Open Access

miR‑181d promotes cell proliferation via the IGF1/PI3K/AKT axis in glioma

  • Authors:
    • Dong Tang
    • Wenhong Gao
    • Jian Yang
    • Junhui Liu
    • Jian Zhao
    • Jian Ge
    • Qianxue Chen
    • Baohui Liu
  • View Affiliations

  • Published online on: August 26, 2020     https://doi.org/10.3892/mmr.2020.11464
  • Pages: 3804-3812
  • Copyright: © Tang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioma is a malignant brain cancer that exhibits high invasive ability and poor prognosis. MicroRNA (miR)‑181d has been reported to be involved in the development of glioma. Therefore, the aim of the present study was to investigate whether miR‑181d affected cellular progression by influencing the insulin like growth factor (IGF1)/PI3K/AKT axis. Western blot analysis was performed to analyze the expression levels of specific proteins, and a Cell Counting Kit‑8 assay was used to assess the proliferative ability of cells. Cell cycle progression and cellular apoptosis were both measured using flow cytometry. The results indicated that miR‑181d promoted cellular proliferation and cell cycle progression, while suppressing cellular apoptosis via the IGF1/PI3K/AKT axis. It was demonstrated that the IGF1 and PI3K/AKT inhibitors reversed these observed functions of miR‑181d. Furthermore, miR‑181d enhanced the growth of glioma xenografts in vivo, promoted cell cycle progression and suppressed cellular apoptosis within glioma xenograft tissues. Therefore, this newly identified miR‑181d/IGF1/PI3K/AKT axis may provide novel insights into the pathogenesis of glioma.

Introduction

Glioma is a malignant brain cancer that exhibits high invasiveness, is typically dispersed throughout the brain, blood vessels and their basement membranes, which are rich in extracellular matrix (13). The DNA alkylating agent temozolomide, when used in combination with radiation therapy, improves the survival rate in patients with glioma (4). However, previous studies have reported that only ~10% of patients undergoing this therapy can survive for 5 years (5,6). Therefore, it is important to discover novel biomarkers that may be clinically useful in the field of neuro-oncology.

MicroRNAs (miRNAs) are small, non-coding RNAs that are 19–24 nucleotides in length, and when combined with target mRNAs, they can function to post-transcriptionally regulate gene expression (7,8). miRNAs have become a hotspot for research in the field of biology, and an increasing number of studies have identified the underlying mechanism of action for numerous miRNAs (miRs) (9,10). miR-181d is a member of the miR-181 family, which includes miR-181a, miR-181b, miR-181c and miR-181d (11,12). miR-181d is strongly expressed in a wide range of cancer tissues, and it plays an important role in the regulation of tumorigenesis, metastasis and apoptosis (13,14). For example, miR-181d can regulate KRAS proto-oncogene to reduce migration, apoptosis and cell cycle transition in glioma cells (15). In addition, miR-181d downregulates methylguanine-methyltransferase to inhibit cancer migration, and it serves as a common biomarker for human glioblastoma (16). However, it has been suggested that miR-181d is upregulated in gliomas and is an indicator of poor prognosis (17). Based on the evidence that miR-181d acts as a prognostic factor and plays a major role in glioma, the present study performed a number of experiments to further assess the expression and function of miR-181d in glioma.

Insulin like growth factor 1 (IGF1) is a member of a family of proteins involved in the regulation of growth and development, and this protein is structurally and functionally similar to insulin (18,19). IGF1 is a cyclic polypeptide consisting of 70 amino acids and it plays vital roles in cellular proliferation and metabolism (20). In colorectal cancer, IGF1 can modulate cell proliferation and migration (21). Moreover, Wang et al (22) revealed that exposure to an IGF1 inhibitor abrogated cellular proliferation and invasion in glioma.

As an important signal transduction pathway, the PI3K/AKT/mTOR signaling pathway plays an important role in cellular proliferation, apoptosis and other processes (23). AKT phosphorylates Bcl2 to initiate apoptosis, inhibits the activity of the proteolytic enzyme Caspase-9 and activates the apoptotic cascade (24). mTOR is a downstream target gene of PI3K/AKT and this protein is indispensable for tumorigenesis (25). Moreover, brusatol regulates cell proliferation or apoptosis via the PI3K/AKT/mTOR signaling pathway in clear cell renal cell carcinoma and hepatocellular carcinoma (26,27). In glioma, oxymatrine induces cell cycle arrest and apoptosis via the PI3K/AKT/mTOR pathway (28). In the present study, a miR-181d mimic, IGF1 inhibitor, PI3K/AKT inhibitor and miR-181d inhibitor were used to treat cells, and the effects of these treatments on cellular proliferation, cell cycle progression and apoptosis were assessed. It was demonstrated that miR-181d promotes cellular proliferation via the PI3K/AKT/mTOR pathway.

Materials and methods

Cell lines and cell culture

The glioma cell line U251 was obtained from the American Type Culture Collection and cultured in a 37°C incubator until the confluence was ~80%. The cells were maintained in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (cat. no. 10270-106; Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin at 37°C in a humidified atmosphere containing 5% CO2.

Cell transfection and selection

To detect the functions of miR-181d in glioma cells, the miR-181d mimic, miR-181d inhibitor and a negative control were synthesized from Shanghai GenePharma Co., Ltd. The miR-181d mimic was a double strand that was formed using a mature miR-181d sequence and the complementary sequence (5′-AACAUUCAUUGUUGUCGGUGGGU-3′), while the miR-181d inhibitor was a single strand consisting of the complementary sequence of the mature miR-181d sequence (5′-UUGUAAGUAACAACAGCCACCCA-3′). The transfection was performed using 5 µl Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and 30 nM miR-181d mimic, 50 nM miR-181d inhibitor or 30 nM negative control that was diluted into 250 µl Opti-MEM/Reduced serum medium (Thermo Fisher Scientific, Inc.). At 48 h post-transfection, the cells were maintained in 400 µg/ml Geneticin (cat. no. G418; Thermo Fisher Scientific, Inc.) to select the cell lines that were stably expressing the miR-181d mimic or miR-181d inhibitor.

Western blot analysis

U251 cells at a density of 70% were washed twice with 100 ml pre-cooled 1X PBS, and RIPA buffer (Beyotime Institute of Biotechnology) containing phosphatase inhibitor was then added. The cells were lysed on ice, centrifuged at 12,000 × g for 10 min at 4°C and the supernatant containing the desired protein was obtained. The xenograft tissues were also lysed using RIPA to obtain total proteins. Total proteins were quantified using the bicinchoninic acid method and the absorbance was measured at 562 nm. A total of 20 µg per lane of total protein were separated by 120 V electrophoresis on a 12% SDS-PAGE for 50 min. The blots were then transferred to PVDF membranes (EMD Millipore) at 90 V for 50 min. After blocking with 5% skimmed milk at room temperature for 1 h, the membranes were incubated with the primary antibodies overnight at 4°C with GAPDH as the internal reference. After washing three times with TBS-Tween 20 (0.05%), the membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit IgG at room temperature for 1 h. Equal amounts (500 µl) of Electrochemical Luminescence Kit (ECL) illuminating liquids A and B (Pierce; Thermo Fisher Scientific, Inc.) were mixed and used to visualize the signal on a Molecular Imager ChemiDoc XRS System (Bio-Rad Laboratories, Inc.) using Tanon MP v1.0.2.0 software (Tanon Science and Technology Co.). The primary antibodies and the secondary antibody are presented in Table I.

Table I.

Details of the antibodies used in the present study.

Table I.

Details of the antibodies used in the present study.

AntibodiesSpeciesCompanyCat. no.Dilution
IGF1RabbitAbcamAb95721:10,000
PI3KRabbitAbcamAb1916061:1,000
p-PI3KRabbitAbcamAb1826511:1,000
AKTRabbitAbcamAb88051:500
p-AKTRabbitAbcamAb384491:1,000
mTORRabbitAbcamAb27321:2,000
p-mTORRabbitAbcamAb1092681:5,000
Bcl2RabbitAbcamAb1828581:2,000
Caspase-3RabbitAbcamAb138471:500
Caspase-8RabbitAbcamAb259011:1,000
Caspase-9RabbitAbcamAb2020681:2,000
Cyclin DRabbitAbcamAb166631:200
Cyclin ERabbitAbcamAb339111:2,000
Ki67RabbitAbcamAb927421:5,000
IDH-1RRabbitAbcamAb945711:1,000
IGFB2RabbitAbcamAb914041:500
PTENRabbitAbcamAb321991:10,000
TERTRabbitAbcamAb1915231:1,000
GAPDHRabbitCST21181:1,000
Goat anti rabbit IgGGoatBio-swamp life sciencePAB1500111:10,000

[i] CST, Cell Signaling Technology, Inc.; p-, phosphorylated; TERT, telomerase reverse transcriptase; IDH-1R, isocitrate dehydrogenase 1 receptor; IGFBP2, insulin like growth factor binding protein 2; IGF1, Insulin like growth factor 1.

Cell Counting Kit-8 (CCK-8) assay

CCK-8 (Bioswamp Life Science Lab) was used to determine cell proliferation according to the manufacturer's instructions. A total of 3×103 U251 cells were seeded into 96-well plates and cultured for 24 h at 37°C in a humidified atmosphere containing 5% CO2. Subsequently, the IGF1 inhibitor (Linsitinib; OSI-906, SelleckChemicals) or the PI3K/AKT inhibitor (LY294002, Sigma-Aldrich; Merck KGaA) were added to the cells and incubated for 24 h at room temperature. The CCK-8 solution (10 µl) was added to the cells, which were then cultured for 4 h at 37°C. The absorbance at 450 nm was subsequently measured using an ELISA plate reader (BioTek Instruments, Inc.).

Cell cycle

Cell cycle progression was detected by flow cytometry. The cells transfected with mir-181d mimic, IGF1 inhibitor, PI3K/AKT inhibitor or mir-181d inhibitor were suspended in a PBS solution containing 10% FBS, and then the cells were fixed in absolute ethyl ethanol at room temperature for 24 h. Next, the samples were centrifuged at 3,000 × g for 30 sec at room temperature and the supernatant was discarded. Subsequently, the cell pellet was suspended in 100 µl RNase A solution (1 mg/ml) and digested at 37°C for 10 min. Cells were then stained at room temperature for 10 min using 400 µl propidium iodide (PI, 50 µg/ml, Nanjing KeyGen Biotech Co., Ltd.) in the dark and analyzed by a FC500 MCL flow cytometer (Beckman Coulter) using MODFIT LT 2.0 (Verity Software House, Inc.). The proportion of cells in G0/G1, S and G2/M stages was evaluated, and all procedures were performed in triplicate.

Cell apoptosis

Cellular apoptosis was detected using the Annexin-V/PI detection kit (Nanjing KeyGen Biotech Co., Ltd.) according to the manufacturer's protocol. The transfected cells were suspended and centrifuged at 1,000 × g for 5 min at room temperature. The supernatant was then removed, and the cells were resuspended in 200 µl binding buffer. The cells were stained with 10 µl Annexin V-FITC and 10 µl PI and then incubated for 30 min at 4°C in the dark. Cellular apoptosis was detected under a FC500 MCL flow cytometer (Beckman Coulter, Inc.) and analyzed using CXP Analysis 2.0 software (Beckman Coulter, Inc.). The apoptotic rate was calculated by the percentage of early and late apoptotic cells.

Animal study

Thirty male BALB/C nude mice (age, 4–6 weeks, weight, 20 g) were purchased from the Experimental Animal Center of the Chinese Academy of Sciences and were prepared for this experiment. All mice were housed at 22±2°C, 45–60% humidity and a 12 h light/day cycle. All mice received food and water ad libitum. For the xenograft tumor model, 5×106 U251 cells were injected subcutaneously into the axilla of the nude mice. After the cells were cultured to the size of ~3 mm, the nude mice were randomly divided into five groups for drug treatment three times a week for a total of three weeks. The five groups included the control, overexpression (10 µg miR-181d-mimic by injection), interference (10 µg miR-181d-inhibitor by injection), IGF-1 inhibitor (20 mg/kg/day Linsitinib, by gavage)and PI3K/AKT inhibitor (100 mg/kg LY294002, by gavage) groups.

After the xenograft model was established, the length (L) and width (W) of the subcutaneous tumors were measured every 2 days using a vernier caliper. Tumor volume was calculated using the formula (L × W2)/2. On the 28th day after seeding the cells, the mice were sacrificed to obtain tumor specimens. The experiment was performed according to the revised guidelines for the Care and Use of Experimental Animals (National Institutes of Health) (29). The experimental program has been approved by the Ethics Committee of Renmin Hospital of Wuhan University (Hubei, China).

Statistical analysis

The data were analyzed using SPSS 16.0 software (IBM Corp.) and GraphPad Prism software (version 6; GraphPad Software, Inc.) and are presented as the mean ± SD. Unpaired Student's t-test and one-way ANOVA followed by Bonferroni post-hoc test were applied to determine the significance of differences among the various groups. P<0.05 was considered to indicate a statistically significant difference. All experiments were performed in triplicate.

Results

miR-181d promotes cellular proliferation via the PI3K/AKT/mTOR signaling pathway in U251 cells

To investigate the functions of miR-181d in glioma cells, a miR-181d mimic, a miR-181d inhibitor and a negative control were synthesized. In addition, IGF1 and PI3K/AKT inhibitors were used to examine whether miR-181d regulated cell proliferation via the PI3K/AKT/mTOR pathway. U251 cells were transfected or treated with miR-181d mimic, IGF1 inhibitor, PI3K/AKT inhibitor or miR-181d inhibitor (Fig. S1). After evaluating the expression of IGF1, it was identified that miR-181d mimic significantly increased (P=0.0039) its expression, while the IGF1, PI3K/AKT and miR-181d inhibitors significantly reduced (P=0.0001, P=0.0002 and P=0.0003, respectively) the expression of IGF1 (Fig. 1A).

The proliferative abilities of the cells were calculated, and it was demonstrated that proliferation was increased after exposure to the miR-181d mimic (P=0.0001), but was inhibited by treatment with the IGF1, PI3K/AKT and miR-181d inhibitors (P<0.0001; Fig. 1B).

In addition, western blot analysis indicated that the miR-181d mimic promoted the expression levels of phosphorylated (p)-PI3K, p-AKT and p-mTOR (P=0.042, P=0.0127 and P=0.0058), while the expression levels of PI3K, AKT and mTOR were unchanged in U251 cells. However, treatment with the IGF1 inhibitor (P=0.0028, P<0.0001 and P=0.0147, respectively), the PI3K/AKT inhibitor (P=0.0011, P<0.0001 and P=0.0055, respectively) and the miR-181d inhibitor (P=0.0014, P=0.0003 and P=0.0009, respectively) suppressed the expression levels of p-PI3K, p-AKT and p-mTOR (Fig. 1C), thus suggesting that miR-181d regulates cellular proliferation via the PI3K/AKT/mTOR pathway.

miR-181d promotes cell cycle progression via the IGF1/PI3K/AKT axis in U251 cells

The cell cycle profile of U251 cells was detected using a flow cytometry-based assay. The proportions of cells in the G0/G1 phase after treatment with negative control, miR-181d mimic, IGF1 inhibitor, PI3K/AKT inhibitor and miR-181d inhibitor were 43.04±1.43, 39.57±0.97, 64.92±1.85, 60.16±2.47 and 66.65±3.46%, respectively. Under these same conditions, the proportions in S phase were 40.21±1.50, 43.61±1.03, 25.38±2.37, 28.49±1.64 and 24.79±2.32%, while the proportions in G2/M phase were 16.75±1.55, 16.82±1.45, 9.70±1.05, 11.35±1.42 and 8.56±0.92%, respectively (Fig. 2A). Compared with the negative control, the proportion of G0/G1 phase cells in the miR-181d mimic group was decreased significant (P=0.0254), while the proportions in the groups treated with the IGF1, PI3K/AKT and miR-181d inhibitors were significantly increased (P<0.0001, P=0.0005 and P=0.0004, respectively). In S phase, compared with the negative control, the proportion of cells was significantly increased in the miR-181d mimic group (P=0.0318), while the proportions in the groups treated with the IGF1, PI3K/AKT and miR-181d inhibitors were significantly reduced (P=0.0008 for all; Fig. 2B).

Proteins associated with cell cycle, including Ki67, isocitrate dehydrogenase 1 receptor (IDH-1R), insulin like growth factor binding protein 2 (IGFBP2), PTEN and telomerase reverse transcriptase (TERT), were assessed by western blotting. The expression levels of Ki67 (P=0.0023), IDH-1R (P=0.0019), IGFBP2 (P=0.0065) and TERT (P=0.0010) were enhanced by miR-181d mimic, while the expression of PTEN was inhibited (P=0.0015). However, the expression levels of Ki67 (P=0.0002, P<0.0001 and P=0.0004, respectively), IDH-1R (P=0.0003, P=0.0008 and P=0.0004, respectively), IGFBP2 (P=0.0002, P=0.0002 and P=0.0005, respectively) and TERT (P=0.0007, P=0.0006 and P=0.0002, respectively) were suppressed, but PTEN expression (P<0.0001, P=0.0006 and P=0.0004, respectively) was increased by treatment with the IGF1 inhibitor, PI3K/AKT inhibitor and miR-181d inhibitor (Fig. 2C). Collectively, these results suggested that the cell cycle was promoted by miR-181d via the IGF1/PI3K/AKT pathway in U251 cells.

miR-181d inhibits cellular apoptosis via the IGF1/PI3K/AKT axis in U251 cells

Flow cytometry was performed to investigate the influence of miR-181d on apoptosis in U251 cells. The apoptotic rates for the negative control, miR-181d mimic, IGF1 inhibitor, PI3K/AKT inhibitor and miR-181d inhibitor groups were 7.08±0.56, 4.90±0.43, 22.22±0.74, 19.07±0.58 and 28.18±0.65, respectively (Fig. 3A). Compared with the negative control, the apoptotic rate was reduced (P=0.0059) by the miR-181d mimic, but was increased by treatment with the IGF1, PI3K/AKT and miR-181d inhibitors (P<0.0001 for all; Fig. 3B).

Apoptotic-related proteins were assessed by western blotting in U251 cells. It was indicated that the miR-181d mimic increased the expression levels of Bcl2, Cyclin D and Cyclin E (P=0.0011, P=0.0013 and P=0.0003, respectively), while it inhibited the expression levels of Caspase-3, Caspase-8 and Caspase-9 (P=0.0073, P<0.0001 and P<0.0001, respectively) in U251 cells. However, cells transfected or treated with IGF1 inhibitor (P=0.0002, P=0.0002 and P=0.0002; and P=0.0354, P=0.0038 and P<0.0001, respectively), PI3K/AKT inhibitor (P=0.0004, P=0.0003 and P=0.0010; and P=0.0067, P=0.0030 and P<0.0001, respectively) and miR-181d inhibitor (P=0.0001, P=0.0002 and P=0.0014; and P=0.0016, P=0.0013 and P<0.0001, respectively) exhibited the opposite trend (Fig. 3C). Thus, the results suggested that miR-181d inhibited cellular apoptosis via the IGF1/PI3K/AKT axis in U251 cells.

miR-181d enhances glioma xenograft growth in vivo

To determine if miR-181d regulated the growth of glioma xenografts via the IGF1/PI3K/AKT axis, U251 cells were subcutaneously injected into nude mice. Xenograft growth was promoted by the miR-181d mimic, but was inhibited by treatment with the IGF1, PI3K/AKT and miR-181d inhibitors (Fig. 4A). Moreover, xenograft weights were calculated, and it was observed that the weights in the miR-181d mimic group were higher compared with the negative control (P=0.0471). The xenograft weights were also decreased in the IGF1 inhibitor (P=0.0430), PI3K/AKT inhibitor (P=0.0459) and miR-181d inhibitor (P=0.0232) groups (Fig. 4B). In addition, the xenograft volumes was significantly increased in the miR-181d mimic group (P=0.0479), but significantly decreased in the IGF1 inhibitor (P=0.0267), PI3K/AKT inhibitor (P=0.0411) and miR-181d inhibitor groups (P=0.0011) compared with the negative group (Fig. 4C). The maximum diameter of the tumors in the miR-181d mimic group was higher compared with the negative control group, while those in the IGF1 inhibitor and miR-181d inhibitor groups were lower (Fig. S2).

miR-181d promotes cell cycle progression and suppresses cellular apoptosis in glioma xenograft tissues

To assess whether miR-181d mediated cell cycle progression via the IGF1/PI3K/AKT axis, the xenograft tissues were lysed using RIPA to obtain total proteins. Similar to the in vitro experiments, the miR-181d mimic enhanced the expression of IGF1 (P=0.0002), while treatment with the IGF1 inhibitor (P=0.0012), PI3K/AKT inhibitor (P=0.0021) and miR-181d inhibitor (P=0.0002) reduced the expression of IGF1 (Fig. 5A).

With regards to the proteins in the PI3K/AKT/mTOR signaling pathway, the expression levels of p-PI3K, p-AKT and p-mTOR were increased by treatment with the miR-181d mimic (P=0.0041, P=0.0025 and P=0.0224, respectively). However, these were reduced by treatment with the IGF1 inhibitor (P=0.0001, P=0.0042 and P=0.0003), PI3K/AKT inhibitor (P=0.0001, P=0.0061 and P=0.0005) and miR-181d inhibitor (P=0.0001, P=0.0020 and P=0.00032; Fig. 5B).

The proteins associated with apoptosis were evaluated by western blotting. The expression levels of Bcl2, Cyclin D and Cyclin E were increased (P=0.0026, P=0.0004 and P=0.0025), while Caspase-3, Caspase-8 and Caspase-9 expression levels were decreased (P=0.0054, P=0.0002 and P=0.0010) in U251 cells transfected with miR-181d mimic. In contrast, treatment with the IGF1, PI3K/AKT and miR-181d inhibitors reduced the expression levels of Bcl2 (P=0.0090, P=0.0072 and P=0.0095, respectively), Cyclin D (P=0.0092, P=0.0014 and P=0.0016) and Cyclin E (P<0.0001, P=0.0002 and P=0.0001), while treatment increased the expression levels of Caspase-3 (P=0.0001, P<0.0001 and P<0.0001), Caspase-8 (P=0.0002, P=0.0003 and P=0.0006) and Caspase-9 (P=0.0003, P<0.0001 and P=0.0002) in U251 cells (Fig. 5C). Therefore, the results suggested that miR-181d inhibited cell apoptosis via the IGF1/PI3K/AKT axis.

For the cell cycle-related proteins, the miR-181d mimic increased the expression levels of Ki67, IDH-1R, IGFBP2 and TERT (P=0.0003, 0.0103, 0.0012 and 0.0006, respectively), but decreased the expression of PTEN (P<0.0001). However, the expression levels of Ki67 (P=0.0001, P=0.0004 and P=0.0001, respectively), IDH-1R (P=0.0161, P=0.0027 and P=0.0019, respectively), IGFBP2 (P=0.0003, P=0.0092 and P=0.0001, respectively) and TERT (P=0.0030, P=0.0005 and P=0.0004, respectively) were reduced by treatment with the IGF1, PI3K/AKT and miR-181d inhibitors, while PTEN expression was increased (P=0.0033, P=0.0036 and P=0.0030, respectively; Fig. 5D).

Discussion

Glioma is a common brain tumor, most commonly caused by genetic abnormalities, with an incidence rate of 3–8 cases per 100,000 worldwide (2). Thus, it is important to investigate novel biomarkers that could be useful for the early diagnosis and treatment of glioma. miRNAs regulate gene expression and protein degradation by binding to the complementary DNA sequences on the 3′untranslated regions of the target mRNAs (7,8). A previous study reported that miR-181d mediated the proliferation, as well as promoted the maturation of dendritic cells (30). Moreover, the present results indicated that miR-181d promoted the expression of IGF1, cellular proliferation and the growth of glioma xenografts.

Ho et al (31) revealed that miR-181d was negatively correlated with the expression of IGF1 in glioma cells. In addition, Chen et al (32) demonstrated that the IGF1/PI3K/AKT pathway was involved in drug-induced cell progression in breast cancer. To investigate whether miR-181d regulated cellular progression via the IGF1/PI3K/AKT axis, the present study assessed cell proliferation, cell cycle progression and cellular apoptosis after treatment with IGF1, PI3K/AKT andmiR-181d inhibitors. Consistent with previous findings (31,32), treatment with the IGF1 and PI3K/AKT inhibitors suppressed the proliferative ability and inhibited the PI3K/AKT/mTOR pathway in U251 cells. Zhang et al (33) reported that miR-181d promoted cell cycle progression in uveal melanoma. In line with this finding, the present results suggested that miR-181d promoted cell cycle progression, while treatment with IGF1 or PI3K/AKT inhibitors suppressed cell cycle progression in U251 cells. In addition, miR-181d mimic inhibited apoptosis while treatment with an IGF1 inhibitor and a PI3K/AKT inhibitor promoted apoptosis in U251 cells. A xenograft model was constructed to assess these results in vivo, and it was observed that the growth of glioma xenografts was improved by miR-181d mimic, while it was suppressed by treatment with IGF1 or PI3K/AKT inhibitors.

In conclusion, the present results indicated that miR-181d promoted cellular proliferation via the PI3K/AKT/mTOR signaling pathway in both U251 cells and xenograft tissues. Furthermore, miR-181d promoted cell cycle progression and suppressed apoptosis via the IGF1/PI3K/AKT axis in xenograft tissues derived from U251 cells. It was also demonstrated that miR-181d enhanced the growth of glioma xenografts in vivo.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The current study was supported by grants from the National Natural Science Foundation of China (grant nos. 81572489 and 81372683) and the Hubei Province Health and Family Planning Scientific Research Project (grant no. WJ2007Q008).

Availability of data and materials

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

Authors' contributions

QC and BL contributed to the conception of the study and analyzed the data. DT performed the experiments and wrote the manuscript. WG, JY and JL analyzed the data and wrote the manuscript. JZ and JG analyzed the data and provided constructive criticism. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal experiments were approved by the Renmin Hospital of Wuhan University Animal Care and Use Committee (approval no. 20180908).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Sayegh ET, Kaur G, Bloch O and Parsa AT: Systematic review of protein biomarkers of invasive behavior in glioblastoma. Mol Neurobiol. 49:1212–1244. 2014. View Article : Google Scholar : PubMed/NCBI

3 

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

4 

Kim GW, Lee DH, Yeon SK, Jeon YH, Yoo J, Lee SW and Kwon SH: Temozolomide-resistant glioblastoma depends on HDAC6 activity through regulation of DNA mismatch repair. Anticancer Res. 39:6731–6741. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Linz U: Commentary on effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial (Lancet Oncol. 2009; 10: 459–466). 2009; 10: 459–466). Cancer. 116:1844–1846. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, 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

7 

Lai EC: Micro RNAs are complementary to 3′UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet. 30:363–364. 2002. View Article : Google Scholar : PubMed/NCBI

8 

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

9 

Adams BD, Kasinski A and Slack FJ: Aberrant regulation and function of microRNAs in cancer. Curr Biol. 24:R762–R776. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Cowland JB, Hother C and Grønbaek K: MicroRNAs and cancer. APMIS. 115:1090–1106. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Chen H, Liu T, Liu J, Feng Y, Wang B, Wang J, Bai J, Zhao W, Shen Y, Wang X, et al: Circ-ANAPC7 is upregulated in acute myeloid leukemia and appears to target the MiR-181 family. Cell Physiol Biochem. 47:1998–2007. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Xia S, Tian H, Fan L and Zheng J: Peripheral blood miR-181-5p serves as a marker for screening patients with osteoarthritis by targeting TNFα. Clin Lab. 63:1819–1825. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Zhu W, Shan X, Wang T, Shu Y and Liu P: miR-181b modulates multidrug resistance by targeting BCL2 in human cancer cell lines. Int J Cancer. 127:2520–2529. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Guo X, Zhu Y and Hong X, Zhang M, Qiu X, Wang Z, Qi Z and Hong X: miR-181d and c-myc-mediated inhibition of CRY2 and FBXL3 reprograms metabolism in colorectal cancer. Cell Death Dis. 8:e29582017. View Article : Google Scholar : PubMed/NCBI

15 

Wang XF, Shi ZM, Wang XR, Cao L, Wang YY, Zhang JX, Yin Y, Luo H, Kang CS, Liu N, et al: MiR-181d acts as a tumor suppressor in glioma by targeting K-ras and Bcl-2. J Cancer Res Clin Oncol. 138:573–584. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Zhang W, Zhang J, Hoadley K, Kushwaha D, Ramakrishnan V, Li S, Kang C, You Y, Jiang C, Song SW, et al: miR-181d: A predictive glioblastoma biomarker that downregulates MGMT expression. Neuro Oncol. 14:712–719. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Chen YY, Ho HL, Lin SC, Ho TDH and Hsu CY: Upregulation of miR-125b, miR-181d, and miR-221 predicts poor prognosis in MGMT promoter-unmethylated glioblastoma patients. Am J Clin Pathol. 149:412–417. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Allard JB and Duan C: IGF-binding proteins: Why do they exist and why are there so many? Front Endocrinol (Lausanne). 9:1172018. View Article : Google Scholar : PubMed/NCBI

19 

Duan C, Ren H and Gao S: Insulin-like growth factors (IGFs), IGF receptors, and IGF-binding proteins: Roles in skeletal muscle growth and differentiation. Gen Comp Endocrinol. 167:344–351. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Nielsen EM, Hansen L, Lajer M, Andersen KL, Echwald SM, Urhammer SA, Hansen T and Pedersen O: A common polymorphism in the promoter of the IGF-I gene associates with increased fasting serum triglyceride levels in glucose-tolerant subjects. Clin Biochem. 37:660–665. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Li Z, Pan W, Shen Y, Chen Z, Zhang L, Zhang Y, Luo Q and Ying X: IGF1/IGF1R and microRNA let-7e down-regulate each other and modulate proliferation and migration of colorectal cancer cells. Cell Cycle. 17:1212–1219. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Wang H, Tang C, Na M, Ma W, Jiang Z, Gu Y, Ma G, Ge H, Shen H and Lin Z: miR-422a inhibits glioma proliferation and invasion by targeting IGF1 and IGF1R. Oncol Res. 25:187–194. 2017. View Article : Google Scholar : PubMed/NCBI

23 

LoRusso PM: Inhibition of the PI3K/AKT/mTOR pathway in solid tumors. J Clin Oncol. 34:3803–3815. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Chen B, Xue Z, Yang G, Shi B, Yang B, Yan Y, Wang X, Han D, Huang Y and Dong W: Akt-signal integration is involved in the differentiation of embryonal carcinoma cells. PLoS One. 8:e648772013. View Article : Google Scholar : PubMed/NCBI

25 

Saiki S, Sasazawa Y, Imamichi Y, Kawajiri S, Fujimaki T, Tanida I, Kobayashi H, Sato F, Sato S, Ishikawa KI, et al: Caffeine induces apoptosis by enhancement of autophagy via PI3K/Akt/mTOR/p70S6K inhibition. Autophagy. 7:176–187. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Liu G, Zhao X, Zhou J, Cheng X, Ye Z and Ji Z: LncRNA TP73-AS1 promotes cell proliferation and inhibits cell apoptosis in clear cell renal cell carcinoma through repressing KISS1 expression and inactivation of PI3K/Akt/mTOR signaling pathway. Cell Physiol Biochem. 48:371–384. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Ye R, Dai N, He Q, Guo P, Xiang Y and Zhang Q, Hong Z and Zhang Q: Comprehensive anti-tumor effect of Brusatol through inhibition of cell viability and promotion of apoptosis caused by autophagy via the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomed Pharmacother. 105:962–973. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Dai Z, Wang L, Wang X, Zhao B, Zhao W, Bhardwaj SS, Ye J, Yin Z, Zhang J and Zhao S: Oxymatrine induces cell cycle arrest and apoptosis and suppresses the invasion of human glioblastoma cells through the EGFR/PI3K/Akt/mTOR signaling pathway and STAT3. Oncol Rep. 40:867–876. 2018.PubMed/NCBI

29 

National Research Council (US) Committee on Recognition and Alleviation of Pain in Laboratory Animals, . Recognition and alleviation of pain in laboratory animals. Washington (DC): National Academies Press (US); 2009

30 

Su XW, Lu G, Leung CK, Liu Q, Li Y, Tsang KS, Zhao SD, Chan DTM, Kung HF and Poon WS: miR-181d regulates human dendritic cell maturation through NF-κB pathway. Cell Prolif. 50:e123582017. View Article : Google Scholar

31 

Ho KH, Chen PH, His E, Shih CM, Chang WC, Cheng CH, Lin CW and Chen KC: Identification of IGF-1-enhanced cytokine expressions targeted by miR-181d in glioblastomas via an integrative miRNA/mRNA regulatory network analysis. Sci Rep. 7:7322017. View Article : Google Scholar : PubMed/NCBI

32 

Chen J, Zeng J, Xin M, Huang W and Chen X: Formononetin induces cell cycle arrest of human breast cancer cells via IGF1/PI3K/Akt pathways in vitro and in vivo. Horm Metab Res. 43:681–686. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Zhang L, He X, Li F, Pan H, Huang X, Wen X, Zhang H, Li B, Ge S, Xu X, et al: The miR-181 family promotes cell cycle by targeting CTDSPL, a phosphatase-like tumor suppressor in uveal melanoma. J Exp Clin Cancer Res. 37:152018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 22 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tang D, Gao W, Yang J, Liu J, Zhao J, Ge J, Chen Q and Liu B: miR‑181d promotes cell proliferation via the IGF1/PI3K/AKT axis in glioma. Mol Med Rep 22: 3804-3812, 2020
APA
Tang, D., Gao, W., Yang, J., Liu, J., Zhao, J., Ge, J. ... Liu, B. (2020). miR‑181d promotes cell proliferation via the IGF1/PI3K/AKT axis in glioma. Molecular Medicine Reports, 22, 3804-3812. https://doi.org/10.3892/mmr.2020.11464
MLA
Tang, D., Gao, W., Yang, J., Liu, J., Zhao, J., Ge, J., Chen, Q., Liu, B."miR‑181d promotes cell proliferation via the IGF1/PI3K/AKT axis in glioma". Molecular Medicine Reports 22.5 (2020): 3804-3812.
Chicago
Tang, D., Gao, W., Yang, J., Liu, J., Zhao, J., Ge, J., Chen, Q., Liu, B."miR‑181d promotes cell proliferation via the IGF1/PI3K/AKT axis in glioma". Molecular Medicine Reports 22, no. 5 (2020): 3804-3812. https://doi.org/10.3892/mmr.2020.11464