Glioma-initiating cells (GIC) have stem-like cell properties thought to be sufficient for recurrence, progression, and drug resistance in glioblastomas. In the present study, we defined miRNA (miR)-340 as a differentially expressed miRNA in human GICs that inhibit GIC-mediated tumorigenesis. Furthermore, we defined tissue plasminogen activator (PLAT) as a critical direct target of miR340 for inhibition. Among miRNAs screened, we found that miR340 expression was decreased in all human GICs and in human glioblastoma tissues, compared with human neural stem cells and normal brain tissues. miR340 overexpression in GICs suppressed their proliferative, invasive, and migratory properties in vitro, triggering cell senescence in vitro and inhibiting GIC-induced tumorigenesis in mouse brains. shRNA-mediated silencing of PLAT in GICs phenocopied the effects of miR340 overexpression in vitro and in vivo, suggesting a potential role for tissue factor in stem-like cell function. Taken together, our results identified miR340 as a tumor suppressor that functions in GIC to enforce PLAT blockade and ablate their stem-like functions. Cancer Res; 75(6); 1123–33. ©2015 AACR.

Glioblastoma multiforme (GBM) is the most common and aggressive malignant brain tumor. Despite the most intensive current therapeutic efforts, the median survival time (∼14 months) has not changed significantly in decades (1). The extremely poor prognosis of patients with GBM is likely because of the presence of glioma-initiating cells (GIC; also known as glioma stem-like cells), which are equivalent of the cancer stem cells (CSC) observed in other cancer types (2–4). CSCs are not only highly resistant to chemo- and radiotherapy (5–7) but are also highly tumorigenic; thus, these cells are thought to be the primary cause of tumor recurrence and progression. To improve the poor prognosis of patients with GBM, it is important to understand the mechanism that GICs are activated to have high tumorigenesis and invasion.

miRNAs (miR) are small, noncoding RNAs of approximately 22 nucleotides in length that mediate the posttranscriptional silencing of specific target mRNAs and that are currently recognized as important regulators of tumorigenesis and development (8). Many miRNAs have been reported to be aberrantly expressed in malignant gliomas and play a role in determining the degree of malignancy. These include miR21, miR221/222, miR124, and miR128 (9–12). These miRNAs can act as either oncogenes or tumor suppressors in gliomas, depending on their effects on cell proliferation and apoptosis. However, there are few studies on integrated functions of miRNAs that significantly regulate tumorigenesis and tumor development initiated by GICs.

In the present study, we comprehensively analyzed the miRNA expression profiles of human GICs (hGIC) and identified miR340 as a novel miRNA that is significantly downregulated in hGICs compared with human neural stem cells (NSC). Functional analyses revealed that miR340 suppressed hGIC proliferation, invasion, and migration in vitro, as well as hGIC tumorigenesis in vivo, in the mouse brain. Furthermore, we defined tissue plasminogen activator (PLAT) gene as a direct target of miR340. These results indicate that GICs with the decreased level of miR340 promote glioblastoma formation in the mouse brain and that miR340 downregulation can induce a variety of malignant processes, such as cell proliferation and diffuse invasion. We also demonstrated that these effects are primarily mediated by increased levels of the target molecule PLAT. These findings reveal miR340 and its target gene PLAT as potentially useful therapeutic candidates for the treatment of glioblastoma.

Animals and chemical reagents

The mice were obtained from CLEA Japan, Inc. The mouse experiments were performed according to protocols approved by the Animal Care and Use Committees of Ehime University (Ehime, Japan) and of Hokkaido University (Hokkaido, Japan). Chemicals and growth factors were purchased from Invitrogen and PeproTech, respectively, except where otherwise indicated.

Glioma-initiating cell culture and cell lines

Seven primary human glioma samples [five GBMs (E1-4, 6), one anaplastic oligodendroglioma (AO), and one diffuse astrocytoma (DA)] were used to prepare the hGICs. These seven samples were obtained from Ehime University Hospital with the patients' consent according to the Research Ethics Committee guidelines and were used in compliance with the research guidelines of the Ehime University Graduate School of Medicine and the Institute for Genetic Medicine of Hokkaido University. Tumor samples were dissociated using a papain dissociation system (Worthington) according to the manufacturer's instructions. The dissociated cells were cultured to form tumor spheres in serum-free DMEM/Ham's F-12 (Wako) supplemented with human basic fibroblast growth factor (bFGF; 10 nmol/L), human EGF (10 nmol/L), heparin (5 μmol/L), N2 supplement (Wako), 10 μg/mL insulin (Wako), GlutaMAX supplement, 100 U/mL penicillin G, and 100 μg/mL streptomycin. For IHC, GICs were cultured on poly-D-lysine (15 μg/mL, Sigma)-coated and fibronectin (1 μg/mL)-coated 8-well chamber slides (Nunc). The human glioma cell lines U251 and U87 (obtained from ATCC) were cultured in DMEM with 10% FBS (Thermo Scientific). The mouse GIC lines NSCL61 and OPCL61, that have been established by overexpression of oncogenic HRasL61 in p53-deficient mouse NSC and oligodendrocyte precursor cell, were cultured as described previously (13, 14). Human NSCs (H9 human embryonic stem cell derived; Invitrogen) were cultured according to the supplier's instructions. The cells were maintained at 37°C under a humidified 5% CO2/95% air atmosphere for all of the experiments. The characterization of human GICs is presented in the Supplementary Fig. S1.

Microarray hybridization and data processing

Total RNA was extracted from GICs, glioma cells, and glioma tissues using the TRIzol Plus RNA Purification System (Invitrogen). The miRNA microarrays were manufactured by Agilent Technologies, and 100 ng total RNA was hybridized using the miRNA Microarray Kit protocol for use with Human miRNA Microarray Release 16.0 or Mouse miRNA Microarray Release 16.0. Hybridization signals were detected using a DNA microarray scanner (Agilent Technologies), and the scanned images were analyzed using Agilent Feature Extraction software.

For the gene expression analyses, total RNA was amplified and labeled with cy3 using a one-color Agilent Low Input Quick Amp Labeling Kit (Agilent Technologies) according to the manufacturer's instructions. Labeled cRNAs were fragmented and hybridized to the Agilent Human GE 8×60K Microarray. After washing, the microarrays were scanned using a DNA microarray scanner. Intensity values for each scanned feature were quantified using Agilent Feature Extraction software, which also performs background subtraction corrections.

Data normalization was conducted using Agilent GeneSpring GX version 11.0.2 software. After normalization, hierarchical sample clustering of the expressed genes was performed using the Euclidean distance and average linkage methods (Agilent GeneSpring GX). We used the freely available GenMAPP 2.1 database (http://www.genmapp.org/) and ConPath Navigator software, in combination with custom software programs, for the pathway analysis.

The microarray data have been submitted to NCBI GEO and available under GSE61078, GSE61079, and GSE61080.

Real-time PCR

Total RNA was extracted from GICs, glioma cells, and glioblastoma and normal brain tissues using Isogen reagent (Nippon Gene). Then, total RNA was reverse-transcribed using MultiScribe Reverse Transcriptase (Applied Biosystems) and MMLV RT (Invitrogen) according to the suppliers' instructions. miR340 expression was analyzed using TaqMan small RNA assays (Applied Biosystems) and a MiniOpticon RT-PCR System (Bio-Rad) according to the suppliers' instructions. PLAT expression was analyzed using FastStart Universal SYBR Green (Roche) with a MiniOpticon RT-PCR System. The PCR conditions were as follows: 10 minutes at 95°C; followed by 40 cycles of 95°C for 15 seconds and 60°C for 1 minute. All of the expression values were normalized against β-actin mRNA expression levels. The RT-PCR primers for PLAT and β-actin are listed in Supplementary Table S1.

Transfections

Either the precursor form of miR340 or a control miRNA was overexpressed in GICs and glioma cells using miRNA lentiviral particles (pEZX-MR03; GeneCopoeia) according to the supplier's instructions. In certain experiments, cells were transfected with a miR340 expression vector (pBApoCMV-Neo-miR340, Takara) using a Nucleofector device (Lonza) and cultured in the presence of neomycin (300 μg/mL; Sigma) for 10 days.

Cell proliferation and cell-cycle analyses

GICs and glioma cells (1×104 cells/well) were transfected with either a control vector or a miR340 expression vector and cultured for 72 hours. Cells were collected on days 1, 2, and 3 and counted using a hemocytometer. Bromodeoxyuridine (BrdUrd) staining was performed as previously described (15). Cell-cycle analysis was performed using propidium iodide staining and FACS analysis (BD FACSCalibur, Cycletest Plus DNA Reagent Kit, BD Biosciences) according to the supplier's instructions.

Cell invasion and migration assays

Cell invasion and migration were assessed using a BioCoat Matrigel Invasion Chamber (Becton-Dickinson) and the BioCoat Cell Culture Inserts (Becton-Dickinson), respectively. Briefly, transfected GICs and glioma cells (5–10×104) were resuspended in DMEM/Ham's F-12 medium, which was supplemented with 1 mg/mL BSA (Sigma), and transferred to the upper chamber of each well. Then, medium containing 10% FBS was added to the lower chamber. After incubation for 12 hours, the cells on the upper membrane surface were mechanically removed. Cells that had invaded or migrated to the lower side of the membrane were fixed, stained with 0.1% crystal violet, and counted the amount of cells in five random fields under a microscope (×400).

Intracranial cell transplantation into the brains of NOD/SCID mice

Control and miR340-overexpressing cells (1×105) were suspended in 5 μL of culture medium and injected into the brains of 6 to 8-week-old female NOD/SCID mice that had been anesthetized with 10% pentobarbital. The stereotactic coordinates of the injection site were 2 mm forward from lambda, 2 mm lateral to the sagittal suture, and 5 mm deep. MRI was taken to confirm the tumor formation before decapitation of the mice.

Brain fixation and histopathology

Dissected mouse brains were fixed in 4% paraformaldehyde at 4°C overnight. After fixation, the brains were embedded in paraffin, cut into 6-μm thick coronal sections, and stained with hematoxylin and eosin (H&E).

IHC

IHC was performed as previously described (15). The following antibodies were used to detect cellular antigens: mouse monoclonal anti-Nestin (1:200; Pharmingen), mouse monoclonal anti-GFAP (1:200; Sigma), mouse monoclonal anti-Sox2 (1:200; Stem Cell Technology), rabbit polyclonal anti-PLAT (1:50; Sigma), and rabbit monoclonal anti-caspase-3 (cleaved; 1:100; Cell Signaling Technology). The antibodies were detected using either Alexa 488-coupled goat anti-rabbit IgG (1:500; Life Technologies) or sheep anti-rabbit IgG (1:1; Nichirei). The nuclei were counterstained with DAPI (1 μg/mL) or hematoxylin.

Senescence-associated-β-galactosidase (SA-β-gal) staining

Cells transfected with either a control or a miR340 overexpression vector were cultured in the presence of 400 μg/mL G418 for 10 days. Then, selected cells were fixed and stained using a SA-β-gal staining kit (Calbiochem) according to the supplier's instructions.

Western blot analysis

Cell or tissue lysates were separated by 10% SDS-PAGE and the proteins were transferred to a polyvinylidene fluoride membrane (Amersham). The membrane was blocked in 5% nonfat milk, and then incubated in an anti-Nestin (1:1000), anti-Sox2 (1:500), anti-GFAP (1:500), anti-PLAT (1:250), or anti-GAPDH antibody (1:2000; Chemicon). After washing, the membrane was incubated in IgG-HRP (1:2000; Santa Cruz Biotechnology). An enhanced chemiluminescence system (Amersham) was used to detect the protein bands.

Vector construction

Full-length human plat was amplified from human glioma cDNA using RT-PCR and KOD-Plus-Ver.2 polymerase (Toyobo) according to the manufacturer's instructions and cloned into the pcDNA3-2×FLAG-c vector, resulting in pcDNA3-hPLAT-2×FLAG-c. The primers used for amplification of the full-length human plat gene are listed in Supplementary Table S2.

To knockdown human plat expression, hairpin sequences were inserted into the psiRNA-h7SKhygro G1 expression vector (InvivoGen) to produce psiRNA-h7SKhygro-platsh. The shRNA target sequence was 5′-GAATTCGATGATGACACTT-3′.

To construct a firefly luciferase-human plat 3′ untranslated region (3′UTR) expression vector, human plat 3′UTR genomic DNA was amplified using the KOD plus DNA polymerase and cloned into the pT7Blue-2 vector (Novagen), according to the manufacturer's instructions. We also amplified a mutant form of the plat 3′UTR genomic DNA, with a deleted miR340-binding site, and cloned it into the pT7Blue-2 vector. The wild and mutant types of plat 3′UTR DNA were inserted with the firefly luciferase cDNA into the pcDNA3.1-hyg vector, resulting in pcDNA3.1-hyg-Luc-PLAT 3′UTR (Luc-PLAT) and pcDNA3.1-hyg-Luc-ΔPLAT 3′UTR (Luc-ΔPLAT), respectively. The primers used to generate plat 3′UTR reporter construct are listed in Supplementary Table S3.

Luciferase assay

hGICs (E3) were transfected with pEF-Rluc (a kind gift from Koji Shimozaki and Shigekazu Nagata), a luciferase-PLAT vector (Luc-PLAT or Luc-ΔPLAT) and a miR expression vector (control vector or pBApoCMV-Neo-miR340), and their luciferase activities were measured as previously described (16).

Statistical analyses

The Mann–Whitney test was used to compare two-paired groups. Kaplan–Meier curves were used to estimate the unadjusted time-to-event variables. Log-rank tests were applied to compare each time-to-event variable between groups. P values less than 0.05 (two sided) were considered significant. All statistical analyses were performed using the StatMate software program.

miR340 is a novel miRNA that is downregulated in GICs

To identify novel miRNAs that are aberrantly expressed in GICs, we analyzed differences in miRNA expression between mouse and human GICs and control NSCs using miRNA microarrays. We identified seven miRNAs that were aberrantly expressed in both human and mouse GICs, three of which were upregulated and four of which were downregulated, compared with NSCs. Among these miRNAs, miR340 was identified as a novel miRNA that was significantly downregulated in all human and mouse GICs and in the human glioma cell lines (Fig. 1A). Using qPCR analysis, we confirmed that miR340 expression was significantly decreased in all examined human and mouse GICs and human glioma cell lines compared with NSCs (Fig. 1B). Furthermore, we determined that miR340 expression was significantly decreased in human GBM tissues compared with normal human brain tissues (Fig. 1C).

Figure 1.

miR340 expression is decreased in mouse and human GICs. A, heatmaps showing the hierarchical clustering of miRNAs expressed in mouse (left) and human (right) GICs and in human glioma cell lines (U87 and U251). miRNAs showing important functions in gliomas are indicated. B, fold changes in miR340 expression in human GICs (E1-4, E6, AO, and DA), human glioma cell lines, and NSCL61 compared with normal NSCs. C, the relative ratios of miR340 expression in 20 GBM tissues compared with five normal brain tissues. Values are expressed as mean ± SD. ***, P < 0.001.

Figure 1.

miR340 expression is decreased in mouse and human GICs. A, heatmaps showing the hierarchical clustering of miRNAs expressed in mouse (left) and human (right) GICs and in human glioma cell lines (U87 and U251). miRNAs showing important functions in gliomas are indicated. B, fold changes in miR340 expression in human GICs (E1-4, E6, AO, and DA), human glioma cell lines, and NSCL61 compared with normal NSCs. C, the relative ratios of miR340 expression in 20 GBM tissues compared with five normal brain tissues. Values are expressed as mean ± SD. ***, P < 0.001.

Close modal

miR340 inhibits cell proliferation, invasion, and migration and promotes cellular senescence in GICs

To examine the function of miR340 in hGICs, we infected hGICs and glioma cell lines with a recombinant lentivirus encoding either GFP alone or GFP and miR340 together. Then, we purified the GFP-positive cells using flow cytometry and evaluated miR340 expression in the infected cells (Supplementary Fig. S2). During the cell culture, we observed that miR340-overexpressing cells became flatter and larger than control cells (Fig. 2A). IHC analysis and Western blot assays revealed that miR340-overexpressing hGICs partially decreased Nestin expression and lost the expression of Sox2, but remained positive for GFAP, an astrocyte marker (Fig. 2B–D). miR340-overexpressing hGICs ceased proliferating during the first 3 days of culture (Fig. 3A). The BrdUrd-incorporation and cell-cycle analyses revealed that miR340 overexpression significantly arrested the cell cycle at the G1–S transition, as indicated by a marked accumulation of cells in the G1 peak and by a reduction of cells in the S phase (Fig. 3B and C). In addition, we examined invasiveness and motility in the miR340-overexpressing hGICs and confirmed that miR340 significantly inhibited both invasion and migration in these cells (Fig. 3D and E). We further demonstrated that miR340 overexpression activated the expression of SA-β-gal, a marker of cellular senescence, in hGICs (Fig. 3F). With the exception of cellular senescence, similar results were observed in the miR340-overexpressing human glioma cell lines. Taken together, these data indicate that miR340 negatively regulates the proliferation and invasiveness of both hGICs and glioma cell lines but induces cellular senescence only in hGICs.

Figure 2.

miR340 overexpression in hGICs changes cell morphology and inhibits expression of stem cell markers, Sox2 and Nestin, but remains positive for GFAP. A, fluorescent images of control (top) and miR340-overexpressing hGICs (bottom). B, immunostaining of Nestin (green) in miR340-overexpressing hGICs (E3, E6) and in the control. C, immunostaining of GFAP (green) and Sox2 (red) in miR340-overexpressing hGICs (E3, E6) and in the control. The nuclei are counterstained with DAPI (blue). D, Western blotting analysis of Nestin, Sox2, and GFAP expression in miR340-overexpressing hGICs (E3) and in the control. Scale bar, 200 μm.

Figure 2.

miR340 overexpression in hGICs changes cell morphology and inhibits expression of stem cell markers, Sox2 and Nestin, but remains positive for GFAP. A, fluorescent images of control (top) and miR340-overexpressing hGICs (bottom). B, immunostaining of Nestin (green) in miR340-overexpressing hGICs (E3, E6) and in the control. C, immunostaining of GFAP (green) and Sox2 (red) in miR340-overexpressing hGICs (E3, E6) and in the control. The nuclei are counterstained with DAPI (blue). D, Western blotting analysis of Nestin, Sox2, and GFAP expression in miR340-overexpressing hGICs (E3) and in the control. Scale bar, 200 μm.

Close modal
Figure 3.

miR340 overexpression in hGICs inhibits cell proliferation, cell invasion, and migration and induces cell-cycle arrest and senescence. A, the cell growth curves of control and miR340-overexpressing hGICs. B, the proportions of BrdUrd-positive cells in control and miR340-overexpressing hGICs. C, DNA histogram plots showing the cell-cycle analysis of hGICs transfected with miR340 or with the control. Cell-cycle distributions were detected 72 hours after transfection, and the ratios of each phase are represented as numerical values (shown in the inset of the figures). D and E, quantification of cell invasion and migration. The amount was expressed as invasion ratios (D) and migration ratios (E) of miR340-overexpressing hGICs compared with control cells. F, SA-β-gal staining (green) in control and miR340-overexpressing hGICs. The nuclei were counterstained with DAPI (blue). Values are expressed as mean ± SD. Scale bar, 50 μm. **, P < 0.01; ***, P < 0.001.

Figure 3.

miR340 overexpression in hGICs inhibits cell proliferation, cell invasion, and migration and induces cell-cycle arrest and senescence. A, the cell growth curves of control and miR340-overexpressing hGICs. B, the proportions of BrdUrd-positive cells in control and miR340-overexpressing hGICs. C, DNA histogram plots showing the cell-cycle analysis of hGICs transfected with miR340 or with the control. Cell-cycle distributions were detected 72 hours after transfection, and the ratios of each phase are represented as numerical values (shown in the inset of the figures). D and E, quantification of cell invasion and migration. The amount was expressed as invasion ratios (D) and migration ratios (E) of miR340-overexpressing hGICs compared with control cells. F, SA-β-gal staining (green) in control and miR340-overexpressing hGICs. The nuclei were counterstained with DAPI (blue). Values are expressed as mean ± SD. Scale bar, 50 μm. **, P < 0.01; ***, P < 0.001.

Close modal

miR340 inhibits GIC-mediated tumorigenesis in vivo

Next, we investigated whether miR340 overexpression inhibits GIC-mediated tumorigenesis in vivo. We transplanted either miR340-overexpressing hGICs or their control hGICs into the brains of immunodeficient mice. The control hGICs without miR340 overexpression continuously formed malignant gliomas with high invasiveness and many mitotic cells with a histopathology closely resembling that of the original tumor, causing death in mice within 48 days (Fig. 4A, top, Fig. 4B–D). In contrast, none of the mice injected with miR340-overexpressing hGICs formed apparent tumors (Fig. 4A, bottom), and these mice survived for more than 80 days (Fig. 4D). Histologic examination 3 days after transplantation of miR340-overexpressing hGICs demonstrated that the injected tumor cells were positive for active form of caspase-3 immunostaining, indicating that miR340 induced GIC apoptosis (Fig. 4E). In contrast, tumor formation of miR340-overexpressing human glioma cell lines in mouse brains was not completely suppressed, resulting in the death of the mice although the mice survived 2 or 4 weeks longer than the control (Supplementary Fig. S3A and S3B). These findings indicate that miR340 is a strong suppressor of tumorigenesis in GICs, although this antitumorigenic effect is less obvious in glioma cell lines.

Figure 4.

miR340 overexpression completely inhibits hGIC tumorigenesis in vivo. A, H&E staining of mouse brains with hGIC-xenograft (bottom) and the magnified images of tumor injection sites (center) 5 weeks after transplanting hGICs (E3 control) or miR340-overexpressing hGICs (E3 miR340). The corresponding MR images taken immediately before the dissection of the mouse brains (right). Dotted circles show the delineation of the extent of the tumor. The tumor volume of E3 control was 75.4±17.8 mm3 and that of E3 miR340 could not be determined because the tumor was undetectable on MRI. B, histopathology showing tumor invasion in the tumor border in control hGICs. C, histopathology showing mitotic cells (arrows) in the tumor of control hGICs. D, survival curves of the mice injected with control hGICs (E3, E6; n = 5; black dotted line) or with miR340-overexpressing hGICs (n = 3; red solid line). E, immunostaining for caspase-3 of the mouse brains dissected 3 days after injection of miR340-overexpressing hGICs (E3; right) or of the control hGICs (left). Scale bars, 1 mm (left) and 0.1 mm (right) in A; 50 μm in B; 20 μm in C, and 25 μm in E.

Figure 4.

miR340 overexpression completely inhibits hGIC tumorigenesis in vivo. A, H&E staining of mouse brains with hGIC-xenograft (bottom) and the magnified images of tumor injection sites (center) 5 weeks after transplanting hGICs (E3 control) or miR340-overexpressing hGICs (E3 miR340). The corresponding MR images taken immediately before the dissection of the mouse brains (right). Dotted circles show the delineation of the extent of the tumor. The tumor volume of E3 control was 75.4±17.8 mm3 and that of E3 miR340 could not be determined because the tumor was undetectable on MRI. B, histopathology showing tumor invasion in the tumor border in control hGICs. C, histopathology showing mitotic cells (arrows) in the tumor of control hGICs. D, survival curves of the mice injected with control hGICs (E3, E6; n = 5; black dotted line) or with miR340-overexpressing hGICs (n = 3; red solid line). E, immunostaining for caspase-3 of the mouse brains dissected 3 days after injection of miR340-overexpressing hGICs (E3; right) or of the control hGICs (left). Scale bars, 1 mm (left) and 0.1 mm (right) in A; 50 μm in B; 20 μm in C, and 25 μm in E.

Close modal

PLAT is a direct target of miR340

To identify functional targets of miR340, we compared the gene expression profiles of miR340-overexpressing hGICs with the profiles of their control GICs (Fig. 5A). We selected candidate target genes that were significantly upregulated or downregulated in miR340-overexpressing hGICs and analyzed these candidates using ConPath Navigator software, which is based on the GenMAPP software program (www.genmapp.org). These data revealed that miR340 regulates the genes related to two signaling pathways. One is the cell-cycle pathway, which involves p21/Cip1 and cyclin A (17, 18). The other is the cell adhesion/ECM remodeling pathway, which includes PLAT and matrix metalloproteases (Supplementary Fig. S4A and S4B; refs. 19, 20). Of the molecules that were strongly regulated by miR340, we focused on PLAT because its expression was significantly decreased in miR340-expressing hGICs (Supplementary Fig. S5A). In addition, the 3′UTRs of both the mouse and human plat mRNAs contained potential miR340 target sequences (Fig. 5B). IHC analysis and Western blot assays confirmed that miR340 overexpression decreased PLAT expression in hGICs (Fig. 5C and D). Using a reporter vector encoding the firefly luciferase gene with the wild-type plat 3′UTR, we demonstrated that miR340 overexpression inhibited luciferase activity in hGICs, whereas a deletion in the predicted binding site of miR340 in the 3′UTR of the plat gene abrogated the aforementioned inhibitory effect of miR340 (Fig. 5E). Taken together, these data strongly indicate that PLAT is a novel direct target of miR340 in hGICs. We confirmed that PLAT was more highly expressed in human glioblastomas than in normal brain tissues at the level of both mRNA (Fig. 5F) and protein (Fig. 5G) and that the tumor cells positive for PLAT immunostaining coexisted with those cells expressing CD15 (SSEA-1, hGIC marker; ref. 21; Supplementary Fig. S5B).

Figure 5.

PLAT is a putative target of miR340. A, heatmaps of genes with significantly altered expression between control hGICs and miR340-overexpressing hGICs and between glioma cell lines (none) and glioma cell lines (miR340). B, sequences of the miR340–binding sites in the mouse and human plat 3′UTRs predicted by TargetScan. Mutant form has a deletion of 20 nucleotides from 588 to 607. C, immunofluorescence showing PLAT expression (green) in the control and miR340–overexpressing hGICs. D, Western blot analysis of PLAT expression levels in control and miR340–overexpressing hGICs (E3). E, relative luciferase activity in miR340-overexpressing hGICs (E3) transfected with either Luc-PLAT or Luc-ΔPLAT (mutant form with deletion in miR340–binding sites). F, PLAT expression levels in 20 GBM tissues and in five normal brain tissues. G, Western blot analysis of PLAT expression levels in human GBM and normal brain tissues. Values are expressed as mean ± SD. **, P < 0.01; ***, P < 0.001.

Figure 5.

PLAT is a putative target of miR340. A, heatmaps of genes with significantly altered expression between control hGICs and miR340-overexpressing hGICs and between glioma cell lines (none) and glioma cell lines (miR340). B, sequences of the miR340–binding sites in the mouse and human plat 3′UTRs predicted by TargetScan. Mutant form has a deletion of 20 nucleotides from 588 to 607. C, immunofluorescence showing PLAT expression (green) in the control and miR340–overexpressing hGICs. D, Western blot analysis of PLAT expression levels in control and miR340–overexpressing hGICs (E3). E, relative luciferase activity in miR340-overexpressing hGICs (E3) transfected with either Luc-PLAT or Luc-ΔPLAT (mutant form with deletion in miR340–binding sites). F, PLAT expression levels in 20 GBM tissues and in five normal brain tissues. G, Western blot analysis of PLAT expression levels in human GBM and normal brain tissues. Values are expressed as mean ± SD. **, P < 0.01; ***, P < 0.001.

Close modal

PLAT knockdown inhibits GIC proliferation, invasion, and migration in vitro and GIC tumor growth in vivo

We subsequently examined the functions of PLAT in hGICs. PLAT knockdown using the plat-sh expression vector (Supplementary Fig. S6A and S6B) inhibited hGIC proliferation and invasion in a manner similar to that induced by miR340 overexpression (Fig. 6A–D). The inhibitory effects were fully recovered by introducing mutant PLAT, in which the shRNA target sequences were deleted (Supplementary Fig. S7). Furthermore, miR340 transfection in hGICs overexpressing the PLAT gene that lacked the 3′UTR recovered the inhibitory effects of miR340 on cell functions (Supplementary Fig. S8). These results indicate that PLAT is a key molecule that promotes malignancy of GIC by directly mediating the functions of miR340 downregulation. In addition, transplanting PLAT-knockdown hGICs into mouse brains resulted in the formation of tiny tumors localized at the injection site, but the growth rate was extremely slow, indicating that PLAT promotes GIC tumor growth in vivo (Fig. 6E).

Figure 6.

PLAT is a functional target of miR340. A, cell growth curves of control and PLAT-shRNA–expressing hGICs (E3 and E6). B, the proportions of BrdUrd-positive cells in control and in PLAT-shRNA–expressing hGICs. C and D, quantification of cell invasion and migration. The amount was expressed as relative invasion (C) and migration (D) ratios of PLAT-shRNA–expressing hGICs compared with control cells. E, H&E staining of whole brain slices 5 weeks after transplantation of PLAT-shRNA–expressing hGICs in the mouse brain. Scale bars, 1 mm (top); 25 μm (bottom). Values are expressed as mean ± SD. **, P < 0.01.

Figure 6.

PLAT is a functional target of miR340. A, cell growth curves of control and PLAT-shRNA–expressing hGICs (E3 and E6). B, the proportions of BrdUrd-positive cells in control and in PLAT-shRNA–expressing hGICs. C and D, quantification of cell invasion and migration. The amount was expressed as relative invasion (C) and migration (D) ratios of PLAT-shRNA–expressing hGICs compared with control cells. E, H&E staining of whole brain slices 5 weeks after transplantation of PLAT-shRNA–expressing hGICs in the mouse brain. Scale bars, 1 mm (top); 25 μm (bottom). Values are expressed as mean ± SD. **, P < 0.01.

Close modal

In the current study, we identified miR340 as a novel miRNA whose expression was significantly lower in both hGICs and glioma cell lines than in NSCs. We also observed that miR340 suppressed not only GIC proliferation and invasion in vitro, but also GIC-initiated tumor formation in mouse brains. These findings indicate that miR340 can act as a suppressor of malignant functions in GICs, particularly gliomagenesis and extensive tumor invasion. Furthermore, we determined that PLAT is the most significant target of miR340 in hGICs from the results of luciferase assays and PLAT-knockdown experiments with plat-sh, which phenocopied the suppressive effects of miR340 overexpression in vitro and in vivo. In addition to PLAT, miR340 overexpression in hGICs decreased the expression of Sox2, c-Met, CD44, and DNMT1, which are miR340 target genes (Supplementary Fig. S9). c-Met and CD44 regulate cell invasion and migration (22, 23), and Sox2 and CD44 play crucial roles in the maintenance of CSC stemness (24). Although miR340 overexpression in hGICs did not markedly decrease Nestin expression, Sox2 and CD44 expression significantly decreased, indicating that miR340 might reduce GIC stemness.

Transplanting miR340-overexpressing hGICs into mouse brains did not result in tumor formation. However, the miR340-overexpressing glioma cell lines generated tumors, although the growth rate was considerably suppressed, finally resulting in tumor-caused death of the mice. The mechanisms underlying the differences in miR340-induced gliomagenesis inhibition in GICs and in glioma cell lines remain unclear. Histopathologically, the transplanted hGICs with miR340 overexpression showed apoptosis in the early stage of transplantation, suggesting that antitumorigenic effects of miR340 in GIC might be partly due to tumor cell apoptosis. Recently, the inhibition of Sox2, which is a stem cell marker, was demonstrated to reduce the tumorigenic potential of human gastric cancer cells (25). In the glioma cell lines, the expression levels of Sox2 were initially quite low; therefore, Sox2 function was likely not highly affected by miR340 in these cell lines. These findings suggest that the tumorigenic mechanisms differ between GICs and more differentiated glioma cell lines.

Interestingly, the present study demonstrated that miR340 overexpression in hGICs promoted senescence in these cells, whereas miR340 introduction into glioma cell lines did not induce senescence (data not shown). Cellular senescence can inhibit tumor formation by modulating the redox state in CSCs and by regulating p27 in glioma cells (26, 27). In addition, GICs derived from wild-type mice expressing the senescence-promoting factor esophageal cancer-related gene 4 (Ecrg4) caused significantly reduced tumor formation in mouse brains compared with cells from Ecrg4-knockdown mice (28). We observed that miR340 overexpression in hGICs decreased DNA methyltransferase 1 (DNMT1) expression (Supplementary Fig. S9D); decreased DNMT1 expression can promote cellular senescence (29). These findings suggest that miR340-induced cellular senescence might explain the reduced tumorigenic activity of miR340-overexpressing GICs.

Currently, many miRNAs that are down-regulated in GSCs have been reported, including miR128, miR124, miR137, miR34a, and miR451 (11), (30–32). The overexpression of these miRNAs inhibits the proliferation and self-renewal, reduces the viability, or induces the differentiation of GSCs but does not cause the irreversible death of GSCs (31–33). Chan and colleagues demonstrated that miR138 acts as a prosurvival oncomiR in GSCs and that functional inhibition of miR138 prevents not only tumorsphere formation in vitro, but also tumorigenesis in vivo by inducing apoptosis and suppressing the proliferation of GSCs (34). It has been recently reported that miR218 inhibits the self-renewal of GSCs by targeting Bmi1 and regulates glioma cell development by Wnt pathways (35). In addition, Wang and colleagues reported that miR33a is highly expressed in GICs and that antagonizing miR33a function in GICs reduces self-renewal and tumor progression in mice by inhibiting the PKA and NOTCH pathways, which promote GIC maintenance (36). Many tumor cell functions, including a high proliferation rate, increased cell motility, and suppressed apoptosis and senescence, may be tightly linked during tumor development. Our findings indicate that miR340 regulates the expression of several key target genes that participate in cell proliferation, invasion, migration, and senescence, including PLAT, c-Met, CD44, Sox2, and DNMT1. Furthermore, p21 and ANXA2 genes, which have important roles in cell proliferation and in cell invasion, respectively, are downstream of PLAT. Both of these genes were also significantly upregulated or downregulated by miR340 overexpression in hGICs (Supplementary Fig. S9), indicating that PLAT plays central roles in cell proliferation, invasion, and migration. The loss of miR340 as a regulator controlling the expression of these genes in GICs promotes the development of these cells into malignant tumors and advances tumor progression (Fig. 7).

Figure 7.

A schematic summary of the function of miR340 and its target genes during the tumorigenesis process in hGICs, including cell cycle, invasiveness, stemness, and senescence. PLAT is a novel direct target gene suppressed by miR340. Other target genes, including c-Met, CD44, Sox2, and DNMT1, are also downregulated by miR340 in hGICs. Of the downstream genes of PLAT, p21 is upregulated, and ANXA2 is downregulated by miR340 overexpression in hGICs.

Figure 7.

A schematic summary of the function of miR340 and its target genes during the tumorigenesis process in hGICs, including cell cycle, invasiveness, stemness, and senescence. PLAT is a novel direct target gene suppressed by miR340. Other target genes, including c-Met, CD44, Sox2, and DNMT1, are also downregulated by miR340 in hGICs. Of the downstream genes of PLAT, p21 is upregulated, and ANXA2 is downregulated by miR340 overexpression in hGICs.

Close modal

The National Cancer Institute's Repository for Molecular Brain Neoplasia Data (REMBRANDT) database (https://caintegrator.nci.nih.gov/rembrandt/; ref. 37) indicates that the prognosis of glioma patients with increased PLAT levels is poorer than that of patients with decreased PLAT levels (Supplementary Fig. S10). These findings suggest that the introduction of miR340, which is capable of directly suppressing PLAT expression, may be a useful therapeutic tool for improving the prognosis of patients with patients. Furthermore, future studies of the functional significance of miR340 target molecules, including PLAT, might provide a greater understanding of the complex mechanisms of GBM development and progression.

No potential conflicts of interest were disclosed.

Conception and design: T. Kondo, J. Tanaka, T. Ohnishi

Development of methodology: T. Kondo

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D. Yamashita, T. Kondo, S. Ohue, M. Ishikawa, R. Matoba, S. Suehiro, S. Kohno, J. Tanaka

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D. Yamashita, T. Kondo, S. Ohue, M. Ishikawa, R. Matoba, T. Ohnishi

Writing, review, and/or revision of the manuscript: D. Yamashita, T. Kondo, S. Ohue, T. Ohnishi

Study supervision: T. Kondo, H. Takahashi, S. Kohno, H. Harada, T. Ohnishi

Other (financial support): T. Kondo

The authors thank Tetsuo Moriguchi for his technical suggestions and Naoki Ohtsu for his technical assistances.

This work was supported in part by funds from the Adaptable and Seamless Technology Transfer Program through target-driven R&D (A-STEP; T. Kondo and R. Matoba) and by Grants-in-Aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan [Program for Scientific Research (C) No. 25462266 to T. Ohnishi and S. Ohue].

1.
Stupp
R
,
Mason
WP
,
van den Bent
MJ
,
Weller
M
,
Fisher
B
,
Taphoorn
MJ
, et al
Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma
.
N Engl J Med
2005
;
352
:
987
96
.
2.
Reya
T
,
Morrison
SJ
,
Clarke
MF
,
Weissman
IL
. 
Stem cells, cancer, and cancer stem cells
.
Nature
2001
;
414
:
105
11
.
3.
Kondo
T
. 
Brain cancer stem-like cells
.
Eur J Cancer
2006
;
42
:
1237
42
.
4.
Visvader
JE
,
Lindeman
GJ
. 
Cancer stem cells in solid tumours: accumulating evidence and unresolved questions
.
Nat Rev Cancer
2008
;
8
:
755
68
.
5.
Jordan
CT
,
Guzman
ML
,
Noble
M
. 
Cancer stem cells
.
N Engl J Med
2006
;
355
:
1253
61
.
6.
Patrawala
L
,
Calhoun
T
,
Schneider-Broussard
R
,
Zhou
J
,
Claypool
K
,
Tang
DG
. 
Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2- cancer cells are similarly tumorigenic
.
Cancer Res
2005
;
65
:
6207
19
.
7.
Bao
S
,
Wu
Q
,
McLendon
RE
,
Hao
Y
,
Shi
Q
,
Hjelmeland
AB
, et al
Glioma stem cells promote radioresistance by preferential activation of the DNA damage response
.
Nature
2006
;
444
:
756
60
.
8.
Calin
GA
,
Croce
CM
. 
MicroRNA signatures in human cancers
.
Nat Rev Cancer
2006
;
6
:
857
66
.
9.
Chan
JA
,
Krichevsky
AM
,
Kosik
KS
. 
MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells
.
Cancer Res
2005
;
65
:
6029
33
.
10.
Gillies
JK
,
Lorimer
IA
. 
Regulation of p27Kip1 by miRNA 221/222 in glioblastoma
.
Cell Cycle
2007
;
6
:
2005
9
.
11.
Silber
J
,
Lim
DA
,
Petritsch
C
,
Persson
AI
,
Maunakea
AK
,
Yu
M
, et al
miR124 and miR137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells
.
BMC Med
2008
;
6
:
14
.
12.
Zhang
Y
,
Chao
T
,
Li
R
,
Liu
W
,
Chen
Y
,
Yan
X
, et al
MicroRNA-128 inhibits glioma cells proliferation by targeting transcription factor E2F3a
.
J Mol Med
2009
;
87
:
43
51
.
13.
Hide
T
,
Takezaki
T
,
Nakatani
Y
,
Nakamura
H
,
Kuratsu
J
,
Kondo
T
. 
Combination of a ptgs2 inhibitor and an epidermal growth factor receptor-signaling inhibitor prevents tumorigenesis of oligodendrocyte lineage-derived glioma-initiating cells
.
Stem Cells
2011
;
29
:
590
9
.
14.
Hide
T
,
Takezaki
T
,
Nakatani
Y
,
Nakamura
H
,
Kuratsu
J
,
Kondo
T
. 
Sox11 prevents tumorigenesis of glioma-initiating cells by inducing neuronal differentiation
.
Cancer Res
2009
;
69
:
7953
9
.
15.
Kondo
T
,
Raff
M
. 
The Id4 HLH protein and the timing of oligodendrocyte differentiation
.
EMBO J
2000
;
19
:
1998
2007
.
16.
Kondo
T
,
Raff
M
. 
Chromatin remodeling and histone modification in the conversion of oligodendrocyte precursors to neural stem cells
.
Genes Dev
2004
;
18
:
2963
72
.
17.
Xiong
Y
,
Hannon
GJ
,
Zhang
H
,
Casso
D
,
Kobayashi
R
,
Beach
D
. 
p21 is a universal inhibitor of cyclin kinases
.
Nature
1993
;
366
:
701
4
.
18.
Paterlini
P
,
De Mitri
MS
,
Martin
C
,
Münnich
A
,
Bréchot
C
. 
A TaqI polymorphism in the human cyclin A gene
.
Nucleic Acids Res
1991
;
19
:
2516
.
19.
Hagège
J
,
Peraldi
MN
,
Rondeau
E
,
Adida
C
,
Delarue
F
,
Medcalf
R
, et al
Plasminogen activator inhibitor-1 deposition in the extracellular matrix of cultured human mesangial cells
.
Am J Pathol
1992
;
141
:
117
28
.
20.
Saus
J
,
Quinones
S
,
Otani
Y
,
Nagase
H
,
Harris
ED
,
Kurkinen
M
. 
The complete primary structure of human matrix metalloproteinase-3. Identity with stromelysin
.
J Biol Chem
1988
;
263
:
6742
5
.
21.
Son
MJ
,
Woolard
K
,
Nam
DH
,
Lee
J
,
Fine
HA
. 
SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma
.
Cell Stem Cell
2009
;
4
:
440
52
.
22.
Joo
KM
,
Jin
J
,
Kim
E
,
Ho Kim
K
,
Kim
Y
,
Gu Kang
B
, et al
MET signaling regulates glioblastoma stem cells
.
Cancer Res
2012
;
72
:
3828
38
.
23.
Merzak
A
,
Koocheckpour
S
,
Pilkington
GJ
. 
CD44 mediates human glioma cell adhesion and invasion in vitro
.
Cancer Res
1994
;
54
:
3988
92
.
24.
Ikushima
H
,
Todo
T
,
Ino
Y
,
Takahashi
M
,
Miyazawa
K
,
Miyazono
K
. 
Autocrine TGF-beta signaling maintains tumorigenicity of glioma-initiating cells through Sry-related HMG-box factors
.
Cell Stem Cell
2009
;
5
:
504
14
.
25.
Hutz
K
,
Mejias-Luque
R
,
Farsakova
K
,
Ogris
M
,
Krebs
S
,
Anton
M
, et al
The stem cell factor SOX2 regulates the tumorigenic potential in human gastric cancer cells
.
Carcinogenesis
2014
;
35
:
942
50
.
26.
Venkataraman
S
,
Alimova
I
,
Fan
R
,
Harris
P
,
Foreman
N
,
Vibhakar
R
. 
MicroRNA 128a increases intracellular ROS level by targeting Bmi-1 and inhibits medulloblastoma cancer cell growth by promoting senescence
.
PLoS ONE
2010
;
5
:
e10748
.
27.
Tchougounova
E
,
Jiang
Y
,
Bråsäter
D
,
Lindberg
N
,
Kastemar
M
,
Asplund
A
, et al
Sox5 can suppress platelet-derived growth factor B-induced glioma development in Ink4a-deficient mice through induction of acute cellular senescence
.
Oncogene
2009
;
28
:
1537
48
.
28.
Kujuro
Y
,
Suzuki
N
,
Kondo
T
. 
Esophageal cancer-related gene 4 is a secreted inducer of cell senescence expressed by aged CNS precursor cells
.
Proc Natl Acad Sci U S A
2010
;
107
:
8259
64
.
29.
Kar
S
,
Deb
M
,
Sengupta
D
,
Shilpi
A
,
Parbin
S
,
Torrisani
J
, et al
An insight into the various regulatory mechanisms modulating human DNA methyltransferase 1 stability and function
.
Epigenetics
2012
;
7
:
994
1007
.
30.
Godlewski
J
,
Nowicki
MO
,
Bronisz
A
,
Williams
S
,
Otsuki
A
,
Nuovo
G
, et al
Targeting of the Bmi-1 oncogene/stem cell renewal factor by microRNA-128 inhibits glioma proliferation and self-renewal
.
Cancer Res
2008
;
68
:
9125
30
.
31.
Guessous
F
,
Zhang
Y
,
Kofman
A
,
Catania
A
,
Li
Y
,
Schiff
D
, et al
MicroRNA-34a is tumor suppressive in brain tumors and glioma stem cells
.
Cell Cycle
2010
;
9
:
1031
36
.
32.
Gal
H
,
Pandi
G
,
Kanner
AA
,
Ram
Z
,
Lithwick-Yanal
G
,
Aarialio
N
, et al
MIR-451 and Imatinib mesylate inhibit tumor growth of glioblastoma stem cells
.
Biochem Biophys Res Commun
2008
;
376
:
86
90
.
33.
Li
Y
,
Guessous
F
,
Zhang
Y
,
Dipierro
C
,
Kefas
B
,
Johnson
E
, et al
MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes
.
Cancer Res
2009
;
69
:
7569
76
.
34.
Chan
XHD
,
Nama
S
,
Gopal
F
,
Rizk
P
,
Ramasamy
S
,
Sundaram
G
, et al
Targeting glioma stem cells by functional inhibition of a prosurvival oncomiR138 in malignant gliomas
.
Cell Reports
2012
;
2
:
591
602
.
35.
Tu
Y
,
Gao
X
,
Li
G
,
Fu
H
,
Cui
D
,
Liu
H
, et al
MicroRNA-218 inhibits glioma invasion, migration, proliferation, and cancer stem-like cell self-renewal by targeting the polycomb group gene Bmi1
.
Cancer Res
2013
;
73
:
6046
55
.
36.
Wang
H
,
Sun
T
,
Hu
J
,
Zhang
R
,
Rao
Y
,
Wang
S
, et al
miR33a promotes glioma-initiating cell self-renewal via PKA and NOTCH pathways
.
J Clin Invest
2014
;
124
:
4489
502
.
37.
Madhavan
S
,
Zenklusen
JC
,
Kotliarov
Y
,
Sahni
H
,
Fine
HA
,
Buetow
K
. 
Rembrandt: helping personalized medicine become a reality through integrative translational research
.
Mol Cancer Res
2009
;
7
:
157
67
.

Supplementary data