MicroRNA‑432 inhibits the aggressiveness of glioblastoma multiforme by directly targeting IGF‑1R

Retraction in: /10.3892/ijmm.2022.5187

  • Authors:
    • Xudong Chen
    • Xufei Zhang
    • Shunjin Sun
    • Meixiao Zhu
  • View Affiliations

  • Published online on: December 16, 2019     https://doi.org/10.3892/ijmm.2019.4429
  • Pages: 597-606
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNA‑432 (miR‑432) has been studied in multiple tumors, but the expression status, biological functions and the mechanism of action of miR‑432 in glioblastoma multiforme (GBM) are yet to be elucidated. In the present study, miR‑432 expression in GBM was determined and its clinical significance was evaluated among patients with GBM. The effects on the malignancy of GBM in vitro and in vivo were examined in detail and the interactions between miR‑432 and insulin‑like growth factor 1 receptor (IGF‑1R) mRNA were then explored. miR‑432 expression in GBM tissue samples and cell lines was measured by reverse transcription‑quantitative (RT‑q)PCR. GBM cell proliferation, apoptosis, migration and invasion in vitro and tumor growth in vivo were evaluated by a Cell Counting Kit‑8 assay, flow‑cytometric analysis, Transwell migration and invasion assays, and a tumor xenograft experiment, respectively. Bioinformatic analysis followed by a luciferase reporter assay, RT‑qPCR and western blotting was applied to demonstrate that IGF‑1R is a direct target gene of miR‑432 in GBM cells. It was found that miR‑432 is downregulated in GBM tumors and cell lines. miR‑432 under expression obviously correlated with the Karnofsky Performance Status score and shorter overall survival among patients with GBM. Exogenous miR‑432 expression significantly reduced proliferation and induced apoptosis of GBM cells. In addition, miR‑432 overexpression impaired the migratory and invasive abilities of GBM cells in vitro and decreased their tumor growth in vivo. Furthermore, IGF‑1R was validated as a direct target gene of miR‑432 in GBM cells. IGF‑1R knockdown imitated the tumor‑suppressive actions of miR‑432 overexpression in GBM cells. Rescue experiments proved IGF‑1R downregulation to be essential for the effects of miR‑432 on GBM cells. The results of the present study revealed a tumor‑suppressive role of the miR‑432‑IGF‑1R axis in GBM cells and this axis may have implications for GBM therapy.

Introduction

Gliomas represent the most common and prevalent malignant tumors in the human central nervous system (1). Glioblastoma multiforme (GBM), also known as World Health Organization grade IV glioma, is the most common and deadly glioma type (2). The treatments for GBM, including surgical resection, chemoradiotherapy, gene therapy and immunotherapy, have improved rapidly lately (3). Unfortunately, the prognosis of patients with GBM remains unsatisfactory, with a median survival duration of only 9-12 months (4). Metastasis, recurrence, unlimited proliferation, fast diffuse infiltration and significant apoptosis resistance are considered the major factors responsible for the poor prognosis of patients with GBM (5,6). The mechanisms underlying the initiation and progression of GBM have not been fully elucidated, and this is another reason for the lack of improvement in therapeutic outcomes (7). Thus, an urgent task for scientists in this field is to further elucidate the molecular pathogenesis of GBM and to identify effective therapeutic methods in order to improve the survival of patients with this fatal disease.

MicroRNAs (miRNAs/miRs) are a family of endogenous noncoding short RNAs 17-24 nucleotides long (8). miRNAs are regarded as a novel group of gene expression regulators and they modulate gene expression by directly binding to partially complimentary sequences in the 3′ untranslated regions (3′-UTRs) of their target mRNAs, thereby causing translation suppression and/or degradation of the mRNAs (9). A considerable number of studies have identified aberrant expression of miRNAs in human cancers, including GBM. For instance, miR-139-3p (10), miR-454-3p (11) and miR-559 (12) are weakly expressed in GBM; on the contrary, miR-141-3p (13), miR-500a-3p (14), and miR-4516 (15) are overexpressed in GBM. The changes in miRNA expression exert crucial effects on nearly all pathological phenomena during GBM formation and progression, e.g., rapid cell proliferation, an aberrant cell cycle, insufficient apoptosis, excessive angiogenesis, epithelial-mesenchymal transition, and metastasis (16-18). Therefore, further research into the specific participation of aberrantly expressed miRNAs in GBM may reveal useful targets for anticancer therapies.

miR-432 has been studied in multiple tumors (19-23). For instance, miR-432 expression is low in lung adenocarcinoma tissues and cell lines. Decreased miR-432 expression significantly correlates with the clinical stage among patients with lung adenocarcinoma. Patients with this tumor under expressing miR-432 show worse overall survival than patients with lung adenocarcinoma featuring high miR-432 expression (19). In addition, downregulation of miR-432 has been proven in osteosarcoma (20), hepatocellular carcinoma (21), neuroblastoma (22) and prostate cancer (23). However, the expression status, biological functions and the mechanism of action of miR-432 in GBM are yet to be elucidated. Accordingly, miR-432 was selected as the object of study. This study aimed to measure miR-432 expression in GBM and to evaluate its clinical significance among patients with GBM. The effects of miR-432 on the malignancy of GBM in vitro and in vivo were examined in detail and the interactions between miR-432 and IGF-1R mRNA were then explored. Collectively, the present findings suggest that miR-432 may act as a tumor-suppressive miRNA on GBM progression by directly targeting IGF-1R mRNA. The present results also show the potential of miR-432 as a therapeutic target in this aggressive tumor.

Materials and methods

Patients and tissue samples

GBM tissue samples and corresponding adjacent normal tissues were collected from 51 patients with GBM in The Sixth Affiliated Hospital of Wenzhou Medical University between January 2013 and February 2014. Patient characteristics are shown in Table I. All these patients were treated with surgical resection and had not received preoperative chemoradiotherapy, gene therapy, immunotherapy, or other anticancer treatments. All the tissue samples were quickly immersed in liquid nitrogen and then stored at −80°C.

Table I

Relationship between miR-432 expression and clinical parameters of patients with glioblastoma.

Table I

Relationship between miR-432 expression and clinical parameters of patients with glioblastoma.

Clinical parametersmiR-432 expression level
P-value
LowHigh
Sex0.579
 Male1512
 Female1113
Age0.083
 <55 years136
 ≥55 years1319
Extension of resection0.565
 Subtotal118
 Total1517
KPS
 ≥808160.025
 <80189

[i] KPS, Karnofsky performance score; miR, microRNA.

Cell culture

A total of three GBM cell lines (T98, U138 and U251) were obtained from Shanghai Institute of Cell Biology Cell Bank, the Chinese Academy of Sciences. These cell lines were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% of fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc.). Normal human astrocytes (NHAs; ScienCell Research Laboratories, Inc.) were grown in the astrocyte medium (ScienCell Research Laboratories, Inc.) supplemented with 10% of FBS. All the cell lines were cultured at 37°C in a humidified incubator supplied with 5% of CO2.

A transfection experiment

An miR-432 agomir (agomir-432) and negative control agomir (agomir-NC) were purchased from Shanghai GenePharma Co., Ltd. A specific small interfering RNA (siRNA) targeting IGF-1R (si-IGF-1R) and a negative control siRNA (si-NC) were synthesized by Guangzhou RioBio, Co., Ltd. An IGF-1R overexpression plasmid lacking its 3'-UTR (pcDNA3.1-IGF-1R; hereafter: pc-IGF-1R) was constructed by Shanghai GeneChem Co., Ltd., and the empty pcDNA3.1 vector served as a negative control. T98 and U251 cells were seeded in 6-well plates at a density of 7×105/well and transfected with the agomir (50 nM), siRNA (100 pmol) or plasmids (4 µg) using the Lipofectamine 2000 Reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Reverse-transcription quantitative polymerase chain reaction (RT-qPCR), flow-cytometric analysis and Transwell assays were conducted 48 h post-transfection. Cell Counting Kit-8 (CCK-8) assay and western blotting were performed 24 and 72 h after transfection.

Extraction of total RNA and RT-qPCR

The TRIzol® Reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was employed for total-RNA extraction. For quantification of miR-432 expression, the cDNA was synthesized from the total RNA using the miRcute miRNA First-Strand cDNA Synthesis kit (Tiangen Biotech Co., Ltd.). The temperature protocol for RT was as follows: 37°C for 60 min, 95°C for 5 min and kept at 4°C. The expression of miR-432 was measured by qPCR with miRcute miRNA qPCR Detection kit SYBR Green (Tiangen Biotech Co., Ltd.). The thermocycling conditions were as follows: 95°C for 2 min, 95°C for 10 sec, 55°C for 30 sec and 72°C for 30 sec, for 40 cycles. To analyze IGF-1R mRNA expression, RT and subsequent qPCR were carried out respectively using the PrimeScript RT Reagent kit and SYBR Premix Ex Taq™ kit (both from Takara Biotechnology, Co., Ltd.). The temperature protocol for RT was as follows: 37°C for 15 min and 85°C for 5 sec. The thermocycling 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. U6 small nuclear RNA and GAPDH served as the internal controls for miR-432 and IGF-1R mRNA, respectively. The 2−ΔΔCq method (24) was employed to calculate the relative gene expression.

The primers were designed as follows: miR-432 forward, 5′-AAC GAG ACG ACG ACA GAC-3′ and reverse, 5′-CTT GGA GTA GGT CAT TGG GT-3′; U6 forward, 5′-GCT TCG GCA GCA CAT ATA CTA AAA T-3′ and reverse, 5′-CGC TTC ACG AAT TTG CGT GTC AT-3′; IGF-1R forward, 5′-AGG ATA TTG GGC TTT ACA ACC TG-3′ and reverse, 5′-GAG GTA ACA GAG GTC AGC ATT TT-3′; and GAPDH forward, 5′-CGG AGT CAA CGG ATT TGG TCG TAT-3′ and reverse, 5′-AGC CTT CTC CAT GGT GGT GAA GAC-3′.

CCK-8 assay

At 24 h post-transfection, T98 and U251 cells were harvested for counting. The cells were seeded in 96-well plates at an initial density of 2×103 cells per well. The cells were next cultured at 37°C with four predetermined time points for analysis: 0, 24, 48 and 72 h after seeding. At every selected time point, 10 µl the CCK-8 reagent (Dojindo Molecular Technologies, Inc.) was added into each well and incubation was conducted at 37°C and 5% CO2 for another 2 h. Optical density (OD) at 450 nm wavelength was measured on an automatic multiwell spectrophotometer (Bio-Rad Laboratories, Inc.). The growth curve was constructed according to the time points and OD values.

Flow-cytometric analysis of apoptosis

Cells were harvested at 48 h post-transfection, washed twice with cold phosphate-buffered saline (PBS) and subjected to apoptosis analysis with the Annexin V-Fluorescein Isothiocyanate (FITC) Apoptosis Detection kit (Biolegend, Inc.). After that, the cells were resuspended in 100 µl of 1X binding buffer, followed by the addition of 5 µl Annexin V-FITC and 5 µl a propidium iodide solution (that came with the kit) into each tube. The cells were incubated in a dark at room temperature for 15 min and apoptotic cells were detected by flow cytometry (FACScan; BD Biosciences; Becton, Dickinson and Company). Data were analyzed with CellQuest Pro 4.0.2 software (BD Biosciences).

Transwell migration and invasion assays

Matrigel (Corning, Inc.) mixed with the culture medium was used to precoat the Transwell chambers (Corning, Inc.) containing polycarbonate membranes with 8-µm pore size. This step was required for the invasion assay but not for the migration assay. For both assays, transfected cells were harvested, counted and resuspended in FBS-free DMEM. A total of 5×104 cells were added into the upper chambers. In the lower chambers, DMEM supplemented with 20% of FBS was added and served as the chemotactic factor for the cells. After 24 h cultivation, cells remaining on the inner side of the membrane were carefully wiped off with a cotton swab. The cells present on the outer side of the membranes were fixed in 4% paraformaldehyde at room temperature for 30 min, stained with 0.05% crystal violet at room temperature for 30 min and images were captured. The migratory or invading cells were counted under an inverted light microscope (Olympus Corporation). The migratory and invasive abilities were expressed as the average number of migratory or invading cells in six randomly chosen visual fields per chamber.

Tumor xenograft experiment

T98 cells transfected with agomir-432 or agomir-NC were collected in the logarithmic phase of growth, centrifuged at 716 × g for 5 min at room temperature and dispersed to obtain a single-cell DMEM suspension. The flank of four-week-old BALB/c nude mice (19-21 g; n=8; Shanghai Laboratory Animal Center; Chinese Academy of Sciences) was subcutaneously injected with 2×106 cells transfected with the above-mentioned nucleic acids. Each group contained four nude mice. The animals were maintained under specific pathogen-free conditions (25°C, 50% humidity, 10-h light/14-h dark cycle) and ab libitum food/water access. Tumor width and length were measured at 2-day intervals with a Vernier caliper, and the tumor volume was calculated via the following formula: Volume (mm3)=0.5 × width2 (mm2) × length (mm). The study period for these mice was 4 weeks. All mice were anesthetized with an intraperitoneal injection of 30 mg/kg pentobarbital sodium (25). Subsequent to anesthesia, all the mice (n=8) were euthanized by cervical dislocation. Finally, the tumor xeno-grafts were excised, imaged, weighed and stored for further analysis.

The animal health and behavior were monitored every day. Symptoms, such as abnormal feeding, weight loss, ascites, cachexia and the size of tumor was >2 cm during the experiments, were set as humane endpoints for the present study; however, no animal was sacrificed before the completion of the 4 weeks experiment as a result of displaying any of these symptoms.

Bioinformatic analysis and a luciferase reporter assay

A total of three miRNA target prediction databases were searched to find the potential target gene of miR-432, including TargetScan 7.1 (http://www.targetscan.org/), starBase 3.0 (http://starbase.sysu.edu.cn/index.php) and miRDB (http://mirdb.org/).

The wild-type (wt) 3′-UTR fragment of IGF-1R containing the predicted miR-432-binding site was amplified by Shanghai GenePharma Co., Ltd., and inserted into the pmirGLO dual-luciferase vector (Promega Corporation) to generate the pmirGLO-IGF-1R-wt-3′-UTR plasmid. The plasmid pmirGLO-IGF-1R-mutant (mut)-3′-UTR was created in the same way. T98 and U251 cells were seeded in 24-well plates at a density of 1.0×105/well and transiently cotransfected with a mixture of either agomir-432 or agomir-NC plus either pmirGLO-IGF-1R-wt-3′-UTR or pmirGLO-IGF-1R-mut-3′-UTR using the Lipofectamine 2000 Reagent. The luciferase activities were quantified 48 h after the transfection via a Dual-Luciferase Reporter Assay System (Promega Corporation). Renilla luciferase activity served for normalization of the data.

Western blotting

Isolation of total protein was carried out with RIPA buffer supplemented with a protease inhibitor: Phenylmethanesulfonyl fluoride (both from Beyotime Institute of Biotechnology). The Bicinchoninic Acid Assay kit (Beyotime Institute of Biotechnology) was used for quantification of protein concentrations. Equal amounts of protein (30 µg) were loaded onto a SDS 10% polyacrylamide gel for electrophoresis. After that, the separated proteins in the gel were transferred to polyvinylidene difluoride membranes (EMD Millipore), followed by blocking with a 5% solution of defatted milk powder at room temperature for 2 h and incubation overnight at 4°C with primary antibodies. After three washes, a horseradish peroxidase (HRP)-conjugated secondary antibody (1:5,000; cat. no. ab205718; Abcam) was incubated with the membranes at room temperature for 2 h, after which the membrane was processed by means of the Immobilon Western Chemiluminescent HRP Substrate kit (EMD Millipore) for detecting the protein signals. Analysis was performed with Quantity One software version 4.62 (Bio-Rad Laboratories, Inc.). The primary antibodies included a mouse anti-human IGF-1R antibody (cat. no. ab182408; Abcam) and a mouse anti-human GAPDH antibody (cat. no. ab128915; Abcam). All the primary antibodies were applied at 1:1,000.

Statistical analysis

All the results are shown as the mean ± standard deviation from three independent experiments. The correlation between the clinical parameters of patients with GBM and miR-432 expression was determined with the χ2 test. Survival analysis was performed by the Kaplan-Meier method and log-rank test. A comparison between two groups was conducted with the t test, whereas the differences among multiple groups were assessed by one-way analysis of variance with Tukey's post hoc test. Spearman's correlation analysis was performed to confirm the expression correlation between miR-432 and IGF-1R mRNA in the GBM tissue samples. All statistical analyses were carried out in the SPSS software (version 16.0; SPSS Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-432 expression is low in GBM tissues and cell lines

To determine the expression profile of miR-432 in GBM, its expression was analyzed in 51 pairs of GBM tissue samples and corresponding adjacent-normal-tissue samples. The RT-qPCR data indicated that the expression of miR-432 was significantly decreased in GBM tissue samples compared with in the corresponding adjacent normal tissues (P<0.05; Fig. 1A). In addition, RT-qPCR was carried out for quantification of miR-432 expression in GBM cell lines (T98, U138 and U251) and NHAs. There was significant downregulation of miR-432 in all three GBM cell lines as compared with NHAs (P<0.05; Fig. 1B). These results suggested that miR-432 is under expressed in GBM and this downregulation may be related to tumor progression.

To understand the clinical value of miR-432, all the 51 patients with GBM were classified into two groups (low and high miR-432 expression) based on the median value of miR-432 expression among the 51 GBM tumors. The analysis revealed a significant association between decreased miR-432 expression and the Karnofsky Performance Status score (P=0.025; Table I). In addition, patients with GBM in the low miR-432 expression group showed significantly shorter overall survival than the patients in the high miR-432 expression group (P=0.029; Fig. 1C). These findings suggested that miR-432 is a promising prognostic biomarker of GBM.

miR-432 overexpression suppresses GBM cell proliferation, migration and invasion and promotes GBM cell apoptosis in vitro

The expression of miR-432 was lower in cell lines T98 and U251 than in U138 cells. Therefore, the two cell lines were chosen for further experiments on the involvement of miR-432 in the malignancy of GBM. To this end, either agomir-432 or agomir-NC was transfected into T98 and U251 cells. The success of transfection was confirmed by RT-qPCR (Fig. 2A). A CCK-8 assay was performed to clarify whether the upregulation of miR-432 affects GBM cell proliferation. The proliferative ability of T98 and U251 cells significantly decreased after agomir-432 transfection (P<0.05; Fig. 2B). In addition, the apoptotic rate of T98 and U251 cells was significantly raised by miR-432 upregulation (P<0.05; Fig. 2C). Transwell migration and invasion assays were conducted to determine the influence of miR-432 overexpression on the metastasis of GBM cells. The result meant that the exogenous miR-432 expression decreased the migration (Fig. 2D) and invasiveness (Fig. 2E) of T98 and U251 cells. These observations revealed the tumor-suppressive role of miR-432 in GBM.

miR-432 directly targets IGF-1R mRNA in GBM cells

According to the miRNA target prediction databases, the 3′-UTR of IGF-1R mRNA contains complementary sequences for miR-432 (Fig. 3A). IGF-1R was selected for further experimental validation because IGF-1R exerts crucial actions on the initiation and progression of GBM (26-33). To confirm the binding of miR-432 to the 3′-UTR of IGF-1R mRNA, the luciferase reporter assay was performed on T98 and U251 cells after cotransfection with either agomir-432 or agomir-NC and either pmirGLO-IGF-1R-wt-3′-UTR or pmirGLO-IGF-1R-mut-3′-UTR. The results suggested that the transfection with the agomir-432 significantly decreased the luciferase activity generated by the plasmid carrying the wt miR-432-binding site (P<0.01; Fig. 3B). By contrast, the increase in miR-432 expression did not reduce the luciferase activity generated by the plasmid containing the mut IGF-1R 3′-UTR fragment.

RT-qPCR and western blotting were conducted to test whether endogenous IGF-1R expression can be changed by miR-432 upregulation in GBM cells. As expected, the mRNA (Fig. 3C) and protein (Fig. 3D) levels of IGF-1R significantly decreased in T98 and U251 cells after the forced miR-432 over-expression (P<0.05). Additionally, the expression of IGF-1R mRNA was analyzed by RT-qPCR in the 51 pairs of GBM tissue samples and corresponding adjacent-normal-tissue samples. IGF-1R mRNA was found to be significantly overexpressed in GBM tissue samples relative to the adjacent normal tissues (P<0.01; Fig. 3E). Moreover, an inverse correlation between the expression levels of miR-432 and IGF-1R mRNA in GBM tissues was confirmed via Spearman's correlation analysis (Fig. 3F; R2=0.3969, P<0.0001). These results collectively proved IGF-1R to be a direct target gene of miR-432 in GBM cells.

IGF-1R knockdown has effects similar to those of miR-432 overexpression in relation to GBM progression

Given that IGF-1R was validated as a direct target gene of miR-432, the present study supposed that an IGF-1R knockdown may imitate the consequences of miR-432 overexpression in GBM cells. To test this hypothesis, loss-of-function assays on T98 and U251 cells were performed through transfection with either si-IGF-1R or si-NC. The expression of the IGF-1R protein significantly diminished in T98 and U251 cells after the transfection with si-IGF-1R, as demonstrated by western blotting (P<0.01; Fig. 4A). In terms of cellular functions, cell proliferation significantly slowed down when IGF-1R was knocked down in T98 and U251 cells from 48 h (P<0.05; Fig. 4B). Next, it was demonstrated that the proportion of apoptotic cells was increased among IGF-1R-deficient T98 and U251 cells compared with in the si-NC group (Fig. 4C). Additionally, the knockdown of IGF-1R significantly impaired the migration (P<0.05; Fig. 4D) and invasiveness (P<0.01; Fig. 4E) of T98 and U251 cells. Collectively, these results meant that the IGF-1R knockdown could mimic the effects of miR-432 overexpression in GBM cells and confirmed that IGF-1R downregulation may be responsible for the actions of miR-432 on GBM progression.

Restoration of IGF-1R expression attenuates the influence of miR-432 on GBM cells

Rescue experiments were carried out to further elucidate whether IGF-1R downregulation mediates the effects of miR-432 in GBM cells. Firstly, the efficiency of IGF-1R overexpression plasmid lacking its 3′-UTR (pc-IGF-1R) was determined via RT-qPCR analysis (Fig. 5A). T98 and U251 cells were cotransfected with agomir-432 and either the IGF-1R overexpression plasmid lacking its 3′-UTR (pc-IGF-1R) or the empty pcDNA3.1 vector. Following the cotransfection, western-blotting data indicated that the decrease of IGF-1R protein expression in miR-432-overexpressing T98 and U251 cells was reversed by the cotransfection with pc-IGF-1R (Fig. 5B). Regarding cellular functions, restoration of IGF-1R expression attenuated the influence of miR-432 overexpression on the proliferation (Fig. 5C), apoptosis (Fig. 5D), migration (Fig. 5E) and invasiveness (Fig. 5F) of T98 and U251 cells. To sum up, the tumor-suppressive effects of miR-432 upregulation on the malignancy of GBM cells were mediated by IGF-1R downregulation.

miR-432 inhibits the tumor growth of GBM cells in vivo

The tumor xenograft experiment was conducted to confirm the in vitro finding that miR-432 may perform tumor-suppressive functions in GBM. T98 cells transfected with either agomir-432 or agomir-NC were subcutaneously inoculated into nude mice. The volume of tumor xenografts was decreased in the agomir-432 group compared with in the agomir-NC group (Fig. 6A and B). In addition, the average weight of tumor xenografts derived from agomir-432-transfected T98 cells was significantly decreased compared with in the agomir-NC group (P<0.01; Fig. 6C). When compared with the agomir-NC group, miR-432 was still overexpressed (Fig. 6D), while the mRNA (Fig. 6E) and protein (Fig. 6F) levels of IGF-1R were significantly decreased in the tumor xenografts of the agomir-432 group (P<0.05). These data were suggestive of a strong inhibitory effect of the miR-432-IGF-1R axis on the tumor growth of GBM cells in vivo.

Discussion

Emerging evidence suggests that miRNAs are aber-rantly expressed in GBM and have either an oncogenic or tumor-suppressive role (34-36). The deregulation of miRNAs is closely related to the malignant progression of GBM (16,17,37). Therefore, identifying the functions of miRNAs in GBM onset and progression may point to effective therapeutic targets for the management of this cancer. In this study, the expression status of miR-432 in GBM was determined, the clinical value of miR-432 for patients with GBM was assessed, the specific functions of miR-432 in GBM progression were explored and the underlying mechanism was investigated. Taken together, for the first time to the best of our knowledge, the present study demonstrated the tumor-suppressive activities of miR-432 in GBM and suggested that miR-432 may find applications in anti-GBM therapies.

miR-432 serves as a tumor-suppressive miRNA in multiple types of human cancer. For instance, exogenous miR-432 expression attenuates lung adenocarcinoma cell proliferation, promotes the apoptosis of these cells and increases their sensitivity to cisplatin chemotherapy; these effects are mediated by direct targeting of E2F transcription factor 3 mRNA and of AXL receptor tyrosine kinase mRNA (19). miR-432 overexpression restricts the proliferation and invasiveness of osteosarcoma cells by inhibiting the expression of metastasis-associated in colon cancer 1 (20). miR-432 directly targets three mRNAs (low-density lipoprotein receptor-related protein 6, tripartite-motif-containing 29 and pygopus homolog 2), decreases their expression, and deactivates the WNT-β-catenin pathway in hepatocellular carcinoma, thereby suppressing tumor growth in vitro and in vivo (21). miR-432 also plays a tumor-suppressive role in neuroblastoma (22) and prostate cancer (23). Nevertheless, few studies have been conducted on the specific involvement of miR-432 in the malignant characteristics of GBM. In the present study, the results revealed that miR-432 is only weakly expressed in GBM tumors and cell lines. In addition, lower miR-432 expression closely correlated with the Karnofsky Performance Status score among the 51 patients with GBM. The patients with GBM in the low miR-432 expression group showed decreased overall survival compared with the patients in the high miR-432 expression group. miR-432 inhibits GBM cell proliferation, promotes GBM cell apoptosis, impairs GBM cell migration and invasion in vitro and slows the tumor growth of GBM cells in vivo.

The mechanisms behind the tumor-suppressive action of miR-432 on GBM progression were explored in the present study at the molecular level. The mechanism investigation identified IGF-1R as a direct target gene of miR-432 in GBM cells. IGF-1R, a transmembrane tyrosine kinase receptor of the insulin receptor family, turned out to be overexpressed in GBM. In addition, upregulation of IGF-1R has been confirmed to be an independent prognostic indicator of shorter survival among patients with GBM (26). IGF-1R promotes GBM formation and progression by enhancing a wide range of aggressive characteristics, including rapid cell proliferation, dysregulation of the cell cycle, apoptosis downregulation, a high glucose-metabolic ability, high motility, metastasis, chemotherapy and radiotherapy resistance and tumorigenesis (27-33). In the present study, the results make it clear that the tumor-suppressive actions of miR-432 on the malignancy of GBM cells in vitro and in vivo are partly mediated by the decrease in IGF-1R expression. Hence, IGF-1R knockdown due to miR-432 restoration could be an effective therapeutic strategy against GBM.

Three limitations are included in the present study. Firstly, expression of miR-432 was not knocked down and subsequently the influences of miR-432 silencing on GBM progression were not explored in detail. Secondly, the effects of miR-432 on GBM metastasis in vitro were not tested. Lastly, the sample size was small. The authors' future investigations will resolve these limitations.

In conclusion, the present study revealed that miR-432 expression is low in GBM and this downregulation is associated with poor clinical outcomes among the patients. Exogenously expressed miR-432 inhibited the malignancy of GBM cells in vitro and in vivo by directly targeting IGF-1R mRNA, thereby downregulating IGF-1R. Thus, these results suggest that miR-432 or IGF-1R may be a target for the treatment of patients with GBM.

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

MZ and XC designed this study, and performed RT-qPCR analysis and CCK-8 assay. XZ and SS conducted flow-cytometric analysis, Transwell assays, the tumor xenograft experiment, luciferase reporter assay and western blotting. MZ analyzed the data. All authors made a significant contribution to the findings and methods. They read and approved the final draft.

Ethics approval and consent to participate

The study protocol was approved by the Medical Ethics Committee of The Sixth Affiliated Hospital of Wenzhou Medical University and was carried out in compliance with the principles outlined in the Declaration of Helsinki. Written informed consent was obtained from all the patients enrolled in this study. The Institutional Animal Care and Use Committee of The Sixth Affiliated Hospital of Wenzhou Medical University approved all the animal experiments. All experimental steps were in accordance with the Animal Protection Law of the People's Republic of China-2009 for experimental animals.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

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

2 

Thorne AH, Zanca C and Furnari F: Epidermal growth factor receptor targeting and challenges in glioblastoma. Neuro Oncol. 18:914–918. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Van Meir EG, Hadjipanayis CG, Norden AD, Shu HK, Wen PY and Olson JJ: Exciting new advances in neuro-oncology: The avenue to a cure for malignant glioma. CA Cancer J Clin. 60:166–193. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, Hahn WC, Ligon KL, Louis DN, Brennan C, et al: Malignant astrocytic glioma: Genetics, biology, and paths to treatment. Genes Dev. 21:2683–2710. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Zhuang W, Qin Z and Liang Z: The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim Biophys Sin (Shanghai). 41:341–351. 2009. View Article : Google Scholar

7 

Wang Y and Jiang T: Understanding high grade glioma: Molecular mechanism, therapy and comprehensive management. Cancer Lett. 331:139–146. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. 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 

Shi L, Yuan Y and Li HY: MicroRNA-139-3p suppresses growth and metastasis of glioblastoma via inhibition of NIN1/RPNI2 binding protein 1 homolog. Eur Rev Med Pharmacol Sci. 23:4264–4274. 2019.PubMed/NCBI

11 

Zuo J, Yu H, Xie P, Liu W, Wang K and Ni H: miR-454-3p exerts tumor-suppressive functions by down-regulation of NFATc2 in glioblastoma. Gene. 710:233–239. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Yang F, Zhang C, Xu C, Fu F, Han D and Li H: MicroRNA-559 plays an inhibitory role in the malignant progression of glio-blastoma cells by directly targeting metadherin. Onco Targets Ther. 12:4415–4426. 2019. View Article : Google Scholar :

13 

Zhou X, Wu W, Zeng A, Nie E, Jin X, Yu T, Zhi T, Jiang K, Wang Y, Zhang J and You Y: MicroRNA-141-3p promotes glioma cell growth and temozolomide resistance by directly targeting p53. Oncotarget. 8:71080–71094. 2017.PubMed/NCBI

14 

Liu Z, Su D, Qi X and Ma J: MiR-500a-5p promotes glioblastoma cell proliferation, migration and invasion by targeting chromodomain helicase DNA binding protein 5. Mol Med Rep. 18:2689–2696. 2018.PubMed/NCBI

15 

Cui T, Bell EH, McElroy J, Becker AP, Gulati PM, Geurts M, Mladkova N, Gray A, Liu K, Yang L, et al: miR-4516 predicts poor prognosis and functions as a novel oncogene via targeting PTPN14 in human glioblastoma. Oncogene. 38:2923–2936. 2019. View Article : Google Scholar :

16 

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

17 

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

18 

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

19 

Chen L, Kong G, Zhang C, Dong H, Yang C, Song G, Guo C, Wang L and Yu H: MicroRNA-432 functions as a tumor suppressor gene through targeting E2F3 and AXL in lung adenocarcinoma. Oncotarget. 7:20041–20053. 2016.PubMed/NCBI

20 

Lv D, Zhen Z and Huang D: MicroRNA-432 is downregulated in osteosarcoma and inhibits cell proliferation and invasion by directly targeting metastasis-associated in colon cancer-1. Exp Ther Med. 17:919–926. 2019.PubMed/NCBI

21 

Jiang N, Chen WJ, Zhang JW, Xu C, Zeng XC, Zhang T, Li Y and Wang GY: Downregulation of miR-432 activates Wnt/β-catenin signaling and promotes human hepatocellular carcinoma proliferation. Oncotarget. 6:7866–7879. 2015.PubMed/NCBI

22 

Das E and Bhattacharyya NP: MicroRNA-432 contributes to dopamine cocktail and retinoic acid induced differentiation of human neuroblastoma cells by targeting NESTIN and RCOR1 genes. FEBS Lett. 588:1706–1714. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Li JB, Liu F, Zhang BP, Bai WK, Cheng W, Zhang YH and Yu LJ: LncRNA625 modulates prostate cancer cells proliferation and apoptosis through regulating the Wnt/β-catenin pathway by targeting miR-432. Eur Rev Med Pharmacol Sci. 21:2586–2595. 2017.PubMed/NCBI

24 

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

25 

Baradaran Rahimi V, Askari VR, Tajani AS, Hosseini A and Rakhshandeh H: Evaluation of the sleep-prolonging effect of lagenaria vulgaris and cucurbita pepo extracts on pentobar-bital-induced sleep and possible mechanisms of action. Medicina (Kaunas). 54:pii: E55. 2018.

26 

Maris C, D'Haene N, Trépant AL, Le Mercier M, Sauvage S, Allard J, Rorive S, Demetter P, Decaestecker C and Salmon I: IGF-IR: A new prognostic biomarker for human glioblastoma. Br J Cancer. 113:729–737. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Shi ZM, Wang XF, Qian X, Tao T, Wang L, Chen QD, Wang XR, Cao L, Wang YY, Zhang JX, et al: MiRNA-181b suppresses IGF-1R and functions as a tumor suppressor gene in gliomas. RNA. 19:552–560. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Y, Chen X, Lian H, Liu J, Zhou B, Han S, Peng B, Yin J, Liu W and He X: MicroRNA-503 acts as a tumor suppressor in glioblastoma for multiple antitumor effects by targeting IGF-1R. Oncology reports. 31:1445–1452. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Guo T, Feng Y, Liu Q, Yang X, Jiang T, Chen Y and Zhang Q: MicroRNA-320a suppresses in GBM patients and modulates glioma cell functions by targeting IGF-1R. Tumour Biol. 35:11269–11275. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Wang B, Sun F, Dong N, Sun Z, Diao Y, Zheng C, Sun J, Yang Y and Jiang D: MicroRNA-7 directly targets insulin-like growth factor 1 receptor to inhibit cellular growth and glucose metabolism in gliomas. Diagn Pathol. 9:2112014. View Article : Google Scholar : PubMed/NCBI

31 

Lian HW, Zhou Y, Jian ZH and Liu RZ: MiR-323-5p acts as a tumor suppressor by targeting the insulin-like growth factor 1 receptor in human glioma cells. Asian Pac J Cancer Prev. 15:10181–10185. 2014. View Article : Google Scholar

32 

Lin YC, Hou SC, Hung CM, Lin JN, Chen WC, Ho CT, Kuo SC and Way TD: Inhibition of the insulin-like growth factor 1 receptor by CHM-1 blocks proliferation of glioblastoma multiforme cells. Chem Biol Interact. 231:119–126. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Ma Y, Tang N, Thompson RC, Mobley BC, Clark SW, Sarkaria JN and Wang J: InsR/IGF1R pathway mediates resistance to EGFR inhibitors in glioblastoma. Clin Cancer Res. 22:1767–1776. 2016. View Article : Google Scholar :

34 

Yu J, Wu SW and Wu WP: A tumor-suppressive microRNA, miRNA-485-5p, inhibits glioma cell proliferation and invasion by down-regulating TPD52L2. Am J Transl Res. 9:3336–3344. 2017.PubMed/NCBI

35 

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

36 

Cheng ZX, Yin WB and Wang ZY: MicroRNA-132 induces temozolomide resistance and promotes the formation of cancer stem cell phenotypes by targeting tumor suppressor candidate 3 in glioblastoma. Int J Mol Med. 40:1307–1314. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Zhang Y, Cruickshanks N, Pahuski M, Yuan F, Dutta A, Schiff D, Purow B and Abounader R: Noncoding RNAs in glioblastoma. Glioblastoma. De Vleeschouwer S: Codon Publications; Brisbane, AU: 2017, View Article : Google Scholar

Related Articles

Journal Cover

February-2020
Volume 45 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen X, Zhang X, Sun S and Zhu M: MicroRNA‑432 inhibits the aggressiveness of glioblastoma multiforme by directly targeting IGF‑1R Retraction in /10.3892/ijmm.2022.5187. Int J Mol Med 45: 597-606, 2020
APA
Chen, X., Zhang, X., Sun, S., & Zhu, M. (2020). MicroRNA‑432 inhibits the aggressiveness of glioblastoma multiforme by directly targeting IGF‑1R Retraction in /10.3892/ijmm.2022.5187. International Journal of Molecular Medicine, 45, 597-606. https://doi.org/10.3892/ijmm.2019.4429
MLA
Chen, X., Zhang, X., Sun, S., Zhu, M."MicroRNA‑432 inhibits the aggressiveness of glioblastoma multiforme by directly targeting IGF‑1R Retraction in /10.3892/ijmm.2022.5187". International Journal of Molecular Medicine 45.2 (2020): 597-606.
Chicago
Chen, X., Zhang, X., Sun, S., Zhu, M."MicroRNA‑432 inhibits the aggressiveness of glioblastoma multiforme by directly targeting IGF‑1R Retraction in /10.3892/ijmm.2022.5187". International Journal of Molecular Medicine 45, no. 2 (2020): 597-606. https://doi.org/10.3892/ijmm.2019.4429