Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Sep 1.
Published in final edited form as: Epigenomics. 2011 Apr;3(2):193–205. doi: 10.2217/epi.11.9

Histone methylation in myelodysplastic syndromes

Yue Wei 1,, Irene Gañán-Gómez 1,2, Sophie Salazar-Dimicoli 1, Sara L McCay 1, Guillermo Garcia-Manero 1
PMCID: PMC3758889  NIHMSID: NIHMS456738  PMID: 22122281

Abstract

Histone methylation is a type of epigenetic modification that is critical for the regulation of gene expression. Numerous studies have demonstrated that abnormalities of this newly characterized epigenetic modification are involved in the development of multiple diseases, including cancer. There is also emerging evidence for a link between histone methylation and the pathogenesis of myeloid neoplasms, including myelodysplastic syndromes (MDS). This article provides an overview of recent progress in the studies of histone methylation in myeloid malignancies, with an emphasis on MDS. We cover each type of histone methylation modification and their regulatory mechanisms, as well as their abnormalities in MDS or potential connections to MDS. We also summarize the recent progress in the development of inhibitors targeting histone methylation and their applications as potential therapeutic agents.

Keywords: histone demethylase inhibitor, histone methylation, histone methyltransferase, myelodysplastic syndrome


Myelodysplastic syndromes (MDS) are a very complex group of myeloid neoplasms with diverse molecular backgrounds and natural histories [1]. With a median age of presentation being 70–75 years, the incidence of MDS in the USA and other western societies increases as the population ages [2]. Besides age, risk factors for MDS also include exposure to chemicals, cigarette smoking and prior exposure to chemotherapy or radiation therapy [3]. Indeed, cases of therapy-related MDS (tMDS) have an extremely poor prognosis and tend to be refractory to conventional therapeutic interventions. Overall, MDS is becoming one of the most common forms of myeloid neoplasms in older individuals [2].

Myelodysplastic syndromes have profound heterogeneity in genetic backgrounds, cytogenetic features, morphologic presentation and clinical course. Due to these complexities, the diagnosis and prognosis of MDS is extremely heterogeneous. At the present time, diagnosis of MDS is usually based on the 2008 WHO classification system [4]. Prognosis of MDS is calculated using a number of scoring systems, with the International Prognostic Scoring System (IPSS) as the one used most frequently [5]. This system divides MDS patients into four different subsets: low, intermediate-1, intermediate-2 and high risk.

Although there is an approximately 25% of incidence of transforming to AML [6], most patients with lower risk MDS (low or intermediate-1) die from causes intrinsic to MDS and not from transformation to acute myelogenous leukemia (AML) [7]. In addition, while karyotype is one of the most important criteria in different classification systems for MDS, with a large portion of MDS patients displaying clonal chromosome abnormalities, such as 5q-deletions and monosomy 7 [8]. In sharp contrast to AML, balanced chromosomal structural abnormalities, including translocations and inversions, are rarely detected in MDS. Overall, these observations suggest that there are unique pathophysiological mechanisms for MDS that are different from AML.

Besides cytogenetic changes, other known genetic lesions identified in MDS include copy number changes (genetic amplifications or deletions) and mutations that alter the sequence or expression of individual genes. These newly identified lesions in MDS have been summarized by Bejar et al. in a recent review, which include mutations within individual genes such as TP53, TET2, ASXL1, RUNX1, IDH1 and IDH2, as well as members involved in tyrosine kinase pathway [9]. In addition, it has been demonstrated that MDS is also characterized by frequent epigenetic abnormalities, resulting in aberrant gene expression [10].

Overview of epigenetic regulation and histone methylation in MDS

The term epigenetic modification refers to different forms of biochemical modification of DNA or histone proteins (core histone H2A, H2B, H3 and H4, as well as the linker histone H1) in chromatin that affect gene expression without altering the DNA sequence [11]. While the importance of epigenetic regulation has been demonstrated in different types of malignancies [10,1214], cancer is now viewed as the result of the acquisition of both genetic and epigenetic abnormalities that cause aberrant gene expression. More importantly, from a cancer therapeutic perspective, in contrast to genetic alterations, epigenetic abnormalities can be reversed, allowing for potential therapeutic interventions.

Among different types of epigenetic modifications, DNA methylation and histone acetylation have been well characterized in various neoplasms, including MDS [10]. Aberrant DNA CpG methylation is very common in MDS [15,16] and aberrant expression as well as genetic lesions of genes encoding DNA methylation regulators has also been reported in MDS. These include overexpression of DNA methyltransferase 1 (DNMT1) and DNMT3A [17], as well as mutations on the gene encoding TET2, an enzyme involved in cytosine demethylation [18,19]. Two DNA hypomethylating agents, 5-azacitidine and 5-aza-2′-deoxycitidine, are approved in the USA for patients with MDS and have shown activity in patients with higher disease risk [20]. For histone acetylation, among the various structurally different, naturally occurring as well as synthetic histone deacetylase inhibitors (HDACis), vorinostat (SAHA), LBH589, depsipeptide, MGCD-0103 and several others have shown clinical activity in trials involving MDS [2124].

In comparison to DNA methylation and histone acetylation, histone methylation has just recently been appreciated as an important epigenetic mechanism of gene regulation, especially after the discovery of the dynamic nature of histone modification and the identification of the family of histone demethylases [25]. Methylation on chromatin histone has been identified on lysine (K) and arginine (R) residues of histone H3 and H4. Documented methylated lysine residues include H3 lysine 4 (H3K4), H3K9, H3K27, H3K36, H3K79 and H4K20 and each lysine can be mono-, di- and tri-methylated (me1, me2 and me3), respectively [26,27]. Methylation of arginine of histone has been detected on H4 arginine 3 (H4R3), H3R2, H3R8, H3R17 and H3R26, which can be mono-, asymmetric-di- and symmetric-di-methylated [28]. Like other forms of epigenetic modification, histone methylation plays an important role in the regulation of gene transcription as well as the development of disease, by influencing the compaction of chromatin histone tails and the binding of proteins such as transcription factors that recognize histone marks [29,30]. In these methyl histone recognizing/interacting proteins, several metylated histone recognizing domains have been identified. Examples of these domains include the chromo domain in HP1 and CHD1 proteins in recognizing the methylated H3K4 [31]; the Tudor domain in p53BP1 and JMJD2A recognizing H3K9 or H3K4 methylation [32]; the MBT domain in CGI-72 recognizing H3K4 or H4K20 methylation [33]; and the PHD domain used by protein ING2 for gene repression [34]. In contrast to other epigenetic modifications, the effect of histone methylation on transcription (activating vs repressive) is not only dependent on the location of each substrate lysine or arginine, but also on the degree of methylation (mono-, di- or tri-methylation) on each substrate residue. This mechanism provides unique fine-tuning regulation of gene expression. Albeit the subtle differences of the effects on transcription associated within each subtype of methylation, generally, methylation of H3K4, H3K36 and possibly also H3K79, are positively associated with active transcription, whereas H3K9, H3K27 and H4K20 methylation tend to negatively regulate transcription [35]. Histone methylation has also been shown to be dynamically regulated. While the methylated status of histone lysine and arginine is maintained by histone methyltransferases (HMTs), the newly discovered family of histone demethylases (HDMs), particularly histone lysine demethylases (KDMs) [25,27,30,3637], catalyze the removal of methyl groups.

Inappropriate levels of histone methylation and lesions of histone methylation regulators have both been identified in different types of myeloid neoplasms [38,39]. Evidence has recently emerged that abnormalities of histone methylation may also contribute to the development of MDS [4042]. To demonstrate the progress in this rapid evolving field, we will discuss the known and potential associations of each type of histone methylation with MDS. We will summarize the aberrant levels of histone methylation on important MDS-related genes, as well as the genetic lesions and functional abnormalities of HMTs, HDMs and their cofactors or interacting proteins. We realize that evidence on the direct involvement of regulation of histone methylation in MDS is still very limited. Molecular research in MDS is hampered owing to the heterogeneity of the disease, the lack of confirmed cell lines or widely available animal models, as well as the limited number of malignant cells that are available. Therefore, in this article, some of the potential roles the discussion of certain types of histone methylation and their regulators in MDS are still hypothetical and sometimes are based on the results achieved from studies performed in other types of myeloid neoplasms, particularly AML.

Abnormalities of histone methylation associated with MDS

Activating histone lysine methylation marks

H3K4 methylation

Methylated H3K4 is mostly concentrated in the proximity of transcription starting sites (TSS) and are associated with activation of transcription [35]. Based on this, our group used chromatin immunoprecipitation (CHIP) combined with deep sequencing techniques to profile the H3K4 trimethylation signature in primary MDS bone marrow cells. With this approach, we have identified a list of aberrantly activated genes associated with differentially high levels of promoter H3K4 trimethylation in MDS [43]. Of interest, a majority of these genes thus identified are involved in NF-κB activation. Further studies need to be performed to dissect the role of these gene products during the pathogenesis of MDS and to characterize the potential interplay between the regulation of H3K4 trimethylation and NF-κB signaling.

In comparison to other types of myeloid neoplasms, including AML, lesions of H3K4 methylation regulators identified in MDS are relatively rare. At the present time, there is only indirect evidence suggesting that two H3K4 methylation regulators, the methyltransferase mixed lineage leukemia (MLL) and the demethylases LSD1, might be involved in MDS. MLL is a H3K4 histone methyltransferase belonging to the family of SET domain proteins [44,45]. In MDS, the translocation of MLL gene locus 11q23 is very rare, much less frequent than AML. However, MLL partial tandem duplications (MLL-PTDs) have been described in both primary MDS and tMDS [46,47]. Of interest, the incidence of MLL-PTD increases from de novo MDS to secondary AML (s-AML) [47], suggesting that MLL may be involved in the progression of MDS to AML. Functionally, MLL has been reported to bind to several Hox gene promoters and to play a crucial role in the positive regulation of Hox genes, including HoxA9 and Meis1, in the hematopoietic system [48]. While overexpression of Hox genes is commonly detected in the bone marrow cells of MDS [49], it might be a reasonable hypothesis that the positive regulation of certain Hox genes by MLL-regulated H3K4 methylation could be involved in the development of MDS. However, more clinical as well as experimental evidence is still needed to support this hypothesis.

LSD1 is the first histone demethylase characterized and also the only none JmjC-domain histone lysine demethylase identified so far [36]. LSD1 protein is located in a transcriptional repressive complex containing CoREST, HDAC1 and HDAC2, which has the activity to remove methyl groups from mono- and di-methyl-H3K4 [50]. Inhibition of CoREST and LSD1 perturbs differentiation of erythroid, megakaryocytic and granulocytic cells as well as primary erythroid progenitors [51,52]. This suggests that LSD1 and the H3K4 methylation regulated by this histone demethylase are important for hematopoiesis. While no genetic lesion or abnormal expression of LSD1 has been reported in MDS, several important transcription regulators that are potentially involved in the pathogenesis of MDS are known to be functionally related to LSD1 and LSD1 mediated H3K4 demethylase activity. For instance, GFI1 is a transcription repressor that has been reported to be expressed at a higher level in high-risk MDS than in low-risk patients [53]. The LSD1 related H3K4 demethylase activity has been demonstrated to be recruited by GFI1 and to mediate the gene repressing activity of GFI1 on its target genes in vivo. LSD1 depletion derepresses GFI targets in lineage-specific patterns, which is accompanied by enhanced H3K4 methylation at the respective promoter [51]. Another transcription regulator that is required for hematopoiesis and is functionally related to LSD1 is TAL1 [52]. TAL1 is upregulated in Shwachman–Diamond syndrome, an inherited bone marrow disorder characterized by cytopenia and a high propensity to develop MDS [54]. LSD1 and its demethylase activity negatively regulate TAL1-mediated transcription of P4.2 gene in undifferentiated, but not in differentiated, murine erythroleukemia (MEL) cells [52] and this dynamic interaction between TAL1 and LSD1 may determine the onset of erythroid differentiation programs [52]. LSD1 may also cooperate with ASXL1 in transcriptional repression [55]. ASXL1 has been found to be mutated in MDS and chronic myelomonocytic leukemia (CMML) [5658]. These connections between the LSD1 mediated H3K4 demethylation and the functions of several transcriptional factors, which are known to be involved in hematopoiesis and/or the development of MDS, suggest that LSD1 may also have a role in MDS pathogenesis. However, direct molecular and clinical evidence are still in need to support this hypothesis.

H3K36 methylation

In general, histone H3K36 methylation is associated with actively transcribed genes [35]. Although no specific gene that carries aberrant level of H3K36 has been documented in MDS, fusion of H3K36 methyltransferase encoding gene NSD1/NSD3 and NUP98 have been detected in AML, MDS and tMDS [59,60]. Functionally, fusion protein NUP98–NSD1 activates and maintains a high methylated status of H3K36 on HoxA9 gene during in vitro self-renewal of myeloid stem cells, [59], whereas inhibition of the H3K36-methyltransferase activity of NSD1 prevents the activation of HoxA9 and myeloid progenitor immortalization [61]. These results link deregulated H3K36 methylation to the transcriptional regulation of the HoxA locus and potentially to the development of myeloid neoplasms. Whether this chimeric gene products and H3K36 methylation are involved in the pathogenesis of MDS still needs to be investigated. The known histone demethylases that target H3K36 methylation are FBXL10 and FBXL11 [62]. Although no genetic lesion or aberrant expression of these two genes has been reported in MDS, FBXL10 has been found to interact with the leukemia associated with the oncogene BCL6 product in the BCL6-interacting corepressor transcriptional repressing complex [63]. Related to this, high-resolution comparative genomic hybridization combined with FISH analysis has detected the amplification of BCL6 gene in MDS [64]. Therefore, it would be interesting to examine the interaction between BCL6 and FBXL10 as well as the possible gene silencing mediated by this protein complex in MDS.

H3K79 methylation

Unlike other methylated lysine residues that are located at the N-terminal tail of histones, H3K79 is uniquely located within a loop in the globular domain of histone H3 and is exposed on the nucleosome surface. Although some studies have associated H3K79 methylation with gene activation [35,65], the effect of this histone modification on transcription is still not completely understood.

DOT1L is an H3K79 specific methyltransferase, which unlike most other histone methyltransferases, lacks the SET domain [66,67]. There are two potential associations between DOT1L mediated H3K79 methylation to the pathogenesis of MDS. First, DOTL1 has been found to be involved in the pathobiology of the majority of hematopoietic neoplasms that are characterized by oncogenic fusion proteins including MLL-AF9, MLL-AF10, MLL-ENL, CALM-AF10 and MLL-AF4 [68]. This interaction contributes to the activation of Hox family genes such as HoxA5, HoxA9 and Meis1, which in many cases, are associated with increase of H3K79 methylation on gene promoters [68,69]. Although these fusion genes are very rarely detected in MDS, one of them, MLL-AF4, has been reported in MDS [70]. The second potential association between DOT1L and MDS is based on a mouse model lacking functional DOT1L protein [71]. These mice displayed defects in early erythropoiesis, accompanied by affected expression of GATA2 and PU.1 genes and altered H3K79 methylation on their promoters [71]. GATA2 and PU.1 are important transcription factors for the differentiation of hematopoietic progenitors of erythroid and myeloid lineages [72], the two hematopoietic lineages frequently affected in MDS. Of importance, GATA2 has been detected to be aberrantly expressed in MDS [73], whereas PU.1 is known to interact with EVI1 oncogene product [74]. It is also important to know that the MDS–EVI1 fusion gene encodes a PR (PRD1–BF1–RIZ1) domain containing protein which belongs to the SET domain protein superfamily, whereas the EVI1 protein lacking a PR domain is often overexpressed in myeloid neoplasms, including MDS [75].

Repressive histone lysine methylation marks

H3K27 methylation

Numerous studies have demonstrated that methylation on H3K27 correlates with gene repression [35]. In pluripotent embryonic stem cells, the repressive mark H3K27me3 can coexist with and usually dominates, the active mark of H3K4me3 [74]. This so called ‘bivalent domain’ promoter status holds a repressed but poised state for their targets, which are often key differentiation and development regulatory genes. Therefore this bivalent status is thought to enable embryonic stem cells to rapidly execute multiple differentiation programs, including the differentiation toward blood-forming precursors [7678]. In AML blast cells, the ‘bivalent’ promoter status has also been identified for p15INK4b tumor suppressor gene, which can be changed to a more repressed status with the loss of H3K4me3 and the exclusive H3K27me3 mark [79]. This change is correlated with DNA methylation [79]. In MDS, gene silencing and hypermethylation of the p15INK4b promoter are also both frequent [80,81]. Therefore, although MDS may not share most mechanisms of pathogenesis with AML, it is still possible that in MDS, similar to the silencing of p15INK4b in AML, the hypermethylation of p15INK4b correlates with a high H3K27me3 level on its promoter in MDS. Status of histone metylation, particularly the level of H3K27 methylation, on p15INK4b promoter in MDS patients should be examined. Another tumor suppressor gene that could be epigenetically repressed in MDS is CTNNA1. The CTNNA1 gene is located on chromosome 5q and is frequently repressed in MDS [82]. The correlation between DNA hypermethylation and enrichment of H3K27 methylation has been detected for CTNNA1 gene in AML cell lines [83]. In MDS, DNA methylation of CTNNA1 was only found in high-risk, but not low-risk patients, whereas high histone H3K27me3 level has not been detected for this gene in MDS [83,84]. Therefore, although DNA methylation and H3K27 methylation may both contribute to the silencing of key tumor suppressors such as p15INK4b and CTNNA1 in AML, whether a similar mechanism could be true for the same gene silencing in MDS still needs to be examined.

EZH2 encodes a H3K27 methyltransferase, which is the catalytic unit of the polycomb repressive complex 2 (PRC2) [85]. The EZH2 gene is located on 7q36.1, a common locus of chromosomal deletion correlated with a poor prognosis in MDS [86]. Missense mutations of EZH2 gene that introduce premature truncated forms of this protein have recently been discovered in MDS bone marrow cells [40]. Particularly, most of these mutations cause the alteration or truncation in the SET domain, the essential methyltransferase functional domain of EZH2 [40], suggesting the importance of the histone methylation regulatory activity of EZH2 in the pathogenesis of MDS. However, more recently, overexpression of EZH2 has also been reported in MDS, which is linked to poor prognosis [87]. At the same time, overexpression of EZH2 in breast cancer has been demonstrated to mediate the downregulation of DNA damage repair, which leads to the expansion of tumor initiating cells [88]. Therefore, the exact role of EZH2 in MDS needs to be further characterized.

There are two H3K27 demethylases potentially involved in the development of MDS, which are UTX and JMJD3 [89,90]. Somatic mutations of the UTX gene has been reported in CMML and MDS and is associated with increase level of H3K27me3 [41,42]. Another member of the UTX family, JMJD3, is overexpressed during the senescence of mouse embryonic fibroblasts [91] and is accompanied by the recruitment of the protein MLL to the INK4/ARF locus, provoking the dissociation of PRC2 and activation of the INK4/ARF gene [91]. Related to these results, it has been recently reported that the expression of p16INK4A is upregulated in MDS but not AML [92]. Taken together, these results suggest that during the pathogenesis of MDS, overexpression of JMJD3 could potentially down-regulate the H3K27 methylation of p16INK4A promoter, which could cause the deregulation of this gene. However, there is also reports of the silencing and hypermethylation of p16INK4A promoter in MDS, particularly in pediatric primary MDS [93]. Therefore, the specific role of p16INK4A gene product, as well as the potential regulation of this gene by JMJD3, in the development of MDS still needs further study.

H3K9 methylation

Methylation on H3K9 is a well conserved mark of transcriptional silencing. Although H3K9 methylation has been shown to control DNA methylation in filamentous fungi and plants [94], there is still no direct interplay between H3K9 methylation and DNA methylation documented in mammals. In MDS, the rearrangements of 3q26 are relatively frequent and often lead to the activation of oncogene EVI1 [95]. In various experimental systems, including mouse models, the activation of EVI1 has been found to lead to myeloid dysplasia, supporting the contribution of EVI1 to the pathogenesis of MDS [96]. In relation to H3K9 methylation, EVI1 has been shown to interact with H3K9 methyltransferases SUV39H1 and G9a [97]. These interactions are important for bone marrow immortalization [97]. In another study using AML cells, the H3K9 methyltransferase activity of SUV39H1 has been demonstrated to silence p15INK4B and E-cadherin genes [98], which are two tumor suppressors also frequently silenced in MDS [99,100]. Whether there is also a interaction between SUV39H1/G9a and EVI1 in MDS and if so whether this interaction affects the H3K9 methylation as well as the expression of the tumor suppressors such as p15INK4B and E-cadherin in this disease, are still unclear and therefore require further study.

H4K20 methylation

Methylation on H4K20 is a mark for heterochromatin [35]. H4K20 methylation has also been reported to affect DNA damage checkpoint by recruiting the checkpoint protein 53BP1 [101]. Methyltransferases of H4K20 that have been identified in mammals include SUV420H1/2 [102] and SETD8 [103]. For demethylase, the JmjC domain protein PHF8, has been shown to catalyze the removal of methylation from both H3K9 and H4K20 [104]. Interestingly, the recruitment of PHF8 to chromatin occurs through the interaction between the methyl-lysine recognizing the PHD domain of PHF8 and methylated H3K4 [105], highlighting the mutual regulation between H3K4 and H3K9 or H4K20 methylation.

Although no specific mutation or abnormalities involving H4K20 methyltransferase or demethylase has been identified in MDS, potential involvement of H4K20 methylation in MDS has been implicated in the knockout mouse model of Arid4a and Arid4b genes [106]. These mice show hematopoietic phenotypes, include ineffective hematopoiesis, followed by transition to CMML-like myelodysplastic/myeloproliferative disorder and then transformation to AML [106]. Of interest, there is increase of histone methylation of H3K4, H3K9 and H4K20 in the bone marrow cells of the knockout mice [106], suggesting the involvement of H4K20 methylation in MDS.

Histone arginine methylation marks

In comparison to histone lysine methylation, reports on characterization of different forms of histone arginine methylation in MDS are more limited. Members of the family of protein arginine methyltransferases (PRMT) in human, which can catalyze the arginine methylation of histone and nonhistone proteins, are PRMT1–11 [107]. Among them, PRMT1, 2, 4, 5 and 6 have been associated with the methylation of histone arginine [107]. Of interest, the potential pathogenetic role of PRMTs in MDS and other myeloid neoplasms may be partially mediated through the interactions between PRMT and histone lysine methyltransferases. For instance, H4R3 methyltransferase PRMT1, an essential regulator enhancing the self-renewal of primary hematopoietic cells, is a component of the oncogenic transcriptional complex containing H3K4 methyltransferase MLL [108]. Another H3R2 methyltransferase PRMT6 has been found to inhibit the recruitment of the H3K4 methytransferase subunit WDR5 to its target and repress the H3K4 trimethylation of the target genes such as HoxA genes [109,110]. It should be noted that PRMTs also target nonhistone proteins, which can also contribute to the pathogenesis of MDS. For instance, besides H3R4, PRMT1 also methylates arginine R206 and R210 on RUNX1 protein [111]. RUNX1 is an important transcription factor frequently mutated in MDS [46]. When CD34+ cells differentiate into myeloid cells, the expression of PRMT1 is upregulated, which is accompanied with high arginine methylation on RUNX1 protein [111]. For the removal of methyl unit, as of now only JMJD6 has been demonstrated to be a histone arginine demethylase [112], which removes methylation from H3R2 and H4R3 [112]. No specific defect on JMJD6 has been identified in MDS and other forms of myeloid neoplasms yet.

Drug development targeting histone methylation

Owing to the importance of histone methylation in gene regulation and its involvement in cancer, the discoveries of potential drugs targeting regulators of histone methylation are now being actively pursued. However, there are still quite a few limitations that hamper the development of therapeutic HMT and HDM inhibitors. One major limitation is the lack of crystallographic structural information of HMTs and HDMs, which is usually required to study the interaction between the inhibitors and its targeted proteins. The other important issue is the problem of specificity. For instance, S-adenosyl-L-methionine (SAM) and analogues are used as cofactor not only by HMTs, but also by DNA methyltransferases and other enzymes [113]. Similarly, current JmjC domain HDM inhibitors also lack subtype specificities. These make the inhibition HMTs via targeting SAM cofactors and the use of JmjC domain HDM inhibitors problematic in therapeutic applications. Therefore, in comparison to the targeting of DNA methylation and histone deacetylation, development of histone methylation inhibitors is still in its infancy and the number of inhibitors reported to target histone methylation is quite limited. Among the compounds reported as HMT/HDM inhibitors, a few have been demonstrated to have potential activities in myeloid neoplasms.

Inhibitors for HMT

3-deazaneplanocin A (DZNep) is a potent inhibitor of S-adenosylhomocysteine (AdoHcy) hydrolase, which in turn causes by-product inhibition of SAM-dependent methyltransferases, including H3K27 methyltransferase EZH2 [114]. DZNep has been demonstrated to cause pharmacologic disruption of EZH2-related histone methylation and associated anticancer activity in several types of cancers, including in AML cell lines and primary AML cells [115]. This is accompanied by the activation of several tumor suppressor genes together with repression of cyline E and HoxA9 [115]. When combined with pan-HDACi panobiostat, DZNep synergistically improved the survival of immunodeficient mice with HL-60 leukemia [115]. These findings suggest that targeting EZH2 mediated H3K27 methylation by DZNep could be a potential effective epigenetic therapy against myeloid neoplasms. However, as motioned above, there is the issue of specificity, since DZNep, as a compound targeting the SAM analogues, may also affect the activities of multiple SAM-dependent methyltransferases other than EZH2. The other complication involved in the potential application of DZNep in MDS is that the definite role of EZH2 in MDS is still unclear, since at the same time of the identification of missense mutation of EZH2 in MDS, this gene has also been found in some cases to be overexpressed in MDS and other types of cancer [87,116].

Several compounds have shown inhibition against HMTs that regulate histone methylation on residues other than H3K27. These include the H3K9 methyltransferase inhibitors chaetocin (targeting SUV39H1) [117], BIX-01294 (targeting G9a) [118] as well as the PRMT1 inhibitors such as arginine methyltransferase inhibitor 1 (AMI-1) derivatives [119] and RM65 [120]. Particularly, among these compounds, G9a inhibitor BIX-01294 has shown potential biological effect, which could activate IP-10 gene, a potential disease gene in idiopathic pulmonary fibrosis [121].

Inhibitors for HDM

A series of inhibitors targeting the non-JmjC domain H3K4 demethylase LSD1 have been synthesized by different groups. Most of these compounds are derivatives of monoamine oxidase (MAO) inhibitors, based on the similar oxidative manner between LSD1 and MAO [122]. Among them, a derivative (14e) of tranylcypromine (PCPA) synthesized by Binda et al. has shown to enhance the efficacy of retinoic acid on growth and differentiation of AML cell line NB4 [123].

In comparison to the inhibitors targeting LSD1, reports about the inhibitors against JmjC domain HDMs are quite limited. Several compounds have recently been shown to inhibit the H3K9 demethylase JMJD2A, including a pyridine-2,4-dicarbo-xylin acid derivative, pyridine-2,4-dicarboxylin acid-dimethyl ester [124]. Some of these JMJD2A inhibitors have shown combinational anticancer effects in prostate and colon cancer cell lines when applied with a LSD1 inhibitor NCL-2 [125].

Although some of the HMT/HDM inhibitors described above have shown potential activities in certain myeloid neoplasms such as AML, there is still no report about trials of these compounds in MDS. This could be owing to the currently still limited number of potential inhibitors, the specificity issue of these inhibitors, as well as the complicity of the pathology of MDS, including the lack of representative cell lines and the limited animal models of this disease. Therefore, a potential application of a specific HMT/HDM inhibitor in MDS is dependent on a better characterization of the role of these HMTs/HDMs in the pathogenesis of MDS or at least in a subtype of MDS, in parallel with further development/optimization of the potential HMT/HDM inhibitors. For instance, based on both clinical data and phenotypes of mouse models, EVI1 has been clearly shown to involve in MDS pathogenesis. Recently, H3K9 methyltransferases SUV39H1 and G9a has been demonstrated to interact with EVI1, which could then affect bone marrow immortalization [97]. It is therefore possible that a better characterization of SUV39H1 and/or G9a in MDS, combined with further optimization of their inhibitors such as the above described chaetocin [117] or BIX-01294 [118], may lead to preclinical trials of these compounds on an animal model such as the EVI1 mice which exhibit MDS phenotype, and furthermore, potential applications of these drugs on MDS patients.

Future perspective

There has been a rapid progress towards the understanding about the regulation of histone methylation and its role in human disease. Owing to the complexity of the pathophysiology of MDS, as well as the lack of representative cell lines or animal models, studies addressing histone methylation in MDS are still quite limited and a large portion of the evidence, in regards to the involvement of a specific histone methylation regulator in the pathogenesis of MDS, is still indirect and sometimes based on the predictions from the results in other types of myeloid neoplasms. Despite these limitations, results summarized in this article provide preliminary evidence for the potential involvement of histone methylation in the pathogenesis MDS (Table 1). Therefore, we believe that further investigations of histone methylation in MDS are both important and promising in the years to come. Detailed investigations of histone methylation in MDS should include careful clinical observations about the alterations of a specific HMT, HDM or their cofactor/target in patients, detailed molecular and cellular studies of these gene products in transcription regulation, as well as the generation of appropriate animal models involving specific key histone methylation regulators that could mimic some key features of MDS. Achievements of these clinical and basic studies, together with the progress in the development of the drugs inhibiting HMTs/HDMs, will eventually help us to identify and target essential histone methylation regulators in the treatment of MDS.

Table 1.

Abnormalities of histone methylation identified in myelodysplastic syndromes: histone methyltransferases, histone demethylases and their associated gene products in myelodysplastic syndromes.

Modified residue HMT/HDM involved in MDS Defect of HMT/HDM in MDS Defect of targets/interacting proteins of HMT/HDM in MDS Inhibitor
Activating K methylation
H3K4 MLL (HMT); LSD1 (HDM) Gene amplification (rare) HoxA9, Meis1 (possible targets of MLL): overexpressed in MDS
LSD1 interacting proteins; GFI1: overexpressed in high-risk MDS; TAL1: upregulated in SDS syndrome; ASXL1: loss of function mutation in MDS
Several MAO inhibitor derivatives
H3K36 NSD1/3 (HMT); JHDM1A/B (HDM) NUP98–NS1/3 fusion HoxA9 (possible target of NSD1): overexpressed in MDS; BCL6 (interact with JHDM1a/b): amplification of BCL6 gene in MDS
H3K79 DOT1L (HMT) MLL–AF4 fusion (interact with DOT1L): gene fusion in MDS rare); targets of DOT1L: GATA2: overexpressed in MDS; PU.1: interacting with EVI1
Repressive K methylation
H3K27 EZH2 (HMT); UTX (HDM); JMJD3 (HDM) Mutation/overexpression in MDS; mutations in MDS p16INK4 (target of JMJD3): overexpressed in MDS but not AML DZNep
H3K9 SUV39H1 (HMT); G9A (HMT) EVI1 (interacting protein of SUV39H1 and G9a): overexpressed in MDS; p15INK4b and E-cadherin (targets of SUV39H1): silencing in MDS
H4K20 Level increase in Arid4a/4b KO mice, which have defects that progress into MDS and AML
R methylation
H4R3 PRMT1 PRMT1 enhances self-renewal of hematopoietic cells MLL AMI-1 derivatives, RM65
H3R2 PRMT6 PRMT6 inhibits MLL mediated H3K4 methylation Chaetocin, BIX-01294

AMI-1: Arginine methyltransferase inhibitor 1; AML: Acute myelogenous leukemia; DZNep: 3-deazaneplanocin A; HDM: Histone demethylases; HMT: Histone methyltransferases; K: Lysine residue; KO: Knockout; MAO: Monoamine oxidase; MDS: Myelodysplastic syndromes; MLL: Mixed-lineage leukemia; PRMT: Protein arginine methyltransferases; R: Arginine residue.

Executive summary.

  • Myelodysplastic syndromes (MDS) are a very complex group of myeloid neoplasms and are becoming one of the most common forms of myeloid neoplasms in older individuals.

  • The heterogeneity of MDS makes both diagnosis and molecular studies of this disease complex.

Overview of epigenetic regulation & histone methylation in MDS

  • Epigenetic regulations have been demonstrated to play an important role in the pathogenesis of malignancies. Compared with genetic lesions, abnormalities of epigenetic regulation can potentially be reversed by pharmacological intervention.

  • Among different epigenetic modifications, DNA methylation and histone acetylation are the best characterized. Development of drugs targeting these two types of epigenetic regulation is more advanced, including in MDS.

  • Although both the substrate lysine residues and the degree of methylation affect gene expression, generally, histone methylation on H3K4, H3K36 and possibly also H3K79 is positively associated with active transcription and methylation on H3K9, H3K27 as well as H4K20 tends to negatively regulate transcription.

  • Histone methylation is dynamically regulated by histone methyltransferases and histone demethylases.

Abnormalities of activating histone methylation marks in MDS

  • Summaries are provided for each type of histone methylation regarding the abnormities that have been identified in, or been potentially linked to, MDS. These include altered histone methylation levels and abnormities of its regulators as well as target genes.

Drug development targeting histone methylation

  • Inhibitors targeting histone methyltransferases have been developed and some of them have shown anticancer activity in acute myelogenous leukemia cells.

  • Inhibitors targeting histone demethylases, such as non-Jmjc domain histone demethylase LSD1, have been developed and shown to have a biological effect in acute myelogenous leukemia cells, whereas inhibitors targeting Jmjc domain histone demethylases are still quite limited.

Future perspective

  • Studies of histone methylation in MDS are important and promising. These will help us to identify key histone methylation regulators as prognostic markers or therapeutic targets for the treatment of MDS.

Footnotes

For reprint orders, please contact: reprints@futuremedicine.com

No writing assistance was utilized in the production of this manuscript.

Financial & competing interests disclosure

Yue Wei and Guillermo Garcia-Manero are supported by the Cancer Prevention and Research and Institute of Texas, Individual Investigator Research Awards. 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.

Bibliography

Papers of special note have been highlighted as:

▪ of interest

▪▪ of considerable interest

  • 1▪▪.Nimer SD. Myelodysplastic syndromes. Blood. 2008;111:4841–4851. doi: 10.1182/blood-2007-08-078139. Critically reviews the recent progress in the understanding of and therapies for myelodysplastic syndromes (MDS) [DOI] [PubMed] [Google Scholar]
  • 2.Rollison DE, Howlader N, Smith MT, et al. Epidemiology of myelodysplastic syndromes and chronic myeloproliferative disorders in the United States, 2001–2004, using data from the NAACCR and SEER programs. Blood. 2008;112:45–52. doi: 10.1182/blood-2008-01-134858. [DOI] [PubMed] [Google Scholar]
  • 3.Godley LA, Larson RA. Therapy-related myeloid leukemia. Semin Oncol. 2008;35:418–429. doi: 10.1053/j.seminoncol.2008.04.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tefferi A, Thiele J, Vardiman W. Response: The 2008 World Health Organization diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary myelofibrosis: a paradigm of effective collaboration among clinicians, pathologists, and scientists. Blood. 2008;111:1742. [Google Scholar]
  • 5.Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079–2088. [PubMed] [Google Scholar]
  • 6.Garcia-Manero G, Shan J, Faderl S, et al. A prognostic score for patients with lower risk myelodysplastic syndrome. Leukemia. 2008;22:538–543. doi: 10.1038/sj.leu.2405070. [DOI] [PubMed] [Google Scholar]
  • 7.Dayyani F, Conley AP, Strom SS, et al. Cause of death of patients with lower risk myelodysplastic syndrome. Cancer. 2010;116:2174–2179. doi: 10.1002/cncr.24984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Haase D, Germing U, Schanz J, et al. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood. 2007;110:4385–4395. doi: 10.1182/blood-2007-03-082404. [DOI] [PubMed] [Google Scholar]
  • 9▪.Bejar R, Levine R, Ebert BL. Unraveling the molecular pathophysiology of myelodysplastic syndromes. J Clin Oncol. 2011;29:504–515. doi: 10.1200/JCO.2010.31.1175. Update of the most recent progress on the genetic abnormalities associated with MDS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10▪.Issa JP. Epigenetic changes in the myelodysplastic syndromes. Hematol Oncol Clin North Am. 2010;24:317–330. doi: 10.1016/j.hoc.2010.02.007. Updates the most recent progress on the epigenetic abnormalities associated with MDS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Riddihough G, Zahn LM. Epigenetics. What is epigenetics? Introduction. Science. 2010;330:611. doi: 10.1126/science.330.6004.611. [DOI] [PubMed] [Google Scholar]
  • 12.Swami M. Epigenetics: Demethylation links cell fate and cancer. Nat Rev Cancer. 2010;10:740. doi: 10.1038/nrc2948. [DOI] [PubMed] [Google Scholar]
  • 13.Kaiser J. Genes link epigenetics and cancer. Science. 2010;330:577. doi: 10.1126/science.330.6004.577. [DOI] [PubMed] [Google Scholar]
  • 14.Garcia-Manero G. Progress in myelodysplastic syndromes. Clin Lymphoma Myeloma. 2009;9:S286–S292. doi: 10.3816/CLM.2009.s.025. [DOI] [PubMed] [Google Scholar]
  • 15.Kuendgen A, Lübbert M. Current status of epigenetic treatment in myelodysplastic syndromes. Ann Hematol. 2008;87:601–611. doi: 10.1007/s00277-008-0477-9. [DOI] [PubMed] [Google Scholar]
  • 16.Figueroa ME, Skrabanek L, Li Y, et al. MDS and secondary AML display unique patterns and abundance of aberrant DNA methylation. Blood. 2009;114:3448–3458. doi: 10.1182/blood-2009-01-200519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Länger F, Dingemann J, Kreipe H, Lehmann U. Up-regulation of DNA methyltransferases DNMT1, 3A, and 3B in myelodysplastic syndrome. Leuk Res. 2005;29:325–329. doi: 10.1016/j.leukres.2004.08.004. [DOI] [PubMed] [Google Scholar]
  • 18.Smith AE, Mohamedali AM, Kulasekararaj A, et al. Next-generation sequencing of the TET2 gene in 355 MDS and CMML patients reveals low-abundance mutant clones with early origins, but indicates no definite prognostic value. Blood. 2010;116:3923–3932. doi: 10.1182/blood-2010-03-274704. [DOI] [PubMed] [Google Scholar]
  • 19.Ko M, Huang Y, Jankowska AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature. 2010;468(7325):839–843. doi: 10.1038/nature09586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomized, open-label, Phase III study. Lancet Oncol. 2009;10:223–232. doi: 10.1016/S1470-2045(09)70003-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Garcia-Manero G, Yang H, Bueso-Ramos C, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111:1060–1066. doi: 10.1182/blood-2007-06-098061. [DOI] [PubMed] [Google Scholar]
  • 22.Garcia-Manero G, Assouline S, Cortes J, et al. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood. 2008;112:981–989. doi: 10.1182/blood-2007-10-115873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Klimek VM, Fircanis S, Maslak P, et al. Tolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromes. Clin Cancer Res. 2008;14:826–832. doi: 10.1158/1078-0432.CCR-07-0318. [DOI] [PubMed] [Google Scholar]
  • 24.Giles F, Fischer T, Cortes J, et al. A Phase I study of intravenous LBH589, a novel cinnamic hydroxamic acid analogue histone deacetylase inhibitor, in patients with refractory hematologic malignancies. Clin Cancer Res. 2006;12:4628–4635. doi: 10.1158/1078-0432.CCR-06-0511. [DOI] [PubMed] [Google Scholar]
  • 25▪▪.Klose RJ, Zhang Y. Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol. 2007;8:307–318. doi: 10.1038/nrm2143. Clearly reviews the mechanisms of histone demethylation and provides the insights about the biological functions of histone demethylases. [DOI] [PubMed] [Google Scholar]
  • 26▪.Kouzarides T. Chromatin Modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005. Critically reviews histone modifications and their potential to influence many fundamental biological processes. [DOI] [PubMed] [Google Scholar]
  • 27▪.Lan F, Nottke AC, Shi Y. Mechanisms involved in the regulation of histone lysine demethylases. Curr Opin Cell Biol. 2008;20:316–325. doi: 10.1016/j.ceb.2008.03.004. Focuses on the regulatory mechanisms that modulate histone demethylase recruitment and activity. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28▪▪.Bedford MT. Arginine methylation at a glance. J Cell Sci. 2007;120:4243–4246. doi: 10.1242/jcs.019885. Provides a clear overview about mammalian arginine methyltransferases. [DOI] [PubMed] [Google Scholar]
  • 29▪.Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol. 2005;6:838–849. doi: 10.1038/nrm1761. Summarizes advances in the understanding of how lysine methylation functions in diverse biological processes, and particularly, its potential link to disease. [DOI] [PubMed] [Google Scholar]
  • 30.Albert M, Helin K. Histone methyltransferases in cancer. Sem Cell Develop Biol. 2010;21:209–220. doi: 10.1016/j.semcdb.2009.10.007. [DOI] [PubMed] [Google Scholar]
  • 31.Flanagan JF, Mi LZ, Chruszcz M. Double chromodomains cooperate to recognize the methylated histone H3 tail. Nature. 2005;438:1090–1091. doi: 10.1038/nature04290. [DOI] [PubMed] [Google Scholar]
  • 32.Spektor TM, Rice JC. Identification and characterization of posttranslational modification-specific binding proteins in vivo by mammalian tethered catalysis. Proc Natl Acad Sci USA. 2009;106:14808–14813. doi: 10.1073/pnas.0907799106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Li H, Fischle W, Wang W, et al. Structural basis for lower lysine methylation state-specific readout by MBT repeats of L3MBTL1 and an engineered PHD finger. Mol Cell. 2007;28:677–691. doi: 10.1016/j.molcel.2007.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shi X, Hong T, Walter KL, et al. ING2 PHD domain links histone H3 lysine 4 methylation to active gene repression. Nature. 2006;442:96–99. doi: 10.1038/nature04835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35▪▪.Barski A, Cuddapah S, Cui K, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–837. doi: 10.1016/j.cell.2007.05.009. Provides the very first set of digital-quality data about histone methylation marks in whole genome, which helps to understand the contribution of histone methylation in gene expression and epigenetic inheritance. [DOI] [PubMed] [Google Scholar]
  • 36▪.Shi Y, Lan F, Matson C, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004;119:941–953. doi: 10.1016/j.cell.2004.12.012. Very first identification of a histone demethylase that is conserved from Schizosaccharomyces pombe to human and reveals the dynamic regulation of histone methylation by both histone methylases and demethylases. [DOI] [PubMed] [Google Scholar]
  • 37▪.Tsukada Y, Fang J, Erdjument-Bromage H, et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature. 2006;439:811–816. doi: 10.1038/nature04433. Highlights the identification of the JmjC domain as a novel demethylase signature motif and uncovers a protein demethylation mechanism that is conserved from yeast to human. [DOI] [PubMed] [Google Scholar]
  • 38▪.Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7:823–833. doi: 10.1038/nrc2253. Clearly reviews the link between MLL and the histone methylations regulated by MLL with hematopoietic malignancies. [DOI] [PubMed] [Google Scholar]
  • 39▪.Okada Y, Feng Q, Lin Y, et al. hDOT1L links histone methylation to leukemogenesis. Cell. 2005;121:167–178. doi: 10.1016/j.cell.2005.02.020. Elucidates the involvement of histone (H3K79) methylation in leukemogenesis. [DOI] [PubMed] [Google Scholar]
  • 40▪▪.Nikoloski G, Langemeijer SM, Kuiper RP, et al. Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat Genet. 2010;42:665–667. doi: 10.1038/ng.620. Provides evidence about the genomic mutation of a histone methyltransferase encoding gene (EZH2) in MDS, and highlights the importance of the regulation of histone methylation in this disease. [DOI] [PubMed] [Google Scholar]
  • 41.Jankowska A, Makishima H, Tiu RV, et al. Mutational spectrum in chronic myelomonocytic leukemia includes genes associated with epigenetic regulation such as UTX and EZH2. Presented at: 52nd American Society of Hematology Annual Meeting and Exposition; Orlando, FL, USA. 4–7 December 2010. [Google Scholar]
  • 42.Szpurka H, Jankowska AM, Przychodzen B, et al. UTX mutations and epigenetic changes in MDS/MPN and related myeloid malignancies. Presented at: 52nd American Society of Hematology Annual Meeting and Exposition; Orlando, FL, USA. 4–7 December 2010. [Google Scholar]
  • 43.Wei Y, Chen R, Bueso-Ramos C, et al. Genome-wide CHIP-Seq analysis of histone methylation reveals modulators of NF-κB signaling and the histone demethylase JMJD3 as implicated in disease progression in myelodysplastic syndrome (MDS). Presented at: 51st American Society of Hematology Annual Meeting and Exposition; New Orleans, LA, USA. 4–7 December 2009. [Google Scholar]
  • 44▪.Milne TA, Briggs SD, Brock HW, et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol Cell. 2002;10:1107–1117. doi: 10.1016/s1097-2765(02)00741-4. Provides evidence for the involvement of MLL regulated H3K4 methylation in Hox gene expression. [DOI] [PubMed] [Google Scholar]
  • 45.Malik S, Bhaumik SR. Mixed lineage leukemia: histone H3 Lysine 4 methyltrasferases from yeast to human. FEBS J. 2010;277:1805–1821. doi: 10.1111/j.1742-4658.2010.07607.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dicker F, Haferlach C, Sundermann J, et al. Mutation analysis for RUNX1, MLL-PTD, FLT3-ITD, NPM1 and NRAS in 269 patients with MDS or secondary AML. Leukemia. 2010;24:1528–1532. doi: 10.1038/leu.2010.124. [DOI] [PubMed] [Google Scholar]
  • 47▪.Wang SA, Jabbar K, Lu G, et al. Trisomy 11 in myelodysplastic syndromes defines a unique group of disease with aggressive clinicopathologic features. Leukemia. 2010;24:740–747. doi: 10.1038/leu.2009.289. Highlights the potential involvement of MLL locus in the progress of MDS. [DOI] [PubMed] [Google Scholar]
  • 48.Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukeamia stem cell development. Nat Rev Cancer. 2007;7:823–833. doi: 10.1038/nrc2253. [DOI] [PubMed] [Google Scholar]
  • 49▪.Heinrichs S, Berman JN, Ortiz TM, et al. CD34+ cell selection is required to assess HOXA9 expression levels in patients with myelodysplastic syndrome. Br J Haematol. 2005;130:83–86. doi: 10.1111/j.1365-2141.2005.05555.x. Provides evidence about altered expression of HOXA9 and related genes in the multipotential malignant progenitor cells of MDS. [DOI] [PubMed] [Google Scholar]
  • 50▪▪.Lee MG, Wynder C, Cooch N, Shiekhattar R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature. 2005;437:432–435. doi: 10.1038/nature04021. Highlights an essential role for CoREST in demethylation of H3K4 both in vitro and in vivo. [DOI] [PubMed] [Google Scholar]
  • 51▪.Saleque S, Kim J, Rooke HM, Orkin SH. Epigenetic regulation of hematopoietic differentiation by Gfi-1 and Gfi-1b is mediated by the cofactors CoREST and LSD1. Mol Cell. 2007;27:562–572. doi: 10.1016/j.molcel.2007.06.039. Elucidate the function of Gfi-1 in the regulation of H3K4 methylation through the interaction with LSD1 and CoREST and the effects on hematopoietic differentiation. [DOI] [PubMed] [Google Scholar]
  • 52▪.Hu X, Li X, Valverde K, Fu X, Noguchi C, Qiu Y, Huang S. LSD1-mediated epigenetic modification is required for TAL1 function and hematopoiesis. Proc Natl Acad Sci USA. 2009;106:10141–10146. doi: 10.1073/pnas.0900437106. Reveals that LSD1 regulates TAL1-mediated transcription, which may determine the onset of erythroid differentiation programs. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Huh HJ, Chae SL, Lee M, Hong KS, Mun YC, Seong CM, et al. PU. 1 and GFI1 mRNA expression in myelodysplastic syndrome. Int J Lab Hematol. 2009;31:344–351. doi: 10.1111/j.1751-553X.2008.01056.x. [DOI] [PubMed] [Google Scholar]
  • 54.Rujkijyanont P, Beyene J, Wei K, Khan F, Dror Y. Leukaemia-related gene expression in bone marrow cells from patients with the preleukaemic disorder Shwachman–Diamond syndrome. Br J Haematol. 2007;137:537–544. doi: 10.1111/j.1365-2141.2007.06608.x. [DOI] [PubMed] [Google Scholar]
  • 55.Lee SW, Cho YS, Na JM, et al. ASXL1 represses retinoic acid receptor-mediated transcription through associating with HP1 and LSD1. J Biol Chem. 2010;285:18–29. doi: 10.1074/jbc.M109.065862. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 56.Gelsi-Boyer V, Trouplin V, Roquain J, et al. ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia. Br J Haematol. 2010;151:365–375. doi: 10.1111/j.1365-2141.2010.08381.x. [DOI] [PubMed] [Google Scholar]
  • 57.Gelsi-Boyer V, Trouplin V, Adélaïde J, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009;145:788–800. doi: 10.1111/j.1365-2141.2009.07697.x. [DOI] [PubMed] [Google Scholar]
  • 58.Boultwood J, Perry J, Pellagatti A, et al. Frequent mutation of the polycomb-associated gene ASXL1 in the myelodysplastic syndromes and in acute myeloid leukemia. Leukemia. 2010;24:1062–1065. doi: 10.1038/leu.2010.20. [DOI] [PubMed] [Google Scholar]
  • 59▪.La Starza R, Gorello P, Rosati R, et al. Cryptic insertion producing two NUP98/NSD1 chimeric transcripts in adult refractory anemia with an excess of blasts. Genes Chromosomes Cancer. 2004;41:395–399. doi: 10.1002/gcc.20103. First report of MDS harboring an NUP98/NSD1 fusion gene. [DOI] [PubMed] [Google Scholar]
  • 60▪.Taketani T, Taki T, Nakamura H, Taniwaki M, Masuda J, Hayashi Y. NUP98-NSD3 fusion gene in radiation-associated myelodysplastic syndrome with t(8;11)(p11;p15) and expression pattern of NSD family genes. Cancer Genet Cytogenet. 2009;190:108–112. doi: 10.1016/j.cancergencyto.2008.12.008. Provides evidence that NSD3/NUP98 fusion gene might be related to t-MDS. [DOI] [PubMed] [Google Scholar]
  • 61▪▪.Wang GG, Cai L, Pasillas MP, Kamps MP. NUP98-NSD1 links H3K36 methylation to Hox-A gene activation and leukaemogenesis. Nat Cell Biol. 2007;9:804–812. doi: 10.1038/ncb1608. Provides first link between deregulated histone (H3K36) methylation to tumorigenesis and between NSD1 to transcriptional regulation of the Hox-A locus. [DOI] [PubMed] [Google Scholar]
  • 62.He J, Kallin EM, Tsukada Y, Zhang Y. The H3K36 demethylase Jhdm1b/Kdm2b regulates cell proliferation and senescence through p15(Ink4b) Nat Struct Mol Biol. 2008;15:1169–1175. doi: 10.1038/nsmb.1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gearhart MD, Corcoran CM, Wamstad JA, Bardwell VJ. Polycomb group and SCF ubiquitin ligases are found in a novel BCOR complex that is recruited to BCL6 targets. Mol Cell Biol. 2006;26:6880–6889. doi: 10.1128/MCB.00630-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Martínez-Ramírez A, Urioste M, Melchor L, et al. Analysis of myelodysplastic syndromes with complex karyotypes by high-resolution comparative genomic hybridization and subtelomeric CGH array. Genes Chromosomes Cancer. 2005;42:287–298. doi: 10.1002/gcc.20154. [DOI] [PubMed] [Google Scholar]
  • 65.Vakoc CR, Sachdeva MM, Wang H, Blobel GA. Profile of histone lysine methylation across transcribed mammalian chromatin. Mol Cell Biol. 2006;26:9185–9195. doi: 10.1128/MCB.01529-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Feng Q, Wang H, Ng HH, et al. Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain. Curr Biol. 2002;12:1052–1058. doi: 10.1016/s0960-9822(02)00901-6. [DOI] [PubMed] [Google Scholar]
  • 67.Janzen CJ, Hake SB, Lowell JE, Cross GA. Selective di- or trimethylation of histone H3 lysine 79 by two DOT1 homologs is important for cell cycle regulation in Trypanosoma brucei. Mol Cell. 2006;23:497–507. doi: 10.1016/j.molcel.2006.06.027. [DOI] [PubMed] [Google Scholar]
  • 68▪.Krivtsov AV, Feng Z, Lemieux ME, et al. H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell. 2008;14:355–368. doi: 10.1016/j.ccr.2008.10.001. Demonstrates the involvement of ectopic H3K79 methylation in hematopoietic malignancies and its importance in the maintenance of MLL-AF4-driven gene expression. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Barry ER, Corry GN, Rasmussen TP. Targeting DOT1L action and interactions in leukemia: the role of DOT1L in transformation and development. Expert Opin Ther Targets. 2010;14:405–418. doi: 10.1517/14728221003623241. [DOI] [PubMed] [Google Scholar]
  • 70▪.Zhu YL, Zhang Y, Zhu P, Yang Y, Du JW, Liu J. Role of molecular screening for common fusion genes in the diagnosis and classification of leukemia. Beijing Da Xue Xue Bao. 2005;37:236–239. Provides evidence of MLL-AF4 fusion in MDS. [PubMed] [Google Scholar]
  • 71▪.Feng Y, Yang Y, Ortega MM, et al. Early mammalian erythropoiesis requires the Dot1L methyltransferase. Blood. 2010;116:4483–4491. doi: 10.1182/blood-2010-03-276501. Highlights the importance of histone (H3K79) methylation in the regulation of hematopoiesis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Chou ST, Khandros E, Bailey LC, et al. Graded repression of PU. 1/Sfpi1 gene transcription by GATA factors regulates hematopoietic cell fate. Blood. 2009;114:983–994. doi: 10.1182/blood-2009-03-207944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lahortiga I, Vázquez I, Agirre X, et al. Molecular heterogeneity in AML/MDS patients with 3q21q26 rearrangements. Genes Chromosomes Cancer. 2004;40:179–189. doi: 10.1002/gcc.20033. [DOI] [PubMed] [Google Scholar]
  • 74.Laricchia-Robbio L, Premanand K, Rinaldi CR, Nucifora G. EVI1 Impairs myelopoiesis by deregulation of PU. 1 function. Cancer Res. 2009;69:1633–1642. doi: 10.1158/0008-5472.CAN-08-2562. [DOI] [PubMed] [Google Scholar]
  • 75▪.Nishikata I, Sasaki H, Iga M, et al. A novel EVI1 gene family, MEL1, lacking a PR domain (MEL1S) is G-CSF–induced myeloid differentiation expressed mainly in t(1;3) (p36;q21)-positive AML and blocks G-CSF-induced myeloid differentiation. Blood. 2003;102:3323–3332. doi: 10.1182/blood-2002-12-3944. Demonstrates that overexpression of the EVI1 lacking the PR domain is one of the causative factors in the pathogenesis of myeloid malignancies. [DOI] [PubMed] [Google Scholar]
  • 76▪▪.Azuara V, Perry P, Sauer S, et al. Chromatin signatures of pluripotent cell lines. Nat Cell Biol. 2006;8:532–538. doi: 10.1038/ncb1403. Provides evidence about the promoter ‘bivalent status’ and its importance for regulating the expression of lineage-specific genes. [DOI] [PubMed] [Google Scholar]
  • 77.Jørgensen HF, Giadrossi S, Casanova M. Stem cells primed for action: polycomb repressive complexes restrain the expression of lineage-specific regulators in embryonic stem cells cell cycle. Cell Cycle. 2006;5:1411–1414. doi: 10.4161/cc.5.13.2927. [DOI] [PubMed] [Google Scholar]
  • 78▪.Mazzarella L, Jørgensen HF, Soza-Ried J, et al. Embryonic stem cell–derived hemangioblasts remain epigenetically plastic and require PRC1 to prevent neural gene expression. Blood. 2011;117:83–87. doi: 10.1182/blood-2010-03-273128. Reinforces the importance of bivalent chromatin and PRC1 for normal hematopoietic differentiation. [DOI] [PubMed] [Google Scholar]
  • 79.Paul TA, Bies J, Small D, Wolff L. Signatures of polycomb repression and reduced H3K4 trimethylation are associated with p15INK4b DNA methylation in AML. Blood. 2010;115:3098–3108. doi: 10.1182/blood-2009-07-233858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Uchida T, Kinoshita T, Nagai H, et al. Hypermethylation of the p15INK4B gene in myelodysplastic syndromes. Blood. 1997;90:1403–1409. [PubMed] [Google Scholar]
  • 81.Quesnel B, Fenaux P. P15INK4b gene methylation and myelodysplastic syndromes. Leuk Lymphoma. 1999;35:437–443. doi: 10.1080/10428199909169608. [DOI] [PubMed] [Google Scholar]
  • 82.Look AT. Molecular pathogenesis of MDS. Hematology Am Soc Hematol Educ Program. 2005:156–160. doi: 10.1182/asheducation-2005.1.156. [DOI] [PubMed] [Google Scholar]
  • 83.Liu TX, Becker MW, Jelinek J, et al. Chromosome 5q deletion and epigenetic suppression of the gene encoding α-catenin (CTNNA1) in myeloid cell transformation. 2007;13:78–83. doi: 10.1038/nm1512. [DOI] [PubMed] [Google Scholar]
  • 84.Ye Y, McDevitt MA, Guo M, et al. Progressive chromatin repression and promoter methylation of CTNNA1 associated with advanced myeloid malignancies. Cancer Res. 2009;69:8482–8490. doi: 10.1158/0008-5472.CAN-09-1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Czermin B, Melfi R, McCabe D, Seitz V, Imhof A, Pirrotta V. Drosophila enhancer of Zeste/ESC complexes have a histone H3 methyltransferase activity that marks chromosomal Polycomb sites. Cell. 2002;11:185–196. doi: 10.1016/s0092-8674(02)00975-3. [DOI] [PubMed] [Google Scholar]
  • 86.Haase D, Germing U, Schanz J, et al. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood. 2007;110:4385–4395. doi: 10.1182/blood-2007-03-082404. [DOI] [PubMed] [Google Scholar]
  • 87▪.Xu F, Li X, Wu L, Zhang Q, et al. Overexpression of the EZH2, RING1 and BMI1 genes is common in myelodysplastic syndromes: relation to adverse epigenetic alteration and poor prognostic scoring. Ann Hematol. 2010 doi: 10.1007/s00277-010-1128-5. (Epub ahead of print). Provides evidence that overexpression of the EZH2 gene is common in MDS and indicate poor prognosis. [DOI] [PubMed] [Google Scholar]
  • 88▪.Chang CJ, Yang JY, Xia W, et al. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-β-catenin signaling. Cancer Cell. 2011;19:86–100. doi: 10.1016/j.ccr.2010.10.035. Provides evidence about the mechanism of the link between EZH2 and breast cancer progression. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Smith ER, Lee MG, Winter B, et al. Drosophila UTX is a histone H3 Lys27 demethylase that colocalizes with the elongating form of RNA polymerase II. Mol Cell Biol. 2008;28:1041–1046. doi: 10.1128/MCB.01504-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Agger K, Cloos PA, Christensen J, et al. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature. 2007;449:731–734. doi: 10.1038/nature06145. [DOI] [PubMed] [Google Scholar]
  • 91▪.Agherbi H, Gaussmann-Wenger A, Verthuy C. Polycomb mediated epigenetic silencing and replication timing at the INK4a/ARF locus during senescence. PLoS One. 2009;4:e5622. doi: 10.1371/journal.pone.0005622. Provides evidence about the involvement of H3K27 demethylase JMJD3 in the regulation of INK4a/ARF expression. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Wang YY, Cen JN, He J, et al. Accelerated cellular senescence in myelodysplastic syndrome. Exp Hematol. 2009;37:1310–1317. doi: 10.1016/j.exphem.2009.09.002. [DOI] [PubMed] [Google Scholar]
  • 93.Rodrigues EF, Santos-Rebouças CB, Gonçalves Pimentel MM, et al. Epigenetic alterations of p15(INK4B) and p16(INK4A) genes in pediatric primary myelodysplastic syndrome. Leuk Lymphoma. 2010;51:1887–1894. doi: 10.3109/10428194.2010.505820. [DOI] [PubMed] [Google Scholar]
  • 94.Tachibana M, Matsumura Y, Fukuda M, Kimura H, Shinkai Y. G9a/GLP complexes independently mediate H3K9 and DNA methylation to silence transcription. EMBO J. 2008;27:2681–2690. doi: 10.1038/emboj.2008.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Voutsadakis IA, Maillard N. Acute myelogenous leukemia with the t(3;12) (q26;p13) translocation: case report and review of the literature. Am J Hematol. 2003;72:135–137. doi: 10.1002/ajh.10248. [DOI] [PubMed] [Google Scholar]
  • 96.Wieser R. The oncogene and developmental regulator EVI1: expression, biochemical properties, and biological functions. Gene. 2007;396:346–357. doi: 10.1016/j.gene.2007.04.012. [DOI] [PubMed] [Google Scholar]
  • 97▪.Spensberger D, Delwel R. A novel interaction between the proto-oncogene Evi1 and histone methyltransferases, SUV39H1 and G9a. FEBS Lett. 2008;582:2761–2767. doi: 10.1016/j.febslet.2008.06.056. Establishs an epigenetic role of EVI1 in cell transformation by interacting with the histone methyltransferase SUV39H1. [DOI] [PubMed] [Google Scholar]
  • 98.Lakshmikuttyamma A, Scott SA, DeCoteau JF, Geyer CR. Reexpression of epigenetically silenced AML tumor suppressor genes by SUV39H1 inhibition. Oncogene. 2010;29:576–588. 2. doi: 10.1038/onc.2009.361. [DOI] [PubMed] [Google Scholar]
  • 99.Aggerholm A, Holm MS, Guldberg P, Olesen LH, Hokland P. Promoter hypermethylation of p15INK4B, HIC1, CDH1, and ER is frequent in myelodysplastic syndrome and predicts poor prognosis in early-stage patients. Eur J Haematol. 2006;76:23–32. doi: 10.1111/j.1600-0609.2005.00559.x. [DOI] [PubMed] [Google Scholar]
  • 100.Grövdal M, Khan R, Aggerholm A, et al. Negative effect of DNA hypermethylation on the outcome of intensive chemotherapy in older patients with high-risk myelodysplastic syndromes and acute myeloid leukemia following myelodysplastic syndrome. Clin Cancer Res. 2007;13:7107–7112. doi: 10.1158/1078-0432.CCR-07-1193. [DOI] [PubMed] [Google Scholar]
  • 101▪.Botuyan MV, Lee J, Ward IM, et al. Structural basis for the methylation state-specific recognition of histone H4–K20 by 53BP1 and Crb2 in DNA repair. Cell. 2006;127:1361–1373. doi: 10.1016/j.cell.2006.10.043. Provides evidence about the involvement of H4K30 methylation in DNA repair. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Schotta G, Lachner M, Sarma K, et al. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 2004;18:1251–1262. doi: 10.1101/gad.300704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Fang J, Feng Q, Ketel CS, et al. Purification and functional characterization of SET8, a nucleosomal histone H4-lysine 20-specific methyltransferase. Curr Biol. 2002;12:1086–1099. doi: 10.1016/s0960-9822(02)00924-7. [DOI] [PubMed] [Google Scholar]
  • 104.Liu W, Tanasa B, Tyurina OV, et al. PHF8 mediates histone H4 lysine 20 demethylation events involved in cell cycle progression. Nature. 2010;466:508–512. doi: 10.1038/nature09272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Feng W, Yonezawa M, Ye J, Jenuwein T, Grummt I. PHF8 activates transcription of rRNA genes through H3K4me3 binding and H3K9me1/2 demethylation. Nat Struct Mol Biol. 2010;17:445–450. doi: 10.1038/nsmb.1778. [DOI] [PubMed] [Google Scholar]
  • 106▪.Wu MY, Tsai TF, Beaudet AL. Deficiency of Rbbp1/Arid4a and Rbbp1l1/Arid4b alters epigenetic modifications and suppresses an imprinting defect in the PWS/AS domain. Genes Dev. 2006;20:2859–2870. doi: 10.1101/gad.1452206. Provides evidence that altered H4K20 methylation is potentially involved in inefficient hematopoiesis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Pal S, Sif S. Interplay between chromatin remodelers and protein arginine methyltransferases. J Cell Physiol. 2007;213:306–315. doi: 10.1002/jcp.21180. [DOI] [PubMed] [Google Scholar]
  • 108▪.Cheung N, Chan LC, Thompson A, Cleary ML, So CW. Protein arginine-methyltransferase-dependent oncogenesis. Nat Cell Biol. 2007;9:1208–1215. doi: 10.1038/ncb1642. For the first time uncovers an essential function of PRMTs in oncogenesis and reveals their potential as novel therapeutic targets in human cancer. [DOI] [PubMed] [Google Scholar]
  • 109▪.Guccione E, Bassi C, Casadio F, et al. Methylation of histone H3R2 by PRMT6 and H3K4 by an MLL complex are mutually exclusive. Nature. 2007;449:933–937. doi: 10.1038/nature06166. Highlights the interaction between regulators of histone lysine and arginine methylation and their potential role in the regulation of transcription. [DOI] [PubMed] [Google Scholar]
  • 110.Hyllus D, Stein C, Schnabel K, et al. PRMT6-mediated methylation of R2 in histone H3 antagonizes H3 K4 trimethylation. Genes Dev. 2007;21:3369–3380. doi: 10.1101/gad.447007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhao X, Jankovic V, Gural A, et al. Methylation of RUNX1 by PRMT1 abrogates SIN3A binding and potentiates its transcriptional activity. Genes Dev. 2008;22:640–653. doi: 10.1101/gad.1632608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Chang B, Chen Y, Zhao Y, Bruick RK. JMJD6 is a histone arginine demethylase. Science. 2007;318:444–47. doi: 10.1126/science.1145801. [DOI] [PubMed] [Google Scholar]
  • 113.Loenen WA. S-adenosylmethionine: jack of all trades and master of everything? Biochem Soc Trans. 2006;34:330–333. doi: 10.1042/BST20060330. [DOI] [PubMed] [Google Scholar]
  • 114.Tan J, Yang X, Zhuang L, et al. Pharmacologic disruption of polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 2007;21:1050–1063. doi: 10.1101/gad.1524107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115▪.Fiskus W, Wang Y, Sreekumar A, et al. Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood. 2009;114:2733–2743. doi: 10.1182/blood-2009-03-213496. Indicates that the combination of EZH2 inhibitor DZNep and panobinostat is a potential effective epigenetic therapy against myeloid malignant cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.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;100:11606–11611. doi: 10.1073/pnas.1933744100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Lakshmikuttyamma A, Scott SA, DeCoteau JF, Geyer CR. Reexpression of epigenetically silenced AML tumor suppressor genes by SUV39H1 inhibition. Oncogene. 2010;29:576–588. doi: 10.1038/onc.2009.361. [DOI] [PubMed] [Google Scholar]
  • 118▪.Kubicek S, O’Sullivan RJ, August EM, et al. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Mol Cell. 2007;25:473–481. doi: 10.1016/j.molcel.2007.01.017. Highlights the potential of H3K9 methyltransferases as therapeutic targets for reactivating expression of hypermethylated genes. [DOI] [PubMed] [Google Scholar]
  • 119.Castellano S, Milite C, Ragno R, et al. Design, synthesis and biological evaluation of carboxy analogues of arginine methyltransferase inhibitor 1 (AMI-1) Chem Med Chem. 2010;5:398–414. doi: 10.1002/cmdc.200900459. [DOI] [PubMed] [Google Scholar]
  • 120.Spannhoff A, Machmur R, Heinke R, et al. A novel arginine methyltransferase inhibitor with cellular activity. Bioorg Med Chem Lett. 2007;17:4150–4153. doi: 10.1016/j.bmcl.2007.05.088. [DOI] [PubMed] [Google Scholar]
  • 121.Coward WR, Watts K, Feghali-Bostwick CA, Jenkins G, Pang L. Repression of IP-10 by interactions between histone deacetylation and hypermethylation in idiopathic pulmonary fibrosis. Mol Cell Biol. 2010;30:2874–2886. doi: 10.1128/MCB.01527-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Spannhoff A, Hauser AT, Heinke R, Sippl W, Jung M. The emerging therapeutic potential of histone methyltransferase and demethylase inhibitors. Chem Med Chem. 2009;4:1568–1582. doi: 10.1002/cmdc.200900301. [DOI] [PubMed] [Google Scholar]
  • 123.Binda C, Valente S, Romanenghi M, et al. Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2. J Am Chem Soc. 2010;132:6827–6833. doi: 10.1021/ja101557k. [DOI] [PubMed] [Google Scholar]
  • 124.Mackeen MM, Kramer HB, Chang KH, et al. Small-molecule-based inhibition of histone demethylation in cells assessed by quantitative mass spectrometry. J Proteome Res. 2010;9:4082–4092. doi: 10.1021/pr100269b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hamada S, Suzuki T, Mino K, et al. Design, synthesis, enzyme-inhibitory activity, and effect on human cancer cells of a novel series of jumonji domain-containing protein 2 histone demethylase inhibitors. J Med Chem. 2010;53:5629–5638. doi: 10.1021/jm1003655. [DOI] [PubMed] [Google Scholar]

RESOURCES