Gliomas are classified by combining histopathologic and molecular features, including isocitrate dehydrogenase (IDH) status. Although IDH-wild-type diffuse astrocytic glioma (DAG) shows a more aggressive phenotype than IDH-mutant type, lack of knowledge regarding relevant molecular drivers for this type of tumor has hindered the development of therapeutic agents. Here, we examined human IDH-wild-type DAGs and a glioma mouse model with a mosaic analysis with double markers (MADM) system, which concurrently lacks p53 and NF1 and spontaneously develops tumors highly comparable with human IDH-wild-type DAG without characteristic molecular features of glioblastoma (DAG-nonMF). During tumor formation, enhancer of zeste homolog (EZH2) and the other polycomb repressive complex 2 (PRC2) components were upregulated even at an early stage of tumorigenesis, together with an increased number of genes with H3K27me3 or H3K27me3 and H3K4me3 bivalent modifications. Among the epigenetically dysregulated genes, frizzled-8 (Fzd8), which is known to be a cancer- and stem cell reprogramming–related gene, was gradually silenced during tumorigenesis. Genetic and pharmacologic inhibition of EZH2 in MADM mice showed reactivation of aberrant H3K27me3 target genes, including Fzd8, together with significant reduction of tumor size. Our study clarifies a pathogenic molecular pathway of IDH-wild-type DAG-nonMF that depends on EZH2 activity and provides a strong rationale for targeting EZH2 as a promising therapeutic approach for this type of glioma.

Significance:

EZH2 is involved in the generation of IDH-wild-type diffuse astrocytic gliomas and is a potential therapeutic target for this type of glioma.

Gliomas represent approximately 80% of malignant central nervous system tumors. The World Health Organization (WHO) classification system subdivides diffuse astrocytic glioma (DAG) and oligodendroglioma into grade II to IV based on histopathologic findings (1, 2). Recent comprehensive studies revealed that WHO grade II and III DAG and oligodendroglioma, called lower grade gliomas (LGG), are classified by combining histopathologic and molecular features including isocitrate dehydrogenase (IDH) status. Mutations in the IDH gene have been found in approximately 80% of LGGs, which show a characteristic feature of relatively favorable prognosis, while IDH-wild-type LGGs display an aggressive phenotype (3–5). Among LGGs, all cases of oligodendroglioma exhibit IDH gene mutation, while DAGs exhibit either wild- or mutant-types IDH gene status. Therefore, LGGs are considered to be divided into IDH-mutant DAGs or oligodendrogliomas (IDH-mutant LGG), or IDH-wild-type DAGs.

Mutant IDH induces characteristic epigenetic alterations, such as extensive DNA hypermethylation (glioma CpG island methylator phenotype, G-CIMP), which may contribute to the initial step of glioma tumorigenesis (6, 7). In contrast, the molecular mechanisms underlying formation of IDH-wild-type DAGs appear to be heterogeneous. Recently, among IDH-wild-type DAGs, tumors with EGFR amplification, or combined whole chromosome 7 gain and whole chromosome 10 loss, or TERT promoter mutation, which are common characteristic molecular features of IDH-wild-type grade IV glioblastomas (GBM), exhibit a poor prognosis comparable with GBMs (8, 9). This type of DAG is termed diffuse astrocytic glioma, IDH-wild-type, with molecular features of glioblastoma, WHO grade IV (DAG-MF). However, the underlying molecular mechanisms related to the relevant molecular drivers for IDH-wild-type DAG without these features (i.e., IDH-wild-type DAG-nonMF) are poorly understood. It is a significant challenge to develop agents that specifically target this type of glioma.

The mouse model mosaic analysis with double markers (MADM) system is a useful technology to investigate the process of tumorigenesis from early to advanced stages (10). Starting from a heterozygous, colorless mouse, the MADM system generates a small number of homozygous mutant cells via inter-chromosomal mitotic recombination and labels them with green fluorescent protein (GFP) and wild-type cells with red fluorescent protein (RFP; ref. 10). In addition to mimicking the physiologic circumstances of tumor initiation, the unequivocal labeling of mutant cells enables the investigation of the entire tumorigenic process.

In this study, we used a glioma mouse model with a MADM system employing a concurrent knock-out of the p53 and NF1 genes, both of which are the most frequently mutated in human IDH-wild-type DAG-nonMF (40% and 30% of these gliomas, respectively). Notably, the pathologic features, and genetic and gene expression profiles of this MADM tumor match well with human IDH-wild-type DAG-nonMF. The resulting tumors showed an upregulation of enhancer of zeste homolog (EZH2), a catalytic component of polycomb repressive complex 2 (PRC2), together with increased H3K27 trimethylation (H3K27me3) on target genes including stem cell reprogramming-related genes, such as Wnt pathway genes. Furthermore, inhibition of EZH2 activity effectively repressed tumor growth both in vitro and in vivo. Our study clarifies a pathogenic molecular pathway, which depends on EZH2/PRC2 activity, and is partly involved in the generation of IDH-wild-type DAG-nonMF.

Animal experiments

MADM mice were generated by crossing MADM-TG, p53 KO, NF1 flox mice, HIPP11GT, MADM-11GT mice and Tg (GFAP-cre) 25Mes mice obtained from The Jackson Laboratory (stock numbers: 017530, 013749 and 004600, respectively). Ezh2flox/flox mice were obtained from RIKEN BRC (RBRC0555 B6-Ezh2 floxed). Oral administration of 160 mg/kg EPZ6438 (27626-3, BPS Bioscience and HY-13803, MedChemExpress) was performed twice daily for 20 days (11). EPZ6438 was formulated as a suspension in 1% carboxymethyl cellulose with 0.2% Tween 80 for oral administration (12). We analyzed MADM tumor mice treated with EPZ6438 or vehicle (1% carboxymethyl cellulose with 0.2% Tween 80) as a control (21 tumors in 8 mice and 14 tumors in 7 mice, respectively). All mice were anesthetized with isoflurane (1%–3% in air) and then placed in a 4.7T AVANCE III Pharma Scan (Bruker). During scanning, the breathing and depth of anesthesia were continuously monitored, with the breathing rate maintained at 80–100 breaths per minute. MRI images were visualized with open-source Osirix software that allowed the volumetric computation of tumors.

Cell sorting of MADM GFP-positive cells

GFP-positive cells with the homozygous deletion of p53 and NF1 genes were collected at P8, P20, and P60 as precancerous cells and at P90 and P120 as tumor cells of the early and late phase, respectively, by immunopanning methods with anti-PDGFRα antibody-coated plates for P8 cells as described previously (13) and cell sorting using a FACS ARIA II (BD Biosciences).

Cell culture of MADM tumor and human glioma cell lines

Cells were derived from various individual MADM tumors. These cells were plated onto flasks, coated with poly-d-lysine (PDL, P6407, Sigma Aldrich) and cultured in Neurobasal medium (21103, Thermo Fisher Scientific) with B27 (17504-044, Thermo Fisher Scientific), Glutamax (35050061, Thermo Fisher Scientific), 10 ng/mL human recombinant PDGF-AA (100-13-A, PeproTech), and 1% penicillin–streptomycin (15140-122, Thermo Fisher Scientific) at 37°C in a humidified incubator with 5% CO2. U87 (IDH-wild-type grade IV) and SW1783 (IDH-wild-type LGG) cell lines were obtained from ATCC. The TM31 (IDH-wild-type LGG) and T98 (IDH-wild-type GBM) cell lines were obtained from RIKEN BRC (RIKEN Cell Bank). Onda10, Onda11, KINGS1 (IDH-wild-type LGG), and U251 (IDH-wild-type GBM) cell lines were obtained from the Japanese Collection of Research Bioresources (JCRB) Cell Bank. GL261 cell line was obtained from the NCI tumor-cell-line repository (Frederick, MD). Normal human astrocytes (NHA) were kindly gifted by Dr. R. Pieper (University of California, San Francisco, San Francisco, CA; ref. 14). All of these cell lines were analyzed within 6 months after resuscitation. These cell lines were authenticated in original cell banks and were tested to be Mycoplasma free when resuscitation. U87, T98, GL261, and NHA cells were cultured with DMEM (044-29765, Wako Pure Chemical Industries, Ltd.) containing 10% FBS and 1% penicillin–streptomycin (15140-122, Thermo Fisher Scientific). TM31, Onda10, and Onda11 cells were cultured with Eagle MEM (M4655, Sigma Aldrich) medium containing 10% FBS. U251 and KINGS1 cells were cultured with RPMI1640 (189-02025, Wako Pure Chemical Industries, Ltd.) containing 5% FBS. All cell lines were maintained at 37°C in a humidified incubator with 5% CO2.

ChIP-chip and ChIP-seq analyses

Chromatin immunoprecipitation (ChIP) assays were performed according to previously published methods with minor modifications (15–17). Cells (up to 1 × 106) were treated with 1% formaldehyde for 8 minutes to crosslink histones to DNA. The cell pellets were resuspended in lysis buffer (1% SDS, 10 mmol/L EDTA, 50 mmol/L Tris-HCl pH 8.1, and protease inhibitor) and sonicated using a Covaris S220 system (Covaris). After diluting the cell lysate 1:10 with dilution buffer (1% Triton, 2 mmol/L EDTA, 150 mmol/L NaCl, 20 mmol/L Tris-HCl pH 8.1), diluted cell lysates were incubated for 16 hours at 4°C with Dynabeads protein G (100-03D, Thermo Fisher Scientific), which were precoated with 2 μL of anti-histone H3 antibodies (ab1791, Abcam) or Dynabeads M280 sheep anti-Mouse IgG (112-01D Thermo Fisher Scientific), which were coated with 2 μL of anti-K4 trimethylated and anti-K27 trimethylated histone H3 antibodies (CMA304 16H10 and CMA323 1E7, respectively; ref. 18). ChIP products were analyzed by SYBR Green ChIP-quantitative PCR (qPCR) using the primer sets shown in Supplementary Table S1. Negative control (NC) region for ChIP-qPCR was selected according to the data of reference (19). We analyzed the ChIP products as probes on a SurePrint G3 mouse promoter array 2 × 400K according to the manufacturer's protocol (ChIP-chip; G4858A; Agilent Technologies; ref. 16). Results were analyzed by a Whitehead Per-Array Neighbourhood Model (modified) to identify bound genes in an Agilent Genomics Workbench (version 7.0, Agilent Technologies). A probe was considered to be bound, if the mean P value of 3 neighboring probes was less than 0.05 and if one of the following requirements was met: (i) the P value of the central probe was less than 0.05 and the P value of at least one neighboring probe was less than 0.1; or (ii) the P value of at least 2 of the neighboring probes were less than 0.01 (20). These data were visualized in GeneSpring GX, version 12.6.0 (Silicon Genetics). Neural stem cells (NSC), astrocytes (AST), and P8 cells were collected from 4–6 mice (a single ChIP-chip experiment). MADM tumor cells were collected from a MADM mouse. Two independent ChIP-chip experiments were performed. We determined genes modified by each chemical modification commonly across two individual experiments as genes with these modifications in examined cells. We also carried out ChIP-seq using fragmented ChIP products (0.5 ng to 10 ng) after preparing DNA libraries for paired-end sequencing using the NEBNext Ultra II DNA library Prep kit for the Illumina HiSeq 2500 next generation sequencing (NGS) system (New England Biolabs) to validate ChIP-chip data. ChIP-seq clean reads were obtained from raw reads by the removal of adaptors and low-quality reads. The clean reads were aligned to the mouse genome (mm9) using bowtie (version 0.12.8) with parameters -a –best –strata -m 3 -v 3 (21). The levels of histone modification at gene promoters were estimated by counting the numbers of reads mapped within a 2-kb region around the transcriptional start site using HTSeq (version 0.10.0; ref. 22). ChIP-seq experiments using anti-H3K4me3 and anti-H3K27me3 antibodies were performed in duplicate in NSCs, ASTs, P8 precancerous cells, and P120 MADM tumor cells.

Database of human glioma samples

o analyze various gene expression patterns in clinical glioma samples, we used the normalized RNA sequencing (RNA-seq) expression data (Illumina Hiseq) from the UCSC cancer genomics browser (https://genome-cancer.ucsc.edu) and the UCSC Xenabrowser (https://xenabrowser.net) in relation to the Cancer Genome Atlas (TCGA) brain lower grade glioma, TCGA glioblastoma multiforme and TCGA pan-cancer (23, 24) and the level-3 preprocessed RNA-seq expression data of grade II and III gliomas in the TCGA portal (https://tcga-data.nci.nih.gov/). Gene set enrichment analysis (GSEA) was performed using the GSEA desktop application (Broad Institute). We analyzed these datasets with R Statistical Software (version 2.14.0; R Foundation for Statistical Computing). The Z score for the expression level of each gene was calculated using the mean expression value and SD among all grade II, III, IV tumors and normal brains or among all genes in each individual.

Clustering for gene expression

mRNA RSEM (RNA-Seq by Expectation Maximization) expression profile and genetic mutation profile were obtained with TCGA GBMLGG data (2015_08_21_stddata Run and 2016_01_28 stddata Run). After stratifying samples according with DAGs-MF and DAGs-nonMF definition, relative changes of expression levels of 544 genes, which correspond to upregulated genes specifically in MADM tumor cells (top 5% upregulated transcripts in MADM tumor cells compared with normal brains) in each tumor sample against mean expression levels among normal samples with log2[(RSEM (g,i) of tumor+0.5)/(mean RSEM (g) of normal samples+0.5)] where “g” and “i” indicate g-th gene and i-th sample. 0.5 was added to all values to avoid dividing by zero counts. These calculated values were directly mapped as heatmap colors. Microarray-based gene expression values of the triplicate MADM tumor samples and the duplicate normal samples were quantile normalized with a background correction using R package limma (25). We detected the most differentially expressed transcripts in MADM tumors compared with those of normal samples. Corresponding mouse gene symbols were converted to human symbols by R package biomaRt (26, 27) and plugged into gene expression data compiled from TCGA data (2015_08_21_stddata Run and 2016_01_28 stddata Run).

Consensus clustering was performed utilizing hierarchical clustering based on Ward and Euclidean correlation algorithms with 1,000 iterations using Bioconductor package ConsensusClusterPlus. We selected 80% item resampling, 90% gene resampling, and a maximum evaluated K of 5. The number of clusters was determined by the relative change in area under a cumulative distribution function curve by consensus clustering.

Statistical analysis

The statistical significance of differences between two groups was analyzed by paired Student t test and Fisher exact test. Microarray data, RSEM, and fragments per kilobase of exon per million reads mapped (FPKM) analyses were performed by paired Student t test, Wilcoxon signed rank test and Wilcoxon rank sum test. Correlation coefficient was calculated with Pearson correlation test. All reported P values were two-sided, with P < 0.05 considered statistically significant.

Data availability

The Gene Expression Omnibus accession code for the ChIP-chip microarray data of NSCs and ASTs along with MADM tumors is GSE110926. Microarray-based gene expression data of MADM tumor tissues and normal brain tissues can be found at GSE91050, while that related to Ezh2Δ/wt MADM tumor tissues is at GSE110927. GSE110926 and GSE110927 are also integrated as GSE110937. ChIP-seq and RNA-seq data are available in the DNA Data Bank of Japan Sequence Read Archive (DDBJ DRA; accession number is DRA008249).

Study approval

All protocols for animal experiments were approved by the Institutional Animal Care and Use Committee of Nagoya University (Nagoya, Japan; 29476). Experiments with an oral administration procedure were also approved by the Committee of Animal Experiments at the National Center for Geriatrics and Gerontology.

Characteristic molecular features of IDH-wild-type DAGs

Using the public database of TCGA, we investigated the molecular profiles of IDH-wild-type DAGs (n = 64 and n = 13, DAG-MF and DAG-nonMF, respectively). Although significantly more genes were mutated in IDH-wild-type DAGs-MF than DAGs-nonMF, EGFR, PTEN, NF1, TTN, and p53 are commonly mutated in more than 10% of both IDH-wild-type DAGs-MF and -nonMF (Fig. 1A and B). Of note, EGFR was the most commonly mutated gene with the highest mutation frequency and variant allele fraction (VAF) in IDH-wild-type DAGs-MF (40% and 55%), while p53 and NF1 were the genes with high mutation frequency and VAF in IDH-wild-type DAGs-nonMF (40% and 46%, 30% and 61%, respectively; Fig. 1C). GSEA indicated that highly expressed RAS–MAPK signaling pathway genes were enriched in IDH-wild-type DAG-nonMFs compared with IDH-wild-type DAG-MFs (P < 0.01, Fig. 1D). Concurrent genetic alterations in both p53 and RAS–MAPK signaling pathway-related genes including NF1, a negative regulator of RAS–MAPK signaling, are more frequent in IDH-wild-type DAGs-nonMF (23.1%) than IDH-wild-type DAGs-MF (3.1%; P < 0.05). These data indicated that genetic alterations of p53 and NF1 genes play important roles in IDH-wild-type DAGs, particularly nonMF, formation.

Figure 1.

Molecular features of human IDH-wild-type DAGs-MF, DAGs-nonMF, and MADM tumor cells. A, Bar graph indicating frequency of cases exhibiting gene mutations in IDH-wild-type DAGs (DAG-MF and -nonMF, n = 64 and n = 13 in TCGA, respectively). Genes whose mutations were found in more than 5% of IDH-wild-type DAG cases are described. B, Boxplots indicating number of genes exhibiting genetic mutations per case in IDH-wild-type DAG-MF cases (light blue box) and IDH-wild-type DAG-nonMF cases (yellow box). Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. C, Dot plots indicating frequency of mutated cases and variant allele fraction in IDH-wild-type DAGs-MF (left) or IDH-wild-type DAGs-nonMF (right) genes whose mutations were found in more than 5% of IDH-wild-type DAGs. The x-axis indicates frequency of cases exhibiting genetic mutations of these genes (%). The y-axis indicates mean variant allele fraction of these gene mutations (%). Red, green, blue, and yellow dots indicate NF1, p53, EGFR, and PTEN genes, respectively. D, GSEA of differentially upregulated genes in IDH-wild-type DAGs-nonMF compared with IDH-wild-type DAGs-MF. Significantly enriched gene sets of target genes of RAS–MAPK pathway and RAS–MAPK pathway genes are described (P < 0.01). Calculated normalized enrichment scores are described in each box. Color of each box corresponds to the calculated normalized enrichment score. E, A schematic diagram of the sequential collection of precancerous cells [post-natal (P) 8, 20, and 60 cells] and tumor cells (P90 and 120 cells) during MADM tumor formation. F, Hematoxylin and eosin staining of MADM tumor tissues (×40). G, Molecular classification of IDH-wild-type DAGs. H, Copy number analysis of genes derived from MADM tumor cells, which correspond to human genes located on chromosome 7 and 10 (753 and 632 genes, respectively). The y-axis indicates normalized log ratios of mean read numbers of each gene (tumor DNA/DNA derived from normal tissue in the same individual). A red line indicates the mean value among points in segments obtained by circular binary segmentation analysis. I, Left, heatmap analysis of the relative gene expression in IDH-wild-type DAGs-MF and IDH-wild-type DAGs-nonMF (light blue box, n = 64 and pink box, n = 13, respectively) compared with normal brains (n = 5). The vertical axis represents 544 human genes, which correspond to genes upregulated specifically in MADM tumor cells among top 5% upregulated transcripts in MADM tumor cells compared with normal brains. Red and blue colored rectangles represent the relative expression levels as indicated in log2-fold change. Right, boxplot of mean RSEM values of these genes in IDH-wild-type DAGs-MF and IDH-wild-type DAGs-nonMF. Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. ***, P < 0.01.

Figure 1.

Molecular features of human IDH-wild-type DAGs-MF, DAGs-nonMF, and MADM tumor cells. A, Bar graph indicating frequency of cases exhibiting gene mutations in IDH-wild-type DAGs (DAG-MF and -nonMF, n = 64 and n = 13 in TCGA, respectively). Genes whose mutations were found in more than 5% of IDH-wild-type DAG cases are described. B, Boxplots indicating number of genes exhibiting genetic mutations per case in IDH-wild-type DAG-MF cases (light blue box) and IDH-wild-type DAG-nonMF cases (yellow box). Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. C, Dot plots indicating frequency of mutated cases and variant allele fraction in IDH-wild-type DAGs-MF (left) or IDH-wild-type DAGs-nonMF (right) genes whose mutations were found in more than 5% of IDH-wild-type DAGs. The x-axis indicates frequency of cases exhibiting genetic mutations of these genes (%). The y-axis indicates mean variant allele fraction of these gene mutations (%). Red, green, blue, and yellow dots indicate NF1, p53, EGFR, and PTEN genes, respectively. D, GSEA of differentially upregulated genes in IDH-wild-type DAGs-nonMF compared with IDH-wild-type DAGs-MF. Significantly enriched gene sets of target genes of RAS–MAPK pathway and RAS–MAPK pathway genes are described (P < 0.01). Calculated normalized enrichment scores are described in each box. Color of each box corresponds to the calculated normalized enrichment score. E, A schematic diagram of the sequential collection of precancerous cells [post-natal (P) 8, 20, and 60 cells] and tumor cells (P90 and 120 cells) during MADM tumor formation. F, Hematoxylin and eosin staining of MADM tumor tissues (×40). G, Molecular classification of IDH-wild-type DAGs. H, Copy number analysis of genes derived from MADM tumor cells, which correspond to human genes located on chromosome 7 and 10 (753 and 632 genes, respectively). The y-axis indicates normalized log ratios of mean read numbers of each gene (tumor DNA/DNA derived from normal tissue in the same individual). A red line indicates the mean value among points in segments obtained by circular binary segmentation analysis. I, Left, heatmap analysis of the relative gene expression in IDH-wild-type DAGs-MF and IDH-wild-type DAGs-nonMF (light blue box, n = 64 and pink box, n = 13, respectively) compared with normal brains (n = 5). The vertical axis represents 544 human genes, which correspond to genes upregulated specifically in MADM tumor cells among top 5% upregulated transcripts in MADM tumor cells compared with normal brains. Red and blue colored rectangles represent the relative expression levels as indicated in log2-fold change. Right, boxplot of mean RSEM values of these genes in IDH-wild-type DAGs-MF and IDH-wild-type DAGs-nonMF. Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. ***, P < 0.01.

Close modal

Gliomas in the MADM mouse model are comparable with IDH-wild-type DAGs-nonMF

To clarify the molecular significances of IDH-wild-type DAGs, a MADM glioma mouse model system was examined. The MADM system generates cells with the homozygous deletion of p53 and NF1 genes (GFP-positive cells), and MADM mice spontaneously develop IDH-wild-type anaplastic gliomas (MADM tumors) whose expression profiles were concordant with the proneural cell type between postnatal days 90 and 120 (P90–P120; Fig. 1E; Supplementary Fig. S1; ref. 13). Pathologic findings showed that MADM tumors are compatible with anaplastic astrocytic glioma without prominent necrosis and microvascular proliferation, which is consistent with a previous study (Fig. 1F; ref. 13). We identified 401 transcripts (top 1% of highly expressed transcripts) that had concordant and markedly higher expression levels in MADM tumor tissues (n = 3) compared with normal brain tissues (n = 2; Supplementary Table S2). TCGA data (n = 581; 194 grade II, 221 grade III, and 166 grade IV) revealed that the expression levels of the 353 human genes that corresponded to the identified 401 mouse transcripts were significantly higher in IDH-wild-type DAGs and GBMs (P < 0.01, Supplementary Fig. S2A) compared with IDH-mutant LGGs. K-means clustering analysis and consensus cumulative distribution function (CDF) analysis revealed that the expression profile of these genes could be used to divide the tissues into two groups dependent on their IDH-mutant or IDH-wild-type status (P < 0.01, Supplementary Fig. S2B). Furthermore, using the expression profiling of the top 10% of highly expressed transcripts (corresponding to 2,411 genes), we obtained concordant findings (Supplementary Fig. S2C and S2D).

Given that IDH-wild-type DAGs contain different subgroups with characteristic molecular features (i.e., IDH-wild-type DAG-MF and -nonMF, Fig. 1G), we investigated the genetic and gene expression profiling of MADM tumors further to identify which IDH-wild-type DAG subgroups are highly comparable with the MADM tumors. Comprehensive genetic analysis revealed that MADM tumors exhibited no EGFR amplification and no combined whole chromosome 7 gain and whole chromosome 10 loss, except for the partial loss of chromosome 10q (Fig. 1H). Sanger sequencing revealed no TERT promoter mutation within the transcription start site ± 1 kb. These data indicated that MADM tumors were not genetically compatible with IDH-wild-type DAG-MF. Next, we selected 544 genes whose expression levels were upregulated specifically in MADM tumor cells but not in a mouse GBM cell line, GL261, which is comparable with human IDH-wild-type GBM (28). Heatmap analysis showed that the expression status of corresponding human 544 genes could be used to clearly divide the IDH-wild-type DAG into two groups (Supplementary Table S3). Expression levels of 544 genes were significantly higher in human IDH-wild-type DAGs-nonMF (n = 13) compared with IDH-wild-type DAGs-MF (n = 64; P < 0.01, Fig. 1I). These data indicate that gliomas in MADM mice are comparable with human IDH-wild-type DAG-nonMF by means of histopathologic and molecular features and that analyzing this mouse model may provide clues to the underlying tumorigenic processes of IDH-wild-type DAG-nonMF.

Elevated expression of PRC2 components in MADM tumor cells and IDH-wild-type DAGs-nonMF

Epigenetic dysregulation occurs at early stages of tumorigenesis and causes altered proliferation and differentiation throughout tumor evolution, including cases of glioma (29). Considering that IDH-wild-type DAGs-nonMF exhibit less genetic alterations in comparison with IDH-wild-type DAGs-MF (Fig. 1B), we focused on epigenetic alterations subsequent to p53 and NF1 gene alterations during MADM tumor formations. We examined the expression level of epigenome-associated genes in MADM tumors. Among 162 epigenome-associated genes, which were previously functionally classified in human gliomas (30), we identified 116 that were substantially expressed and altered at a genetic level in either human LGGs (n = 415) or grade IV GBMs (n = 292; Supplementary Table S4; refs. 5, 30). Among the 116 epigenome-associated genes, Ezh2 was the most upregulated gene in MADM tumors (Supplementary Fig. S3A)

EZH2 is the components of PRC2 for its catalytic activity of H3K27me3 modification along with SUZ12 and EED. EZH2 is highly upregulated in MADM tumor cells compared with those in normal NSCs or astrocytes (AST; Fig. 2A). We evaluated the sequential changes of Ezh2, Suz12, and Eed expressions in GFP-positive precancerous cells and glioma cells collected by FACS sorting at different time points (P20 and P60: precancerous cells, P90 to P120: MADM tumor cells). Expression levels of Ezh2, Suz12, and Eed were consistently highly upregulated from the early precancerous stage (P8, P20) to glioma (P90–P120), resulting in gradually increased level of H3K27me3 in GFP-positive cells (Fig. 2B and C). Immunohistochemistry (IHC) assay of MADM mouse brain tissues revealed the high expression of EZH2 in GFP-positive precancerous cells (i.e., homozygous deletion of p53 and NF1 genes) even in P20 mutant mice brain and tumor tissues in P120 mice, compared with GFP-negative cells (Fig. 2D). Next, we examined the expression status of 116 epigenome-associated genes in human IDH-wild-type DAGs-MF (n = 64) and -nonMF (n = 13) compared with those in normal brains (n = 5; Supplementary Table S4). EZH2 was also the most upregulated gene among them compared with normal brains (Supplementary Fig. S3B and S3C). In addition, all human glioma cell lines examined, such as TM31 (IDH-wild-type DAG-nonMF cell line), Onda10, Onda11, KINGS1 (IDH-wild-type DAG-MF cell lines), T98, U251, and U87 (IDH-wild-type GBM cell lines) showed high protein levels of EZH2 (Supplementary Fig. S3D).

Figure 2.

Elevated expression of PRC2 components in MADM GFP-positive cells and human IDH-wild-type DAGs. A, EZH2 protein expression levels in normal NSCs, ASTs, and MADM tumor cells. β-Actin protein expression was used as an internal control. B, Gene expression levels of Ezh2, Suz12, and Eed in normal mouse brains (white bars) and MADM GFP-positive precancerous cells in P20 and P60 and tumor cells in P90 and P120 (black bars). The y-axis indicates expression levels normalized to Gapdh. Error bars, SD. C, Protein levels of EZH2, SUZ12, EED, and H3K27me3 in normal NSC, AST, and GFP-positive precancerous cells in P8 and P60 and tumor cells in P120. Expression levels of β-actin protein and histone H3 were used as internal controls. D, IHC staining of EZH2 (red) in P20 MADM mouse brains (top left) and MADM tumor (P120; bottom left; bars, 10 μm). White asterisks indicate GFP-positive cells and white arrowheads indicate EZH2-positive cells. Right, relative mean fluorescence intensity of EZH2 in P20 and P120 GFP-positive [GFP (+); black bars] and negative cells [GFP (−); white bars]. EZH2 fluorescence intensity in the nuclei of DAPI-stained cells was averaged in four fields [mean 115 GFP (+) cells and 43 GFP (−) cells]. The y-axis indicates the mean value of EZH2 fluorescence intensities among GFP (+) cells, relative to those among GFP (−) cells. Error bars, SD. E, Correlation between gene expression levels of three core components of PRC2, EZH2, SUZ12, and EED in IDH-wild-type DAGs-MF and IDH-wild-type DAGs-nonMF (n = 64 and n = 13, respectively). The width of lines connecting two components reflects the Pearson correlation coefficient values (r) between the gene expression levels of these connected components. F, Boxplot of relative expression levels of PRC2 target genes (904 genes) in IDH-wild-type DAGs-MF or IDH-wild-type DAGs-nonMF (n = 64 and n = 13, respectively) compared with normal brains (n = 5). The y-axis indicates the mean log ratio of RSEM values in IDH-wild-type DAGs-MF or IDH-wild-type DAGs-nonMF relative to those of normal brains. Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. **, P < 0.05; ***, P < 0.01; ns, not significant.

Figure 2.

Elevated expression of PRC2 components in MADM GFP-positive cells and human IDH-wild-type DAGs. A, EZH2 protein expression levels in normal NSCs, ASTs, and MADM tumor cells. β-Actin protein expression was used as an internal control. B, Gene expression levels of Ezh2, Suz12, and Eed in normal mouse brains (white bars) and MADM GFP-positive precancerous cells in P20 and P60 and tumor cells in P90 and P120 (black bars). The y-axis indicates expression levels normalized to Gapdh. Error bars, SD. C, Protein levels of EZH2, SUZ12, EED, and H3K27me3 in normal NSC, AST, and GFP-positive precancerous cells in P8 and P60 and tumor cells in P120. Expression levels of β-actin protein and histone H3 were used as internal controls. D, IHC staining of EZH2 (red) in P20 MADM mouse brains (top left) and MADM tumor (P120; bottom left; bars, 10 μm). White asterisks indicate GFP-positive cells and white arrowheads indicate EZH2-positive cells. Right, relative mean fluorescence intensity of EZH2 in P20 and P120 GFP-positive [GFP (+); black bars] and negative cells [GFP (−); white bars]. EZH2 fluorescence intensity in the nuclei of DAPI-stained cells was averaged in four fields [mean 115 GFP (+) cells and 43 GFP (−) cells]. The y-axis indicates the mean value of EZH2 fluorescence intensities among GFP (+) cells, relative to those among GFP (−) cells. Error bars, SD. E, Correlation between gene expression levels of three core components of PRC2, EZH2, SUZ12, and EED in IDH-wild-type DAGs-MF and IDH-wild-type DAGs-nonMF (n = 64 and n = 13, respectively). The width of lines connecting two components reflects the Pearson correlation coefficient values (r) between the gene expression levels of these connected components. F, Boxplot of relative expression levels of PRC2 target genes (904 genes) in IDH-wild-type DAGs-MF or IDH-wild-type DAGs-nonMF (n = 64 and n = 13, respectively) compared with normal brains (n = 5). The y-axis indicates the mean log ratio of RSEM values in IDH-wild-type DAGs-MF or IDH-wild-type DAGs-nonMF relative to those of normal brains. Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. **, P < 0.05; ***, P < 0.01; ns, not significant.

Close modal

Recent analysis revealed the nonsimultaneous overexpression of EZH2, EED, and SUZ12 in human gliomas: the frequent overexpression of EZH2 and relatively lower frequencies of EED and SUZ12 overexpression were observed (31), indicating that specific EZH2 functions are independent of the PRC2-repressive function (32) in a certain population of gliomas. Of note, the expression levels of EZH2, SUZ12, and EED were highly concordant with each other in IDH-wild-type DAG-nonMF compared with IDH-wild-type DAG-MF in the TCGA dataset (Fig. 2E). Consistently, lower expression levels of known PRC2 target genes (904 genes; ref. 33) were observed in IDH-wild-type DAG-nonMF compared with IDH-wild-type DAG-MF (P < 0.01, Fig. 2F). These data suggest the PRC2 dysregulation subsequent to genetic alterations of p53 and NF1 genes play pivotal roles in MADM tumor and IDH-wild-type DAGs-nonMF formation as well.

Genetic deletion of Ezh2 suppresses glioma formation

Epigenomic analysis of the MADM mouse model suggested an important role for EZH2/PRC2 during the tumorigenesis of IDH-wild-type DAG-nonMF tumors. Next, we generated Ezh2Δ/wt MADM mice, in which the Ezh2 catalytic SET domain was heterozygously deleted by the Cre/loxP system (34). In MADM mice, the GFAP promoter becomes active in NSC at E13.5 days and regulates Cre expression. In Ezh2Δ/wt MADM tumors (P120), the mRNA expression level of the Ezh2 SET domain region was significantly decreased compared with that in Ezh2 wild-type MADM tumors (P120; Fig. 3A). Consistently, H3K27me3 levels were significantly reduced in Ezh2Δ/wt MADM tumors compared with those of Ezh2 wild-type MADM tumors (P < 0.01; Fig. 3B). Tumor growth in Ezh2Δ/wt MADM mice (n = 14) was significantly suppressed compared with that observed in Ezh2 wild-type MADM mice (n = 10), resulting in significantly prolonged survival (P < 0.05; Fig. 3C).

Figure 3.

Genetic deletion of Ezh2 suppresses glioma formation. A, Bottom, gene expression levels of Ezh2 in Ezh2 wild-type (white bars) and Ezh2Δ/wt (black bars) MADM mice. Top, primers were designed to amplify the CXC domain (#1, as a control region) and SET domain (#2) regions. Error bars, SD. B, Left, IHC staining of H3K27me3 (red) in GFP-positive cells in Ezh2 wild-type and Ezh2Δ/wt MADM tumors (bars, 10 μm). Right, bar graph indicating the fluorescence intensities of H3K27me3 in Ezh2 wild-type and Ezh2Δ/wt MADM tumors relative to that of Ezh2 wild-type MADM tumors. Fluorescence intensities of H3K27me3 in DAPI staining cells were averaged in four fields (mean cell number was 48 for Ezh2 wild-type MADM tumors and 37 for Ezh2Δ/wt MADM tumors). The y-axis indicates the mean value of relative fluorescence intensities of H3K27me3 compared with that in Ezh2 wild-type MADM mice tumors. Error bars, SD. C, Left, hematoxylin and eosin staining of Ezh2 wild-type (P120) and Ezh2Δ/wt MADM mouse brains (P140). Tumor lesions are surrounded by dotted lines. Middle, mean maximum tumor area (mm2) in Ezh2 wild-type and Ezh2Δ/wt MADM mice (n = 3 and n = 5, respectively). Error bars, SD. Right, Kaplan–Meier curves for Ezh2Δ/wt MADM mice (black line; n = 14) and Ezh2 wild-type MADM mice (red line; n = 10). D, Volcano plot illustrating the difference in gene expression between Ezh2 wild-type and Ezh2Δ/wt MADM tumor tissues. Log2-fold change in gene expression values are plotted on the x-axis and calculated adjusted log10P values are plotted on the y-axis. Horizontal dotted line indicates P = 0.05, while vertical dotted lines indicate fold change = 2. E, Boxplot of expression levels of genes significantly upregulated in Ezh2Δ/wt MADM mice (175 genes, fold change >2 and P < 0.05) in normal mouse brains, Ezh2 wild-type MADM mice, and Ezh2Δ/wt MADM mice tumors. Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. F, Expression levels of bivalent (biv) or H3K27me3 (K27) modified genes in either Ezh2 wild-type or Ezh2Δ/wt MADM tumors are shown. The y-axis indicates the normalized expression value (log2). Error bars, 95% CI. G, Significant GO terms of 175 upregulated genes following Ezh2 gene depletion (P < 0.05). The x-axis indicates the 1/P value. The y-axis indicates identified ontology terms. **, P < 0.05; ***, P < 0.01; ns, not significant.

Figure 3.

Genetic deletion of Ezh2 suppresses glioma formation. A, Bottom, gene expression levels of Ezh2 in Ezh2 wild-type (white bars) and Ezh2Δ/wt (black bars) MADM mice. Top, primers were designed to amplify the CXC domain (#1, as a control region) and SET domain (#2) regions. Error bars, SD. B, Left, IHC staining of H3K27me3 (red) in GFP-positive cells in Ezh2 wild-type and Ezh2Δ/wt MADM tumors (bars, 10 μm). Right, bar graph indicating the fluorescence intensities of H3K27me3 in Ezh2 wild-type and Ezh2Δ/wt MADM tumors relative to that of Ezh2 wild-type MADM tumors. Fluorescence intensities of H3K27me3 in DAPI staining cells were averaged in four fields (mean cell number was 48 for Ezh2 wild-type MADM tumors and 37 for Ezh2Δ/wt MADM tumors). The y-axis indicates the mean value of relative fluorescence intensities of H3K27me3 compared with that in Ezh2 wild-type MADM mice tumors. Error bars, SD. C, Left, hematoxylin and eosin staining of Ezh2 wild-type (P120) and Ezh2Δ/wt MADM mouse brains (P140). Tumor lesions are surrounded by dotted lines. Middle, mean maximum tumor area (mm2) in Ezh2 wild-type and Ezh2Δ/wt MADM mice (n = 3 and n = 5, respectively). Error bars, SD. Right, Kaplan–Meier curves for Ezh2Δ/wt MADM mice (black line; n = 14) and Ezh2 wild-type MADM mice (red line; n = 10). D, Volcano plot illustrating the difference in gene expression between Ezh2 wild-type and Ezh2Δ/wt MADM tumor tissues. Log2-fold change in gene expression values are plotted on the x-axis and calculated adjusted log10P values are plotted on the y-axis. Horizontal dotted line indicates P = 0.05, while vertical dotted lines indicate fold change = 2. E, Boxplot of expression levels of genes significantly upregulated in Ezh2Δ/wt MADM mice (175 genes, fold change >2 and P < 0.05) in normal mouse brains, Ezh2 wild-type MADM mice, and Ezh2Δ/wt MADM mice tumors. Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. F, Expression levels of bivalent (biv) or H3K27me3 (K27) modified genes in either Ezh2 wild-type or Ezh2Δ/wt MADM tumors are shown. The y-axis indicates the normalized expression value (log2). Error bars, 95% CI. G, Significant GO terms of 175 upregulated genes following Ezh2 gene depletion (P < 0.05). The x-axis indicates the 1/P value. The y-axis indicates identified ontology terms. **, P < 0.05; ***, P < 0.01; ns, not significant.

Close modal

Gene expression analysis revealed that 175 genes and 261 genes were significantly upregulated and downregulated, respectively, in Ezh2Δ/wt MADM mice compared with Ezh2 wild-type MADM mice (Fig. 3D; Supplementary Table S5). Expression levels of the 175 upregulated genes were significantly lower in Ezh2 wild-type MADM tumors than in normal brain tissues. Heterozygous Ezh2 catalytic inactivation restored the expression of these genes to almost normal levels (P < 0.01; Fig. 3E). We identified genes modified with H3K4me3, H3K27me3, or H3K4me3 and H3K27me3 bivalent modifications in Ezh2 wild-type MADM tumor cells with ChIP-chip (H3K4me3 alone: 4,895 genes, H3K27me3 alone: 690 genes and bivalent: 637 genes; representative genes were listed in Supplementary Table S6). The histone modification status of these genes was reflective of gene expression levels in Ezh2 wild-type MADM tumors (Supplementary Fig. S4A). In addition, repressed genes with H3K27me3 and bivalent H3K4me3 and H3K27me3 modifications in Ezh2 wild-type MADM mice were significantly upregulated in Ezh2Δ/wt MADM mice (P < 0.01, Fig. 3F). We further performed ChIP-seq analysis in NSCs, ASTs, P8 precancerous cells, and P120 MADM tumor cells. The experimental concordances are shown in Supplementary Fig. S4B. The relationship between each modification and corresponding gene expression revealed a significant positive and negative correlation of H3K4me3 and H3K27me3, with mRNA expression, respectively, in all cell types analyzed in duplicate (Supplementary Fig. S4B). GO analysis revealed that the 175 upregulated genes included many neural differentiation-associated genes (Fig. 3G; Supplementary Table S7), while no significant GO terms were found in 261 downregulated genes (P > 0.05).

Depletion of Fzd8 by EZH2-H3K27me3 during tumor formation

Among the 175 significantly upregulated genes caused by Ezh2 gene depletion, Fzd8 showed the lowest expression among genes that were modified by bivalent modification in MADM tumors but were active in P8 cells (Fig. 4A). FZD8, a member of the frizzled protein family of G protein–coupled receptors involved in Wnt signaling, inactivates β-catenin function (35). IHC analysis revealed that FZD8 protein expression levels in Ezh2 wild-type MADM tumors were significantly decreased compared with that in Ezh2Δ/wt MADM tumors (Fig. 4B). In MADM mice, the Fzd8 gene promoter was consistently modified with a H3K4me3 mark in P8 cells, while it acquired H3K27me3 and was modified with bivalent modification in MADM tumor cells (Fig. 4C). The mRNA and protein expression levels of FZD8 were downregulated during tumor formation (Fig. 4D). Ccnd1 is one of the most studied downstream genes of β-catenin transactivation, which promotes cell proliferation (36). During MADM tumor formation, RNA expression level of Ccnd1 gene was upregulated (Fig. 4E).

Figure 4.

Fzd8 is downregulated by H3K27me3 during tumor formation. A, Expression levels of genes that were activated with H3K4me3 in P8 among bivalently modified genes in MADM tumors. The y-axis indicates the mean normalized value among three tumors. B, Left, IHC staining of FZD8 in Ezh2 wild-type and Ezh2Δ/wt MADM tumors (bars, 20 μm). Right, FZD8 fluorescence intensities were averaged in four fields (mean 55 cells). The y-axis indicates the mean FZD8 fluorescence intensity, relative to that in Ezh2 wild-type tumors. Error bars, SD. C, Left, ChIP-chip signals of H3K4me3 (red) and H3K27me3 (blue) in P8 and MADM tumor cells. The y-axis indicates normalized value of log2 [Cy5 (ChIP target)/Cy3 (input)]. Right, relative enrichment value of H3K4me3 or H3K27me3 in the Fzd8 promoter and negative control loci without these modifications (NC; see Materials and Methods) in P8 and MADM tumor cells analyzed by ChIP-qPCR. The y-axis indicates relative enrichment value of each modification to input DNA. The y-axis indicates the relative signal intensity of each histone modification normalized by input DNA. Error bars, SD. D, Left, mRNA expression levels of Fzd8 in P20-P120 cells. The y-axis indicates the expression level normalized to Gapdh. Top right, IHC staining of FZD8 in P20-P90 MADM mice brains (bars, 20 μm). Bottom right, FZD8 fluorescence intensities were averaged in four fields (mean 53 cells). The y-axis indicates the mean FZD8 fluorescence intensity. Error bars, SD. E, mRNA expression levels of Ccnd1 in P20 and P120 cells. The y-axis indicates the expression level of Ccnd1 normalized to Gapdh. Error bars, SD. F, Top, protein expression levels of FZD8 and β-actin in GFP-positive cells treated with either si-negative control (NC), si-Fzd8-1, or si-Fzd8-2. Bottom, cell numbers of P20 cells (#1 and #2) and MADM tumor cells (#1 and #2) treated with either si-NC, si-Fzd8-1, or si-Fzd8-2 for 72 hours. The y-axis indicates the cell proliferation ratio (after/before treatment relative to si-NC–treated cells). Error bars, SD. G, Top, protein expression levels of FZD8 and β-actin in GFP-positive cells after transfection of either pcDNA3.1(+) (NC) or pcDNA3.1 (+)-Fzd8 (pcDNA-Fzd8). Bottom, cell numbers of P20 cells (#1 and #2) and MADM tumor cells (#1 and #2) after transfection of these vectors for 72 hours. The y-axis indicates the cell proliferation ratio (after/before treatment). Error bars, SD. H, Boxplot of RSEM of FZD8 in human LGGs (n = 415) and normal brains (n = 5). Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. **, P < 0.05; ***, P < 0.01; ns, not significant.

Figure 4.

Fzd8 is downregulated by H3K27me3 during tumor formation. A, Expression levels of genes that were activated with H3K4me3 in P8 among bivalently modified genes in MADM tumors. The y-axis indicates the mean normalized value among three tumors. B, Left, IHC staining of FZD8 in Ezh2 wild-type and Ezh2Δ/wt MADM tumors (bars, 20 μm). Right, FZD8 fluorescence intensities were averaged in four fields (mean 55 cells). The y-axis indicates the mean FZD8 fluorescence intensity, relative to that in Ezh2 wild-type tumors. Error bars, SD. C, Left, ChIP-chip signals of H3K4me3 (red) and H3K27me3 (blue) in P8 and MADM tumor cells. The y-axis indicates normalized value of log2 [Cy5 (ChIP target)/Cy3 (input)]. Right, relative enrichment value of H3K4me3 or H3K27me3 in the Fzd8 promoter and negative control loci without these modifications (NC; see Materials and Methods) in P8 and MADM tumor cells analyzed by ChIP-qPCR. The y-axis indicates relative enrichment value of each modification to input DNA. The y-axis indicates the relative signal intensity of each histone modification normalized by input DNA. Error bars, SD. D, Left, mRNA expression levels of Fzd8 in P20-P120 cells. The y-axis indicates the expression level normalized to Gapdh. Top right, IHC staining of FZD8 in P20-P90 MADM mice brains (bars, 20 μm). Bottom right, FZD8 fluorescence intensities were averaged in four fields (mean 53 cells). The y-axis indicates the mean FZD8 fluorescence intensity. Error bars, SD. E, mRNA expression levels of Ccnd1 in P20 and P120 cells. The y-axis indicates the expression level of Ccnd1 normalized to Gapdh. Error bars, SD. F, Top, protein expression levels of FZD8 and β-actin in GFP-positive cells treated with either si-negative control (NC), si-Fzd8-1, or si-Fzd8-2. Bottom, cell numbers of P20 cells (#1 and #2) and MADM tumor cells (#1 and #2) treated with either si-NC, si-Fzd8-1, or si-Fzd8-2 for 72 hours. The y-axis indicates the cell proliferation ratio (after/before treatment relative to si-NC–treated cells). Error bars, SD. G, Top, protein expression levels of FZD8 and β-actin in GFP-positive cells after transfection of either pcDNA3.1(+) (NC) or pcDNA3.1 (+)-Fzd8 (pcDNA-Fzd8). Bottom, cell numbers of P20 cells (#1 and #2) and MADM tumor cells (#1 and #2) after transfection of these vectors for 72 hours. The y-axis indicates the cell proliferation ratio (after/before treatment). Error bars, SD. H, Boxplot of RSEM of FZD8 in human LGGs (n = 415) and normal brains (n = 5). Middle horizontal line inside the box indicates the median. Bottom and top of the box indicate the 25th and 75th percentiles, respectively. **, P < 0.05; ***, P < 0.01; ns, not significant.

Close modal

Depletion of Fzd8 by siRNA increased the proliferation of P20 GFP–positive precancerous cells, while minimal effects were seen in MADM tumor cells, in which Fzd8 had already been repressed (Fig. 4F). In contrast, the overexpression of Fzd8 decreased the proliferation of MADM tumor cells, while no effects were seen in P20 GFP-positive precancerous cells (Fig. 4G). Notably, the expression levels of FZD8 mRNA in human gliomas were lower than those in normal brains (Fig. 4H). Taken together, our data indicate that the dysregulation of EZH2/PRC2 induces aberrant histone modification and may abolish the normal differentiation program via disruption of important mediator pathways, such as FZD8-Wnt–mediated signaling.

Effects of pharmacologic EZH2 inhibition on MADM tumors

Selective EZH2 catalytic inhibitors are effective against hematopoietic malignancy. Therefore, we examined the effects of pharmacologic EZH2 inhibition on MADM tumors by the post-oral (PO) administration of EPZ6438, a selective small-molecule inhibitor of EZH2 (11). EPZ6438 significantly inhibited the cell growth of MADM tumor cells in vitro. EPZ6438 transcriptionally downregulated Ccnd1 expression together with upregulation of FZD8 expression via reduction of H3K27me3 level in MADM tumor cells (Fig. 5A). Of note, EPZ6438 significantly inhibited the cell growth of TM31, a representative human IDH-wild-type DAG-nonMF cell line, which has no EGFR amplification, no combined whole chromosome 7 gain and whole chromosome 10 loss, and no TERT promoter mutation (Fig. 5B; Supplementary Fig. S5). In TM31 cells, EPZ6438 downregulated β-catenin expression together with upregulation of FZD8 expression via reduction of H3K27me3 level (Fig. 5B). In contrast, EPZ6438 had a minimal effect on the growth of human IDH-wild-type DAG-MF cell lines (Onda10, Onda11, KINGS1, and SW1783; all cells have a TERT gene promoter mutation) and human IDH-wild-type GBM cell lines (T98, U251, and U87), although EPZ6438 reduced the H3K27me3 level in these cells and TM31 at the comparable level (Fig. 5C). Furthermore, EPZ6438 had a minimal effect on the growth of a mouse GBM cell line, GL261 (Fig. 5D).

Figure 5.

An EZH2 inhibitor inhibits MADM tumor and human IDH-wild-type DAG-nonMF growth. A, Left, MADM tumor cells were treated with DMSO (control), or 1 or 2.5 μmol/L EPZ6438 for 6 days. Cell viabilities were measured by MTT assay. The y-axis indicates the absorbance measured at 450 nm. Error bars, SD. Middle, protein levels of H3K27me3 and FZD8 in MADM tumor cells treated with DMSO, or 2.5 μmol/L. Protein levels of histone H3 and β-actin were used as internal controls. Right, mRNA expression levels of Fzd8 and Ccnd1 in MADM tumor cells treated with DMSO, or 2.5 μmol/L (white box or black boxes, respectively). The y-axis indicates the expression level normalized to Gapdh. Error bars, SD. B, Left, TM31 cells (IDH-wild-type DAG-nonMF), Onda10, Onda11, KINGS1, and SW1783 cells (IDH-wild-type DAGs-MF), T98, and U251 and U87 cells (IDH-wild-type GBMs) were treated with DMSO (control), or 2.5 or 5 μmol/L EPZ6438 for 6 days. The tumor type of each cell line is described. Cell viabilities were measured by MTT assay. The y-axis indicates the relative absorbance measured at 450 nm compared with those of DMSO-treated cells. Error bars, SD. Right, protein expression levels of FZD8 and β-catenin in TM31 cells treated with DMSO, or 2.5 or 5 μmol/L EPZ6438. β-Actin protein was used as an internal control. C, Protein levels of H3K27me3 in TM31, Onda10 or U251 cells treated with DMSO, or 2.5 or 5 μmol/L EPZ6438. Histone H3 was used as an internal control. D, GL261 (mouse IDH-wild-type GBM) cells were treated with DMSO (control), or 2.5 or 5 μmol/L EPZ6438 for 6 days. Cell viabilities were measured by MTT assay. The y-axis indicates the relative absorbance measured at 450 nm compared with those of DMSO-treated cells. Error bars, SD. E, T2-weighted MRI images of control (top) and EPZ6438-treated mice (bottom) at pre- (P90; left) and posttreatment (P110; right). White arrows, tumor lesions. Right, bar graph of relative volumetric rate calculated by post-/pretreatment tumor volume in control (n = 7) and EPZ6438-treated mice (n = 8). Error bars, SD. F, IHC staining of H3K27me3 (red; left) or FZD8 (red; right) in control (top) or EPZ6438-treated (bottom) MADM mouse brains (bars, 10 μm). Right, bar graph indicating the relative fluorescence intensities of H3K27me3 and FZD8 in EPZ6438-treated mouse brains (black box) compared with those in control mice brains (white box). Both fluorescence intensities of H3K27me3 and FZD8 in DAPI staining cells were averaged in four fields (mean cell number was 41 for H3K27me3 and 32 for FZD8). The y-axis indicates the mean value of relative fluorescence intensities compared with those in control mice brains. Error bars, SD. ***, P < 0.01; ns, not significant.

Figure 5.

An EZH2 inhibitor inhibits MADM tumor and human IDH-wild-type DAG-nonMF growth. A, Left, MADM tumor cells were treated with DMSO (control), or 1 or 2.5 μmol/L EPZ6438 for 6 days. Cell viabilities were measured by MTT assay. The y-axis indicates the absorbance measured at 450 nm. Error bars, SD. Middle, protein levels of H3K27me3 and FZD8 in MADM tumor cells treated with DMSO, or 2.5 μmol/L. Protein levels of histone H3 and β-actin were used as internal controls. Right, mRNA expression levels of Fzd8 and Ccnd1 in MADM tumor cells treated with DMSO, or 2.5 μmol/L (white box or black boxes, respectively). The y-axis indicates the expression level normalized to Gapdh. Error bars, SD. B, Left, TM31 cells (IDH-wild-type DAG-nonMF), Onda10, Onda11, KINGS1, and SW1783 cells (IDH-wild-type DAGs-MF), T98, and U251 and U87 cells (IDH-wild-type GBMs) were treated with DMSO (control), or 2.5 or 5 μmol/L EPZ6438 for 6 days. The tumor type of each cell line is described. Cell viabilities were measured by MTT assay. The y-axis indicates the relative absorbance measured at 450 nm compared with those of DMSO-treated cells. Error bars, SD. Right, protein expression levels of FZD8 and β-catenin in TM31 cells treated with DMSO, or 2.5 or 5 μmol/L EPZ6438. β-Actin protein was used as an internal control. C, Protein levels of H3K27me3 in TM31, Onda10 or U251 cells treated with DMSO, or 2.5 or 5 μmol/L EPZ6438. Histone H3 was used as an internal control. D, GL261 (mouse IDH-wild-type GBM) cells were treated with DMSO (control), or 2.5 or 5 μmol/L EPZ6438 for 6 days. Cell viabilities were measured by MTT assay. The y-axis indicates the relative absorbance measured at 450 nm compared with those of DMSO-treated cells. Error bars, SD. E, T2-weighted MRI images of control (top) and EPZ6438-treated mice (bottom) at pre- (P90; left) and posttreatment (P110; right). White arrows, tumor lesions. Right, bar graph of relative volumetric rate calculated by post-/pretreatment tumor volume in control (n = 7) and EPZ6438-treated mice (n = 8). Error bars, SD. F, IHC staining of H3K27me3 (red; left) or FZD8 (red; right) in control (top) or EPZ6438-treated (bottom) MADM mouse brains (bars, 10 μm). Right, bar graph indicating the relative fluorescence intensities of H3K27me3 and FZD8 in EPZ6438-treated mouse brains (black box) compared with those in control mice brains (white box). Both fluorescence intensities of H3K27me3 and FZD8 in DAPI staining cells were averaged in four fields (mean cell number was 41 for H3K27me3 and 32 for FZD8). The y-axis indicates the mean value of relative fluorescence intensities compared with those in control mice brains. Error bars, SD. ***, P < 0.01; ns, not significant.

Close modal

In MADM mice, substantial tumors can be detected by MRI imaging at P90. We therefore started the treatment of MADM mice from P90 onwards. Treatment arms were the PO administration of EPZ6438 at a dose of 160 mg/kg (n = 8) or PBS as a control (n = 7), twice a day for 20 days (11). Consistent with the genetic inhibition of Ezh2, EPZ6438 treatment significantly suppressed tumor growth compared with controls, along with the downregulation of H3K27me3 and upregulation of FZD8 expression in tumor tissues (Fig. 5E and F).

Precision medicine based on the characteristics of each cancer type is required to deliver the most appropriate treatment. Recent studies, including our own, have extensively demonstrated multiple genetic and epigenetic alterations in glioma formation (3–5, 30, 37). While G-CIMP is a characteristic epigenetic feature of IDH-mutant LGG (6, 7), the molecular basis underlying the tumorigenesis of the often-devastating IDH-wild-type DAG is still largely unclear. In this study, we identified the dysregulation of EZH2/PRC2 in both human IDH-wild-type DAG-nonMF and a MADM glioma mouse model in which the genetic and gene expression profile and histopathologic findings resemble this type of glioma.

In our mouse model, the loss of p53 and NF1 genes may affect the PRC2/EZH2-dependent epigenetic machinery at the early stage of malignancy. Interestingly, Ezh2 expression was not affected in mice with a p53 homozygous deletion alone (Supplementary Fig. S6). Given previous evidence that the genetic loss of NF1 activated RAS/MEK/ERK signaling (38) and that the activation of RAS/MEK/ERK signaling upregulated EZH2 expression (39, 40), we argue that the deletion of NF1 may contribute to the upregulation of EZH2 in our MADM mouse model.

Aberrant activation of the Wnt pathway is involved in gliomagenesis via multiple mechanisms. Various negative regulators of the Wnt pathway, such as FAT Atypical Cadherin 1 (FAT1), and Frizzled-related proteins (FRP) such as Dickkopf (DKK), are downregulated in gliomas (41, 42). WNT5A, a classical component of noncanonical Wnt signaling, is commonly upregulated in gliomas and sometimes promotes its invasiveness (43). In this study, we newly report the aberrant activation of the Wnt pathway via the PRC2/EZH2-mediated epigenetic repression of FZD8. Human FZD8 is expressed in the brain, pancreas, kidney, heart, and muscle (44). It is also involved in hematopoietic stem cell maintenance via activation of the noncanonical Wnt pathway (35). A previous study reported that abundant FZD8 expression in normal pancreas tissues was frequently lost in pancreatic adenocarcinomas (45). Mouse model analysis revealed that oncogenic K-Ras causes a tumorigenic phenotype through the downregulation of noncanonical Wnt/Ca2+ signaling via the repression of FZD8. Restoring FZD8 in K-Ras–mutant pancreatic cells suppressed malignancy, indicating that the suppression of FZD8-mediated Wnt signaling was essential to K-Ras malignancy (45). Because the RAS/MAPK signaling suppressor, NF1, is deleted in the MADM mouse model, suppression of Fzd8 via aberrant bivalent modification may also play a key role in gliomagenesis via activation of Wnt signaling. Indeed, the suppression or overexpression of FZD8 in GFP-positive MADM cells significantly altered cell growth.

The balance between H3K4me3 and H3K27me3 modification levels is crucial for proper regulation of differentiation. It is known that pluripotent embryonic stem (ES) cells and multipotent NSCs exhibit characteristic bivalent modifications of concurrent active H3K4me3 and repressive H3K27me3, which are characteristic modifications of suppressive and poised transcription on a set of important differentiation-associated genes. Dysregulation of these modifications, such as inappropriate activation by the removal of H3K27me3 or constitutive repression by the removal of H3K4me3, may contribute to tumor initiation and/or progression (46). Fzd8 gene was modified with bivalent modification in ES cells and was activated by the removal of H3K27me3 in NSCs (47). During glioma tumorigenesis, repression of Fzd8 by aberrant bivalent modification may play a pivotal role in gliomagenesis via dysregulation of neural differentiation together with promoting of cell proliferation.

A previous study showed the oligodendrocyte precursor cell (OPC) was the cell of origin for glioma in the MADM mouse model (13). Furthermore, the deletion of NF1 led to mutant OPC expansion through increased proliferation and decreased differentiation, while the deletion of p53 impaired OPC senescence, indicating that specific aberrations caused by individual gene mutations and altered signaling events occur at specific times during gliomagenesis (48). In this study, we provide further key evidence that aberrant epigenetic phenomena induced by certain tumor-related genes affect the activation of reprogramming-associated genes during gliomagenesis. Recently, a conceptual framework was proposed whereby epigenetic functional disorders in cancers can be driven by epigenetic modulators, epigenetic modifiers, and epigenetic mediators (49). An epigenetic modulator regulates the function of epigenetic modifiers and leads to the regulation of epigenetic mediators whose gene products regulate proliferation, development, and cellular plasticity. Mutations in modulators and modifiers are often selected for during cancer development (49). Our data shows that aberrant histone modifications were reproducibly induced by concomitant p53 and NF1 genetic loss during the tumorigenic process from precancerous cells to MADM tumors within individual MADM mice.

Although EZH2 is upregulated in malignant gliomas (50), the therapeutic benefit of targeting EZH2 as a novel therapy for gliomas is still under investigation. Prolonged EZH2 depletion has been reported to lead to more aggressive phenotypes of gliomas (51). In contrast, EZH2 inhibitors appear to be effective in certain types of gliomas such as diffuse intrinsic pontine glioma, which is the most lethal form of pediatric glioma (52, 53). The noncanonical roles of EZH2 independent of PRC2 activity and other PRC2 components, should be addressed before targeting EZH2 as a treatment (32, 54). In IDH-wild-type DAGs-nonMF, three core components of PRC2 are more closely associated with each other than in IDH-wild-type DAGs-MF, suggesting the importance of PRC2 activity in this type of glioma.

Comprehensive mRNA analysis also revealed that gliomas can be divided into four subtypes based on gene expression profiling, namely classical, neural, mesenchymal, and proneural subtypes (55). A recent study demonstrated that the high expression of EZH2 was preferentially found in proneural type glioma stem cells and that the inhibition of EZH2 preferentially reduced the growth of this type of cells (56). In MADM tumors, gene expression profiles were concordant with the proneural cell type, as reported previously (13). Given that the genetic and pharmacologic inhibition of EZH2 efficiently suppresses gliomas in MADM mice and a human IDH-wild-type DAG-nonMF cell line, EZH2 chemical inhibitors may be applicable for the treatment of IDH wild-type DAG-nonMF. In contrast, the chemical inhibitor of catalytic activity of EZH2 had the minimal effects on cell proliferation of IDH-wild-type DAGs-MF cell lines (Onda10, Onda11, KINGS1, and SW1783) and IDH-wild-type grade IV GBM cell lines (T98, U251, and U87) although it reduced H3K27me3 levels in these cells at comparable level with that in TM31. This may be explained by the accumulation of other genetic and epigenetic alterations in IDH-wild-type DAGs-MF and GBMs (Supplementary Fig. S7). Specific EZH2 functions, which are independent of the PRC2 repressive function, may contribute to gliomagenesis of IDH-wild-type DAGs-MF and GBMs. Further clinical trials are required to examine this possibility.

To achieve the essential requirements of precision medicine for patient-specific therapy, our combined analyses of human IDH-wild-type DAGs and the MADM mouse system will help improve the stratification of cancers by utilizing biological biomarkers and the development of effective treatment strategies. Our data clarify a pathogenic molecular pathway of IDH-wild-type DAG-nonMF, which provides a strong rationale for targeting EZH2 to treat this subtype of glioma.

F. Ohka has received speakers bureau honoraria from Daiichi Sankyo. No potential conflicts of interest were disclosed by the other authors.

Conception and design: F. Ohka, K. Katsushima, A. Suzumura, H. Zong, A. Natsume, Y. Kondo

Development of methodology: F. Ohka, K. Shinjo, Y. Okuno, K. Katsushima, K. Aoki, N.A. Rayan, H. Kimura, H. Zong

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): F. Ohka, K. Shinjo, S. Deguchi, Y. Okuno, A. Kato, N. Ogiso, K. Aoki, R. Maruyama, S. Takahashi

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): F. Ohka, K. Shinjo, Y. Matsui, Y. Okuno, M. Suzuki, N. Ogiso, K. Aoki, H. Suzuki, N.A. Rayan, S. Prabhakar, J. Göke, T. Shimamura

Writing, review, and/or revision of the manuscript: F. Ohka, Y. Okuno, K. Aoki, S. Sato, A. Natsume, Y. Kondo

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): F. Ohka, Y. Okuno, A. Yamamichi, S. Sato, H. Kimura, H. Zong

Study supervision: A. Suzumura, T. Wakabayashi, H. Zong, A. Natsume, Y. Kondo

This study was performed as a research program of P-CREATE, Japan Agency for Medical Research and Development (18cm0106108h0003 to Y. Kondo), and of the Grant-in-Aid for Scientific Research, the Japan Society for the Promotion of Science (17H03582 to Y. Kondo; 17928985 to A. Natsume).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Louis
DN
,
Perry
A
,
Burger
P
,
Ellison
DW
,
Reifenberger
G
,
von Deimling
A
, et al
International Society Of Neuropathology–Haarlem consensus guidelines for nervous system tumor classification and grading
.
Brain Pathol
2014
;
24
:
429
35
.
2.
Louis
DN
,
Perry
A
,
Reifenberger
G
,
von Deimling
A
,
Figarella-Branger
D
,
Cavenee
WK
, et al
The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary
.
Acta Neuropathol
2016
;
131
:
803
20
.
3.
Brat
DJ
,
Verhaak
RG
,
Aldape
KD
,
Yung
WK
,
Salama
SR
,
Cooper
LA
, et al
Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas
.
N Engl J Med
2015
;
372
:
2481
98
.
4.
Ceccarelli
M
,
Barthel
FP
,
Malta
TM
,
Sabedot
TS
,
Salama
SR
,
Murray
BA
, et al
Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma
.
Cell
2016
;
164
:
550
63
.
5.
Suzuki
H
,
Aoki
K
,
Chiba
K
,
Sato
Y
,
Shiozawa
Y
,
Shiraishi
Y
, et al
Mutational landscape and clonal architecture in grade II and III gliomas
.
Nat Genet
2015
;
47
:
458
68
.
6.
Noushmehr
H
,
Weisenberger
DJ
,
Diefes
K
,
Phillips
HS
,
Pujara
K
,
Berman
BP
, et al
Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma
.
Cancer Cell
2010
;
17
:
510
22
.
7.
Turcan
S
,
Rohle
D
,
Goenka
A
,
Walsh
LA
,
Fang
F
,
Yilmaz
E
, et al
IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype
.
Nature
2012
;
483
:
479
83
.
8.
Stichel
D
,
Ebrahimi
A
,
Reuss
D
,
Schrimpf
D
,
Ono
T
,
Shirahata
M
, et al
Distribution of EGFR amplification, combined chromosome 7 gain and chromosome 10 loss, and TERT promoter mutation in brain tumors and their potential for the reclassification of IDHwt astrocytoma to glioblastoma
.
Acta Neuropathol (Berl)
2018
;
136
:
793
803
.
9.
Brat
DJ
,
Aldape
K
,
Colman
H
,
Holland
EC
,
Louis
DN
,
Jenkins
RB
, et al
cIMPACT-NOW update 3: recommended diagnostic criteria for "Diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV"
.
Acta Neuropathol
2018
;
136
:
805
10
.
10.
Zong
H
,
Espinosa
JS
,
Su
HH
,
Muzumdar
MD
,
Luo
L
. 
Mosaic analysis with double markers in mice
.
Cell
2005
;
121
:
479
92
.
11.
Knutson
SK
,
Kawano
S
,
Minoshima
Y
,
Warholic
NM
,
Huang
KC
,
Xiao
Y
, et al
Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma
.
Mol Cancer Ther
2014
;
13
:
842
54
.
12.
Katsushima
K
,
Natsume
A
,
Ohka
F
,
Shinjo
K
,
Hatanaka
A
,
Ichimura
N
, et al
Targeting the Notch-regulated non-coding RNA TUG1 for glioma treatment
.
Nat Commun
2016
;
7
:
13616
.
13.
Liu
C
,
Sage
JC
,
Miller
MR
,
Verhaak
RG
,
Hippenmeyer
S
,
Vogel
H
, et al
Mosaic analysis with double markers reveals tumor cell of origin in glioma
.
Cell
2011
;
146
:
209
21
.
14.
Sonoda
Y
,
Ozawa
T
,
Hirose
Y
,
Aldape
KD
,
McMahon
M
,
Berger
MS
, et al
Formation of intracranial tumors by genetically modified human astrocytes defines four pathways critical in the development of human anaplastic astrocytoma
.
Cancer Res
2001
;
61
:
4956
60
.
15.
Kondo
Y
,
Shen
L
,
Yan
PS
,
Huang
TH
,
Issa
JP
. 
Chromatin immunoprecipitation microarrays for identification of genes silenced by histone H3 lysine 9 methylation
.
Proc Natl Acad Sci U S A
2004
;
101
:
7398
403
.
16.
Kondo
Y
,
Shen
L
,
Cheng
AS
,
Ahmed
S
,
Boumber
Y
,
Charo
C
, et al
Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation
.
Nat Genet
2008
;
40
:
741
50
.
17.
Maruyama
R
,
Choudhury
S
,
Kowalczyk
A
,
Bessarabova
M
,
Beresford-Smith
B
,
Conway
T
, et al
Epigenetic regulation of cell type-specific expression patterns in the human mammary epithelium
.
PLos Genet
2011
;
7
:
e1001369
.
18.
Kimura
H
,
Hayashi-Takanaka
Y
,
Goto
Y
,
Takizawa
N
,
Nozaki
N
. 
The organization of histone H3 modifications as revealed by a panel of specific monoclonal antibodies
.
Cell Struct Funct
2008
;
33
:
61
73
.
19.
Kumar
V
,
Rayan
NA
,
Muratani
M
,
Lim
S
,
Elanggovan
B
,
Xin
L
, et al
Comprehensive benchmarking reveals H2BK20 acetylation as a distinctive signature of cell-state-specific enhancers and promoters
.
Genome Res
2016
;
26
:
612
23
.
20.
Xie
W
,
Song
C
,
Young
NL
,
Sperling
AS
,
Xu
F
,
Sridharan
R
, et al
Histone h3 lysine 56 acetylation is linked to the core transcriptional network in human embryonic stem cells
.
Mol Cell
2009
;
33
:
417
27
.
21.
Langmead
B
,
Trapnell
C
,
Pop
M
,
Salzberg
SL
. 
Ultrafast and memory-efficient alignment of short DNA sequences to the human genome
.
Genome Biol
2009
;
10
:
R25
.
22.
Anders
S
,
Pyl
PT
,
Huber
W
. 
HTSeq–a Python framework to work with high-throughput sequencing data
.
Bioinformatics
2015
;
31
:
166
9
.
23.
Zhu
J
,
Sanborn
JZ
,
Benz
S
,
Szeto
C
,
Hsu
F
,
Kuhn
RM
, et al
The UCSC Cancer Genomics Browser
.
Nat Methods
2009
;
6
:
239
40
.
24.
Cline
MS
,
Craft
B
,
Swatloski
T
,
Goldman
M
,
Ma
S
,
Haussler
D
, et al
Exploring TCGA Pan-Cancer data at the UCSC Cancer Genomics Browser
.
Sci Rep
2013
;
3
:
2652
.
25.
Ritchie
ME
,
Phipson
B
,
Wu
D
,
Hu
Y
,
Law
CW
,
Shi
W
, et al
limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res
2015
;
43
:
e47
.
26.
Durinck
S
,
Moreau
Y
,
Kasprzyk
A
,
Davis
S
,
De Moor
B
,
Brazma
A
, et al
BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis
.
Bioinformatics
2005
;
21
:
3439
40
.
27.
Durinck
S
,
Spellman
PT
,
Birney
E
,
Huber
W
. 
Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt
.
Nat Protoc
2009
;
4
:
1184
91
.
28.
Jacobs
VL
,
Valdes
PA
,
Hickey
WF
,
De Leo
JA
. 
Current review of in vivo GBM rodent models: emphasis on the CNS-1 tumour model
.
ASN Neuro
2011
;
3
:
e00063
.
29.
Gusyatiner
O
,
Hegi
ME
. 
Glioma epigenetics: from subclassification to novel treatment options
.
Semin Cancer Biol
2017
;
51
:
50
8
.
30.
Brennan
CW
,
Verhaak
RG
,
McKenna
A
,
Campos
B
,
Noushmehr
H
,
Salama
SR
, et al
The somatic genomic landscape of glioblastoma
.
Cell
2013
;
155
:
462
77
.
31.
Li
G
,
Warden
C
,
Zou
Z
,
Neman
J
,
Krueger
JS
,
Jain
A
, et al
Altered expression of polycomb group genes in glioblastoma multiforme
.
PLoS One
2013
;
8
:
e80970
.
32.
Kondo
Y
. 
Targeting histone methyltransferase EZH2 as cancer treatment
.
J Biochem
2014
;
156
:
249
57
.
33.
Cao
Q
,
Wang
X
,
Zhao
M
,
Yang
R
,
Malik
R
,
Qiao
Y
, et al
The central role of EED in the orchestration of polycomb group complexes
.
Nat Commun
2014
;
5
:
3127
.
34.
Hirabayashi
Y
,
Suzki
N
,
Tsuboi
M
,
Endo
TA
,
Toyoda
T
,
Shinga
J
, et al
Polycomb limits the neurogenic competence of neural precursor cells to promote astrogenic fate transition
.
Neuron
2009
;
63
:
600
13
.
35.
Sugimura
R
,
He
XC
,
Venkatraman
A
,
Arai
F
,
Box
A
,
Semerad
C
, et al
Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche
.
Cell
2012
;
150
:
351
65
.
36.
Reya
T
,
Clevers
H
. 
Wnt signalling in stem cells and cancer
.
Nature
2005
;
434
:
843
50
.
37.
Sturm
D
,
Bender
S
,
Jones
DT
,
Lichter
P
,
Grill
J
,
Becher
O
, et al
Paediatric and adult glioblastoma: multiform (epi)genomic culprits emerge
.
Nat Rev Cancer
2014
;
14
:
92
107
.
38.
Simanshu
DK
,
Nissley
DV
,
McCormick
F
. 
RAS proteins and their regulators in human disease
.
Cell
2017
;
170
:
17
33
.
39.
Fujii
S
,
Fukamachi
K
,
Tsuda
H
,
Ito
K
,
Ito
Y
,
Ochiai
A
. 
RAS oncogenic signal upregulates EZH2 in pancreatic cancer
.
Biochem Biophys Res Commun
2012
;
417
:
1074
9
.
40.
Riquelme
E
,
Behrens
C
,
Lin
HY
,
Simon
G
,
Papadimitrakopoulou
V
,
Izzo
J
, et al
Modulation of EZH2 expression by MEK-ERK or PI3K-AKT signaling in lung cancer is dictated by different KRAS oncogene mutations
.
Cancer Res
2016
;
76
:
675
85
.
41.
Morris
LG
,
Kaufman
AM
,
Gong
Y
,
Ramaswami
D
,
Walsh
LA
,
Turcan
S
, et al
Recurrent somatic mutation of FAT1 in multiple human cancers leads to aberrant Wnt activation
.
Nat Genet
2013
;
45
:
253
61
.
42.
Shou
J
,
Ali-Osman
F
,
Multani
AS
,
Pathak
S
,
Fedi
P
,
Srivenugopal
KS
. 
Human Dkk-1, a gene encoding a Wnt antagonist, responds to DNA damage and its overexpression sensitizes brain tumor cells to apoptosis following alkylation damage of DNA
.
Oncogene
2002
;
21
:
878
89
.
43.
Hu
B
,
Wang
Q
,
Wang
YA
,
Hua
S
,
Sauve
CG
,
Ong
D
, et al
Epigenetic activation of WNT5A drives glioblastoma stem cell differentiation and invasive growth
.
Cell
2016
;
167
:
1281
95
.
44.
Saitoh
T
,
Hirai
M
,
Katoh
M
. 
Molecular cloning and characterization of human Frizzled-8 gene on chromosome 10p11.2
.
Int J Oncol
2001
;
18
:
991
6
.
45.
Wang
M-T
,
Holderfield
M
,
Galeas
J
,
Delrosario
R
,
To
MD
,
Balmain
A
, et al
K-Ras promotes tumorigenicity through suppression of non-canonical Wnt signaling
.
Cell
2015
;
163
:
1237
51
.
46.
Bernstein
BE
,
Mikkelsen
TS
,
Xie
X
,
Kamal
M
,
Huebert
DJ
,
Cuff
J
, et al
A bivalent chromatin structure marks key developmental genes in embryonic stem cells
.
Cell
2006
;
125
:
315
26
.
47.
Mikkelsen
TS
,
Ku
M
,
Jaffe
DB
,
Issac
B
,
Lieberman
E
,
Giannoukos
G
, et al
Genome-wide maps of chromatin state in pluripotent and lineage-committed cells
.
Nature
2007
;
448
:
553
60
.
48.
Gonzalez
PP
,
Kim
J
,
Galvao
RP
,
Cruickshanks
N
,
Abounader
R
,
Zong
H
. 
p53 and NF 1 loss plays distinct but complementary roles in glioma initiation and progression
.
Glia
2018
;
66
:
999
1015
.
49.
Feinberg
AP
,
Koldobskiy
MA
,
Gondor
A
. 
Epigenetic modulators, modifiers and mediators in cancer aetiology and progression
.
Nat Rev Genet
2016
;
17
:
284
99
.
50.
Natsume
A
,
Ito
M
,
Katsushima
K
,
Ohka
F
,
Hatanaka
A
,
Shinjo
K
, et al
Chromatin regulator PRC2 is a key regulator of epigenetic plasticity in glioblastoma
.
Cancer Res
2013
;
73
:
4559
70
.
51.
de Vries
NA
,
Hulsman
D
,
Akhtar
W
,
de Jong
J
,
Miles
DC
,
Blom
M
, et al
Prolonged Ezh2 depletion in glioblastoma causes a robust switch in cell fate resulting in tumor progression
.
Cell Rep
2015
;
10
:
383
97
.
52.
Mohammad
F
,
Weissmann
S
,
Leblanc
B
,
Pandey
DP
,
Højfeldt
JW
,
Comet
I
, et al
EZH2 is a potential therapeutic target for H3K27M-mutant pediatric gliomas
.
Nat Med
2017
;
23
:
483
92
.
53.
Piunti
A
,
Hashizume
R
,
Morgan
MA
,
Bartom
ET
,
Horbinski
CM
,
Marshall
SA
, et al
Therapeutic targeting of polycomb and BET bromodomain proteins in diffuse intrinsic pontine gliomas
.
Nat Med
2017
;
23
:
493
500
.
54.
Kim
E
,
Kim
M
,
Woo
DH
,
Shin
Y
,
Shin
J
,
Chang
N
, et al
Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells
.
Cancer Cell
2013
;
23
:
839
52
.
55.
Verhaak
RG
,
Hoadley
KA
,
Purdom
E
,
Wang
V
,
Qi
Y
,
Wilkerson
MD
, et al
Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1
.
Cancer Cell
2010
;
17
:
98
110
.
56.
Jin
X
,
Kim
LJY
,
Wu
Q
,
Wallace
LC
,
Prager
BC
,
Sanvoranart
T
, et al
Targeting glioma stem cells through combined BMI1 and EZH2 inhibition
.
Nat Med
2017
;
23
:
1352
61
.

Supplementary data