Abstract
DOT1L is a unique histone methyltransferase that targets the histone H3 lysine 79 (H3K79) residue for mono-, di- and tri- methylation. Histone H3K79 mono- and di-methylation results in active gene transcription, while H3K79 tri-methylation is associated with gene repression. DOT1L has a critical role in regulating gene transcription, development, cell cycle progression, somatic reprogramming and DNA damage repair. DOT1L interacts with Mixed Lineage Leukemia (MLL) fusion proteins, leading to enhanced H3K79 methylation, maintenance of open chromatin, overexpression of downstream oncogenes and leukemogenesis. Importantly, small molecule DOT1L inhibitors have been recently developed, and one of the DOT1L inhibitors is already under investigation in a Phase I clinical trial in patients with MLL fusion gene-driven leukemia.
Keywords: DOT1L, histone methylation, mixed lineage leukemia, gene transcription, DOT1L inhibitors
Introduction
Post-translational histone modifications have become a focus of research due to their ability to regulate gene transcription by modifying chromatin structure. Histone modifications can interact and crosstalk, forming a complex web of gene regulation called the histone code [1]. The four well-known histone modifications are acetylation [2], methylation [3], phosphorylation [4] and ubiquitination [5].
Histone methylation was the first post-translational histone modification identified by radio-labelling cell extracts [6]. It involves the attachment of a methyl group to a basic amino residue: lysine (K) [7] or arginine (R) [8]. Lysine residue methylation is the best studied, with lysine undergoing mono, di-, or tri-methylation on the -amine group.
Methyl groups are generally thought to have slowest turnover rate out of the four common histone modifications. However, methylation marks on different lysine residues have been shown to have differing turnover rates [9]. Mass spectrometry has identified many lysine residues in core histone proteins to be dynamically methylated and de-methylated [10]. The most comprehensively studied lysine methylation sites are located on the N-terminal tail (H3K4, H3K9, H3K27, H3K36 and H4K20) and in the histone core (H3K79) [11,12].
Cross talk among different histone lysine residue methylation modulates gene transcription with H3K9 methylation overlapping with H3K4 de-methylation in regions of heterochromatin; and euchromatic regions show the opposite with H3K4 methylated and H3K9 demethylated [13].
Histone H3K4 mono-methylation (H3K4me), di-methylation (H3K4me2), tri-methylation (H3K4me3), H3K36me3, H3K79me, H3K79me2, H3K9me and H3K27me are linked to gene transcription [14-17]. Differing levels of methylation at the same histone position has been shown to have different effects with H3K9me2, H3K9me3, H3K27me2, H3K27me3 and H4K20me linked to gene repression [18-20].
Histone methyltransferases and histone demethylases
Histone methyltransferases target either lysine or arginine residues, with the majority belonging to the SET domain methyltransferase protein superfamily [21-24]. SET domain methytransferases function by transferring a methyl group from S-adenosyl-L-methionine (SAM) to the amino group of a lysine residue on the histone or non-histone protein, leaving a methylated lysine residue and S-adeno-L-homocysteine (SAH) as a by-product [21]. Further methyl groups are added progressively to achieve di- and tri- methylation.
Many histone methyltransferases have been shown to be involved in cancer and neurological diseases [25,26]. One of the most well studied is Enhancer of Zest Homologue 2 (EZH2), a histone lysine methyltransferase belonging to the the Polycomb group (PcG) protein family. EZH2 is the active catalystic subunit of the Polycomb Repressive Complex 2 (PRC2), targeting histone H3 lysine 27 for mono, di and tri-methylation [19]. PRC2 is involved in a range of normal cellular processes, including cellular differentiation and stem-cell plasticity. Upregulation of EZH2 is a marker for aggressive prostate and breast cancers [27,28]. Recurrent gain of function mutations have been identified at the Y641, A677 and A687 residues within the EZH2 catalytic domain [29,30]. These mutations alter the substrate specificity of EZH2, increasing the conversion of H3K27 di-methylation to tri-methylation, while wild type EZH2 preferentially converts H3K79 mono-methylation to di-methylation. EZH2 gain of function mutations have been found in follicular lymphoma and Germinal Centre B-Cell like Diffuse Large B-Cell lymphoma [31]. A range of small molecular EZH2 inhibitors have been synthesized and shown good in vitro and in vivo efficacy against lymphoma cells [32-34].
Histone methylation was originally believed to be irreversible until the discovery of Lysine Specific Demethylase 1 (LSD1), also known as KDM1A [8]. Since 2004, a total of 15 lysine demethylases have been discovered [35], and have been separated into 2 families: the LSD family consisting of the amine-oxidase related enzymes LSD1 and LSD2, and the Jumonji C-terminal (JMJC) domain containing family [36,37].
LSD1 converts mono- and di-methylated H3K4 into unmethylated H3K4 [38]. The catalytic mechanism of LSD family demethylases requires a lone electron pair on the lysine ε-nitrogen atom, meaning it cannot demethylate tri-methylated lysines [39]. LSD1 has been shown to require the removal of acetylated lysine residues on histone 3 before H3K4me2 demethylation can efficiently occur, due to LSD1 being a part of a complex that includes histone deacetylases [40,41].
The JMJC protein domain has been found in 31 human proteins with 17 of these demonstrating demethylase activity [42]. The enzymatic mechanism of JMJC demethylases involves two cofactors, Fe(II) and 2-oxoglutarate binding to the JMJC domain and reacting with dioxygen to form an active oxoferryl intermediate that hydroxylates the ζ-methyl groups of the methylated lysine substrate [43]. This results in an unstable lysyl hemiaminal that breaks down to release methyl groups from nitrogen. This mechanism allows the mono-, di- and tri-methylation of lysine. Currently there are no known histone lysine demethylases that target H4K20 and H3K79 methyl marks.
The histone H3K79 methylatransferase: Disruptor of telomeric silencing 1-like (DOT1L)
Disruptor of telomeric silencing 1 (DOT1) was first identified through a genetic screen for proteins whose over-expression would lead to impaired telomeric silencing in yeast [44]. The DOT1 homolog gene, DOT1-like (DOT1L), has been found in a range of species, including drosophila [45], protozoa [46] and mammals [47] with mouse and human versions of DOT1L sharing an 88% similarity at the amino acid level [48,49].
DOT1L is the only known histone methyltransferase that targets the histone H3 lysine 79 (H3K79) position, located on the nucleosome surface instead of the N-terminal tail where epigenetic modifications normally occur [48,49]. It adds methyl groups in a non-progressive manner, requiring DOT1L to dissociate and reassociate to H3K79 as it adds methyl groups to generate mono-methylation (H3K79me), di-methylation (H3K79me2) and tri-methylation (H3K79me3) (Figure 1A).
Figure 1.

Chemical structures of DOT1L inhibitors. A. DOT1L catalyses histone H3K79 methylation by transferring a methyl group from its substrate S-adenosyl-L-methionine (SAM) to the amino group of a lysine residue on the histone. A methylated H3K79 residue and S-adeno-L-homocysteine (SAH) are produced, and DOT1L then dissociates. Additional methyl groups are added in a sequential and similar manner. B. Small molecular DOT1L inhibitors: EPZ004777, EPZ5676, SGC0946 and SYC-522. All are based on SAH backbone and target the SAM binding pocket of DOT1L.
Instead of a SET domain, DOT1L has an AdoMet binding motif similar to arginine and DNA methyltransferases [50]. It is currently the only known non-SET histone methyltransferase protein [48,49]. This makes DOT1L a key target for specific therapeutic treatments, with several small molecular inhibitors developed and one currently in clinical trials [51-53] (Figure 1B).
Study of the crystal structure of DOT1L has shown that the AdoMet binding pocket must be near a lysine binding channel and the C-terminus of the catalytic domain in order for nucleosome binding and enzymatic activity to occur [48]. This active site of DOT1L closely resembles catechol-O-methyltransferases and L-isoaspartyl methyltransferases, which are highly conserved in eukaryotic organisms [48].
Regulatory functions of DOT1L in gene transcription, somatic reprogramming, cell cycle regulation and development
The distribution of all three forms of H3K79 methylation on human histones has been studied using mass spectrometry, demonstrating that H3K79me is the most abundant and correlates with the fraction of histone H3 modified by acetylation [54]. This suggests H3K79 methylation enrichment at active gene transcription sites. Further studies focusing on individual genes in mammalian cells have correlated H3K79me2 with transcriptional activation [55,56]. Subsequent quantitative-chromatin immunoprecipitation (q-ChIP) studies of H3K79 methylation across the human genome reveal that H3K79me3 is present at higher levels in silent gene regions in comparison to active regions, linking it to gene repression [17]. This demonstrates the complex regulatory effects of DOT1L on a wide range of functions in eukaryotic organisms, with H3K79me and H3K79me2 associated with active gene transcription and H3K79me3 with gene repression in human cells.
DOT1L inhibition may enhance reprogramming in a broad range of cell types by facilitating the silencing of lineage-specific programs of gene expression. Inhibition of DOT1L by shRNA or small molecule inhibitors accelerates reprogramming, significantly increasing the yield of pluripotent stem cell colonies, and substitutes for KLF4 and c-Myc [57]. Genome wide analysis of H3K79me2 distribution reveals that DOT1L inhibition accelerates reprogramming due to loss of H3K79me2 among genes fated to be repressed in the pluripotent state [57,58].
Methylation of H3K79, mediated by DOT1 and DOT1L, has been implicated in transcriptional elongation and cell cycle regulation. Nearly 90% of histone H3 in Saccharomyces cerevisiae (S. cerevisiae) bears H3K79me, H3K79me2 or H3K79me3. The level of H3K79me2 fluctuates between different stages of the cell cycle in S. cerevisiae, with a low level of H3K79me2 at the G1 phase, gradually increasing at the S phase and peaking at the G2/M phase, while H3K79me3 remains constant throughout the cell cycle [59]. S. cerevisiae mutants arrested at G1 and G2/M phases showed increased H3K79me3 levels, demonstrating that H3K79 methylation levels increase progressively over time in arrested cells [60].
H3K79me2 associates with gene promoter and coding regions during transcriptional activation of hepatic genes in G0/G1 enriched human liver carcinoma cells [61]. The mechanism responsible for targeted DOT1L binding and consequent H3K79 methylation of actively transcribed genes involves DOT1L binding to the phosphorylated C-terminal domain of actively transcribing RNA polymerase II (RNAP II) [62]. Similarly human liver carcinoma cells arrested at the G2/M phases have increased H3K79 methylation levels compared to G0/G1 enriched cells, showing that this methylation mark is generated independently of gene transcription at the S phase [61].
H3K79 has been shown to control sexual differentiation in silk worms (Bombyx mori), with higher H3K79me2 levels on the insulin-like growth factor II mRNA-binding protein (Imp) gene promoter in males than females [63]. RNAi-mediated depletion of DOT1L results in a total abolishment of male-specific Imp, showing H3K79 methylation regulates sex-specific splicing of Imp mRNA [63].
H3K79 methylation levels in Drosophila correlate to transcriptional activity [55]. The Drosophila ortholog of DOT1, grappa (gpp), has been identified as a dominant suppressor of pair-dependant silencing, necessary for the maintenance phase of Bithorax complex expression [64]. H3K79 methylation expression during embryogenesis is conserved in Drosophila, mouse, rat and human spermatids and also independent of H3K4 and H3K9 methylation [65]. During chromatin reorganisation in spermatids, H3K79 methylation is accompanied by H4 hyperacetylation and may be a prerequisite for proper histone to protamine transition [66].
DOT1L serves further developmental roles in a wide range of species. It is directly regulated during tadpole development in the model system Xenopus tropicalis by thyroid hormone receptor, which binds to a thyroid hormone response element in the DOT1L gene promoter region [67]. Embryos treated with DOT1L specific transcription activator-like effector nuclease show low H3K79 methylation and experience growth difficulties as tadpoles, ultimately leading to tadpole mortality [68].
DOT1L has been shown as a crucial regulator of early mammalian haematopoiesis by regulating the steady state levels of GATA2, a growth factor essential for early erythropoiesis [69], and PU.1, a transcription factor that inhibits erythropoiesis and promotes myelopoiesis [70]. DOT1L knockout mice display reduced GATA2 and increased PU.1 levels and early embryonic death due to anaemia, indicating that DOT1L is essential for embryonic development and prenatal haematopoiesis [71]. Conditional DOT1L targeting strategies show DOT1L also playing a role in adult haematopoiesis maintenance, with DOT1L deletion in mice resulting in pancytopenia and failure of hematopoietic homeostasis [72,73]. Taken together, these findings demonstrate that DOT1L plays a critical regulatory role in gene transcription, somatic reprogramming, cell cycle, development and haematopoiesis, via H3K79 methylation.
The role of DOT1L in DNA damage response
DOT1 was identified in a screen of radiation sensitive yeast mutants for DNA damage checkpoint defects, with DOT1 yeast mutants exposed to ionizing radiation becoming defective at the G1 and intra-S phase checkpoint [74]. Checkpoint mediated arrest at the pachytene stage in dmc1 and zip1 S. cerevisiae mutants was also shown to be DOT1-dependent, with loss of DOT1 leading to continued meiosis and the generation of unviable cells [75].
In the human osteosarcoma cell line U2OS, in vitro experiments established that the double-stranded DNA break repair protein 53BP1 bound most efficiently to H3K20me2 [76]. This was confirmed using a stable DOT1L knockdown model system, which showed that H3K79 methylation was not critical for 53BP1 recruitment to DNA damage sites [77]. However, H3K79me2 was shown to be required in an alternate pathway of 53BP1 recruitment in response to DNA damage during the G1 and G2/M cell cycle phases, when H3K20me2 levels dropped [78]. In addition, suppression of DOT1L inhibited recruitment of 53BP1 to sites of DNA damage in 293T cells [79]. Thus both H3K79 and H3K20 methylation are capable of 53BP1 recruitment in response to DNA damage repair, with each methylation covering different stages of the cell cycle.
In mouse embryonic fibroblast cells treated with UV, DOT1L reduction leads to UV hypersensitivity and reduced recovery of transcription initiation. DOT1L was found to promote an open chromatin structure to reactivate RNA Pol II-mediated transcription after DNA damage and was not involved in the nuclear excision repair pathway [80]. In addition, DOT1 was also shown to be required for cell cycle arrest in response to double stranded DNA breaks [75]. Thus, DOT1L-mediated H3K79 methylation may play a critical role in DNA damage signalling.
DOT1L and mixed lineage leukaemia
In humans, DOT1L is involved in the oncogenesis of several leukaemia subtypes, mostly characterised by chromosomal translocations involving the mixed lineage leukaemia (MLL) gene. MLL chromosomal translocations involving the cytogenetic band 11q23 produce a wide array of fusion proteins associated with Acute Lymophoblastic Leukaemia (ALL), Acute Myeloid Leukaemia (AML) and Mixed Lineage Leukaemia [81-83].
DOT1L has been found in the Elongation Assisting Proteins (EAP) complex along with RNA PII, factors AF4, AF6, AF9, AF10 or ENL [84-87], all known to be involved in chromosomal translocation-induced fusion with the MLL protein. DOT1L and AF10 have been isolated from yeast and mammalian two-hybrid assays as binding partners of ENL, which is a MLL fusion partner with transactivation abilities and known to associate with AF4 [88,89]. AF4 stimulates kinase elongation by P-TEFb and interacts with AF9 and AF10 to recruit DOT1L to the Pol II elongation complex at ectopic loci, resulting in aberrant gene expression and MLL leukaemogenesis [84,90]. The MLL-AF6 fusion protein requires H3K79 methylation for the maintenance of MLL-AF6 target oncogenic gene expression, with gene expression analysis and ChIP-sequencing finding high levels of H3K79me2 at MLL target gene promoters [87]. Abnormally high levels of H3K79 methylation on MLL target gene promoters in MLL leukaemia cells are indicative of aberrant DOT1L activity in these leukaemia cells due to MLL fusion oncoproteins recruiting DOT1L, causing overexpression of MLL target genes [91]. This has further been shown with inducible expression of the MLL-ENL fusion gene activating H3K79me2 on MLL target gene promoters, while inhibiting DOT1L binding leads to the MLL-ENL fusion gene losing its transforming ability [89].
A notable family of genes dis-regulated in leukaemia is the homeobox (HOX) gene family, which is highly expressed in multipotent haematopoietic stem cells (HSCs) but down-regulated once the HSCs have become differentiated [92,93]. Epigenetic regulation of HOX loci is modulated by the MLL protein which introduces the H3K4me3 histone mark, resulting in HOX transcriptional activation [94]. DOT1L-AF10 interaction activates HOXA9 gene transcription and plays an important part in MLL-AF10-mediated leukaemogenesis, with AF10 being a DOT1L cofactor [95]. DOT1L also contributes to clathrin assembly lymphoid myeloid leukaemia protein (CALM)-AF10-mediated leukaemic transformation by preventing nuclear export of CALM-AF10 and by up-regulating HOXA5 gene expression through H3K79 methylation [96]. Consequently inhibiting DOT1L results in suppression of MLL-AF10 and CALM–AF10 mediated transformation by down-regulating leukaemogenic genes such as HOXA and Meis1 [97].
H3K79 methylation by DOT1L is also crucial for the expression of critical MLL-AF4 target oncogenes such as HOXA in human MLL-AF4 leukaemia cells [85]. Experiments with conditional DOT1L knockout mice have found that H3K79me2 drives MLL-AF9 fusion gene-mediated leukaemogenesis, and DOT1L is required for up-regulation of HOXA and Meis1 gene expression and consequent initiation and maintenance of MLL-AF9-induced leukaemogenesis [86]. However, HOXA9/Meis1 and E2A-HLF MLL cell lines do not require DOT1L for oncogenic transformation [72], illustrating a crucial role for DOT1L in driving leukaemogenesis with specific MLL translocation oncogenes.
The mechanisms underlying DOT1L interaction with MLL-fusion proteins are still unclear, but studies have focused on identifying DOT1L protein binding sites. A 10 amino acid region of human DOT1L (865-874) has been identified as the AF9/ENL binding site, with four conserved hydrophobic residues within the binding site being shown to be essential for interactions with the C-terminal binding domain of AF9/ENL [98]. DOT1L binding to MLL-AF9 has been shown at three sites by nuclear magnetic resonance. Structure-guided point mutations of these binding sites lead to a graded reduction in DOT1L recruitment to MLL-AF9, with differential loss of H3K79me2 and H3K79me3 at MLL-AF9 target genes [99].
The YEATS domain of AF9 recognises H3K9 acetylation, providing a link between elevated H3K9 acetylation and DOT1L recruitment to target gene promoters [100]. Furthermore DOT1L inhibits chromatin localization of a repressive complex composed of the histone H3 deacetylase SIRT1 and the H3K9 methyltransferase SUV39H1, thus maintaining an open chromatin state with elevated H3K9 acetylation and H3K79 methylation and minimal H3K9 methylation at MLL fusion target gene promoters (Figure 2) [101].
Figure 2.

Model of possible MLL-AF9 and DOT1L-mediated gene transcription mechanism in MLL-driven leukaemia. A. SUV39H1 and SIRT1 increase H3K9 methylation and decrease H3K9 acetylation, preventing MLL-AF9 and Elongation Assisting Proteins (EAP) complex binding to gene promoters. B. DOT1L inhibits SIRT1 and SUV39H1 chromatin localization, thereby maintaining an open chromatin state with elevated H3K9 acetylation and H3K79 methylation and minimal H3K9 methylation at MLL fusion protein target gene promoters. MLL-AF9 forms a protein complex with DOT1L, recognises elevated H3K9 acetylation via its YEATS domain, and recruits the EAP complex containing RNA Pol II and other transcription factors at MLL fusion protein target gene promoters, leading to transcriptional activation.
DOT1L in other cancers
The complex regulatory role of DOT1L in MLL-driven leukaemia and physiologically normal eukaryotic organisms has led to interest in its possible function in various other cancer types. RNAi-mediated depletion of DOT1L in A549 and NCI-H1299 lung cancer cells resulted in chromosomal mis-segregation, cell-cycle arrest at the G1 phase and senescence [102]. Overexpression of DOT1L reversed these RNAi-mediated phenotype changes in lung cancer cells.
DOT1L increased the tumorigenic potential of colorectal cancer cells by inducing NANOG, SOX2 and Pou5F1 gene expression [103]. High DOT1L gene expression and consequently increased H3K79me2 levels are predictors of poor patient survival [103], indicating DOT1L having a tumorigenic role in colorectal cancer.
DOT1L has also been recently linked to breast cancer with a study of a genomic database of over 1000 patient samples showing that higher levels of DOT1L expression correlated with breast cancer compared to normal breast tissues [104]. High DOT1L levels in breast cancer tissues correlated with approximately 20 pro-proliferative genes from the PAM50 gene set [104]. DOT1L interaction with c-Myc-p300, has been suggested to be critical for promoting EMT/CSC leading to an aggressive phenotype in breast cancer [105].
Chromatin immunoprecipitation assays have linked H3K4 and H3K79 methylation and H3 acetylation to the ability of the c-Myc transcription factor to recognise and bind to target gene promoters [106]. H3K4me2, H3K4me3 and H3K79me2 are generally associated with transcription machinery and these methylation marks precede and are independent of c-Myc binding at target gene promoters [107]. In contrast, H3K79 methylation has been shown to be non-essential for the maintenance or activation of Wnt pathway target gene expression in human colon adenocarcinoma cell lines, with H3K79 methylation showing no elevation in comparison to normal colon tissue [108].
Thus H3K79 methylation is a critical histone modification in a range of cancer types. The dynamic interplays among different chromatin post-transcription modifications control gene expression, and provide novel opportunities for targeted combination therapies in multiple cancer types.
DOT1L inhibitors
Molecular targeting of specific histone methyltransferase is emerging as a new direction for cancer therapy. Of the known histone methyltransferase enzymes, DOT1L is a promising target due to it being the only known H3K79 histone methyltransferase, its unique non-SET catalytic domain and its role in promoting and maintaining MLL leukaemogenesis [48,49].
The first DOT1L specific small molecular inhibitor was EPZ004777, designed by Epizyme using a traditional ligand-based approach based on the DOT1L substrate SAM and the product SAH [51]. EPZ004777 displayed specificity against DOT1L with little reactivity against a panel of eight other histone methyltransferases. Modifications to EPZ00477 lead to the synthesis of EPZ5676 [52] and SGC0946 (Figure 1B) [109].
SGC0946 features an additional bromine atom at position 7 targeting a hydrophobic cleft present in DOT1L to improve the DOT1L inhibitor’s binding affinity [109]. Another novel DOT1L inhibitor, SYC-522, was synthesized based on the structure of SAH with additional urea group, and showed specificity for DOT1L when tested against three representative histone methytransferases: PRMT1, PRMT4 and SUV39H1 [110]. These four DOT1L specific small molecular inhibitors are designed to occupy the SAM binding pocket, inducing DOT1L conformational changes and leading to the opening of a hydrophobic pocket outside of the SAM binding domain [52].
Anticancer efficacy of DOT1L inhibitors
The anticancer efficacy of allosteric DOT1L inhibitors has been tested in vitro and in vivo. The first reported DOT1L inhibitor, EPZ004777, demonstrated an IC50 of 400 ± 100 pM to inhibit DOT1L enzymatic activity in a biochemical radionucleotide assay [51]. Treatment with EPZ004777 caused apoptosis in MLL-rearranged leukaemia cells in vitro and blocked leukaemia progression in mice by suppressing the expression of HOXA cluster genes and Meis1 [51]. MLL-AF6 transformed mouse bone marrow cells also demonstrated a dose-dependent reduction in H3K79me2 and a reduction in cell number when treated with 10 µM EPZ004777 for 10 days [87]. DOT1L inhibition reduced the number of MLL-AF6 transformed cells at the S-phase, and increased apoptotic cell death [87]. In vivo administration of EPZ004777 by subcutaneously implanted osmotic pumps over 14 days resulted in a dose-dependent increase in the survival of mice xenografted with MV4-11 leukemia cells [51]. Pre-treatment of mice with EPZ004777 also decreased the in vivo spleen-colony-forming ability of MLL-AF10/CALM-AF10 transformed bone marrow cells [51].
EPZ5676 demonstrated a superior enzyme inhibition Ki value of ≤ 0.08 Nm compared to EPZ004777. EPZ5676 had an IC50 of 3 nM and 5 nM in MV4-11 and HL60 leukaemia cells, respectively [52]. It demonstrated synergistic anticancer effects when used in combination with cytarabine and daunorubicin to treat MOLM-13 and MV4-11 MLL-rearrangement leukaemia cells lines [111]. Continuous intravenous treatment of rats xenografted with MV4-11 cells with EPZ5676 for 21 days, leaded to dose-dependent leukemia regression with a 70 mg/kg dose leading to complete regression [52].
SGC0946 showed an IC50 of 8.8 ± 1.6 nM in reducing H3K79 methylation in cells, representing a significant improvement over EPZ004777, which exhibited an IC50 of 84 ± 20 nM [109]. Treatment of MLL-rearranged MV4-11 and THP1 leukemia cells with the DOT1L inhibitor SYC-522 led to cell cycle arrest and cell differentiation, and treatment of primary MLL-rearranged AML cells resulted in up to 50% decrease in colony formation and promotion of monocytic differentiation [110]. SYC-522 was also tested in combination with existing chemotherapeutics: mitoxantrone, etoposide or cytarabine. Pretreatment with SYC-522 sensitized primary MLL-rearranged leukaemia cells to treatment with all three chemotherapy agents [112]. In addition, combination therapy with the SIRT1 activator SRT1720 and the DOT1L inhibitor EPZ004777 demonstrated enhanced anti-proliferative activity against MLL-rearranged leukemia cells by supressing HOXA7 and Meis1 in vivo [101]. DOT1L inhibitors have also demonstrated anticancer effects against solid tumor cells. SYC-522 and EPZ004777 induced differentiation and inhibited proliferation, self-renewal and metastatic potential in a range of DOT1L overexpressing breast cancer cells [104].
Currently all reported DOT1L inhibitors have the common substructure of adenosine, making them competitive to the DOT1L enzyme substrate SAM and resulting in overall poor pharmacokinetic properties [51,53]. This indicates that improvements in the metabolic stability of DOT1L inhibitors are required before their use for human patient treatment.
Conclusions
Epigenetic modifications in cells play a major role in regulating the expression of genes important for development, haematopoiesis and cancer. Histone H3K79 methylation mediated solely by DOT1L is involved in somatic reprogramming, cell cycle progression, DNA damage response and tumorigenesis. Aberrant H3K79 methylation is associated with various types of aggressive MLL fusion gene-driven leukaemia. P-TEFb, ENL, AF4, AF6 AF9 and AF10 proteins have all been shown to be involved in the recruitment of DOT1L to MLL target gene promoters. H3K79me2 has been shown to be critical to MLL target gene expression via maintenance of open chromatin states around MLL target genes, with loss of H3K79me2 leading to loss of tumorigenicity in MLL-driven leukaemia. In addition, DOT1L dis-regulation has been linked to poor patient prognosis in lung, colorectal and breast cancers. These observations raise the possibility of DOT1L playing a major role in other forms of cancer and will be a subject for future inquiry.
The unique structural features of DOT1L have made it an emerging drug target with multiple allosteric small molecular inhibitors, based on the SAH structure showing selective inhibitory effects and low IC50 concentrations in vitro. Further chemical modifications to the first small molecular DOT1L inhibitor, EPZ004777, have led to the generation of EPZ5676 with improvements in metabolic stability and anticancer efficacy. These small molecular DOT1L inhibitors have shown synergy when used in combination with existing chemotherapeutics. Importantly, EPZ5676 is currently in Phase 1 clinical trials in leukemia patients (NCT02141828).
A limiting factor in DOT1L inhibitor discovery has been the complex biochemical assays necessary to determine loss of H3K79 methylation. Two new assays involving nanoparticle proximity and fluorescence polarisation respectively have been created for future DOT1L inhibitor screening and lead optimisation [113].
An alternative strategy for targeting DOT1L in MLL would be to target the DOT1L binding sites of the MLL-fusion proteins [98]. Targeting these MLL-AF9 and AF9/ENL binding sites in DOT1L could potentially result in fewer adverse effects than the conventional approach of targeting DOT1L enzymatic activity. Inhibitors targeting these binding sites could also have improved pharmacokinetic properties compared to existing DOT1L inhibitors based on SAH. Thus future DOT1L inhibitors have the potential to deliver better patient outcomes in treating MLL leukemia and potentially other forms of cancers where H3K79 methylation dis-regulation is present.
Acknowledgements
The authors were supported by National Health & Medical Research Council (NHMRC) and Cancer Council New South Wales grants. T.L. is a recipient of an Australian Research Council Future Fellowship. Children’s Cancer Institute Australia is affiliated with UNSW Australia and Sydney Children’s Hospital.
Disclosure of conflict of interest
None.
Abbreviations
- H3K79
histone H3 lysine 79
- MLL
mixed lineage leukemia
- H3K4me
histone H3 lysine 4 mono-methylation
- H3K4me2
histone H3 lysine 4 di-methylation
- H3K4me3
histone H3 lysine 4 tri-methylation
- H3K36me3
histone H3 lysine 36 tri-methylation
- H3K79me
histone H3 lysine 79 mono-methylation
- H3K79me2
histone H3 lysine 79 di-methylation
- H3K79me3
histone H3 lysine 79 tri-methylation
- H3K27me
histone H3 lysine 27 mono-methylation
- SET
(Su(var), Enhancer of zeste, Trithorax)
- SAM
S-adenosyl-L-methionine
- SAH
S-adeno-L-homocysteine
- EZH2
Enhancer of Zest Homologue 2
- LSD1
Lysine Specific Demethylase 1
- q-ChIP
quantitative-chromatin immunoprecipitation
- AdoMet
S-Adenosyl methionine
- ALL
Acute Lymophoblastic Leukaemia
References
- 1.Strahl B, Allis CD. The language of covalent histone modifications. Nature. 2000;403:41–45. doi: 10.1038/47412. [DOI] [PubMed] [Google Scholar]
- 2.Parbin S, Kar S, Shilpi A, Sengupta D, Deb M, Rath SK, Patra SK. Histone deacetylases: a saga of perturbed acetylation homeostasis in cancer. J Histochem Cytochem. 2014;62:11–33. doi: 10.1369/0022155413506582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Leung K, Cheng VW, Mok SW, Tsui SK. The Involvement of DNA Methylation and Histone Modification on the Epigenetic Regulation of Embryonic Stem cells and Induced Pluripotent Stem Cells. Curr Stem Cell Res Ther. 2014;9:388–95. doi: 10.2174/1574888x09666140507154005. [DOI] [PubMed] [Google Scholar]
- 4.Pérez-Cadahía B, Drobic B, Khan P, Shivashankar CC, Davie JR. Current understanding and importance of histone phosphorylation in regulating chromatin biology. Curr Opin Drug Discov Devel. 2010;13:613–622. [PubMed] [Google Scholar]
- 5.Cao J, Yan Q. Histone ubiquitination and deubiquitination in transcription, DNA damage response, and cancer. Front Oncol. 2012;2:26. doi: 10.3389/fonc.2012.00026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Allfrey VG, Faulkner R, Mirsky AE. Aceylation and Methylation of Histones and their possible role in the Regulation of RNA. Proc Natl Acad Sci U S A. 1964;51:786–794. doi: 10.1073/pnas.51.5.786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Murray K. The occurrence of ε-N-methyl lysine in histones. Biochemistry. 1964;3:10–15. doi: 10.1021/bi00889a003. [DOI] [PubMed] [Google Scholar]
- 8.Byvoet P, Shepherd GR, Hardin JM, Noland BJ. The distribution and turnover of labelled methyl groups in histone fractions of cultured mammalian cells. Arch Biochem Biophys. 1972;148:558–567. doi: 10.1016/0003-9861(72)90174-9. [DOI] [PubMed] [Google Scholar]
- 9.Zee B, Levin RS, Xu B, LeRoy G, Wingreen NS, Garcia BA. In vivo residue-specific histone methylation dynamics. J Biol Chem. 2010;285:3341–3350. doi: 10.1074/jbc.M109.063784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Young N, Dimaggio PA, Garcia BA. The significance, development and progress of highthroughput combinatorial histone code analysis. Cell Mol Life Sci. 2010;67:3983–4000. doi: 10.1007/s00018-010-0475-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang YR, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 2001;15:2343–2360. doi: 10.1101/gad.927301. [DOI] [PubMed] [Google Scholar]
- 12.Greer E, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet. 2012;13:343–357. doi: 10.1038/nrg3173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Noma K, Allis CD, Grewal SI. Transitions in distinct histone H3 methylation patterns at the heterochromatin domain boundaries. Science. 2001;293:1150–1155. doi: 10.1126/science.1064150. [DOI] [PubMed] [Google Scholar]
- 14.Bernstein BE, Humphrey EL, Erlich RL, Schneider R, Bouman P, Liu JS, Kouzarides T, Schreiber SL. Methylation of histone H3 Lys4 in coding regions of active genes. Proc Natl Acad Sci U S A. 2002;99:8695–8700. doi: 10.1073/pnas.082249499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bannister A, Schneider R, Myers FA, Thorne AW, Crane-Robinson C, Kouzarides T. Spatial distribution of di- and tri-methyl lysine 36 of histone H3 at active genes. J Biol Chem. 2005;280:17732–17736. doi: 10.1074/jbc.M500796200. [DOI] [PubMed] [Google Scholar]
- 16.Mohan M, Herz HM, Takahashi YH, Lin C, Lai KC, Zhang Y, Washburn MP, Florens L, Shilatifard A. Linking H3K79 trimethylation to Wnt signaling through a novel Dot1-containing complex (DotCom) Genes Dev. 2010;24:574–589. doi: 10.1101/gad.1898410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Barski A, Cuddapah S, Cui K, Roh TY, Schones DE, Wang Z, Wei G, Chepelev I, Zhao K. Highresolution profiling of histone methylations in the human genome. Cell. 2007;129:823–837. doi: 10.1016/j.cell.2007.05.009. [DOI] [PubMed] [Google Scholar]
- 18.Snowden A, Gregory PD, Case CC, Pabo CO. Gene-specific targeting of H3K9 methylation is sufficient for initiating repression in vivo. Curr Biol. 2002;12:2159–2166. doi: 10.1016/s0960-9822(02)01391-x. [DOI] [PubMed] [Google Scholar]
- 19.Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298:1039–43. doi: 10.1126/science.1076997. [DOI] [PubMed] [Google Scholar]
- 20.Nishioka K, Rice JC, Sarma K, Erdjument-Bromage H, Werner J, Wang Y, Chuikov S, Valenzuela P, Tempst P, Steward R, Lis JT, Allis CD, Reinberg D. PR-Set7 is a nucleosome-specific methyltransferase that modifies lysine 20 of histone H4 and is associated with silent chromatin. Mol Cell. 2002;9:1201–1213. doi: 10.1016/s1097-2765(02)00548-8. [DOI] [PubMed] [Google Scholar]
- 21.Dillon S, Zhang X, Trievel RC, Cheng X. The SETdomain protein superfamily: protein lysine methyltransferases. Genome Biol. 2005;6:227. doi: 10.1186/gb-2005-6-8-227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nishioka K, Chuikov S, Sarma K, Erdjument-Bromage H, Allis CD, Tempst P, Reinberg D. Set9, a novel histone H3 methyltransferase that facilitates transcription by precluding histone tail modifications required for heterochromatin formation. Genes Dev. 2002;16:479–489. doi: 10.1101/gad.967202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wang H, Cao R, Xia L, Erdjument-Bromage H, Borchers C, Tempst P, Zhang Y. Purification and functional characterization of a histone H3-lysine 4-specific methyltransferase. Mol Cell. 2001;8:1207–1217. doi: 10.1016/s1097-2765(01)00405-1. [DOI] [PubMed] [Google Scholar]
- 24.Schultz D, Ayyanathan K, Negorev D, Maul GG, Rauscher FJ 3rd. SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zincfinger proteins. Genes Dev. 2002;16:919–932. doi: 10.1101/gad.973302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hua K, Wang MY, Chen MW, Wei LH, Chen CK, Ko CH, Jeng YM, Sung PL, Jan YH, Hsiao M, Kuo ML, Yen ML. The H3K9 methyltransferase G9a is a marker of aggressive ovarian cancer that promotes peritoneal metastasis. Mol Cancer. 2014;13:189. doi: 10.1186/1476-4598-13-189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bae W, Hennighausen L. Canonical and noncanonical roles of the histone methyltransferase EZH2 in mammary development and cancer. Mol Cell Endocrinol. 2014;382:593–597. doi: 10.1016/j.mce.2013.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Xu K, Wu ZJ, Groner AC, He HH, Cai C, Lis RT, Wu X, Stack EC, Loda M, Liu T, Xu H, Cato L, Thornton JE, Gregory RI, Morrissey C, Vessella RL, Montironi R, Magi-Galluzzi C, Kantoff PW, Balk SP, Liu XS, Brown M. EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent. Science. 2012;338:1465–1469. doi: 10.1126/science.1227604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RG, Otte AP, Hayes DF, Sabel MS, Livant D, Weiss SJ, Rubin MA, Chinnaiyan AM. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A. 2003;100:11606–11611. doi: 10.1073/pnas.1933744100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Majer C, Jin L, Scott MP, Knutson SK, Kuntz KW, Keilhack H, Smith JJ, Moyer MP, Richon VM, Copeland RA, Wigle TJ. A687V EZH2 is a gain-of-function mutation found in lymphoma patients. FEBS Lett. 2012;586:3448–3451. doi: 10.1016/j.febslet.2012.07.066. [DOI] [PubMed] [Google Scholar]
- 30.Sneeringer C, Scott MP, Kuntz KW, Knutson SK, Pollock RM, Richon VM, Copeland RA. Coordinated activities of wild-type plus mutant EZH2 drive tumor-associated hypertrimethylation of lysine 27 on histone H3 (H3K27) in human B-cell lymphomas. Proc Natl Acad Sci U S A. 2010;107:20980–20985. doi: 10.1073/pnas.1012525107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wigle T, Knutson SK, Jin L, Kuntz KW, Pollock RM, Richon VM, Copeland RA, Scott MP. The Y641C mutation of EZH2 alters substrate specificity for histone H3 lysine 27 methylation states. FEBS Lett. 2011;585:3011–3014. doi: 10.1016/j.febslet.2011.08.018. [DOI] [PubMed] [Google Scholar]
- 32.Konze K, Ma A, Li F, Barsyte-Lovejoy D, Parton T, Macnevin CJ, Liu F, Gao C, Huang XP, Kuznetsova E, Rougie M, Jiang A, Pattenden SG, Norris JL, James LI, Roth BL, Brown PJ, Frye SV, Arrowsmith CH, Hahn KM, Wang GG, Vedadi M, Jin J. An orally bioavailable chemical probe of the Lysine Methyltransferases EZH2 and EZH1. ACS Chem Biol. 2013;8:1324–1334. doi: 10.1021/cb400133j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.McCabe M, Ott HM, Ganji G, Korenchuk S, Thompson C, Van Aller GS, Liu Y, Graves AP, Della Pietra A 3rd, Diaz E, LaFrance LV, Mellinger M, Duquenne C, Tian X, Kruger RG, McHugh CF, Brandt M, Miller WH, Dhanak D, Verma SK, Tummino PJ, Creasy CL. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature. 2012;492:108–112. doi: 10.1038/nature11606. [DOI] [PubMed] [Google Scholar]
- 34.Knutson S, Wigle TJ, Warholic NM, Sneeringer CJ, Allain CJ, Klaus CR, Sacks JD, Raimondi A, Majer CR, Song J, Scott MP, Jin L, Smith JJ, Olhava EJ, Chesworth R, Moyer MP, Richon VM, Copeland RA, Keilhack H, Pollock RM, Kuntz KW. A Selective Inhibitor of EZH2 Blocks H3K27 Methylation and Kills Mutant Lymphoma Cells. Nature Chem Biol. 2012;8:890–896. doi: 10.1038/nchembio.1084. [DOI] [PubMed] [Google Scholar]
- 35.Højfeldt J, Agger K, Helin K. Histone lysine demethylases as targets for anticancer therapy. Nat Rev Drug Discov. 2013;12:917–930. doi: 10.1038/nrd4154. [DOI] [PubMed] [Google Scholar]
- 36.Allis C, Berger SL, Cote J, Dent S, Jenuwien T, Kouzarides T, Pillus L, Reinberg D, Shi Y, Shiekhattar R, Shilatifard A, Workman J, Zhang Y. New nomenclature for chromatin-modifying enzymes. Cell. 2007;131:633–636. doi: 10.1016/j.cell.2007.10.039. [DOI] [PubMed] [Google Scholar]
- 37.Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Tempst P, Wong J, Zhang Y. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell. 2006;125:483–495. doi: 10.1016/j.cell.2006.03.027. [DOI] [PubMed] [Google Scholar]
- 38.Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA, Shi Y. Histone demethylation mediated by the nuclear amine oxidase homologue LSD1. Cell. 2004;119:941–53. doi: 10.1016/j.cell.2004.12.012. [DOI] [PubMed] [Google Scholar]
- 39.Forneris F, Binda C, Vanoni MA, Mattevi A, Battaglioli E. Histone demethylation catalysed by LSD1 is a flavin-dependent oxidative process. FEBS Lett. 2005;579:2203–2207. doi: 10.1016/j.febslet.2005.03.015. [DOI] [PubMed] [Google Scholar]
- 40.Forneris F, Binda C, Vanoni MA, Battaglioli E, Mattevi A. Human histone demethylase LSD1 reads the histone code. J Biol Chem. 2005;280:41360–41365. doi: 10.1074/jbc.M509549200. [DOI] [PubMed] [Google Scholar]
- 41.Forneris F, Binda C, Dall’Aglio A, Fraaije MW, Battaglioli E, Mattevi A. A highly specific mechanism of histone H3-K4 recognition by histone demethylase LSD1. J Biol Chem. 2006;281:35289–35295. doi: 10.1074/jbc.M607411200. [DOI] [PubMed] [Google Scholar]
- 42.Kooistra S, Helin K. Molecular mechanisms and potential functions of histone demethylases. Nat Rev Mol Cell Biol. 2012;13:297–311. doi: 10.1038/nrm3327. [DOI] [PubMed] [Google Scholar]
- 43.McDonough M, Loenarz C, Chowdhury R, Clifton IJ & Schofield CJ. Structural studies on human 2-oxoglutarate dependent oxygenases. Curr Opin Struc Biol. 2010;20:659–672. doi: 10.1016/j.sbi.2010.08.006. [DOI] [PubMed] [Google Scholar]
- 44.Singer M, Kahana A, Wolf AJ, Meisinger LL, Peterson SE, Goggin C, Mahowald M, Gottschling DE. Identification of high-copy disruptors of telomeric silencing in Saccharomyces cerevisiae. Genetics. 1998;150:613–632. doi: 10.1093/genetics/150.2.613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.List O, Togawa T, Tsuda M, Matsuo T, Elard L, Aigaki T. Overexpression of grappa encoding a histone methyltransferase enhances stress resistance in Drosophila. Hereditas. 2009;146:19–28. doi: 10.1111/j.1601-5223.2008.02080.x. [DOI] [PubMed] [Google Scholar]
- 46.Jansen C, Hake SB, Lowell JE, Cross GA. Selective di- or trimethylation of histone H3 lysine 76 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]
- 47.Jones B, Su H, Bhat A, Lei H, Bajko J, Hevi S, Baltus GA, Kadam S, Zhai H, Valdez R, Gonzalo S, Zhang Y, Li E, Chen T. The histone H3K79 methyltransferase Dot1L is essential for mammalian development and heterochromatin structure. PLoS Genet. 2008;4:e1000190. doi: 10.1371/journal.pgen.1000190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Min J, Feng Q, Li Z, Zhang Y, Xu RM. Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase. Cell. 2003;112:711–723. doi: 10.1016/s0092-8674(03)00114-4. [DOI] [PubMed] [Google Scholar]
- 49.Feng Q, Wang H, Ng HH, Erdjument-Bromage H, Tempst P, Struhl K, Zhang Y. 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]
- 50.Sawada K, Yang Z, Horton JR, Collins RE, Zhang X, Cheng X. Structure of the conserved core of the yeast Dot1p, a nucleosomal histone H3 lysine 79 methyltransferase. J Biol Chem. 2004;279:43296–43306. doi: 10.1074/jbc.M405902200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Daigle S, Olhava EJ, Therkelsen CA, Majer CR, Sneeringer CJ, Song J, Johnston LD, Scott MP, Smith JJ, Xiao Y, Jin L, Kuntz KW, Chesworth R, Moyer MP, Bernt KM, Tseng JC, Kung AL, Armstrong SA, Copeland RA, Richon VM, Pollock RM. Selective Killing of Mixed Lineage Leukemia Cells by a Potent Small-Molecule DOT1L Inhibitor. Cancer Cell. 2011;20:53–65. doi: 10.1016/j.ccr.2011.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Daigle S, Olhava EJ, Therkelsen CA, Basavapathruni A, Jin L, Boriack-Sjodin PA, Allain CJ, Klaus CR, Raimondi A, Scott MP, Waters NJ, Chesworth R, Moyer MP, Copeland RA, Richon VM, Pollock RM. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood. 2013;122:1017–1025. doi: 10.1182/blood-2013-04-497644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Anglin J, Song Y. A Medicinal Chemistry Perspective for Targeting Histone H3 Lysine-79 Methyltransferase DOT1L. J Med Chem. 2013;56:8972–8983. doi: 10.1021/jm4007752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhang K, Siino JS, Jones PR, Yau PM, Bradbury EM. A mass spectrometric “Western blot” to evaluate the correlations between histone methylation and histone acetylation. Proteomics. 2004;4:3765–3775. doi: 10.1002/pmic.200400819. [DOI] [PubMed] [Google Scholar]
- 55.Schübeler D, MacAlpine DM, Scalzo D, Wirbelauer C, Kooperberg C, van Leeuwen F, Gottschling DE, O’Neill LP, Turner BM, Delrow J, Bell SP, Groudine M. The histone modification pattern of active genes revealed through genome-wide chromatin analysis of a higher eukaryote. Genes Dev. 2004;18:1263–1271. doi: 10.1101/gad.1198204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Morillon A, Karabetsou N, Nair A, Mellor J. Dynamic lysine methylation on histone H3 defines the regulatory phase of gene transcription. Mol Cell. 2005;18:723–734. doi: 10.1016/j.molcel.2005.05.009. [DOI] [PubMed] [Google Scholar]
- 57.Onder T, Kara N, Cherry A, Sinha AU, Zhu N, Bernt KM, Cahan P, Marcarci BO, Unternaehrer J, Gupta PB, Lander ES, Armstrong SA, Daley GQ. Chromatin-modifying enzymes as modulators of reprogramming. Nature. 2012;483:598–602. doi: 10.1038/nature10953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cattaneo P, Kunderfranco P, Greco C, Guffanti A, Stirparo GG, Rusconi F, Rizzi R, Di Pasquale E, Locatelli SL, Latronico MV, Bearzi C, Papait R, Condorelli G. DOT1L-mediated H3K79me2 modification critically regulates gene expression during cardiomyocyte differentiation. Cell Death Differ. 2014 doi: 10.1038/cdd.2014.199. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schulze J, Jackson J, Nakanishi S, Gardner JM, Hentrich T, Haug J, Johnston M, Jaspersen SL, Kobor MS, Shilatifard A. Linking cell cycle to histone modifications: SBF and H2B monoubiquitination machinery and cell-cycle regulation of H3K79 dimethylation. Mol Cell. 2009;35:626–641. doi: 10.1016/j.molcel.2009.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.De Vos D, Frederiks F, Terweij M, van Welsem T, Verzijlbergen KF, Iachina E, de Graaf EL, Altelaar AF, Oudgenoeg G, Heck AJ, Krijgsveld J, Bakker BM, van Leeuwen F. Progressive methylation of ageing histones by Dot1 functions as a timer. EMBO Rep. 2011;12:956–962. doi: 10.1038/embor.2011.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kouskouti A, Talianidis I. Histone modifications defining active genes persist after transcriptional and mitotic inactivation. EMBO J. 2005;24:347–357. doi: 10.1038/sj.emboj.7600516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kim S, Jung I, Lee H, Kang K, Kim M, Jeong K, Kwon CS, Han YM, Kim YS, Kim D, Lee D. Human Histone H3K79 Methyltransferase DOT1L Methyltransferase Binds Actively Transcribing RNA Polymerase II to Regulate Gene Expression. J Biol Chem. 2012;287:39698–39709. doi: 10.1074/jbc.M112.384057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Suzuki M, Ito H, Aoki F. Effects of RNAi-Mediated Knockdown of Histone Methyltransferases on the Sex-Specific mRNA Expression of Imp in the Silkworm Bombyx mori. Int J Mol Sci. 2014;15:6772–6792. doi: 10.3390/ijms15046772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Shanower G, Muller M, Blanton JL, Honti V, Gyurkovics H, Schedl P. Characterization of the grappa gene, the Drosophila histone H3 lysine 79 methyltransferase. Genetics. 2005;169:173–184. doi: 10.1534/genetics.104.033191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Dottermusch-Heidel C, Gärtner SM, Tegeder I, Rathke C, Barckmann B, Bartkuhn M, Bhushan S, Steger K, Meinhardt A, Renkawitz-Pohl R. H3K79 methylation: a new conserved mark that accompanies H4 hyperacetylation prior to histone-to-protamine transition in Drosophila and rat. Biol Open. 2014;3:444–452. doi: 10.1242/bio.20147302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Dottermusch-Heidel C, Klaus ES, Gonzalez NH, Bhushan S, Meinhardt A, Bergmann M, Renkawitz-Pohl R, Rathke C, Steger K. H3K79 methylation directly precedes the histone-toprotamine transition in mammalian spermatids and is sensitive to bacterial infections. Andrology. 2014;2:655–665. doi: 10.1111/j.2047-2927.2014.00248.x. [DOI] [PubMed] [Google Scholar]
- 67.Matsuura K, Fujimoto K, Das B, Fu L, Lu CD, Shi YB. Histone H3K79 methyltransferase Dot1L is directly activated by thyroid hormone receptor during Xenopus metamorphosis. Cell Biosci. 2012;2:25. doi: 10.1186/2045-3701-2-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wen L, Fu L, Guo X, Chen Y, Shi YB. Histone methyltransferase Dot1L plays a role in postembryonic development in Xenopus tropicalis. FASEB J. 2014:385–393. doi: 10.1096/fj.14-252171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tsai F, Keller G, Kuo FC, Weiss M, Chen J, Rosenblatt M, Alt FW, Orkin SH. An early haematopoietic defect in mice lacking the transcription factor GATA-2. Nature. 1994;371:221–6. doi: 10.1038/371221a0. [DOI] [PubMed] [Google Scholar]
- 70.Back J, Dierich A, Bronn C, Kastner P, Chan S. PU. 1 determines the self-renewal capacity of erythroid progenitor cells. Blood. 2004;130:3615–3623. doi: 10.1182/blood-2003-11-4089. [DOI] [PubMed] [Google Scholar]
- 71.Feng Y, Yang Y, Ortega MM, Copeland JN, Zhang M, Jacob JB, Fields TA, Vivian JL, Fields PE. Early mammalian erythropoiesis requires the Dot1L methyltransferase. Blood. 2010;116:4483–4491. doi: 10.1182/blood-2010-03-276501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jo S, Granowicz EM, Maillard I, Thomas D, Hess JL. Requirement for Dot1l in murine postnatal hematopoiesis and leukemogenesis by MLL translocation. Blood. 2011;117:4759–4768. doi: 10.1182/blood-2010-12-327668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Nguyen A, He J, Taranova O, Zhang Y. Essential role of DOT1L in maintaining normal adult hematopoiesis. Cell Res. 2011;21:1370–1373. doi: 10.1038/cr.2011.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wysocki R, Javaheri A, Allard S, Sha F, Côté J, Kron SJ. Role of Dot1-dependent histone H3 methylation in G1 and S phase DNA damage checkpoint functions of Rad9. Mol Cell Biol. 2005;25:8430–8443. doi: 10.1128/MCB.25.19.8430-8443.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.San-Segundo P, Roeder GS. Role for the silencing protein Dot1 in meiotic checkpoint control. Mol Biol Cell. 2000;11:3601–3615. doi: 10.1091/mbc.11.10.3601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Botuyan M, Lee J, Ward IM, Kim JE, Thompson JR, Chen J, Mer G. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.FitzGerald J, Moureau S, Drogaris P, O’Connell E, Abshiru N, Verreault A, Thibault P, Grenon M, Lowndes NF. Regulation of the DNA damage response and gene expression by the Dot1L histone methyltransferase and the 53Bp1 tumour suppressor. PLoS One. 2011;6:e14714. doi: 10.1371/journal.pone.0014714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wakeman T, Wang Q, Feng J, Wang XF. Bat3 facilitates H3K79 dimethylation by DOT1L and promotes DNA damage-induced 53BP1 foci at G1/G2 cell-cycle phases. EMBO J. 2012;31:2169–2181. doi: 10.1038/emboj.2012.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huyen Y, Zgheib O, Ditullio RA Jr, Gorgoulis VG, Zacharatos P, Petty TJ, Sheston EA, Mellert HS, Stavridi ES, Halazonetis TD. Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks. Nature. 2004;432:406–411. doi: 10.1038/nature03114. [DOI] [PubMed] [Google Scholar]
- 80.Oksenych V, Zhovmer A, Ziani S, Mari PO, Eberova J, Nardo T, Stefanini M, Giglia-Mari G, Jean-Marc Egly JM, Frédéric Coin F. Histone Methyltransferase DOT1L Drives Recovery of Gene Expression after a Genotoxic Attack. PLoS Genet. 2013;9:e1003611. doi: 10.1371/journal.pgen.1003611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Biondi A, Cimino G, Pieters R, Pui CH. Biological and therapeutic aspects of infant leukemia. Blood. 2000;96:24–33. [PubMed] [Google Scholar]
- 82.Ziemin-van der Poel S, McCabe NR, Gill HJ, Espinosa R 3rd, Patel Y, Harden A, Rubinelli P, Smith SD, LeBeau MM, Rowley JD, et al. Identification of a gene, MLL, that spans the breakpoint in 11q23 translocations associated with human leukemia. Proc Natl Acad Sci U S A. 1991;88:10735–9. doi: 10.1073/pnas.88.23.10735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dimartino JF, Cleary ML. Mll rearrangements in haematological malignancies: lessons from clinical and biological studies. Br J Haematol. 1999;106:614–26. doi: 10.1046/j.1365-2141.1999.01439.x. [DOI] [PubMed] [Google Scholar]
- 84.Bitoun E, Oliver PL, Davies KE. The mixed-lineage leukemia fusion partner AF4 stimulates RNA polymerase II transcriptional elongation and mediates coordinated chromatin remodeling. Hum Mol Genet. 2007;16:92–106. doi: 10.1093/hmg/ddl444. [DOI] [PubMed] [Google Scholar]
- 85.Krivtsov A, Feng Z, Lemieux ME, Faber J, Vempati S, Sinha AU, Xia X, Jesneck J, Bracken AP, Silverman LB, Kutok JL, Kung AL, Armstrong SA. H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell. 2008;14:355–368. doi: 10.1016/j.ccr.2008.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Nguyen A, Taranova O, He J, Zhang Y. DOT1L, the H3K79 methyltransferase, is required for MLL-AF9-mediated leukemogenesis. Blood. 2011;117:6912–6922. doi: 10.1182/blood-2011-02-334359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Deshpande A, Chen L, Fazio M, Sinha AU, Bernt KM, Banka D, Dias S, Chang J, Olhava EJ, Daigle SR, Richon VM, Pollock RM, Armstrong SA. Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l. Blood. 2013;121:2533–2541. doi: 10.1182/blood-2012-11-465120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zeisig D, Bittner CB, Zeisig BB, Garcia-Cuellar MP, Hess JL, Slany RK. The eleven-nineteenleukemia protein ENL connects nuclear MLL fusion partners with chromatin. Oncogene. 2005;24:5525–5532. doi: 10.1038/sj.onc.1208699. [DOI] [PubMed] [Google Scholar]
- 89.Okada Y, Feng Q, Lin Y, Jiang Q, Li Y, Coffield VM, Su L, Xu G, Zhang Y. hDOT1L links histone methylation to leukemogenesis. Cell. 2005;121:167–178. doi: 10.1016/j.cell.2005.02.020. [DOI] [PubMed] [Google Scholar]
- 90.Barry E, 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]
- 91.Deshpande A, Bradner J, Armstrong SA. Chromatin modifications as therapeutic targets in MLL-rearranged leukemia. Trends Immunol. 2012;33:563–570. doi: 10.1016/j.it.2012.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Buske C, Feuring-Buske M, Abramovich C, Spiekermann K, Eaves CJ, Coulombel L, Sauvageau G, Hogge DE, Humphries RK. Deregulated expression of HOXB4 enhances the primitive growth activity of human hematopoietic cells. Blood. 2002;100:862–8. doi: 10.1182/blood-2002-01-0220. [DOI] [PubMed] [Google Scholar]
- 93.Thorsteinsdottir U, Mamo A, Kroon E, Jerome L, Bijl J, Lawrence HJ, Humphries K, Sauvageau G. Overexpression of the myeloid leukemia-associated Hoxa9 gene in bone marrow cells induces stem cell expansion. Blood. 2002;99:121–129. doi: 10.1182/blood.v99.1.121. [DOI] [PubMed] [Google Scholar]
- 94.Milne T, Briggs SD, Brock HW, Martin ME, Gibbs D, Allis CD, Hess JL. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol Cell. 2002;10:1107–17. doi: 10.1016/s1097-2765(02)00741-4. [DOI] [PubMed] [Google Scholar]
- 95.Deshpande A, Deshpande A, Sinha AU, Chen L, Chang J, Cihan A, Fazio M, Chen CW, Zhu N, Koche R, Dzhekieva L, Ibáñez G, Dias S, Banka D, Krivtsov A, Luo M, Roeder RG, Bradner JE, Bernt KM, Armstrong SA. AF10 regulates progressive H3K79 methylation and HOX gene expression in diverse AML subtypes. Cancer Cell. 2014;26:896–908. doi: 10.1016/j.ccell.2014.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Okada Y, Jiang Q, Lemieux M, Jeannotte L, Su L, Zhang Y. Leukaemic transformation by CALM-AF10 involves upregulation of Hoxa5 by hDOT1L. Nat Cell Bio. 2006;8:1017–1024. doi: 10.1038/ncb1464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chen L, Deshpande AJ, Banka D, Bernt KM, Dias S, Buske C, Olhava EJ, Daigle SR, Richon VM, Pollock RM, Armstrong SA. Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1l. Leukemia. 2013;27:812–822. doi: 10.1038/leu.2012.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Shen C, Jo SY, Liao C, Hess JL, Nikolovska-Coleska Z. Targeting recruitment of disruptor of telomeric silencing 1-like (DOT1L): characterizing the interactions between DOT1L and mixed lineage leukemia (MLL) fusion proteins. J Biol Chem. 2013;288:30585–30596. doi: 10.1074/jbc.M113.457135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kuntimaddi A, Achille NJ, Thorpe J, Lokken AA, Singh R, Hemenway CS, Adli M, Zeleznik-Le NJ, Bushweller JH. Degree of Recruitment of DOT1L to MLL-AF9 Defines Level of H3K79 Di- and Tri-methylation on Target Genes and Transformation Potential. Cell Rep. 2015;11:808–820. doi: 10.1016/j.celrep.2015.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Li Y, Wen H, Xi Y, Tanaka K, Wang H, Peng D, Ren Y, Jin Q, Dent SY, Li W, Li H, Shi X. AF9 YEATS domain links histone acetylation to DOT1L-mediated H3K79 methylation. Cell. 2014;159:558–571. doi: 10.1016/j.cell.2014.09.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Chen C, Koche RP, Sinha AU, Deshpande AJ, Zhu N, Eng R, Doench JG, Xu H, Chu SH, Qi J, Wang X, Delaney C, Bernt KM, Root DE, Hahn WC, Bradner JE, Armstrong SA. DOT1L inhibits SIRT1-mediated epigenetic silencing to maintain leukemic gene expression in MLLrearranged leukemia. Nat Med. 2015;21:335–343. doi: 10.1038/nm.3832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Kim W, Kim R, Park G, Park JW, Kim JE. Deficiency of H3K79 histone methyltransferase Dot1-like protein (DOT1L) inhibits cell proliferation. J Biol Chem. 2012;287:5588–5599. doi: 10.1074/jbc.M111.328138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kryczek I, Lin Y, Nagarsheth N, Peng D, Zhao L, Zhao E, Vatan L, Szeliga W, Dou Y, Owens S, Zgodzinski W, Majewski M, Wallner G, Fang J, Huang E, Zou W. IL-22(+)CD4(+) T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L. Immunity. 2014;40:772–784. doi: 10.1016/j.immuni.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhang L, Deng L, Chen F, Yao Y, Wu B, Wei L, Mo Q, Song Y. Inhibition of histone H3K79 methylation selectively inhibits proliferation, self-renewal and metastatic potential of breast cancer. Oncotarget. 2014;5:10665–10677. doi: 10.18632/oncotarget.2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Cho M, Park JH, Choi HJ, Park MK, Won HY, Park YJ, Lee CH, Oh SH, Song YS, Kim HS, Oh YH, Lee JY, Kong G. DOT1L cooperates with the c-Myc-p300 complex to epigenetically derepress CDH1 transcription factors in breast cancer progression. Nat Commun. 2015;6:7821. doi: 10.1038/ncomms8821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Guccione E, Martinato F, Finocchiaro G, Luzi L, Tizzoni L, Dall’ Olio V, Zardo G, Nervi C, Bernard L, Amati B. Myc-binding-site recognition in the human genome is determined by chromatin context. Nat Cell Biol. 2006;8:764–770. doi: 10.1038/ncb1434. [DOI] [PubMed] [Google Scholar]
- 107.Kim T, Barrera LO, Zheng M, Qu C, Singer MA, Richmond TA, Wu Y, Green RD, Ren B. A highresolution map of active promoters in the human genome. Nature. 2005;436:876–880. doi: 10.1038/nature03877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Gibbons G, Owens SR, Fearon ER, Nikolovska-Coleska Z. Regulation of Wnt signaling target gene expression by the histone methyltransferase DOT1L. ACS Chem Biol. 2015;10:109–114. doi: 10.1021/cb500668u. [DOI] [PubMed] [Google Scholar]
- 109.Yu W, Chory EJ, Wernimont AK, Tempel W, Scopton A, Federation A, Marineau JJ, Qi J, Barsyte-Lovejoy D, Yi J, Marcellus R, Iacob RE, Engen JR, Griffin C, Aman A, Wienholds E, Li F, Pineda J, Estiu G, Shatseva T, Hajian T, Al-Awar R, Dick JE, Vedadi M, Brown PJ, Arrowsmith CH, Bradner JE, Schapira M. Catalytic site remodelling of the DOT1L methyltransferase by selective inhibitors. Nat Commun. 2012;3:1288. doi: 10.1038/ncomms2304. [DOI] [PubMed] [Google Scholar]
- 110.Anglin J, Deng L, Yao Y, Cai G, Liu Z, Jiang H, Cheng G, Chen P, Dong S, Song Y. Synthesis and structure-activity relationship investigation of adenosine-containing inhibitors of histone methyltransferase DOT1L. J Med Chem. 2012;55:8066–8074. doi: 10.1021/jm300917h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Klaus C, Iwanowicz D, Johnston LD, Campbell CA, Smith JJ, Moyer MP, Copeland RA, Olhava EJ, Porter Scott M, Pollock RM, Daigle SR, Raimondi A. DOT1L Inhibitor EPZ-5676 Displays Synergistic Antiproliferative Activity in Combination with Standard of Care Drugs and Hypomethylating Agents in MLL-Rearranged Leukemia Cells. J Pharmacol Exp Ther. 2014;350:646–56. doi: 10.1124/jpet.114.214577. [DOI] [PubMed] [Google Scholar]
- 112.Liu W, Deng L, Song Y, Redell M. DOT1L Inhibition Sensitizes MLL-Rearranged AML to Chemotherapy. PLoS One. 2014;9:e98270. doi: 10.1371/journal.pone.0098270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yi J, Federation AJ, Qi J, Dhe-Paganon S, Hadler M, Xu X, St Pierre R, Varca AC, Wu L, Marineau JJ, Smith WB, Souza A, Chory EJ, Armstrong SA, Bradner JE. Structure-guided DOT1L probe optimization by label-free ligand displacement. ACS Chem Biol. 2015;10:667–674. doi: 10.1021/cb500796d. [DOI] [PMC free article] [PubMed] [Google Scholar]
