and reverse, 5′-CAGGAAATGATGCGGTTG-3′; and GAPDH forward, 5′-CGGAGTCAACGGATTTGGTCGTAT-3′ and reverse, 5′-AGCCTTCTCCATGGTGGTGAAGAC-3′. Relative gene expression was calculated using the 2−ΔΔCq method (25).

MTT cell proliferation assay

Following transfection for 24 h, cells (2,000 cells/well) were seeded into 96-well plates, and the extent of proliferation was detected at room temperature by performing an MTT assay at 0, 24, 48 and 72 h. Briefly, a total of 20 µl MTT solution (5 mg/ml; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was added to each well. The culture plates were incubated at 37°C for 4 h. Subsequently, the supernatant containing the MTT solution was discarded and 150 µl dimethyl-sulfoxide was added into each well to dissolve the formazan precipitate. The absorbance was read at a wavelength of 490 nm using an ELISA microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Cell invasion assay

Transfected cells were trypsinized, collected and washed with FBS-free DMEM/F-12 medium at 48 h post-transfection. A total of 1×105 cells were suspended in 200 µl FBS-free DMEM/F-12 medium and seeded in the upper Transwell chambers (8 µm; Corning Inc., Corning, NY, USA) coated with Matrigel (BD Biosciences, San Jose, CA, USA). DMEM/F-12 medium (500 µl) containing 10% FBS was added into the lower chamber. Following 24 h incubation at 37°C with 5% CO2, the non-invading cells on the upper membrane were scraped off with cotton swabs. The invasive cells were fixed with 95% ethanol for 15 min and stained with 0.5% crystal violet for 15 min, both at room temperature. The number of invasive cells was counted in at least five randomly selected fields using an IX51 inverted microscope (magnification, ×200; Olympus Corporation, Tokyo, Japan).

Bioinformatics analysis

TargetScan (version 7.2; http://www.targetscan.org) and PicTar (http://pictar.mdc-berlin.de) were used to predict the potential targets of miR-655.

Luciferase report assay

Human MTDH 3′-UTRs containing putative wild-type (WT) or mutated (Mut) binding sites for miR-655 were amplified by Shanghai GenePharma Co., Ltd., cloned into the psiCHECK-2 reporter vector (Promega Corporation, Madison, WI, USA), and designated MTDH-3′-UTR-WT or MTDH-3′-UTR-Mut. Cells were seeded into 24-well plates (1.5×105 cells/well) 1 day prior to transfection and incubated at 37°C with 5% CO2. miR-655 mimics (50 pmol) or miR-NC (50 pmol) were co-transfected into cells with either MTDH-3′-UTR-WT (100 ng) or MTDH-3′-UTR-Mut (100 ng) using Lipofectamine 2000, and incubated at 37°C with 5% CO2 for 48 h, according to the manufacturer's protocol. Luciferase activities were evaluated using the Dual Luciferase Assay kit (Promega Corporation), according to the manufacturer's protocol. Firefly luciferase activities were normalized to Renilla luciferase activities.

Western blot analysis

The Total Protein Extraction kit (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China) was used to extract total protein from tissue samples (200 mg) or cells (1×106). Subsequently, the concentration of total protein was quantified using a Bicinchoninic Acid Assay kit (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China). Equivalent amounts of protein (30 µg) were separated by 10% SDS-PAGE and electroblotted onto polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat milk in TBS containing 0.1% Tween-20 (TBST) at room temperature for 1 h and incubated with the following primary antibodies overnight at 4°C: Mouse anti-human MTDH antibody (1:1,000; cat. no. sc-517220; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), mouse anti-human PTEN antibody (1:1,000; cat. no. ab77161; Abcam, Cambridge, UK), mouse anti-human RAC-α serine/threonine-AKT antibody (1:1,000; cat. no. sc-56878), mouse anti-human phosphorylated (p)-AKT antibody (1:1,000; cat. no. sc-271966; both Santa Cruz Biotechnology, Inc.) and mouse anti-human GAPDH antibody (1:1,000; cat. no. ab125247; Abcam). Membranes were washed three times with TBST, followed by incubation with horseradish peroxidase-conjugated goat anti-mouse secondary antibody (1:5,000; cat. no. ab205719; Abcam). Following extensive washes with TBST, protein signals were visualized with an Enhanced Chemiluminescence Detection System (Pierce; Thermo Fisher Scientific, Inc.). GAPDH was used as a loading control for normalization of protein expression levels. Densitometric analysis was performed using Quantity One software version 4.62 (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Statistical analysis

Data are presented as the mean ± standard deviation, based on the results of at least three repeated experiments. Analyses were performed with SPSS 19.0 software (IBM Corporation, Armonk, NY, USA). Differences between two groups were analyzed with a two-tailed Student's t-test. One-way analysis of variance followed by Student-Newman-Keuls post hoc test was performed to investigate the differences between more than two groups. Spearman's correlation analysis was used to examine the correlation between miR-655 and MTDH mRNA expression levels in OSCC tissues. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-655 expression is downregulated in OSCC tissues and cell lines

To examine the role of miR-655 in OSCC, miR-655 expression levels were determined in a total of 26 pairs of OSCC tissues and adjacent non-tumoral tissues using RT-qPCR. Compared with expression in the adjacent non-tumor tissues, miR-655 expression was significantly lower in OSCC tissues (Fig. 1A; P<0.05). In addition, miR-655 expression levels were examined in three OSCC cell lines (Tca8113, CAL-27 and SCC-9) and NHOK cells. The data indicated that miR-665 expression was significantly lower in OSCC cell lines compared with expression in NHOK cells (Fig. 1B; P<0.05). These results suggested that the expression of miR-655 is significantly reduced in OSCC; this reduced expression may be related to OSCC progression.

miR-655 inhibits Tca8113 and CAL-27 cell proliferation and invasion

Tca8113 and CAL-27 cells exhibited relatively low miR-655 expression among the three OSCC cell lines; therefore, these cells were selected for subsequent experiments. To explore the effects of miR-655 in OSCC, miR-655 mimics were transfected into Tca8113 and CAL-27 cells to increase miR-655 expression levels. RT-qPCR analysis confirmed that miR-655 was significantly increased in Tca8113 and CAL-27 cells transfected with miR-655 mimics compared expression levels in cells transfected with miR-NC (Fig. 2A; P<0.05).

MTT and cell invasion assays were performed to examine the effects of miR-655 overexpression on proliferation and invasion, respectively, of Tca8113 and CAL-27 cells. The results indicated that transfection with miR-655 mimics led to a significant reduction in proliferation and invasion compared with the respective miR-NC groups (Fig. 2B and C, respectively; P<0.05). These results suggested that miR-655 may have tumor suppressive roles in OSCC growth and metastasis.

miR-655 directly targets and downregulates MTDH mRNA expression in OSCC cells

The biological roles of miRNAs in human malignancies depend on their specific targets; therefore, bioinformatics analysis was performed to predict the potential targets of miR-655. A total of 780 conserved sites were revealed, including A disintegrin metallopeptidase domain-containing protein 10; pituitary tumor-transforming 1 interacting protein; and membrane associated guanylate kinase, WW and PDZ domain containing 2. Among these candidates, MTDH was predicted as a major target of miR-655 (Fig. 3A), which has been previously reported to be involved in OSCC occurrence and development (2629). To confirm this hypothesis, luciferase reporter assays were performed to determine whether the 3′-UTR of MTDH was directly targeted by miR-655 in OSCC cells. Luciferase activity was significantly reduced in MTDH-3′-UTR-WT + miR655 mimics transfected Tca8113 and CAL-27 cells compared with cells co-transfected with MTDH-3′-UTR-WT + miR-NC (Fig. 3B; P<0.05), whereas no significant differences in luciferase activities were identified in cells co-transfected with miR-655 mimics or miR-NC and MTDH-3′-UTR-Mut. Furthermore, RT-qPCR and western blot assays demonstrated that overexpression of miR-655 significantly reduced the expression levels of MTDH mRNA and protein (Fig. 3C and D, respectively; P<0.05) in Tca8113 and CAL-27 cells compared with miR-NC transfected cells. These data indicated that MTDH is a direct target of miR-655 in OSCC cells.

miR-655 expression is inversely correlated with MTDH mRNA expression levels in OSCC tissues

To further explore the association between miR-655 and MTDH in OSCC, MTDH mRNA expression levels were determined in the 26 pairs of OSCC tissues and adjacent non-tumoral tissues. RT-qPCR analysis revealed that MTDH mRNA expression was significantly higher in OSCC tissues compared with the expression levels in the adjacent non-tumoral tissues (Fig. 4A; P<0.05). In addition, western blot analysis was performed to detect MTDH protein levels in OSCC tissues and adjacent non-tumor tissues, which demonstrated that the protein expression level of MTDH was notably higher in OSCC tissues compared to that in adjacent non-tumor tissues (Fig. 4B). Furthermore, the expression of miR-655 exhibited an inverse correlation with MTDH mRNA expression in OSCC tissues (Fig. 4C; r=−0.5949; P<0.05). These results further indicated that MTDH may be a novel target of miR-655 in OSCC.

Inhibiting MTDH is crucial for the inhibitory effects of miR-655 expression in OSCC cells

Rescue experiments were performed to further determine whether the suppressive roles of miR-655 were mediated through MTDH in OSCC cells. The MTDH overexpression vector pCMV-MTDH or the pCMV empty vector was transfected into Tca8113 and CAL-27 cells. RT-qPCR analysis was performed to confirm that MTDH mRNA expression was significantly increased in Tca8113 and CAL-27 cells that were transfected with pCMV-MTDH compared with pCMV empty vector-transfected cells (Fig. 5A; P<0.05). To perform rescue experiments, Tca8113 and CAL-27 cells were co-transfected with pCMV-MTDH or pCMV empty vector along with miR-655 mimics. Western blot analysis revealed that the MTDH protein expression that was decreased by miR-655 mimics was recovered in cells co-transfected with pCMV-MTDH (Fig. 5B; P<0.05). Subsequent MTT and cell invasion assays demonstrated that the restoration of MTDH expression reversed the suppressive effects of miR-655 overexpression on cell proliferation (Fig. 5C and D; P<0.05) and invasion (Fig. 5E; P<0.05). These results suggested that the tumor-suppressive roles of miR-655 in OSCC cells may be mediated, at least part, by the inhibition of MTDH expression.

miR-655 overexpression inactivates the PTEN/AKT pathway in OSCC cells

MTDH has previously been reported to contribute to the regulation of the PTEN/AKT signaling pathway (23,24). To examine whether miR-655 was able to affect the PTEN/AKT pathway, the protein expression levels of PTEN, p-AKT and AKT were detected in Tca8113 and CAL-27 cells co-transfected with miR-655 mimics and either pCMV-MTDH or pCMV empty vector. In miR-655 mimics + pCMV empty vector co-transfected cells, there was a notable increase in PTEN expression and a notable decrease in p-AKT expression compared with miR-NC transfected cells (Fig. 6); total AKT expression appeared to be unaffected. Conversely, co-transfection with pCMV-MTDH restored the reduced expression of PTEN and p-AKT caused by miR-655 overexpression. These results suggested that miR-655 may inhibit the activation of the PTEN/AKT signaling pathway in OSCC through the negative regulation of MTDH expression.

Discussion

Numerous studies have indicated that miRNAs are important regulators for a variety of biological processes, and their dysregulation is closely related to cancer formation and progression (3032). Therefore, the identification of aberrantly expressed miRNAs may provide important insight into the diagnosis and therapy of patients with OSCC. In the present study, miR-655 expression in OSCC was investigated as well as the roles and underlying mechanisms associated with miR-655 in the progression of OSCC. To the best of our knowledge, the present study is the first to demonstrate that miR-655 may have exhibited tumor suppressive roles in OSCC by directly targeting MTDH and regulating the PTEN/AKT signaling pathway, which suggested that miR-655 may represent an effective therapeutic agent in the treatment of patients with OSCC.

In the present study, miR-655 expression was demonstrated to be significantly lower in OSCC tissues and cell lines compared with expression levels in adjacent non-tumor tissues and NHOK cells. miR-655 has been previously reported to be dysregulated in several types of tumors. For example, miR-655 expressed in low levels in hepatocellular carcinoma tissues, and its expression levels were strongly correlated with tumor size, portal vein tumor thrombosis status, microvascular invasion, tumor-node-metastasis stage and lymph node metastasis (19,20). Patients with hepatocellular carcinoma with low miR-655 expression levels exhibited lower survival time compared with those patients with high miR-655 levels, and multivariate analysis identified miR-655 as an independent risk factor for patients with hepatocellular carcinoma (19). In triple-negative breast cancer, the expression of miR-655 was reduced in tumoral tissues and was significantly associated with lymph node metastasis (21). In esophageal squamous cell carcinoma, miR-655 expression was lower in tumoral tissues compared with expression in adjacent non-tumoral tissues and was negatively correlated with of lymph node metastases (22). These findings suggested that miR-655 expression may be frequently reduced in human cancers and should be investigated as a potential biomarker for the diagnosis and prognosis of patients with these specific cancer types.

The results from the present study further demonstrated that ectopic expression of miR-655 reduced the proliferative and invasive abilities of OSCC cells. Dysregulation of miR-655 expression was implicated in the occurrence and development of several types of human malignancies. For instance, overexpression of miR-655 inhibited cell growth and metastasis in hepatocellular carcinoma (20). Another study reported that upregulation of miR-655 decreased migration, invasion and epithelial-to-mesenchymal transition of triple-negative breast cancer cells (21). miR-655 overexpression was reported to restrict the migratory capacity of esophageal squamous cell carcinoma cells (22). It was also previously reported that miR-655 upregulation prohibited cell proliferation and motility, and promoted apoptosis in pituitary tumor cells (33). These findings suggested that miR-655 may be a novel and promising therapeutic candidate target for anticancer treatment.

Many direct targets of miR-655 have been validated, including ADAM metallopeptidase domain 10 in hepatocellular carcinoma (20), paired-related homeobox 1 in triple-negative breast cancer (21), pituitary tumor-transforming 1 in esophageal squamous cell carcinoma (22) and membrane-associated guanylate kinase, WW and PDZ domain-containing 2 in lung adenocarcinoma (34). In the present study, MTDH was identified as a direct target gene of miR-655 in OSCC. MTDH is located on chromosome 8q22 (35); it has been reported to be highly expressed in various types of human cancer, including glioma (36), breast cancer (37), gastric cancer (38), bladder cancer (39) and cervical cancer (40). In OSCC, both mRNA and protein levels of MTDH were overexpressed in tumor tissues and their overexpression was positively correlated with differentiation, clinical stage, T classification and lymph node metastasis. OSCC patients with high MTDH levels exhibited shorter overall survival rates relative to those patients with low MTDH expression (26,27). In addition, MTDH was previously confirmed as an independent prognostic factor for overall survival rates in OSCC patients (26,27). Highly expressed MTDH was reported to serve important roles in the onset and the progression of OSCC by affecting tumor cell growth, colony formation, migration and invasion (26,28,29). MTDH has previously been reported to be involved in the regulation of the PTEN/AKT signaling pathway (23,24). It has been well established that activation of the PTEN/AKT pathway contributes to OSCC development (41,42). In the present study, it was demonstrated that restoration of MTDH expression inactivated the PTEN/AKT pathway in OSCC cells via regulation of MTDH. These findings suggested that MTDH is an effective candidate for molecular targeted therapy for OSCC.

To the best of our knowledge, the present study is the first to demonstrate that miR-655 expression was reduced in OSCC tissues and cell lines. In addition, miR-655 overexpression suppressed cell proliferation and invasion in OSCC by directly targeting MTDH and regulating the PTEN/AKT pathway. These results may improve our understanding of OSCC pathogenesis and may also provide a theoretical basis for the identification of miR-655 as a potential tumor suppressor in OSCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

HJ and QW designed the research; QW, LL and YL performed functional experiments. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Yidu Central Hospital of Weifang (Weifang, China) and was performed in accordance with The Declaration of Helsinki and the guidelines of the Ethics Committee of Yidu Central Hospital of Weifang. Written informed consent was obtained from all patients for the use of their clinical tissues, prior to enrolment in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Min A, Zhu C, Peng S, Rajthala S, Costea DE and Sapkota D: MicroRNAs as important players and biomarkers in oral carcinogenesis. Biomed Res Int. 2015:1869042015. View Article : Google Scholar : PubMed/NCBI

2 

Warnakulasuriya S: Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 45:309–316. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Zaravinos A: An updated overview of HPV-associated head and neck carcinomas. Oncotarget. 5:3956–3969. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Leemans CR, Braakhuis BJ and Brakenhoff RH: The molecular biology of head and neck cancer. Nat Rev Cancer. 11:9–22. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Perez-Sayans M, Suarez-Penaranda JM, Padin-Iruegas ME, Gayoso-Diz P, Reis-De Almeida M, Barros-Angueira F, Gandara-Vila P, Blanco-Carrion A and Garcia-Garcia A: The loss of p16 expression worsens the prognosis of OSCC. Appl Immunohistochem Mol Morphol. 23:724–732. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Liang L, Zhang T, Kong Q, Liang J and Liao G: A meta-analysis on selective versus comprehensive neck dissection in oral squamous cell carcinoma patients with clinically node-positive neck. Oral Oncol. 51:1076–1081. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Moreno-Moya JM, Vilella F and Simon C: MicroRNA: Key gene expression regulators. Fertil Steril. 101:1516–1523. 2014. View Article : Google Scholar : PubMed/NCBI

8 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar : PubMed/NCBI

9 

Vasudevan S, Tong Y and Steitz JA: Switching from repression to activation: microRNAs can up-regulate translation. Science. 318:1931–1934. 2007. View Article : Google Scholar : PubMed/NCBI

10 

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

11 

Wang YJ, Zhang ZF, Fan SH, Zhuang J, Shan Q, Han XR, Wen X, Li MQ, Hu B, Sun CH, et al: MicroRNA-433 inhibits oral squamous cell carcinoma cells by targeting FAK. Oncotarget. 8:100227–100241. 2017.PubMed/NCBI

12 

Yang D, Du G, Xu A, Xi X and Li D: Expression of miR-149-3p inhibits proliferation, migration and invasion of bladder cancer by targeting S100A4. Am J Cancer Res. 7:2209–2219. 2017.PubMed/NCBI

13 

Zabaglia LM, Bartolomeu NC, Dos Santos MP, Peruquetti RL, Chen E, de Arruda Cardoso, Smith M, Payao SLM and Rasmussen LT: Decreased MicroRNA miR-181c expression associated with gastric cancer. J Gastrointest Cancer. 49:97–101. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Celano M, Rosignolo F, Maggisano V, Pecce V, Iannone M, Russo D and Bulotta S: MicroRNAs as biomarkers in thyroid carcinoma. Int J Genomics. 2017:64965702017. View Article : Google Scholar : PubMed/NCBI

15 

Kang M, Shi J, Peng N and He S: MicroRNA-211 promotes non-small-cell lung cancer proliferation and invasion by targeting MxA. Onco Targets Ther. 10:5667–5675. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD, Gerald WL and Massague J: Endogenous human microRNAs that suppress breast cancer metastasis. Nature. 451:147–152. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Cui F, Li X, Zhu X, Huang L, Huang Y, Mao C, Yan Q, Zhu J, Zhao W and Shi H: MiR-125b inhibits tumor growth and promotes apoptosis of cervical cancer cells by targeting phosphoinositide 3-kinase catalytic subunit delta. Cell Physiol Biochem. 30:1310–1318. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Zuo QF, Zhang R, Li BS, Zhao YL, Zhuang Y, Yu T, Gong L, Li S, Xiao B and Zou QM: MicroRNA-141 inhibits tumor growth and metastasis in gastric cancer by directly targeting transcriptional co-activator with PDZ-binding motif, TAZ. Cell Death Dis. 6:e16232015. View Article : Google Scholar : PubMed/NCBI

19 

Zhao XQ, Liang B, Jiang K and Zhang HY: Down-regulation of miR-655-3p predicts worse clinical outcome in patients suffering from hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 21:748–752. 2017.PubMed/NCBI

20 

Wu G, Zheng K, Xia S, Wang Y, Meng X, Qin X and Cheng Y: MicroRNA-655-3p functions as a tumor suppressor by regulating ADAM10 and β-catenin pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 35:892016. View Article : Google Scholar : PubMed/NCBI

21 

Lv ZD, Kong B, Liu XP, Jin LY, Dong Q, Li FN and Wang HB: miR-655 suppresses epithelial-to-mesenchymal transition by targeting Prrx1 in triple-negative breast cancer. J Cell Mol Med. 20:864–873. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Wang Y, Zang W, Du Y, Ma Y, Li M, Li P, Chen X, Wang T, Dong Z and Zhao G: Mir-655 up-regulation suppresses cell invasion by targeting pituitary tumor-transforming gene-1 in esophageal squamous cell carcinoma. J Transl Med. 11:3012013. View Article : Google Scholar : PubMed/NCBI

23 

Li J, Li C, Li H, Zhang T, Hao X, Chang J and Xu Y: MicroRNA30a5p suppresses tumor cell proliferation of human renal cancer via the MTDH/PTEN/AKT pathway. Int J Mol Med. 41:1021–1029. 2018.PubMed/NCBI

24 

Li L and Zhang H: MicroRNA-379 inhibits cell proliferation and invasion in glioma via targeting metadherin and regulating PTEN/AKT pathway. Mol Med Rep. 17:4049–4056. 2018.PubMed/NCBI

25 

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

26 

Xia X, Du R, Zhao L, Sun W and Wang X: Expression of AEG-1 and microvessel density correlates with metastasis and prognosis of oral squamous cell carcinoma. Hum Pathol. 45:858–865. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Seyedmajidi M, Sohanian S, Abbaszadeh H, Moslemi D and Bijani A: Astrocyte elevated gene 1 (AEG-1): A promising candidate for molecular targeted therapy in oral squamous cell carcinomas. Asian Pac J Cancer Prev. 18:3301–3305. 2017.PubMed/NCBI

28 

Wang Y, Wang T, Sun Y, Sun W and Wang X: Astrocyte elevated gene-1 promotes tumour growth and invasion by inducing EMT in oral squamous cell carcinoma. Sci Rep. 7:154472017. View Article : Google Scholar : PubMed/NCBI

29 

Wang YP, Liu IJ, Chiang CP and Wu HC: Astrocyte elevated gene-1 is associated with metastasis in head and neck squamous cell carcinoma through p65 phosphorylation and upregulation of MMP1. Mol Cancer. 12:1092013. View Article : Google Scholar : PubMed/NCBI

30 

Garzon R, Calin GA and Croce CM: MicroRNAs in Cancer. Annu Rev Med. 60:167–179. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Garzon R and Marcucci G: Potential of microRNAs for cancer diagnostics, prognostication and therapy. Curr Opin Oncol. 24:655–659. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Schickel R, Boyerinas B, Park SM and Peter ME: MicroRNAs: Key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene. 27:5959–5974. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Liang HQ, Wang RJ, Diao CF, Li JW, Su JL and Zhang S: The PTTG1-targeting miRNAs miR-329, miR-300, miR-381 and miR-655 inhibit pituitary tumor cell tumorigenesis and are involved in a p53/PTTG1 regulation feedback loop. Oncotarget. 6:29413–29427. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Kitamura K, Seike M, Okano T, Matsuda K, Miyanaga A, Mizutani H, Noro R, Minegishi Y, Kubota K and Gemma A: MiR-134/487b/655 cluster regulates TGF-beta-induced epithelial-mesenchymal transition and drug resistance to gefitinib by targeting MAGI2 in lung adenocarcinoma cells. Mol Cancer Ther. 13:444–453. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Su ZZ, Kang DC, Chen Y, Pekarskaya O, Chao W, Volsky DJ and Fisher PB: Identification and cloning of human astrocyte genes displaying elevated expression after infection with HIV-1 or exposure to HIV-1 envelope glycoprotein by rapid subtraction hybridization, RaSH. Oncogene. 21:3592–3602. 2002. View Article : Google Scholar : PubMed/NCBI

36 

He Z, He M, Wang C, Xu B, Tong L, He J, Sun B, Wei L and Chu M: Prognostic significance of astrocyte elevated gene-1 in human astrocytomas. Int J Clin Exp Pathol. 7:5038–5044. 2014.PubMed/NCBI

37 

Li J, Zhang N, Song LB, Liao WT, Jiang LL, Gong LY, Wu J, Yuan J, Zhang HZ, Zeng MS and Li M: Astrocyte elevated gene-1 is a novel prognostic marker for breast cancer progression and overall patient survival. Clin Cancer Res. 14:3319–3326. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Dong L, Qin S, Li Y, Zhao L, Dong S, Wang Y, Zhang C and Han S: High expression of astrocyte elevated gene-1 is associated with clinical staging, metastasis and unfavorable prognosis in gastric carcinoma. Tumour Biol. 36:2169–2178. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Nikpour M, Emadi-Baygi M, Fischer U, Niegisch G, Schulz WA and Nikpour P: MTDH/AEG-1 contributes to central features of the neoplastic phenotype in bladder cancer. Urol Oncol. 32:670–677. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Yu JQ, Zhou Q, Zhu H, Zheng FY and Chen ZW: Overexpression of astrocyte elevated gene-1 (AEG-1) in cervical cancer and its correlation with angiogenesis. Asian Pac J Cancer Prev. 16:2277–2281. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Alyasiri NS, Mehdi SJ, Alam MS, Ali A, Mandal AK, Gupta S, Singh I and Rizvi MM: PTEN-mediated AKT activation contributes to the reduced apoptosis among Indian oral squamous cell carcinoma patients. J Cancer Res Clin Oncol. 138:103–109. 2012. View Article : Google Scholar : PubMed/NCBI

42 

Gan YH and Zhang S: PTEN/AKT pathway involved in histone deacetylases inhibitor induced cell growth inhibition and apoptosis of oral squamous cell carcinoma cells. Oral Oncol. 45:e150–e154. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 18 Issue 3

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
Wang Q, Lv L, Li Y and Ji H: MicroRNA‑655 suppresses cell proliferation and invasion in oral squamous cell carcinoma by directly targeting metadherin and regulating the PTEN/AKT pathway Retraction in /10.3892/mmr.2022.12897. Mol Med Rep 18: 3106-3114, 2018
APA
Wang, Q., Lv, L., Li, Y., & Ji, H. (2018). MicroRNA‑655 suppresses cell proliferation and invasion in oral squamous cell carcinoma by directly targeting metadherin and regulating the PTEN/AKT pathway Retraction in /10.3892/mmr.2022.12897. Molecular Medicine Reports, 18, 3106-3114. https://doi.org/10.3892/mmr.2018.9292
MLA
Wang, Q., Lv, L., Li, Y., Ji, H."MicroRNA‑655 suppresses cell proliferation and invasion in oral squamous cell carcinoma by directly targeting metadherin and regulating the PTEN/AKT pathway Retraction in /10.3892/mmr.2022.12897". Molecular Medicine Reports 18.3 (2018): 3106-3114.
Chicago
Wang, Q., Lv, L., Li, Y., Ji, H."MicroRNA‑655 suppresses cell proliferation and invasion in oral squamous cell carcinoma by directly targeting metadherin and regulating the PTEN/AKT pathway Retraction in /10.3892/mmr.2022.12897". Molecular Medicine Reports 18, no. 3 (2018): 3106-3114. https://doi.org/10.3892/mmr.2018.9292