Review

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

Epigenetic modification in gliomas: role of the histone methyltransferase EZH2 1.

Introduction

2.

EZH2

3.

The role of EZH2 in glioma

4.

Overview of DNA methylation

5.

The interaction of DNA methylation with EZH2 in gliomas

6.

The regulation of EZH2 by miRNAs in glioma

7.

Expert opinion

Er-Bao Bian, Jia Li, Xiao-Jun He, Gang Zong, Tao Jiang, Jun Li & Bing Zhao† †

The Second Affiliated Hospital of Anhui Medical University, Department of Neurosurgery, Hefei, China

Introduction: Gliomas are characterized by increased anaplasia, malignization, proliferation and invasion. They exhibit high resistance to standard treatment with combinations of radiotherapy and chemotherapy. They are currently the most common primary malignancy tumors in the brain that is related to a high mortality rate. Recently, increasing evidence suggests that EZH2 is involved in a number of glioma cell processes, including proliferation, apoptosis, invasion and angiogenesis. Areas covered: In this review, we emphasize the role of EZH2 in gliomas. We also address that EZH2 interacting with DNA methylation mediates transcriptional repression of specific genes in gliomas, and the regulation of EZH2 by microRNAs in gliomas. Expert opinion: Although the exact role of EZH2 in gliomas has not been fully elucidated, to understand the role of EZH2 proteins in epigenetic modification will provide valuable insights into the causes of gliomas, and pave the way to the potential future applications of EZH2 in the treatment of gliomas. Keywords: EZH2, glioma, methylation, microRNAs Expert Opin. Ther. Targets (2014) 18(10):1197-1206

1.

Introduction

Gliomas are the most common form of CNS primary tumors, accounting for > 50% of all primary CNS tumors [1]. Malignant gliomas possess a sequence of characteristics: transformation from a cell of origin, increased proliferative combined with abrogation of cell cycle control, clonal evolution, acquisition of invasive ability and activation of angiogenesis signal pathways [2]. Gliomas are tumors of neuroepithelial tissues, including several types that are astrocytic, oligodendroglial, oligoastrocytic, ependymal, choroid plexus-derived or others, according to the cell type with which they share [3]. Gliomas are clinicopathologically categorized into grades I -- IV according to the 2007 classification by the WHO [4]. Histological evaluation remains the gold standard for glioma diagnosis and serves as a criterion to estimate the prognosis of patients [5]. Among all gliomas cases diagnosed, most of them belong to grade IV, identified as high-grade astrocytoma or glioblastoma (GBM) with marked nuclear atypia and high mitotic rates [6,7]. Primary GBM arise de novo, however, secondary GBM derives from pre-existing low-grade astrocytomas [8]. Most patients with these glioma progress to GBM, with current treatment protocols, patients with GBM have a 5-year survival rate of no > 30% [9]. Despite aggressive therapeutic modalities are surgical resection combined with radiation and temozolomide chemotherapy, the prognostic statistics for GBM patients is discouraging [10-12]. GBM is highly infiltrative, however, complete elimination of the tumor by surgery is usually impossible. Further, radiation and chemotherapy can kill most of the remaining tumor cells, those that typically reoccur [13]. 10.1517/14728222.2014.941807 © 2014 Informa UK, Ltd. ISSN 1472-8222, e-ISSN 1744-7631 All rights reserved: reproduction in whole or in part not permitted

1197

E-B Bian et al.

Article highlights. . . .

The role of EZH2 in glioma. The interaction of DNA methylation with EZH2 in gliomas. The regulation of EZH2 by microRNAs in gliomas.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

This box summarizes key points contained in the article.

Accumulating evidences suggests that the presence of a subpopulation of cells with the potential to initiate and maintain growth of gliomas might be pivotal for their resistance to conventional therapies [14]. Very recently, GBM is thought to be derived from a small population of GBM stem cells (GSCs) [15-17]. GSCs and normal neural stem cells appear to share common features, including the expression of neural stem cell markers, the capacity for self-renewal and long-term proliferation, the formation of neurospheres and the capability of differentiating into multiple lineages. Intriguingly, recent studies revealed that in addition to GSCs differentiating into non-GSCs, non-GSCs can become GSCs or tumor-initiating cells and exhibit an enhanced ability to form neurospheres, thereby suggesting that the GSC state may be plastic [18,19]. This phenotypic plasticity between the GSC and non-GSC states may be regulated by the signaling pathways, genetic and epigenetic mechanisms. Recently, increasing evidences indicate that EZH2 have been implicated in gliomas initiation, progression, and importantly, in the formation, maintenance and plasticity of GSCs. A rapidly growing body of data describes the importance of EZH2 in gliomas, whereas most recent reports focus on the interplay between EZH2 and noncoding RNAs. Therefore, EZH2 may emerge as a potential therapeutic target in gliomas and in the development of novel treatment strategies that target both genetic and epigenetic mechanisms of tumorigenesis. With the knowledge that EZH2 is associated with aggressive gliomas, understanding how EZH2 promotes disease progression has gradually made possible the development of epigenetic drugs that directly or indirectly target EZH2. Herein, this review aims to provide a comprehensive overview of EZH2 involvement in gliomas pathogenesis and progression, with specific emphasis on the regulatory network of the interaction of EZH2 with microRNAs (miRNAs) and DNA methylation. 2.

EZH2

Human EZH2 gene is located on the long arm of chromosome 7 at position 7q35 and encodes a 746 amino acid protein that has histone methyltransferase (HMT) activity [20]. Besides its HMT activity, EZH2 protein contains several domains with potential functions (Figure 1). The major domains include CXC, cysteine-rich domain; noncoding RNA-binding domain; SANT (SWI3, ADA2, N-CoR) and TFIIIB DNA1198

binding domain; SET, Su(var)3-9, enhancer of zeste, trithorax domain [21]. The N-terminal SET domains provide binding sites for assembly with the required partner subunits. In addition, both the C-terminal SET domain and the adjacent cysteine-rich CXC domain are required for HMT activity [22-24]. The SET domain of EZH2 is identified as the signature of methyltransferases, which leads to trimethylation (me3) of histone 3 (H3) at lysine 27 (K27), as well as at lysine 9 (K9) albeit to a much lesser extent [25]. When the polycomb repressive complex 2 (PRC2) complex is recruited to chromatin, the HMT EZH2 catalyses the H3K27me3, which results in subsequent recruitment of the PRC1 complex that monoubiquitylates the lysine 119 of histone H2A (H2AK119ub1) to prevent RNA polymerase II-dependent transcriptional elongation and consolidate transcriptional repression [26,27]. More recently, a study reported that EZH2 mutations may either cause gain-of-function or complete loss of HMT activity in lymphoma and myeloid neoplasms [28]. In addition, somatic activating mutations in the catalytic (SET) domain of EZH2 have been identified in follicular lymphoma and germinal center B-cell diffuse large B-cell lymphoma, leading to increased H3K27me3 [29-33]. It is interesting to point out that the role of transcriptional regulation of EZH2 in epigenetics is complex. EZH2, identified as the core member of PRC2, combines with embryonic ectoderm development (EED) and SUZ12 and plays the role of HTMase to catalyze histone H3k27 and H3K9 methylation, and represses transcription at the chromatin level. Another role of EZH2 is direct interaction with DNA methyltransferases (DNMTs) to catalyze the methylation of promoter region in downstream genes. In addition, several reports have shown that EZH2 may function in target gene activation as a multifaceted molecule. 3.

The role of EZH2 in glioma

EZH2 functions as a chromatin-modifying enzyme, which plays a critical role in epigenetic silencing of genes (especially TSGs) through complex mechanisms (Figure 2). Increasing evidences have appeared that the links between EZH2 and tumorigenesis have been found in diverse type of cancers: breast, prostate, bladder, colon, lung, pancreatic, sarcoma and lymphomas [34-41]. The common phenomenon is that the levels of EZH2 expression are abnormally upregulated in cancer tissues compared with normal tissues, and overexpression of EZH2 is related to advanced stages of disease and a poor clinical outcome. Recently, a number of evidences suggest that EZH2 plays a central role in gliomas. EZH2 was more expressed in GBM than in low-grade gliomas and highly expressed in U87 human glioma cells [42]. The knockdown of EZH2 expression by using RNA interference in U87 human glioma cells resulted in apoptosis and a cell cycle arrest in the G0/G1phase. Moreover, silencing of EZH2 altered the mitochondrial membrane potential and promoted the release of cytochrome c from the mitochondria. In addition, the reduced expression of EZH2 altered the Bax

Expert Opin. Ther. Targets (2014) 18(10)

Epigenetic modification in gliomas

EID EZH2

Domain I SANT

Domain II ncRBC

EED

CXC CXC

SANT

RNA

SET SET

SUZ12

Figure 1. Characterized domains with potential functions are displayed for EZH2. Both the C-terminal SET domain and the adjacent cysteine-rich CXC domain are required for histone methyltransferase activity. Robust methyltransferase requires EZH2 assembly with both EED and SUZ12 of domain II, and domains required for binding these noncatalytic subunits are indicated. Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

EED: Embryonic ectoderm development; ncRBD: Noncoding RNA-binding domain; SANT: SWI3, ADA2, N-CoR.

GSC self-renewal

c-myc

STAT3

Apoptosis

Caspase9 Caspase3

Bax Bcl-2

EZH2

H3K27m

Puripotency

AXL

Proliferation

Invasion

Figure 2. EZH2 regulates the biology of gliomas by controlling of different pathways. Knockdown of EZH2 alters expression of apoptosis associated-genes and reduces transcription of AXL receptor kinase in gliomas. In addition, EZH2 regulates the biology of GSC by control of global levels of H3K27 trimethylation. AXL: AXL receptor kinase; GSC: Glioblastoma stem cell.

and Bcl-2 protein levels and led to the activation of caspase 9 and caspase 3 [43]. Smits et al. reported that inhibition of EZH2 in vivo by systemic 3-deazaneplanocin A (DZNep) administration in a U87-Fluc-mCherry GBM xenograft mouse imaging model resulted in reduced tumor growth [44]. Glioma progression is associated with EZH2 overexpression and PRC2 target gene silencing; EZH2 inhibits differentiation and activates cancer-, cell cycle- and cellular movement-related genes. In addition, EZH2 is both a prognostic factor and a promising therapy target in brain tumors [45]. Ott et al. demonstrated that EZH2 is expressed in human glioma and correlates with malignancy, whereas silencing of EZH2 reduced the proliferation and invasion of glioma cells. Microarray analyses of U87MG glioma cells after EZH2 knockdown demonstrated a strong transcriptional reduction of the AXL receptor kinase (AXL). Furthermore, histone modification appeared to be involved in the positive regulation of AXL by EZH2. Silencing AXL

mimicked the anti-invasive effects of EZH2 knockdown. Finally, AXL expression is found in human gliomas with high EZH2 expression [46]. Recently, both Bmi1 and EZH2 expressions in glioma tissues were significantly higher than those in corresponding nonneoplastic brain tissues. Additionally, the upregulations of Bmi1 and EZH2 proteins were both significantly related to advanced WHO grades and low Karnofsky. Moreover, the overall survival of patients with high Bmi1 protein expression or high EZH2 protein expression was obviously lower than those with low expressions [47]. Abdouh et al. reported that Bmi1 is expressed in human GBM tumors and highly enriched in CD133-positive cells. Stable Bmi1 knockdown using short hairpin RNA (shRNA)-expressing lentiviruses inhibited clonogenic potential in vitro and tumor formation in vivo. Bmi1 prevents CD133-positive cell apoptosis and/or differentiation into neurons and astrocytes, depending on the cellular

Expert Opin. Ther. Targets (2014) 18(10)

1199

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

E-B Bian et al.

context. Bmi1 represses alternate tumor suppressor pathways that attempt to compensate for INK4A/ARF/P53 deletion and PI(3)K/AKT hyperactivity [48]. More interestingly, glioma patients with combined overexpression of Bmi1 and EZH2 proteins had the shortest overall survival. Furthermore, expression of Bmi1 and EZH2 was independent prognostic factor for overall survival in glioma patients [47]. Natsume et al. reported that the biological interconversion between GSCs and differentiated non-GSCs is functionally plastic and accompanied by gain or loss of PRC2-mediated histone H3 K27 trimethylation on pluripotency [18]. H3K27M inhibits the enzymatic activity of the PRC2 through interaction with the EZH2 subunit. In addition, transgenes containing lysine-tomethionine substitutions at other known methylated lysines (H3K9 and H3K36) are sufficient to cause specific reduction in methylation through inhibition of SET-domain enzymes. Recent studies have identified a Lys 27-to-methionine (K27M) mutation at one allele of H3F3A, one of the two genes encoding histone H3 variant H3.3, in 60% of high-grade pediatric glioma cases. The levels of H3K27 di- and trimethylation (H3K27me2 and H3K27me3) are reduced globally in H3.3K27M patient samples due to the expression of the H3.3K27M mutant allele. Remarkably, H3K27me3 and EZH2 (the catalytic subunit of H3K27 methyltransferase) at chromatin are dramatically increased locally at hundreds of gene loci in H3.3K27M patient cells. Moreover, the gain of H3K27me3 and EZH2 at gene promoters alters the expression of genes. In addition, the development associated genes (e.g., Nanog, Wnt1, BMP5) together with alterations in the subcellular localization of EZH2 [18,49]. A recent study showed that EZH2 is highly expressed in murine and human GSC. Treatment with suberoylanilide hydroxamic acid caused significant upregulation of PRC2 predicted target genes, GSC disruption and decreased expression of EZH2 and of the stem cell marker CD133. Inhibition of EZH2 expression by shRNA was related to a significant decrease in the proliferation of glioma cells. Inhibition of EZH2, the main component of the PRC2, also impaired GBM tumor growth [42,48]. Targeted pharmacologic disruption of EZH2 by the S-adenosylhomocysteine hydrolase inhibitor DZNep, or its specific downregulation by shRNA, strongly impairs GBM cancer stem cell (CSC) self-renewal in vitro and tumor-initiating capacity in vivo. In DZNep-treated GBM CSCs, the expression of c-myc, known as a direct target of EZH2 in GBM CSCs, strongly repressed upon EZH2 depletion [50]. EZH2 binds to and methylates STAT3, resulting in enhanced STAT3 activity by increased tyrosine phosphorylation of STAT3. EZH2 knockdown greatly decreased global levels of H3K27 trimethylation in GSCs and reduced the levels of p-STAT3 in GSCs. P-STAT3 in GSCs was also rapidly decreased by treatment with either DZNep or GSK126 [51]. These results suggest that the inhibition of EZH2 reverses the silencing of Polycomb target genes and diminishes STAT3 activity. 1200

4.

Overview of DNA methylation

DNA methylation is an enzyme-mediated chemical modification of DNA by the addition of a methyl group from S-adenosyl-L-methionine substrates to the 5-position of cytosine (5-methylcytosine, 5mC) [52]. In humans and other mammals, DNA methylation is catalyzed by two general classes of DNMTs: maintenance DNMT1 and de novo DNMT3A and DNMT3B [53,54]. DNMT1, which maintains the existing methylation patterns following DNA replication, and DNMT3A and DNMT3B, de novo enzymes that prefer unmethylated and hemimethylated DNA and establish new DNA methylation patterns early in development [55,56]. DNA methylation can regulate gene expression directly, by repressing the binding of key transcription factors, and indirectly, by recruiting methyl-CpG-binding domain (MBD) proteins to the promoter of gene. So far, six members of the methyl-CpGbinding proteins family have been identified: MECP2, MBD1, MBD2, MBD3, MBD4 and Kaiso [57]. MECP2 recognizes methylated CpG sites and, when cooperation with DNMT1, forms a complex to copy the parental DNA methylation to the daughter DNA strands during cell division [58]. In addition, DNMTs interact with polycomb group (PcG) protein EZH2 to methylate EZH2-binding promoters, suggesting that epigenetic transcriptional repression complex is closely connected in regulating the expression of gene [58]. DNA demethylation is regulated by the ten--eleven translocation (TET) family. To date, three members of the TETs family, including TET1 TET2 TET3, have been identified in mammals. TETs convert 5mC to 5-hydroxymethylcytosine (5hmC), provide a way for dysregulation of DNA methylation [59]. TETs can promote to transcriptional repression, and to a minor extent also transcriptional activation, and that the majority of TETs-mediated transcriptional effects are independent of conversion of 5mC to 5hmC [60,61]. TET1 and TET3, but not TET2, is frequently found in chromatinbinding proteins such as DNMT1 and 5mC-binding proteins (MBDs), which is involved in regulating the expression of genes [60]. TET1 contributes to silencing of a subgroup of genes by facilitating recruitment of PRC2 to CpG-rich target gene promoters [62,63]. Another mechanism by which transcriptional repression of target genes are mediated is by the interaction of TET1 with the Sin3A co-repressor complex [62]. TETs regulate gene expression by preventing the gene promoter from hypermethylation mediated by DNMTs [64].

The interaction of DNA methylation with EZH2 in gliomas

5.

DNA hypermethylation is frequently recruited by PcGmediated repressive mark to a number of tumor suppressor genes that are epigenetic silencing in cancer cells, suggesting that links EZH2 with DNMTs has established a role for this protein during the induction and targeting of DNA

Expert Opin. Ther. Targets (2014) 18(10)

Epigenetic modification in gliomas

A.

Transcrptional repression CpG H3K27m Me EZH2

DNMTs

B.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

EZH2

5hmc

5mc TETs

C.

Transcrptional activation

EZH2

MeCP2 Transcrptional repression

Methylated CpGs

Figure 3. miRNAs mediate the signal pathways of gliomas by targeting EZH2. EZH2 interacts with miRNAs and other upstream and downstream molecules. EZH2 is under the negative control of several tumor-suppressive miRNAs and inhibits cell cycle regulators and molecules involved in gliomas. 5hmC: 5-hydroxymethylcytosine; 5mC: 5-methylcytosine; DNMTs: DNA methyltransferases; miRNA: MicroRNAs; TET: Ten--eleven translocation.

methylation [65-67]. The expression levels of DNMT1 and DNMT3B were strongly upregulated in malignant glioma tissue compared with normal brain samples, whereas DNMT1 and DNMT3B knockdown resulted in the restored expression of 308 genes that also contained promoter region hypermethylation [68]. EZH2 interacts with DNMTs and associates with DNMT activity. Binding of DNMTs to several EZH2-repressed genes depends on the presence of EZH2. Furthermore, EZH2 is required for DNA methylation of EZH2-target promoters, suggesting that EZH2 serves as a recruitment platform for DNMTs, thus highlighting a previously unrecognized direct connection between two key epigenetic repression systems [66]. Both MeCP2 and EZH2 have recently been shown to be recruited and appear to be involved in the maintenance of PPARg repression in mouse stellate cells undergoing liver fibrogenesis [69]. Consistently, MeCP2 and EZH2 levels inversely correlate with PPARg expression in the colorectal cancer cells. MeCP2 and EZH2 silencing re-activate PPARg, confirming their crucial role in PPARg epigenetic repression [70]. Cartron et al. reported that U251 cells presenting a low level of EZH2 induced by the knockdown of EZH2. EZH2 downregulation affected the TET1/EZH2 interactions. The downexpression of EZH2 decreased TET1 recruitment on the HOXD12 genes. The decrease of TET1/EZH2 recruitment on the HOXD12 gene in cells treated with siRNA-EZH2 produced an increase in methylation of the HOX12D gene. These results

suggest that EZH2 plays a role as anchors for TET1 recruitment on the HOXD12 genes [71]. These findings suggest that the interaction of EHZ2, known as an epigenetic driver, with several specific genes associated with regulation of DNA methylation mediated transcriptional repression of genes (Figure 3).

The regulation of EZH2 by miRNAs in glioma

6.

miRNAs are now recognized as single-stranded noncoding RNA molecules, about 19 -- 25 nucleotides in length [72]. miRNA regulates gene expression by the sequence-selective targeting of mRNAs, promoting degradation and inhibiting translation of cellular mRNAs, depending on the degree of complementarity with target mRNA sequences [72-76]. As much knowledge about the biological characteristics of the miRNA has been accumulated, miRNA has been found to be closely associated with cellular events, including cell proliferation, apoptosis, differentiation, development and tumorigenesis [77-79]. Increasing evidences suggest that miRNAs are pivotal players in glioma biology exhibiting oncogenic or tumorsuppressive functions. The gliomas-specific expression pattern of miRNAs highlights their potential use as diagnostic biomarkers and target for the treatment of gliomas. The interactions of the epigenetic machinery modulate important cellular pathways leading to oncogenesis.

Expert Opin. Ther. Targets (2014) 18(10)

1201

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

E-B Bian et al.

Downregulation of miR-101 was observed in primary glioma samples of different WHO grades, whereas upregulation of miR-101 by pre-miR-101 resulted in a significant decrease in the migration of U87 glioma cells. Pre-miR-101 notably repressed EZH2 protein expression and reduced the levels of trimethylation of H3K27, suggesting inhibition of EZH2 function. Inhibition of EZH2 by pre-miR-101 attenuated GBM cell growth, migration/invasion and GBM-induced endothelial tubule formation [44]. It is known that EZH2 and EED, known as the member of PRC2 complexes, are direct targets of miR-101. MiR-101 downregulated the expression of cytoplasmic polyadenylation element-binding protein 1 (CPEB1) through reversing the methylation status of the CPEB1 promoter by regulating the presence on the promoter of the methylation-related histones H3K4me2, H3K27me3, H3K9me3 and H4K20me3. CPEB1 is hypomethylated and overexpressed in glioma cells and tissues. The knockdown of CPEB1 reduced cell senescence by regulating the expression or distribution of p53 in glioma cells. The epigenetic regulation of H3K27me3 on CPEB1 promoter is mediated by EZH2 and EED. CPEB1 is also regulated directly by miR-101 [80]. Guo et al. reported that the expression of miR-708 was downregulated in GBM cells. The overexpression of miR-708 inhibited invasion in the human GBM cell lines. Furthermore, EZH2 was reduced when miR-708 was overexpressed in GBM cells [81]. EZH2 plays an important role in promoting invasive of several types of tumor cells [82,83]. These findings suggest that the regulation of EZH2 provides support for miR-708 as a tumor suppressor that targets cell invasion. Qiu et al. have demonstrated that miR-138 is reduced in both GBM clinical specimens and cell lines, and is effective to inhibit EZH2 expression. Moreover, high levels of miR-138 are associated with long overall and progression-free survival of GBM patients. Ectopic expression of miR-138 effectively inhibits GBM cell proliferation in vitro and tumorigenicity in vivo through inducing cell cycles G1/S arrest. In addition miR-138 acquires tumor inhibition through directly targeting EZH2, CDK6, E2F2 and E2F3. Furthermore, an EZH2-mediated signal loop, EZH2CDK4/6-pRb-E2F1, is probably involved in GBM tumorigenicity, and this loop can be blocked by miR-138 [84]. These findings suggest that the 3¢-untranslated region (3¢-UTR) of EZH2 targeted directly by miRNAs is involved in regulating the growth, migration and invasion of glioma cells. Lee et al. reported that transfection of let-7 reduced expression of pan-RAS, N-RAS and K-RAS in the GBM cells. Let-7 also reduced the proliferation and migration of the cells, and reduced the sizes of the tumors produced after transplantation into nude mice [85]. Overexpression of let-7 by transfection of let-7 precursors decreased EZH2 expression and repressed clonogenic ability and sphere-forming capacity of prostate cancer cells, which was consistent with inhibition of EZH2 3¢-UTR luciferase activity [86]. The expression of miR-98 in glioma tissues was significantly lower than that in normal brain tissues. Moreover, forced expression of miR-98 accelerated the 1202

inhibition of glioma cell invasion [87]. In Eca109 cells, overexpression of miR-98 significantly inhibited the migration and invasion of esophageal squamous cell carcinoma cells, which was reversed by transfection of EZH2. MiR-98 was validated as bona fide regulators of EZH2 expression. In particular, miR-98 was underexpressed in relapsed patient samples, strongly suggesting an important role for the miR-98 and EZH2 axis in nasopharyngeal carcinoma (NPC) biology [88]. MiR-124 is dramatically downregulated in human GBM tumor cell lines and clinical specimen of glioma, and is negatively correlated with the tumor pathological grading [89,90]. MiR-124 could directly target the 3¢-UTR of EZH2 mRNA, and suppress mRNA and protein expressions of EZH2 in hepatocellular carcinoma [91]. MiR-137 expression is significantly downregulated in a cohort of 35 oligodendroglial tumors and 9 glioma cell lines compared with normal brains. Lower miR-137 expression is associated with shorter progressive-free and overall survival. Restoration of miR-137 expression significantly suppressed cell growth, anchorage-independent growth, as well as invasion of GBM cells [92]. The decreased invasion caused by miR-137 overexpression could be phenocopied by small-interfering RNA knockdown of EZH2 [93]. The patients displayed a significantly decreased expression of miR-137 levels, as well as significantly increased expression of the validated downstream target EZH2 [94]. Forced expression of miR-26a in glioma cells significantly increased both growth rate and colony formation in vitro and tumor growth and angiogenesis in vivo, while reduced expression of miR-26a played opposite roles [95]. MiR-26a strongly reduced the expression of EZH2 oncogene in NPC cells. In addition, miR-26a suppressed the expression of c-myc, the cyclin D3 and E2, and the cyclindependent kinase CDK4 and CDK6 while enhancing the expression of CDK inhibitors p14(ARF) and p21(CIP1) in an EZH2-dependent manner [96]. These results suggest that miRNAs may be involved in controlling the biology of gliomas by targeting EZH2 (Figure 4). 7.

Expert opinion

It is speculated that epigenetic control of EZH2 provides a potential target for the treatment of gliomas. In this review, we summarize our current view of the role EZH2 in the epigenetic modifications and that EZH2 is involved in cellular homeostasis affecting cell proliferation, cell growth, invasion and apoptosis, while their disruption results in plasticity of GSCs. Gene repression mediated by EZH2 acts in a highly interconnected and coordinated manner, epigenetically regulating a diverse cohort of genes involved in gliomas. Together, understanding the regulatory mechanisms of EZH2 and the function of EZH2 target genes will shed light on the development of novel glioma treatments targeting EZH2-mediated gliomas to suppress oncogenic and stemness-associated pathways, which will pave the way to prevention and remission of gliomas. Several miRNAs bind directly to the 3¢-UTR of EZH2 through which they exert their function in gliomas.

Expert Opin. Ther. Targets (2014) 18(10)

Epigenetic modification in gliomas

miR-708 miR-101

C-myc CyclinD3 E2 CDK4 CDK6

miR-138

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

miR-26a

Let-7 miR-98 miR-124 miR-137

EZH2 p14 p21

CDK4/6-PRb-E2F1 CPEB

P53

Figure 4. The interaction of EZH2 with several specific genes mediated by DNA methylation regulates transcriptional repression or activation in gliomas. A. EZH2 mediates H3K27 methylation and recruits DNMT1, resulting in further modifications such as maintenance of DNA methylation that is responsible for stable repression of specific genes. B. The oxidation of 5mC to 5hmC by ten--eleven translocation (TET) proteins may therefore ease the repressive effects of 5mC. In addition, TETs binding may prevent access to EZH2, further contributing to the maintenance of an unmethylated promoter. C. DNA methylation at promoter regions is normally associated with silenced genes, where binding of MeCP2, EZH2 promotes to 5mC, mediating gene repression. 5mC: 5-methylcytosine; CPEB: Cytoplasmic polyadenylation element-binding protein; DNMT1: DNA methyltransferase-1.

MiRNAs are capable of regulating EZH2 expression at both transcriptional and posttranscriptional levels. Through the interaction with EZH2 they affect H3K27 methylation and in consequence they are involved in the biology of gliomas. In addition, EZH2 interacted with DNA methylation related to genes such as DNMTs, TETs, MeCP2, mediates transcriptional repression of glioma-mediated genes. EZH2 has a mediator role in transcriptional repression of genes, and if an EZH2 role exists and is experimentally verified, epigenetic modifications will also affect gliomas biology by forming important epigenetic regulatory network in which EZH2 plays the role of the pathway-stabilizing factor. Although there has been progress in generating EZH2 inhibitors for treatment of cancer, a more deeply understanding of the multifaceted biological roles of EZH2 is yet to be clarified, in order to promote the development of pharmacological agents. Inhibitors of EZH2, including GSK-A, GSK126, GSK-343 and DZNep, represent the beginning of a promising era that on-target epigenetic drugs can be applied to clinics and be effectively treated in EZH2-high tumors. In addition, Hypericin is a potent inhibitor of glioma growth by inducing unique epigenetic downregulations of EZH2, remodeling chromatin structure. UNC1999, the first orally bioavailable inhibitor that was highly selective for EZH2, may be more efficient and effective for treatment of gliomas. Although increasing evidences support the epigenetic biomarkers and therapeutic potential of EZH2 for gliomas,

there are largely unknown about its regulatory mechanism. For example, it is needed to clarify why the expression of EZH2 is upregulated in gliomas, and what the underlying mechanisms are? What is the relationship between the DNA methylation and miRNA about EZH2? Is histone modification such as histone acetylation involved in regulation of the expression of EZH2 in gliomas? It is also not clear whether targeted EZH2 protein has a beneficial effect for treatment of gliomas. More experiments are needed to unravel how DNA methylation together with other repression mechanisms such as miRNA or histone acetylation, generates distinct patterns of EZH2 silencing in gliomas. Better understanding of the epigenetics involved in gliomas by using in vitro in cells, in vivo in rodents and gliomas specimen with advanced techniques, such as microarray and next-generation sequencing, may provide a big step toward epigenetic drug translating into clinical applications treatment of gliomas.

Declaration of interest The authors were supported by the National Science Foundation of China (No.81072066). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Expert Opin. Ther. Targets (2014) 18(10)

1203

E-B Bian et al.

Bibliography

25.

13.

Weller M, Stupp R, Hegi M, et al. Individualized targeted therapy for glioblastoma: fact or fiction? Cancer J 2012;18(1):40-4

Lachner M, O’Carroll D, Rea S, et al. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature 2001;410(6824):116-20

1.

Hamza MA, Gilbert M. Targeted therapy in gliomas. Curr Oncol Rep 2014;16(4):379

2.

Louis DN. Molecular pathology of malignant gliomas. Annu Rev Pathol 2006;1:97-117

26.

14.

Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 2007;114(2):97-109

Hadjipanayis CG, Van Meir EG. Tumor initiating cells in malignant gliomas: biology and implications for therapy. J Mol Med (Berl) 2009;87(4):363-74

Sauvageau M, Sauvageau G. Polycomb group proteins: multi-faceted regulators of somatic stem cells and cancer. Cell Stem Cell 2010;7(3):299-313

27.

15.

Kim YH, Nobusawa S, Mittelbronn M, et al. Molecular classification of lowgrade diffuse gliomas. Am J Pathol 2010;177(6):2708-14

Cheng L, Bao S, Rich JN. Potential therapeutic implications of cancer stem cells in glioblastoma. Biochem Pharmacol 2010;80(5):654-65

Wang H, Wang L, Erdjument-Bromage H, et al. Role of histone H2A ubiquitination in Polycomb silencing. Nature 2004;431(7010):873-8

28.

16.

Vermeulen L, de Sousa e Melo F, Richel DJ, et al. The developing cancer stem-cell model: clinical challenges and opportunities. Lancet Oncol 2012;13(2):e83-9

Chase A, Cross NC. Aberrations of EZH2 in cancer. Clin Cancer Res 2011;17(9):2613-18

29.

McCabe MT, Graves AP, Ganji G, et al. Mutation of A677 in histone methyltransferase EZH2 in human B-cell lymphoma promotes hypertrimethylation of histone H3 on lysine 27 (H3K27). Proc Natl Acad Sci USA 2012;109(8):2989-94

3.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

Drosophila Polycomb group repressor complex. Cell 2002;111(2):197-208

astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 2005;366(9490):985-90

Papers of special note have been highlighted as either of interest () or of considerable interest () to readers.

4.

5.

6.

7.

8.

9.

10.

11.

12.

1204

Gupta K, Salunke P. Molecular markers of glioma: an update on recent progress and perspectives. J Cancer Res Clin Oncol 2012;138(12):1971-81 Gupta M, Djalilvand A, Brat DJ. Clarifying the diffuse gliomas: an update on the morphologic features and markers that discriminate oligodendroglioma from astrocytoma. Am J Clin Pathol 2005;124(5):755-68 Perry A. Pathology of low-grade gliomas: an update of emerging concepts. Neuro Oncol 2003;5(3):168-78 Benjamin R, Capparella J, Brown A. Classification of glioblastoma multiforme in adults by molecular genetics. Cancer J 2003;9(2):82-90 Dolecek TA, Propp JM, Stroup NE, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005 -- 2009. Neuro Oncol 2012;14(Suppl 5):v1-49 De Witt Hamer PC, Robles SG, Zwinderman AH, et al. Impact of intraoperative stimulation brain mapping on glioma surgery outcome: a meta-analysis. J Clin Oncol 2012;30(20):2559-65

17.

Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 2008;8(10):755-68

18.

Natsume A, Ito M, Katsushima K, et al. Chromatin regulator PRC2 is a key regulator of epigenetic plasticity in glioblastoma. Cancer Res 2013;73(14):4559-70

30.

Morin RD, Johnson NA, Severson TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet 2010;42(2):181-5

19.

Vescovi AL, Galli R, Reynolds BA. Brain tumour stem cells. Nat Rev Cancer 2006;6(6):425-36

31.

20.

Cardoso C, Mignon C, Hetet G, et al. The human EZH2 gene: genomic organisation and revised mapping in 7q35 within the critical region for malignant myeloid disorders. Eur J Hum Genet 2000;8(3):174-80

Pasqualucci L, Trifonov V, Fabbri G, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat Genet 2011;43(9):830-7

32.

Wigle TJ, Knutson SK, Jin L, et al. The Y641C mutation of EZH2 alters substrate specificity for histone H3 lysine 27 methylation states. FEBS Lett 2011;585(19):3011-14

33.

Yap DB, Chu J, Berg T, et al. Somatic mutations at EZH2 Y641 act dominantly through a mechanism of selectively altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood 2011;117(8):2451-9

34.

Arisan S, Buyuktuncer ED, Palavan-Unsal N, et al. Increased expression of EZH2, a polycomb group protein, in bladder carcinoma. Urol Int 2005;75(3):252-7

35.

Kleer CG, Cao Q, Varambally S, et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA 2003;100(20):11606-11

21.

Margueron R, Reinberg D. The Polycomb complex PRC2 and its mark in life. Nature 2011;469(7330):343-9

22.

Cao R, Wang L, Wang H, et al. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science 2002;298(5595):1039-43

23.

Kuzmichev A, Nishioka K, Erdjument-Bromage H, et al. Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein. Genes Dev 2002;16(22):2893-905

Smith JS, Chang EF, Lamborn KR, et al. Role of extent of resection in the longterm outcome of low-grade hemispheric gliomas. J Clin Oncol 2008;26(8):1338-45 van den Bent MJ, Afra D, de Witte O, et al. Long-term efficacy of early versus delayed radiotherapy for low-grade

24.

Muller J, Hart CM, Francis N J, et al. Histone methyltransferase activity of a

Expert Opin. Ther. Targets (2014) 18(10)

Epigenetic modification in gliomas

36.

Matsukawa Y, Semba S, Kato H, et al. Expression of the enhancer of zeste homolog 2 is correlated with poor prognosis in human gastric cancer. Cancer Sci 2006;97(6):484-91

46.

Ott M, Litzenburger UM, Sahm F, et al. Promotion of glioblastoma cell motility by enhancer of zeste homolog 2 (EZH2) is mediated by AXL receptor kinase. PLoS One 2012;7(10):e47663

58.

Kimura H, Shiota K. Methyl-CpG-binding protein, MeCP2, is a target molecule for maintenance DNA methyltransferase, Dnmt1. J Biol Chem 2003;278(7):4806-12

37.

Raman JD, Mongan NP, Tickoo SK, et al. Increased expression of the polycomb group gene, EZH2, in transitional cell carcinoma of the bladder. Clin Cancer Res 2005;11(24 Pt 1): 8570-6

47.

Wu Z, Wang Q, Wang L, et al. Combined aberrant expression of Bmi1 and EZH2 is predictive of poor prognosis in glioma patients. J Neurol Sci 2013;335(1-2):191-6 This paper describes that expression of EZH2 was independent prognostic factor for overall survival in glioma patients.

59.

Cimmino L, Abdel-Wahab O, Levine RL, et al. TET family proteins and their role in stem cell differentiation and transformation. Cell Stem Cell 2011;9(3):193-204

60.

Abdouh M, Facchino S, Chatoo W, et al. BMI1 sustains human glioblastoma multiforme stem cell renewal. J Neurosci 2009;29(28):8884-96

Frauer C, Rottach A, Meilinger D, et al. Different binding properties and function of CXXC zinc finger domains in Dnmt1 and Tet1. PLoS One 2011;6(2):e16627

61.

Chan KM, Fang D, Gan H, et al. The histone H3.3K27M mutation in pediatric glioma reprograms H3K27 methylation and gene expression. Genes Dev 2013;27(9):985-90

Iyer L M, Tahiliani M, Rao A, et al. Prediction of novel families of enzymes involved in oxidative and other complex modifications of bases in nucleic acids. Cell Cycle 2009;8(11):1698-710

62.

Suva ML, Riggi N, Janiszewska M, et al. EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res 2009;69(24):9211-18

Williams K, Christensen J, Pedersen MT, et al. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 2011;473(7347):343-8

63.

Wu H, D’Alessio AC, Ito S, et al. Dual functions of Tet1 in transcriptional regulation in mouse embryonic stem cells. Nature 2011;473(7347):389-93

64.

Freudenberg JM, Ghosh S, Lackford BL, et al. Acute depletion of Tet1-dependent 5-hydroxymethylcytosine levels impairs LIF/Stat3 signaling and results in loss of embryonic stem cell identity. Nucleic Acids Res 2012;40(8):3364-77

65.

Rauch T, Wang Z, Zhang X, et al. Homeobox gene methylation in lung cancer studied by genome-wide analysis with a microarray-based methylated CpG island recovery assay. Proc Natl Acad Sci USA 2007;104(13):5527-32

66.

Vire E, Brenner C, Deplus R, et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature 2006;439(7078):871-4 Binding of DNA methyltransferases (DNMTs) to several EZH2-repressed genes depends on the presence of EZH2, suggesting that EZH2 serves as a recruitment platform for DNMTs epigenetic repression of gene expression.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

38.

39.

40.

41.

42.

43.

44.

..

45.

Sudo T, Utsunomiya T, Mimori K, et al. Clinicopathological significance of EZH2 mRNA expression in patients with hepatocellular carcinoma. Br J Cancer 2005;92(9):1754-8 Varambally S, Dhanasekaran SM, Zhou M, et al. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature 2002;419(6907):624-9 Watanabe H, Soejima K, Yasuda H, et al. Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer Cell Int 2008;8:15

.

48.

49.

50.

Weikert S, Christoph F, Kollermann J, et al. Expression levels of the EZH2 polycomb transcriptional repressor correlate with aggressiveness and invasive potential of bladder carcinomas. Int J Mol Med 2005;16(2):349-53

51.

Orzan F, Pellegatta S, Poliani PL, et al. Enhancer of Zeste 2 (EZH2) is upregulated in malignant gliomas and in glioma stem-like cells. Neuropathol Appl Neurobiol 2011;37(4):381-94

52.

Zhang R, Wang R, Chang H, et al. Downregulation of Ezh2 expression by RNA interference induces cell cycle arrest in the G0/G1 phase and apoptosis in U87 human glioma cells. Oncol Rep 2012;28(6):2278-84 Smits M, Nilsson J, Mir SE, et al. miR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis. Oncotarget 2010;1(8):710-20 U87-Fluc-mCherry glioblastoma xenograft mouse by systemic 3-deazaneplanocin A administration inhibited tumor growth. Crea F, Hurt EM, Farrar WL. Clinical significance of Polycomb gene expression in brain tumors. Mol Cancer 2010;9:265

Kim E, Kim M, Woo DH, et al. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell 2013;23(6):839-52 Weng YL, An R, Shin J, et al. DNA modifications and neurological disorders. Neurotherapeutics 2013;10(4):556-67

53.

Rottach A, Leonhardt H, Spada F. DNA methylation-mediated epigenetic control. J Cell Biochem 2009;108(1):43-51

54.

Zhao YE, Wu LP. RAPD-SCAR marker and genetic relationship analysis of three Demodex species (Acari: demodicidae). Parasitol Res 2012;110(6):2395-402

55.

Brenner C, Fuks F. DNA methyltransferases: facts, clues, mysteries. Curr Top Microbiol Immunol 2006;301:45-66

56.

Turek-Plewa J, Jagodzinski PP. The role of mammalian DNA methyltransferases in the regulation of gene expression. Cell Mol Biol Lett 2005;10(4):631-47

57.

Roloff TC, Ropers HH, Nuber UA. Comparative study of methyl-CpG-binding domain proteins. BMC Genomics 2003;4(1):1

Expert Opin. Ther. Targets (2014) 18(10)

..

67.

Widschwendter M, Fiegl H, Egle D, et al. Epigenetic stem cell signature in cancer. Nat Genet 2007;39(2):157-8

68.

Kreth S, Thon N, Eigenbrod S, et al. O-methylguanine-DNA methyltransferase (MGMT) mRNA expression predicts

1205

E-B Bian et al.

outcome in malignant glioma independent of MGMT promoter methylation. PLoS One 2011;6(2):e17156 69.

Expert Opin. Ther. Targets Downloaded from informahealthcare.com by Universitat de Girona on 10/24/14 For personal use only.

70.

71.

Mann J, Chu DC, Maxwell A, et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology 2010;138(2):705-14; 714 e1-4 Pancione M, Sabatino L, Fucci A, et al. Epigenetic silencing of peroxisome proliferator-activated receptor gamma is a biomarker for colorectal cancer progression and adverse patients’ outcome. PLoS One 2010;5(12):e14229

Filipowicz W, Jaskiewicz L, Kolb FA, et al. Post-transcriptional gene silencing by siRNAs and miRNAs. Curr Opin Struct Biol 2005;15(3):331-41

73.

He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 2004;5(7):522-31

75.

Guo P, Lan J, Ge J, et al. miR-708 acts as a tumor suppressor in human glioblastoma cells. Oncol Rep 2013;30(2):870-6

82.

Rao ZY, Cai MY, Yang GF, et al. EZH2 supports ovarian carcinoma cell invasion and/or metastasis via regulation of TGF-beta1 and is a predictor of outcome in ovarian carcinoma patients. Carcinogenesis 2010;31(9):1576-83

83.

84.

Kozomara A, Griffiths-Jones S. miRBase: integrating microRNA annotation and deep-sequencing data. Nucleic Acids Res 2011;39(Database issue):D152-7 Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogenesis, function and decay. Nat Rev Genet 2010;11(9):597-610

76.

Sontheimer EJ, Carthew RW. Silence from within: endogenous siRNAs and miRNAs. Cell 2005;122(1):9-12

77.

Hong L, Yang Z, Ma J, et al. Function of miRNA in controlling drug resistance of human cancers. Curr Drug Targets 2013;14(10):1118-27

78.

Pencheva N, Tavazoie SF. Control of metastatic progression by microRNA regulatory networks. Nat Cell Biol 2013;15(6):546-54

79.

Yates LA, Norbury CJ, Gilbert RJ. The long and short of microRNA. Cell 2013;153(3):516-19

80.

Xiaoping L, Zhibin Y, Wenjuan L, et al. CPEB1, a histone-modified hypomethylated gene, is regulated by

1206

81.

Cartron PF, Nadaradjane A, Lepape F, et al. Identification of TET1 partners that control Its DNA-demethylating function. Genes Cancer 2013;4(5-6):235-41

72.

74.

..

miR-101 and involved in cell senescence in glioma. Cell Death Dis 2013;4:e675 MiR-101 regulated the expression of cytoplasmic polyadenylation elementbinding protein 1 (CPEB1) through reversing the methylation status of the CPEB1 promoter by regulating the EZH2-related histones H3K27me3, suggesting that the interaction of microRNA with EZH2 regulation of gene expression.

85.

86.

Ren G, Baritaki S, Marathe H, et al. Polycomb protein EZH2 regulates tumor invasion via the transcriptional repression of the metastasis suppressor RKIP in breast and prostate cancer. Cancer Res 2012;72(12):3091-104 Qiu S, Huang D, Yin D, et al. Suppression of tumorigenicity by microRNA-138 through inhibition of EZH2-CDK4/6-pRb-E2F1 signal loop in glioblastoma multiforme. Biochim Biophys Acta 2013;1832(10):1697-707 Lee ST, Chu K, Oh H J, et al. Let-7 microRNA inhibits the proliferation of human glioblastoma cells. J Neurooncol 2011;102(1):19-24 Kong D, Heath E, Chen W, et al. Loss of let-7 up-regulates EZH2 in prostate cancer consistent with the acquisition of cancer stem cell signatures that are attenuated by BR-DIM. PLoS One 2012;7(3):e33729

87.

Chen Z, Cheng Q, Ma Z, et al. Overexpression of RKIP inhibits cell invasion in glioma cell lines through upregulation of miR-98. Biomed Res Int 2013;2013:695179

88.

Huang SD, Yuan Y, Zhuang CW, et al. MicroRNA-98 and microRNA-214 posttranscriptionally regulate enhancer of zeste homolog 2 and inhibit migration and invasion in human esophageal squamous cell carcinoma. Mol Cancer 2012;11:51

Expert Opin. Ther. Targets (2014) 18(10)

89.

An L, Liu Y, Wu A, et al. microRNA-124 inhibits migration and invasion by down-regulating ROCK1 in glioma. PLoS One 2013;8(7):e69478

90.

Lv Z, Yang L. MiR-124 inhibits the growth of glioblastoma through the downregulation of SOS1. Mol Med Rep 2013;8(2):345-9

91.

Zheng F, Liao YJ, Cai MY, et al. The putative tumour suppressor microRNA-124 modulates hepatocellular carcinoma cell aggressiveness by repressing ROCK2 and EZH2. Gut 2012;61(2):278-89

92.

Li KK, Yang L, Pang JC, et al. MIR-137 suppresses growth and invasion, is downregulated in oligodendroglial tumors and targets CSE1L. Brain Pathol 2013;23(4):426-39

93.

Luo C, Tetteh PW, Merz PR, et al. miR-137 inhibits the invasion of melanoma cells through downregulation of multiple oncogenic target genes. J Invest Dermatol 2013;133(3):768-75

94.

Willemsen MH, Valles A, Kirkels LA, et al. Chromosome 1p21.3 microdeletions comprising DPYD and MIR137 are associated with intellectual disability. J Med Genet 2011;48(12):810-18

95.

Qian X, Zhao P, Li W, et al. MicroRNA-26a promotes tumor growth and angiogenesis in glioma by directly targeting prohibitin. CNS Neurosci Ther 2013;19(10):804-12

96.

Lu J, He ML, Wang L, et al. MiR-26a inhibits cell growth and tumorigenesis of nasopharyngeal carcinoma through repression of EZH2. Cancer Res 2011;71(1):225-33

Affiliation Er-Bao Bian1,2, Jia Li1,2, Xiao-Jun He1,2, Gang Zong1,2, Tao Jiang1,2, Jun Li3 & Bing Zhao†1,2 † Author for correspondence 1 The Second Affiliated Hospital of Anhui Medical University, Department of Neurosurgery, Hefei 230601, China E-mail: [email protected] 2 Anhui Medical University, Cerebral Vascular Disease Research Center, Hefei 230601, China 3 Anhui Medical University, School of Pharmacy, Hefei 230032, China

Epigenetic modification in gliomas: role of the histone methyltransferase EZH2.

Gliomas are characterized by increased anaplasia, malignization, proliferation and invasion. They exhibit high resistance to standard treatment with c...
429KB Sizes 2 Downloads 5 Views