MicroRNA‑876‑5p inhibits the progression of glioblastoma multiforme by directly targeting Forkhead box M1

Retraction in: /10.3892/or.2022.8360

  • Authors:
    • Lina Wang
    • Jia Lu
    • Hong Zhang
    • Xueman Lyu
    • Zhigang Sun
  • View Affiliations

  • Published online on: October 17, 2018     https://doi.org/10.3892/or.2018.6804
  • Pages: 702-710
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Considerable evidence has suggested that microRNAs (miRNAs) are dysregulated in glioblastoma multiforme (GBM), and their dysregulation may modulate the aggressiveness of GBM. Therefore, miRNAs with dysregulated expression are potential therapeutic targets for the treatment of GBM. miRNA‑876‑5p (miR‑876‑5p) has recently been identified to be aberrantly expressed and serve an important role in hepatocellular carcinoma and lung cancer. However, its expression pattern and functional significance in GBM remains largely unknown. Therefore, the present study detected miR‑876‑5p expression in GBM, examined the biological roles of miR‑876‑5p in GBM progression and explored its underlying mechanism. The present study demonstrated that miR‑876‑5p expression was significantly downregulated in GBM tissues and cell lines. Overexpression of miR‑876‑5p restricted the proliferation, induced the apoptosis and reduced the migration and invasion capabilities of GBM cells. In GBM cells, Forkhead box M1 (FOXM1) was identified as a direct target of miR‑876‑5p. FOXM1 was overexpressed in clinical GBM tissues, and its overexpression was inversely correlated with miR‑876‑5p level. Small interfering RNA‑mediated knockdown of FOXM1 exhibited effects similar to those of miR‑876‑5p overexpression in GBM cells. The tumour suppressive roles of miR‑876‑5p overexpression in GBM cells were significantly reversed by FOXM1 reintroduction. Overall, the present results revealed that miR‑876‑5p may inhibit the development of GBM by directly targeting FOXM1, suggesting that this miRNA may be a potential therapeutic target in patients, for the management of GBM.

Introduction

Glioma, which originates from neural epithelium, is the most common primary brain tumour in adults (1). On the basis of malignancy, gliomas are classified into four histopathologic grades, namely, WHO grades I–IV (2). Glioblastoma multiforme (GBM), a WHO grade IV glioma, represents ~70% of all glioma cases (3). Surgical resection followed by postoperative chemotherapy and radiotherapy is currently the primary therapeutic technique used to treat GBM patients (4). Although significant advancement has been achieved in GBM diagnosis and therapy, the treatment outcome of GBM patients remains unsatisfactory, with an estimated 5-year survival rate of <3% (5). The aggressive characteristics of GBM, including rapid growth and strong invasiveness, are the primary causes of poor prognosis (6). Activation of oncogenes, inactivation of tumour suppressor genes and chromosomal abnormalities are implicated in the oncogenesis and development of GBM; however, the detailed mechanisms underlying GBM formation and progression remain to be elucidated (7). Therefore, further studies on the mechanisms underlying the tumourigenicity and development of GBM are essential, and may facilitate the identification of novel diagnostic and therapeutic strategies for patients carrying this malignant tumour.

MicroRNAs (miRNAs) are a series of endogenous and noncoding short RNAs consisting of ~18-4 nucleotides (8). miRNAs target the 3′-untranslated regions (3′-UTRs) of their target genes in a sequence-specific manner to inhibit translation and/or cause degradation of messenger RNAs (mRNAs) (9). According to miRBase (www.mirbase.org/index.shtml, Release 21), 1881 precursor and 2588 mature miRNAs have been identified in the human genome. These mature miRNAs may modulate the expression of ~30% of all human protein-coding genes (10). In recent years, an expanding number of studies have documented that miRNAs are differentially expressed in many malignancies, including GBM (1113). Highly expressed miRNAs in GBM may play oncogenic roles through negative regulation of tumour suppressor genes (14). Conversely, decreased expression of various miRNAs may serve tumour suppressive roles in GBM progression by directly targeting and inhibiting oncogenes (15). Functional studies have revealed that miRNA dysregulation is involved in GBM progression and regulates various pathological processes, including cell proliferation, cycle, apoptosis, epithelial-mesenchymal transition, metastasis and angiogenesis (1618). Hence, investigation on the roles of dysregulated miRNAs in GBM is critical, and may lead to the possible development of promising therapeutic agents for GBM treatment.

miR-876-5p has previously been identified to be aberrantly expressed and play an important role in hepatocellular carcinoma (19,20) and lung cancer (21). However, its expression pattern and functional significance in GBM remains largely unknown. Therefore, the present study detected miR-876-5p expression in GBM, examined the biological roles of miR-876-5p in GBM progression and explored its underlying mechanism. FOXM1, a member of the Forkhead superfamily of transcription factors, was predicted as a putative target of miR-876-5p. FOXM1 is upregulated in numerous types of human cancer, such as osteosarcoma (22), hepatocellular carcinoma (23), pancreatic (24), colorectal (25) and breast cancer (26). FOXM1 serves oncogenic roles and is implicated in the regulation of various biological behaviors, including cell proliferation, cell cycle statue, DNA damage repair, tissue homeostasis, angiogenesis and metastasis (27,28). Due to its previously demonstrated crucial role in tumorigenesis and tumor development, FOXM1 was selected for further experimental verification of its role in the mediation of biological functions of miR-876-5p in GBM cells.

Materials and methods

Collection of tissue samples

GBM and adjacent normal tissues were collected from 26 patients (15 males and 11 females; age range: 37–65 years old) who were newly diagnosed as GBM and treated with surgical resection in China-Japan Union Hospital of Jilin University (Changchun, China) between July 2014 and May 2017. None of these patients received radiotherapy or chemotherapy prior to surgery. Tissue samples were collected immediately after surgical resection, frozen in liquid nitrogen and stored at −80°C until further use. The present study was approved by the Ethics Committee of China-Japan Union Hospital of Jilin University. Written informed consent was obtained from all enrolled patients prior to surgery.

Cell lines

A total of four human GBM cell lines, including U138, U251, T98, and LN229, were purchased from Shanghai Cell Bank of the Chinese Academy of Sciences (Shanghai, China), and were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 mg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Normal human astrocytes (NHAs) were obtained from ScienCell Research Laboratories (Carlsbad, CA, USA), and were cultured in astrocyte medium (Sciencell Research Laboratories) supplemented with 10% FBS. All cells were grown at 37°C under normoxic conditions of 95% air and 5% CO2.

Transfection

Cells were plated into 6-well plates one day prior to transfection. Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) was used in cell transfection according to the manufacturer's protocol. miR-876-5p mimics, miRNA mimics negative control (miR-NC), small interfering RNA (siRNA) targeting FOXM1 (FOXM1 siRNA) and negative control siRNA (NC siRNA) were purchased from Shanghai GenePharma Co., Ltd. (Shanghai, China). The miR-876-5p mimics sequence was 5′-UGGAUUUCUUUGUGAAUCACCA-3′ and the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. The FOXM1 siRNA sequence was 5′-GGACCACUUUCCCUACUUUTT-3′ and the NC siRNA sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. The FOXM1 overexpression plasmid pCMV-FOXM1 and empty pCMV plasmid were constructed by the Chinese Academy of Sciences (Changchun, China). Cells were transfected with miRNA mimics (100 pmol), siRNA (100 pmol) or plasmids (4 µg), and transfected cells were incubated at 37°C with 5% CO2 and then subjected to the evaluation of transfection efficiency. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis was used to assess the transfection efficiency of miR-876-5p mimics. The efficiencies of siRNA and plasmid transfection were determined through western blot analysis. Forty-eight hours after transfection, RT-qPCR analysis was performed. Cell Counting Kit-8 (CCK-8) assay and flow cytometry assay were carried out at 24 and 48 h post-transfection, respectively. In vitro migration and cell invasion assays were conducted 48 h following transfection. After 72 h incubation, western blot analysis was performed to determine the FOXM1 protein expression.

RT-qPCR

Total RNA was isolated from tissue specimens or cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. For the detection of miR-876-5p expression, total RNA was used for complementary DNA (cDNA) synthesis using a TaqMan MicroRNA Reverse Transcription Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). The temperature protocol for reverse transcription was as follows: 16°C for 30 min, 42°C for 30 min and 85°C for 5 min. Quantitative polymerase chain reaction (qPCR) was conducted using a TaqMan MicroRNA PCR kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). The cycling conditions for qPCR were as follows: 50°C for 2 min, 95°C for 10 min; 40 cycles of denaturation at 95°C for 15 sec; and annealing/extension at 60°C for 60 sec. For FOXM1 mRNA expression determination, reverse transcription was performed using a Prime-Script RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). The temperature protocol for reverse transcription was as follows: 37°C for 15 min and 85°C for 5 sec. The synthesized cDNA was subjected to qPCR using a SYBR Premix Ex Taq™ Kit (Takara Biotechnology Co., Ltd.). The cycling conditions 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. miR-876-5p and FOXM1 expression levels were normalized with reference to U6 snRNA and GAPDH, respectively. The primers were designed as follows: Forward, 5′-AGGACUUCUCCCUCCUCCCAG-3′ and reverse, 5′-UCCUCUUCUCCCUCCAGGGAG-3′ for miR-876-5p; forward, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ and reverse, 5′-CGCTTCACGAATTTGCGTGTCAT-3′ for U6; forward, 5′-GAAGAACTCCATCCGCCACA-3′ and reverse, 5′-GCCTTAAACACCTGGTCCAATGTC-3′ for FOXM1 and forward and 5′-TGGATTTGGACGCATTGGTC-3′ and reverse 5′-TTTGCACTGGTACGTGTTGATA-3′ for GAPDH. Relative gene expression levels were analysed using the 2−ΔΔCq method (29).

CCK-8 assay

Cells were transfected and cultured at 37°C with 5% CO2 for 24 h. Following the culture period, transfected cells were collected and plated into 96-well plates at an initial density of 3×103 cells/well. A CCK-8 assay was performed to detect cell proliferation at different time points (0, 1, 2 and 3 days). A tostal of 10 µl CCK-8 solution (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added into each well of the plate, and then the cells were incubated at 37°C for a further 2 h. The absorbance of each well was detected at a wavelength of 450 nm (A450) by a microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Flow cytometry assay

An Annexin V fluorescein isothiocyanate (FITC) apoptosis detection kit (Biolegend, San Diego, CA, USA) was utilized to assess cell apoptosis rate. In brief, transfected cells were incubated at 37°C under 5% CO2 for 48 h. Subsequently, the transfected cells were harvested, washed twice with phosphate-buffered saline (PBS) and suspended in 100 µl binding buffer. Following this, the transfected cells were incubated with Annexin V-FITC (5 µl) and propidium iodide (5 µl) at room temperature in the dark. Following incubation for 15 min, a flow cytometer (FACScan; BD Biosciences, Franklin Lakes, NJ, USA) was used to acquire data on cell apoptosis rate. The data was analysed with CellQuest version 5.1 (BD Biosciences).

In vitro migration and cell invasion assays

In vitro migration and cell invasion assays were performed to detect cell migration and invasion using 24-well Transwell chambers coated without or with Matrigel (both from BD Biosciences), respectively. Cells were collected 48 h after transfection and suspended in FBS-free DMEM. The upper compartments of the Transwell chambers were loaded with 200 µl of cell suspension containing 1×105 transfected cells. DMEM (500 µl) supplemented with 20% FBS was placed in the lower compartments. After 24 h of incubation, cells remaining on the upper surface were removed using a cotton swab, whereas the migrated and invaded cells were fixed in 100% methanol at room temperature for 15 min, stained with 0.1% crystal violet at room temperature for 15 min, washed with PBS and air-dried. Migration and invasion capacities were quantified by counting the number of migrated and invaded cells in five randomly selected microscopic fields seen under an inverted microscope (×200 magnification; IX83; Olympus Corp., Tokyo, Japan).

Bioinformatics analysis

The putative targets of miR-876-5p were predicted using TargetScan (www.targetscan.org), microRNA.org (www.microrna.org) and miRDB (www.mirdb.org). FOXM1 was predicted as a major target of miR-876-5p, and was selected for further verification experiments.

Luciferase reporter assay

The 3′-UTR fragments of FOXM1 containing the wild-type (Wt) or mutant (Mut) miR-876-5p targeting sequences were chemically produced by Shanghai GenePharma Co., Ltd., and inserted into the pmirGLO luciferase reporter vector (Promega Corp., Madison, WI, USA). The constructed luciferase plasmids were defined as pmirGLO-Wt-FOXM1-3′-UTR and pmirGLO-Mut-FOXM1-3′-UTR, respectively. For reporter assays, cells were inoculated into 24-well plates at a density of 1×105 cells/well. After overnight incubation, miR-876-5p mimics or miR-NC were transfected into cells containing pmirGLO-Wt-FOXM1-3′-UTR or pmirGLO-Mut-FOXM1-3′-UTR with Lipofectamine 2000, according to the manufacturer's protocol. Subsequent to 48 h incubation, transfected cells were harvested and the luciferase activity was analysed using the Dual-Luciferase Reporter Assay System (Promega Corp.). Renilla luciferase activity was used for normalization.

Western blot analysis

Cells or homogenized tissues were lysed using a Total Protein Extraction Kit (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China). Protein concentration was measured by a BCA protein assay kit (Pierce; Thermo Fisher Scientific, Inc.). The same amounts of proteins (30 µg) were subjected to 10% SDS-PAGE gel electrophoresis and then transferred onto polyvinylidene difluoride membranes (Thermo Fisher Scientific, Inc.). The membranes were subsequently blocked with 5% fat-free milk diluted in Tris-buffered saline containing 0.1% Tween-20 (TBST) for 2 h at room temperature and incubated overnight at 4°C with primary antibodies. Following washing with TBST three times, the membranes were further immersed in goat anti-mouse horseradish peroxidase-conjugated IgG secondary antibody (dilution 1:5,000; cat. no. ab205719; Abcam, Cambridge, UK) at room temperature for 2 h. Subsequent to three washes with TBST, chemiluminescence detection was conducted using an ECL Protein Detection Kit (Pierce; Thermo Fisher Scientific, Inc.). Primary antibodies used in the present study included mouse anti-human monoclonal FOXM1 (dilution 1:500; cat. no. sc-271746; Santa Cruz Biotechnology Inc., Dallas, TX, USA) and mouse anti-human monoclonal GAPDH (dilution 1:500; cat. no. ab8245; Abcam). GAPDH was used as endogenous control. Protein expression was quantified using Quantity One software version 4.62 (BioRad Laboratories, Inc., Hercules, CA, USA).

Statistical analysis

Data are presented as the mean ± standard deviation of three independent experiments. Differences between groups were determined using two-tailed Student's t-test or one-way analysis of variance (ANOVA) for multiple comparisons. The Student-Newman-Keuls method was used as a post hoc test following ANOVA. The association between miR-876-5p and FOXM1 mRNA levels in GBM tissues was assessed using Spearman's correlation analysis. SPSS software, version 16.0 (SPSS, Inc., Chicago, IL, USA) was used for statistical analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-876-5p level decreases in GBM tissues and cell lines

To illustrate the potential relevance of miR-876-5p in GBM, the present study first detected miR-876-5p expression in GBM and adjacent normal tissues obtained from 26 patients. RT-qPCR data demonstrated that miR-876-5p expression was significantly downregulated in GBM tissues compared with adjacent normal tissues (Fig. 1A; P<0.05). In addition, the expression levels of miR-876-5p in four GBM cell lines (U138, U251, T98 and LN229) and normal human astrocytes (NHAs) were examined. The miR-876-5p expression in all four tested GBM cell lines was decreased compared with in NHAs (Fig. 1B; P<0.05). U251 and T98 cell lines exhibited relatively lower miR-876-5p expression compared with the two other GBM cell lines; hence, U251 and T98 cell lines were selected for subsequent experiments. These observations suggested that decreased expression of miR-876-5p was associated with the development and progression of GBM.

miR-876-5p overexpression inhibits proliferation and induces apoptosis of GBM cells

To investigate the functional role of miR-876-5p in GBM, the present study transfected U251 and T98 cells with miR-876-5p mimics that resulted in increased endogenous miR-876-5p expression (Fig. 2A; P<0.05). The impact of miR-876-5p overexpression on GBM cell proliferation was determined using a CCK-8 assay. The results indicated that transfection of miR-876-5p mimics significantly reduced the proliferation of U251 and T98 cells (Fig. 2B; P<0.05). A flow cytometry assay was performed to measure the apoptosis rate of U251 and T98 cells treated with miR-876-5p mimics or miR-NC. miR-876-5p overexpression significantly increased the apoptosis rate of U251 and T98 cells compared with miR-NC groups (Fig. 2C; P<0.05). These results suggested that miR-876-5p served a tumour suppressive role in GBM cell proliferation and apoptosis.

miR-876-5p restricts the migration and invasion of GBM cells

In vitro migration and invasion assays were performed to further examine the potential role of miR-876-5p in metastasis of GBM cells. As presented in Fig. 3A, the migratory ability of the miR-876-5p mimics-transfected U251 and T98 cells was significantly suppressed compared with miR-NC-transfected cells (P<0.05). Additionally, overexpression of miR-876-5p expression resulted in the reduced number of invaded U251 and T98 cells compared with miR-NC groups (Fig. 3B; P<0.05). Taken together, miR-876-5p overexpression inhibited the metastasis of GBM.

FOXM1 is a direct target gene of miR-876-5p in GBM cells

To elucidate the mechanism of miR-876-5p activity in GBM, the present study employed bioinformatics analysis to predict the putative targets of miR-876-5p. It was demonstrated that the 3′-UTR of FOXM1 matched the seed sequences of miR-876-5p (Fig. 4A). FOXM1 has been well documented to play crucial roles in GBM progression (3034) and thus was selected for further verification. To determine whether miR-876-5p could directly target the 3′-UTR of FOXM1, luciferase reporter plasmids were constructed and were transfected into U251 and T98 cells containing miR-876-5p mimics or miR-NC. The results of the luciferase reporter assay indicated that upregulation of miR-876-5p significantly reduced the luciferase activity of the plasmid carrying the wild type (Wt) 3′-UTR of FOXM1 in U251 and T98 cells (P<0.05). Conversely, the luciferase activity was unaltered in cells transfected with plasmid harbouring the mutant (Mut) 3′-UTR of FOXM1 (Fig. 4B).

To further explore the association between miR-876-5p and FOXM1 in GBM, the present study measured FOXM1 expression levels in 26 pairs of GBM tissues and adjacent normal tissues. RT-qPCR analysis revealed that FOXM1 expression in GBM tissues was significantly upregulated compared with adjacent normal tissues (Fig. 4C; P<0.05). In addition, a negative correlation between miR-876-5p and FOXM1 mRNA levels was identified in GBM specimens (Fig. 4D; r=−0.5589, P=0.0030). Furthermore, enforced miR-876-5p expression suppressed FOXM1 expression in U251 and T98 cells at the mRNA (Fig. 4E; P<0.05) and protein (Fig. 4F; P<0.05) levels. The results collectively suggested that FOXM1 was a direct target gene of miR-876-5p in GBM cells.

Suppression of FOXM1 imitates the effects of miR-876-5p overexpression in the malignant phenotype of GBM cells

FOXM1 was confirmed to be a direct target of miR-876-5p in GBM cells; hence, the authors hypothesized that inhibition of FOXM1 could imitate the suppressive roles of miR-876-5p in GBM cells. To confirm this hypothesis, U251 and T98 cells were transfected with NC siRNA or FOXM1 siRNA, and western blot analysis was performed in order to validate the transfection efficiency. As expected, FOXM1 protein was efficiently knocked down in U251 and T98 cells treated with FOXM1 siRNA (Fig. 5A; P<0.05). Functional experiments revealed that siRNA-mediated knockdown of FOXM1 inhibited the proliferation (Fig. 5B; P<0.05) and promoted the apoptosis (Fig. 5C; P<0.05) of U251 and T98 cells. In vitro migration and invasion assays revealed that FOXM1 suppression remarkably restricted both the migration (Fig. 5D; P<0.05) and invasion (Fig. 5E; P<0.05) capabilities of U251 and T98 cells. These results demonstrated that FOXM1 downregulation could mimic the tumour suppressive role of miR-876-5p in GBM cells, further suggesting that FOXM1 is a functional downstream target of miR-876-5p in GBM.

FOXM1 reintroduction abrogates the effects of miR-876-5p overexpression in GBM cells

Rescue experiments were conducted to determine whether FOXM1 mediates the tumour suppressive role of miR-876-5p in GBM cells. The present study co-transfected miR-876-5p mimics with empty pCMV or pCMV-FOXM1 plasmid lacking a 3′-UTR into U251 and T98 cells; subsequently, western blot analysis was performed to detect FOXM1 protein expression. As presented in Fig. 6A, the downregulated FOXM1 protein expression caused by miR-876-5p overexpression was restored in U251 and T98 cells following co-transfection with pCMV-FOXM1 (P<0.05). Furthermore, CCK-8 assays revealed that restoration of FOXM1 expression partially rescued the miR-876-5p-mediated suppression of cell proliferation (Fig. 6B; P<0.05). Similarly, the flow cytometry assay revealed that while miR-876-5p promoted apoptosis of U251 and T98 cells, this effect was abolished by forced FOXM1 expression (Fig. 6C; P<0.05). Furthermore, FOXM1 upregulation partially abrogated the suppressive effects of miR-876-5p overexpression on migration (Fig. 6D; P<0.05) and invasion (Fig. 6E; P<0.05) of U251 and T98 cells. These results collectively demonstrated that miR-876-5p may inhibit the progression of GBM, at least partly through downregulation of FOXM1 expression.

Discussion

Considerable evidence has suggested that miRNAs are dysregulated in GBM, and their dysregulation may modulate the aggressiveness of GBM (3537). Thus, miRNAs with dysregulated expression are potential therapeutic targets against GBM. In the present study, the expression levels of miR-876-5p were lower in GBM tissues and cell lines compared with adjacent normal tissues and NHAs. Upregulation of miR-876-5p repressed GBM cell proliferation, increased cell apoptosis and decreased cell migration and invasion in vitro. In addition, FOXM1 was validated as a direct target of miR-876-5p in GBM cells. These results suggested that miR-876-5p serves as a tumour suppressor in GBM by directly targeting FOXM1. Investigations regarding the expression and role of miR-876-5p have provided important information that strengthens understanding of GBM oncogenesis and progression, which may promote the development of novel therapeutic strategies.

miR-876-5p is aberrantly expressed and contributes to the malignant phenotype of several types of human cancer. For example, miR-876-5p is downregulated in hepatocellular carcinoma tissues and cell lines. Reduced miR-876-5p expression is significantly associated with venous infiltration, high tumour grade and advanced tumour stage. Hepatocellular carcinoma patients with low miR-876-5p expression have poorer overall and disease-free survival rates compared with those with high miR-876-5p expression levels (19). Functional assays have revealed that miR-876-5p re-expression represses cell proliferation, migration, invasion and epithelial-mesenchymal transition in hepatocellular carcinoma (19,20). Furthermore, miR-876-5p expression is reduced in lung cancer tissues and cell lines. Resumption of miR-876-5p expression attenuates cell metastasis and epithelial-mesenchymal transition of lung cancer in vitro and reduces tumour metastasis in vivo (21). These findings suggest that re-expression of miR-876-5p is a promising therapeutic method to manage symptoms in patients with these malignancies.

Several genes, including B cell lymphoma-6 corepressor-like 1 (19), DNA methyltransferase 3 α (20) and bone morphogenetic protein 4 (21), have been validated as direct targets of miR-876-5p. To illustrate the mechanisms underlying the cellular response to miR-876-5p, the present study determined whether FOXM1 is a direct target gene of miR-876-5p in GBM. Firstly, bioinformatics analysis predicted that the 3′-UTR of FOXM1 matches the seed sequence of miR-876-5p. Secondly, luciferase reporter assays revealed that miR-876-5p could directly target the 3′-UTR of FOXM1 in GBM cells. Thirdly, the results of RT-qPCR and western blot analysis demonstrated that miR-876-5p upregulation decreased FOXM1 expression in GBM cells at the mRNA and protein levels. Furthermore, FOXM1 was upregulated in GBM tissues, and its upregulation was inversely correlated with miR-876-5p levels. Inhibition of FOXM1 exerted effects similar to those of miR-876-5p overexpression on GBM cells. FOXM1 restoration partially abolished the suppressive effects of miR-876-5p overexpression on GBM cells. Identification of cancer-associated miRNAs and their target genes is crucial in clarifying the roles of miRNA in tumour initiation and progression and may lead to the development of effective therapeutic strategies.

In GBM, FOXM1 is also overexpressed in tumour tissues and cell lines. Considerable evidence supports the importance of FOXM1 in the GBM genesis and development through regulation of multiple biological behaviours, including cell proliferation, cell cycle, apoptosis, metastasis, epithelial to mesenchymal transition, chemosensitivity and angiogenesis (3034). Previously, accumulating studies reported that FOXM1 may be regulated by multiple miRNAs in various cancers, such as miR-197 in cervical cancer (38), miR-761 in colorectal cancer (25), miR-320 in glioma, and miR-630 in gastric cancer (39). In the present study, for the first time to the best of the authors knowledge, it was demonstrated that FOXM1 was negatively regulated by miR-876-5p in GBM, and therefore inhibited the progression and development of GBM. Therefore, the use of miR876-5p-based therapy targeting FOXM1 expression may be a potential method for GBM treatment, to block rapid growth and metastasis.

In conclusion, the results of the present study demonstrated that miR-876-5p was downregulated in GBM tissues and cell lines. miR-876-5p restoration impeded the development of GBM by directly targeting FOXM1. These results may provide a novel insight into the carcinogenesis and progression of GBM and serve as a basis in identification of miRNA-based targeted therapies against GBM.

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

XL and ZS designed the study and carried out bioinformatics analysis and statistical analysis. LW conducted transfection, CCK-8 and flow cytometry assays. JL and HZ performed RT-qPCR, in vitro migration and cell invasion assays, western blot analysis and luciferase reporter assay. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of China-Japan Union Hospital of Jilin University, and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of China-Japan Union Hospital of Jilin University. Written informed consent was obtained from all patients for the use of their clinical tissues.

Patient consent for publication

Written informed consent was obtained.

Competing interests

The authors declare that they have no competing interests.

References

1 

Caldarella A and Barchielli A: Glioblastoma in the canton of Zurich, Switzerland revisited: 2005 to 2009. Cancer. 122:37402016. View Article : Google Scholar : PubMed/NCBI

2 

Komori T, Sasaki H and Yoshida K: Revised WHO classification of tumours of the central nervous system: Summary of the revision and perspective. No Shinkei Geka. 44:625–635. 2016.(In Japanese). PubMed/NCBI

3 

Ohgaki H and Kleihues P: Genetic pathways to primary and secondary glioblastoma. Am J Pathol. 170:1445–1453. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Rigamonti A, Imbesi F, Silvani A, Grimod G, Prone V, Gaviani P, Ardizzoia A, Milanesi I, Arienti V, Agostoni E, et al: Pattern of care and outcome in elderly patients with glioblastoma: Data in 151 patients from 3 Lombardia Hospitals. J Neurol Sci. 378:3–8. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Komotar RJ, Otten ML, Moise G and Connolly ES Jr: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma-A critical review. Clin Med Oncol. 2:421–422. 2008.PubMed/NCBI

6 

Khosla D: Concurrent therapy to enhance radiotherapeutic outcomes in glioblastoma. Ann Transl Med. 4:542016.PubMed/NCBI

7 

Marumoto T and Saya H: Molecular biology of glioma. Adv Exp Med Biol. 746:2–11. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Griffiths-Jones S, Grocock RJ, van Dongen S, Bateman A and Enright AJ: miRBase: microRNA sequences, targets and gene nomenclature. Nucleic Acids Res. 34:D140–D144. 2006. View Article : Google Scholar : PubMed/NCBI

9 

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

10 

Chen K and Rajewsky N: The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. 8:93–103. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Piwecka M, Rolle K, Belter A, Barciszewska AM, Żywicki M, Michalak M, Nowak S, Naskręt-Barciszewska MZ and Barciszewski J: Comprehensive analysis of microRNA expression profile in malignant glioma tissues. Mol Oncol. 9:1324–1340. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Hayes J, Peruzzi PP and Lawler S: MicroRNAs in cancer: Biomarkers, functions and therapy. Trends Mol Med. 20:460–469. 2014. View Article : Google Scholar : PubMed/NCBI

13 

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

14 

Gu JJ, Fan KC, Zhang JH, Chen HJ and Wang SS: Suppression of microRNA-130b inhibits glioma cell proliferation and invasion, and induces apoptosis by PTEN/AKT signaling. Int J Mol Med. 41:284–292. 2018.PubMed/NCBI

15 

Gomez Zubieta DM, Hamood MA, Beydoun R, Pall AE and Kondapalli KC: MicroRNA-135a regulates NHE9 to inhibit proliferation and migration of glioblastoma cells. Cell Commun Signal. 15:552017. View Article : Google Scholar : PubMed/NCBI

16 

Cheng ZX, Song YX, Wang ZY, Wang Y and Dong Y: miR-144-3p serves as a tumor suppressor by targeting FZD7 and predicts the prognosis of human glioblastoma. Eur Rev Med Pharmacol Sci. 21:4079–4086. 2017.PubMed/NCBI

17 

Xu X, Cai N, Zhi T, Bao Z, Wang D, Liu Y, Jiang K, Fan L, Ji J and Liu N: MicroRNA-1179 inhibits glioblastoma cell proliferation and cell cycle progression via directly targeting E2F transcription factor 5. Am J Cancer Res. 7:1680–1692. 2017.PubMed/NCBI

18 

Sun J, Zheng G, Gu Z and Guo Z: MiR-137 inhibits proliferation and angiogenesis of human glioblastoma cells by targeting EZH2. J Neurooncol. 122:481–489. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Xu Q, Zhu Q, Zhou Z, Wang Y, Liu X, Yin G, Tong X and Tu K: MicroRNA-876-5p inhibits epithelial-mesenchymal transition and metastasis of hepatocellular carcinoma by targeting BCL6 corepressor like 1. Biomed Pharmacother. 103:645–652. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Wang Y, Xie Y, Li X, Lin J, Zhang S, Li Z, Huo L and Gong R: MiR-876-5p acts as an inhibitor in hepatocellular carcinoma progression by targeting DNMT3A. Pathol Res Pract. 214:2024–2030. 2018. View Article : Google Scholar

21 

Bao L, Lv L, Feng J, Chen Y, Wang X, Han S and Zhao H: MiR-876-5p suppresses epithelial-mesenchymal transition of lung cancer by directly down-regulating bone morphogenetic protein 4. J Biosci. 42:671–681. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Duan N, Hu X, Yang X, Cheng H and Zhang W: MicroRNA-370 directly targets FOXM1 to inhibit cell growth and metastasis in osteosarcoma cells. Int J Clin Exp Pathol. 8:10250–10260. 2015.PubMed/NCBI

23 

Song BN and Chu IS: A gene expression signature of FOXM1 predicts the prognosis of hepatocellular carcinoma. Exp Mol Med. 50:e4182018. View Article : Google Scholar : PubMed/NCBI

24 

Huang C, Du J and Xie K: FOX M1 and its oncogenic signaling in pancreatic cancer pathogenesis. Biochim Biophys Acta. 1845:104–116. 2014.PubMed/NCBI

25 

Cao S, Lin L, Xia X and Wu H: MicroRNA-761 promotes the sensitivity of colorectal cancer cells to 5-Fluorouracil through targeting FOXM1. Oncotarget. 9:321–331. 2017.PubMed/NCBI

26 

Siraj AK, Pratheeshkumar P, Parvathareddy SK, Qadri Z, Thangavel S, Ahmed S, Al-Dayel F, Tulbah A, Ajarim D and Al-Kuraya KS: FoxM1 is an independent poor prognostic marker and therapeutic target for advanced Middle Eastern breast cancer. Oncotarget. 9:17466–17482. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Hou Y, Li W, Sheng Y, Li L, Huang Y, Zhang Z, Zhu T, Peace D, Quigley JG, Wu W, et al: The transcription factor Foxm1 is essential for the quiescence and maintenance of hematopoietic stem cells. Nat Immunol. 16:810–818. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Lu XF, Zeng, Liang WQ, Chen CF, Sun SM and Lin HY: FoxM1 is a promising candidate target in the treatment of breast cancer. Oncotarget. 9:842–852. 2017.PubMed/NCBI

29 

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

30 

Bowman A and Nusse R: Location, location, location: FoxM1 mediates beta-catenin nuclear translocation and promotes glioma tumorigenesis. Cancer Cell. 20:415–416. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Zhang N, Wei P, Gong A, Chiu WT, Lee HT, Colman H, Huang H, Xue J, Liu M, Wang Y, et al: FoxM1 promotes beta-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis. Cancer Cell. 20:427–442. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Li Y, Zhang S and Huang S: FoxM1: A potential drug target for glioma. Future Oncol. 8:223–226. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Zhang N, Wu X, Yang L, Xiao F, Zhang H, Zhou A, Huang Z and Huang S: FoxM1 inhibition sensitizes resistant glioblastoma cells to temozolomide by downregulating the expression of DNA-repair gene Rad51. Clin Cancer Res. 18:5961–5971. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Wang Z, Zhang S, Siu TL and Huang S: Glioblastoma multiforme formation and EMT: Role of FoxM1 transcription factor. Curr Pharm Des. 21:1268–1271. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Huang SW, Ali ND, Zhong L and Shi J: MicroRNAs as biomarkers for human glioblastoma: Progress and potential. Acta Pharmacol Sin. 39:1405–1413. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Banelli B, Forlani A, Allemanni G, Morabito A, Pistillo MP and Romani M: MicroRNA in glioblastoma: An overview. Int J Genomics. 2017:76390842017. View Article : Google Scholar : PubMed/NCBI

37 

Ahir BK, Ozer H, Engelhard HH and Lakka SS: MicroRNAs in glioblastoma pathogenesis and therapy: A comprehensive review. Crit Rev Oncol Hematol. 120:22–33. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Hu Q, Du K, Mao X and Ning S: miR-197 is downregulated in cervical carcinogenesis and suppresses cell proliferation and invasion through targeting forkhead box M1. Oncol Lett. 15:10063–10069. 2018.PubMed/NCBI

39 

Feng J, Wang X, Zhu W, Chen S and Feng C: MicroRNA-630 suppresses epithelial-to-mesenchymal transition by regulating FoxM1 in gastric cancer cells. Biochemistry. 82:707–714. 2017.PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

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
Wang L, Lu J, Zhang H, Lyu X and Sun Z: MicroRNA‑876‑5p inhibits the progression of glioblastoma multiforme by directly targeting Forkhead box M1 Retraction in /10.3892/or.2022.8360. Oncol Rep 41: 702-710, 2019
APA
Wang, L., Lu, J., Zhang, H., Lyu, X., & Sun, Z. (2019). MicroRNA‑876‑5p inhibits the progression of glioblastoma multiforme by directly targeting Forkhead box M1 Retraction in /10.3892/or.2022.8360. Oncology Reports, 41, 702-710. https://doi.org/10.3892/or.2018.6804
MLA
Wang, L., Lu, J., Zhang, H., Lyu, X., Sun, Z."MicroRNA‑876‑5p inhibits the progression of glioblastoma multiforme by directly targeting Forkhead box M1 Retraction in /10.3892/or.2022.8360". Oncology Reports 41.1 (2019): 702-710.
Chicago
Wang, L., Lu, J., Zhang, H., Lyu, X., Sun, Z."MicroRNA‑876‑5p inhibits the progression of glioblastoma multiforme by directly targeting Forkhead box M1 Retraction in /10.3892/or.2022.8360". Oncology Reports 41, no. 1 (2019): 702-710. https://doi.org/10.3892/or.2018.6804