Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 12(9): 2120-2139, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35789380

ABSTRACT

Polycomb repressive complex 2 (PRC2) has oncogenic and tumor-suppressive roles in cancer. There is clinical success of targeting this complex in PRC2-dependent cancers, but an unmet therapeutic need exists in PRC2-loss cancer. PRC2-inactivating mutations are a hallmark feature of high-grade malignant peripheral nerve sheath tumor (MPNST), an aggressive sarcoma with poor prognosis and no effective targeted therapy. Through RNAi screening in MPNST, we found that PRC2 inactivation increases sensitivity to genetic or small-molecule inhibition of DNA methyltransferase 1 (DNMT1), which results in enhanced cytotoxicity and antitumor response. Mechanistically, PRC2 inactivation amplifies DNMT inhibitor-mediated expression of retrotransposons, subsequent viral mimicry response, and robust cell death in part through a protein kinase R (PKR)-dependent double-stranded RNA sensor. Collectively, our observations posit DNA methylation as a safeguard against antitumorigenic cell-fate decisions in PRC2-loss cancer to promote cancer pathogenesis, which can be therapeutically exploited by DNMT1-targeted therapy. SIGNIFICANCE: PRC2 inactivation drives oncogenesis in various cancers, but therapeutically targeting PRC2 loss has remained challenging. Here we show that PRC2-inactivating mutations set up a tumor context-specific liability for therapeutic intervention via DNMT1 inhibitors, which leads to innate immune signaling mediated by sensing of derepressed retrotransposons and accompanied by enhanced cytotoxicity. See related commentary by Guil and Esteller, p. 2020. This article is highlighted in the In This Issue feature, p. 2007.


Subject(s)
Antineoplastic Agents , Neoplasms , Neurofibrosarcoma , Carcinogenesis/genetics , Humans , Mutation , Neoplasms/genetics , Neurofibrosarcoma/diagnosis , Neurofibrosarcoma/genetics , Neurofibrosarcoma/pathology , Polycomb Repressive Complex 2/genetics , Retroelements
2.
Commun Biol ; 5(1): 528, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35654826

ABSTRACT

The DNA methylation status of the X-chromosome in cancer cells is often overlooked because of computational difficulties. Most of the CpG islands on the X-chromosome are mono-allelically methylated in normal female cells and only present as a single copy in male cells. We treated two colorectal cancer cell lines from a male (HCT116) and a female (RKO) with increasing doses of a DNA methyltransferase 1 (DNMT1)-specific inhibitor (GSK3685032/GSK5032) over several months to remove as much non-essential CpG methylation as possible. Profiling of the remaining DNA methylome revealed an unexpected, enriched retention of DNA methylation on the X-chromosome. Strikingly, the identified retained X-chromosome DNA methylation patterns accurately predicted de novo DNA hypermethylation in colon cancer patient methylomes in the TCGA COAD/READ cohort. These results suggest that a re-examination of tumors for X-linked DNA methylation changes may enable greater understanding of the importance of epigenetic silencing of cancer related genes.


Subject(s)
DNA Methylation , Neoplasms , CpG Islands , DNA , Female , Humans , Male , Neoplasms/genetics , X Chromosome
3.
Structure ; 30(6): 793-802.e5, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35395178

ABSTRACT

DNMT1 maintains the parental DNA methylation pattern on newly replicated hemimethylated DNA. The failure of this maintenance process causes aberrant DNA methylation that affects transcription and contributes to the development and progression of cancers such as acute myeloid leukemia. Here, we structurally characterized a set of newly discovered DNMT1-selective, reversible, non-nucleoside inhibitors that bear a core 3,5-dicyanopyridine moiety, as exemplified by GSK3735967, to better understand their mechanism of inhibition. All of the dicyanopydridine-containing inhibitors examined intercalate into the hemimethylated DNA between two CpG base pairs through the DNA minor groove, resulting in conformational movement of the DNMT1 active-site loop. In addition, GSK3735967 introduces two new binding sites, where it interacts with and stabilizes the displaced DNMT1 active-site loop and it occupies an open aromatic cage in which trimethylated histone H4 lysine 20 is expected to bind. Our work represents a substantial step in generating potent, selective, and non-nucleoside inhibitors of DNMT1.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methylation , Binding Sites , Catalytic Domain , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism
4.
Nat Cancer ; 2(10): 1002-1017, 2021 10.
Article in English | MEDLINE | ID: mdl-34790902

ABSTRACT

DNA methylation, a key epigenetic driver of transcriptional silencing, is universally dysregulated in cancer. Reversal of DNA methylation by hypomethylating agents, such as the cytidine analogs decitabine or azacytidine, has demonstrated clinical benefit in hematologic malignancies. These nucleoside analogs are incorporated into replicating DNA where they inhibit DNA cytosine methyltransferases DNMT1, DNMT3A and DNMT3B through irreversible covalent interactions. These agents induce notable toxicity to normal blood cells thus limiting their clinical doses. Herein we report the discovery of GSK3685032, a potent first-in-class DNMT1-selective inhibitor that was shown via crystallographic studies to compete with the active-site loop of DNMT1 for penetration into hemi-methylated DNA between two CpG base pairs. GSK3685032 induces robust loss of DNA methylation, transcriptional activation and cancer cell growth inhibition in vitro. Due to improved in vivo tolerability compared with decitabine, GSK3685032 yields superior tumor regression and survival mouse models of acute myeloid leukemia.


Subject(s)
Azacitidine , Leukemia, Myeloid, Acute , Animals , Azacitidine/pharmacology , DNA/metabolism , DNA Methylation , DNA Modification Methylases/genetics , Decitabine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Mice
5.
Nat Struct Mol Biol ; 28(7): 594-603, 2021 07.
Article in English | MEDLINE | ID: mdl-34140676

ABSTRACT

DNA methylation plays a critical role during development, particularly in repressing retrotransposons. The mammalian methylation landscape is dependent on the combined activities of the canonical maintenance enzyme Dnmt1 and the de novo Dnmts, 3a and 3b. Here, we demonstrate that Dnmt1 displays de novo methylation activity in vitro and in vivo with specific retrotransposon targeting. We used whole-genome bisulfite and long-read Nanopore sequencing in genetically engineered methylation-depleted mouse embryonic stem cells to provide an in-depth assessment and quantification of this activity. Utilizing additional knockout lines and molecular characterization, we show that the de novo methylation activity of Dnmt1 depends on Uhrf1, and its genomic recruitment overlaps with regions that enrich for Uhrf1, Trim28 and H3K9 trimethylation. Our data demonstrate that Dnmt1 can catalyze DNA methylation in both a de novo and maintenance context, especially at retrotransposons, where this mechanism may provide additional stability for long-term repression and epigenetic propagation throughout development.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , DNA Transposable Elements/genetics , Embryonic Development/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Gene Knockout Techniques , Genome/genetics , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Tripartite Motif-Containing Protein 28/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Whole Genome Sequencing , DNA Methyltransferase 3B
6.
Haematologica ; 106(7): 1979-1987, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32586904

ABSTRACT

Pharmacological induction of fetal hemoglobin (HbF) expression is an effective therapeutic strategy for the management of beta-hemoglobinopathies such as sickle cell disease. DNA methyltransferase (DNMT) inhibitors 5-azacytidine (5-aza) and 5-aza-2'-deoxycytidine (decitabine) have been shown to induce fetal hemoglobin expression in both preclinical models and clinical studies, but are not currently approved for the management of hemoglobinopathies. We report here the discovery of a novel class of orally bioavailable DNMT1-selective inhibitors as exemplified by GSK3482364. This molecule potently inhibits the methyltransferase activity of DNMT1, but not DNMT family members DNMT3A or DNMT3B. In contrast with cytidine analog DNMT inhibitors, the DNMT1 inhibitory mechanism of GSK3482364 does not require DNA incorporation and is reversible. In cultured human erythroid progenitor cells (EPCs), GSK3482364 decreased overall DNA methylation resulting in de-repression of the gamma globin genes HBG1 and HBG2 and increased HbF expression. In a transgenic mouse model of sickle cell disease, orally administered GSK3482364 caused significant increases in both HbF levels and in the percentage HbF-expressing erythrocytes, with good overall tolerability. We conclude that in these preclinical models, selective, reversible inhibition of DNMT1 is sufficient for the induction of HbF, and is well-tolerated. We anticipate that GSK3482364 will be a useful tool molecule for the further study of selective DNMT1 inhibition both in vitro and in vivo.


Subject(s)
Anemia, Sickle Cell , Fetal Hemoglobin , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Azacitidine/pharmacology , DNA Methylation , Fetal Hemoglobin/genetics , Mice , gamma-Globins/genetics
7.
ACS Med Chem Lett ; 11(2): 133-140, 2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32071679

ABSTRACT

We report herein the discovery of isoxazole amides as potent and selective SET and MYND Domain-Containing Protein 3 (SMYD3) inhibitors. Elucidation of the structure-activity relationship of the high-throughput screening (HTS) lead compound 1 provided potent and selective SMYD3 inhibitors. The SAR optimization, cocrystal structures of small molecules with SMYD3, and mode of inhibition (MOI) characterization of compounds are described. The synthesis and biological and pharmacokinetic profiles of compounds are also presented.

8.
Cancer Cell ; 36(1): 100-114.e25, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31257072

ABSTRACT

Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/deficiency , Alternative Splicing , Antineoplastic Agents/chemistry , Biomarkers , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/chemistry , Humans , Methylation , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Protein-Arginine N-Methyltransferases/chemistry , Substrate Specificity
9.
Haematologica ; 104(6): 1156-1167, 2019 06.
Article in English | MEDLINE | ID: mdl-30514804

ABSTRACT

Lysine specific demethylase 1 (LSD1) is a histone modifying enzyme that suppresses gene expression through demethylation of lysine 4 on histone H3. The anti-tumor activity of GSK2879552 and GSK-LSD1, potent, selective irreversible inactivators of LSD1, has previously been described. Inhibition of LSD1 results in a cytostatic growth inhibitory effect in a range of acute myeloid leukemia cell lines. To enhance the therapeutic potential of LSD1 inhibition in this disease setting, a combination of LSD1 inhibition and all-trans retinoic acid was explored. All-trans retinoic acid is currently approved for use in acute promyelocytic leukemia in which it promotes differentiation of abnormal blast cells into normal white blood cells. Combined treatment with all-trans retinoic acid and GSK2879552 results in synergistic effects on cell proliferation, markers of differentiation, and, most importantly, cytotoxicity. Ultimately the combination potential for LSD1 inhibition and ATRA will require validation in acute myeloid leukemia patients, and clinical studies to assess this are currently underway.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Histone Demethylases/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Tretinoin/pharmacology , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Benzoates/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclopropanes/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Treatment Outcome , Tretinoin/administration & dosage
10.
Sci Rep ; 8(1): 9711, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29946150

ABSTRACT

Evasion of the potent tumour suppressor activity of p53 is one of the hurdles that must be overcome for cancer cells to escape normal regulation of cellular proliferation and survival. In addition to frequent loss of function mutations, p53 wild-type activity can also be suppressed post-translationally through several mechanisms, including the activity of PRMT5. Here we describe broad anti-proliferative activity of potent, selective, reversible inhibitors of protein arginine methyltransferase 5 (PRMT5) including GSK3326595 in human cancer cell lines representing both hematologic and solid malignancies. Interestingly, PRMT5 inhibition activates the p53 pathway via the induction of alternative splicing of MDM4. The MDM4 isoform switch and subsequent p53 activation are critical determinants of the response to PRMT5 inhibition suggesting that the integrity of the p53-MDM4 regulatory axis defines a subset of patients that could benefit from treatment with GSK3326595.


Subject(s)
Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins/metabolism , RNA Splicing/genetics , Tumor Suppressor Protein p53/metabolism , Alternative Splicing/genetics , Antineoplastic Agents , Arginine/analogs & derivatives , Arginine/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/pharmacology , Humans , Nuclear Proteins/genetics , Protein Isoforms/genetics , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , snRNP Core Proteins/metabolism
11.
J Pharmacol Exp Ther ; 363(3): 336-347, 2017 12.
Article in English | MEDLINE | ID: mdl-28928122

ABSTRACT

Decreased erythropoietin (EPO) production, shortened erythrocyte survival, and other factors reducing the response to EPO contribute to anemia in patients who have a variety of underlying pathologies such as chronic kidney disease. Treatment with recombinant human EPO (rHuEPO) at supraphysiologic concentrations has proven to be efficacious. However, it does not ameliorate the condition in all patients, and it presents its own risks, including cardiovascular complications. The transcription factors hypoxia-inducible factor (HIF) 1α and HIF2α control the physiologic response to hypoxia and invoke a program of increased erythropoiesis. Levels of HIFα are modulated by oxygen tension via the action of a family of HIF-prolyl hydroxylases (PHDs), which tag HIFα for proteasomal degradation. Inhibition of these PHDs simulates conditions of mild hypoxia, leading to a potentially more physiologic erythropoietic response and presenting a potential alternative to high doses of rHuEPO. Here we describe the discovery and characterization of GSK1278863 [2-(1,3-dicyclohexyl-6-hydroxy-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamido) acetic acid], a pyrimidinetrione-glycinamide low nanomolar inhibitor of PHDs 1-3 that stabilizes HIFα in cell lines, resulting in the production of increased levels of EPO. In normal mice, a single dose of GSK1278863 induced significant increases in circulating plasma EPO but only minimal increases in plasma vascular endothelial growth factor (VEGF-A) concentrations. GSK1278863 significantly increased reticulocytes and red cell mass parameters in preclinical species after once-daily oral administration and has demonstrated an acceptable nonclinical toxicity profile, supporting continued clinical development. GSK1278863 is currently in phase 3 clinical trials for treatment of anemia in patients with chronic kidney disease.


Subject(s)
Barbiturates/pharmacology , Drugs, Investigational/pharmacology , Enzyme Inhibitors/pharmacology , Erythropoiesis/drug effects , Erythropoietin/agonists , Glycine/analogs & derivatives , Hematinics/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Animals , Barbiturates/administration & dosage , Barbiturates/adverse effects , Barbiturates/pharmacokinetics , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Dogs , Dose-Response Relationship, Drug , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Glycine/administration & dosage , Glycine/adverse effects , Glycine/pharmacokinetics , Glycine/pharmacology , Hematinics/administration & dosage , Hematinics/adverse effects , Hematinics/pharmacokinetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Protein Stability/drug effects , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Toxicity Tests, Chronic
12.
Cancer Cell ; 28(1): 57-69, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26175415

ABSTRACT

Epigenetic dysregulation has emerged as an important mechanism in cancer. Alterations in epigenetic machinery have become a major focus for targeted therapies. The current report describes the discovery and biological activity of a cyclopropylamine containing inhibitor of Lysine Demethylase 1 (LSD1), GSK2879552. This small molecule is a potent, selective, orally bioavailable, mechanism-based irreversible inactivator of LSD1. A proliferation screen of cell lines representing a number of tumor types indicated that small cell lung carcinoma (SCLC) is sensitive to LSD1 inhibition. The subset of SCLC lines and primary samples that undergo growth inhibition in response to GSK2879552 exhibit DNA hypomethylation of a signature set of probes, suggesting this may be used as a predictive biomarker of activity.


Subject(s)
Antineoplastic Agents/administration & dosage , Benzoates/administration & dosage , Cyclopropanes/administration & dosage , DNA Methylation/drug effects , Enzyme Inhibitors/administration & dosage , Histone Demethylases/antagonists & inhibitors , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclopropanes/pharmacology , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Molecular Sequence Data , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Xenograft Model Antitumor Assays
13.
Nat Chem Biol ; 11(6): 432-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915199

ABSTRACT

Protein arginine methyltransferase-5 (PRMT5) is reported to have a role in diverse cellular processes, including tumorigenesis, and its overexpression is observed in cell lines and primary patient samples derived from lymphomas, particularly mantle cell lymphoma (MCL). Here we describe the identification and characterization of a potent and selective inhibitor of PRMT5 with antiproliferative effects in both in vitro and in vivo models of MCL. EPZ015666 (GSK3235025) is an orally available inhibitor of PRMT5 enzymatic activity in biochemical assays with a half-maximal inhibitory concentration (IC50) of 22 nM and broad selectivity against a panel of other histone methyltransferases. Treatment of MCL cell lines with EPZ015666 led to inhibition of SmD3 methylation and cell death, with IC50 values in the nanomolar range. Oral dosing with EPZ015666 demonstrated dose-dependent antitumor activity in multiple MCL xenograft models. EPZ015666 represents a validated chemical probe for further study of PRMT5 biology and arginine methylation in cancer and other diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Isoquinolines/pharmacology , Lymphoma, Mantle-Cell/pathology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Isoquinolines/chemistry , Isoquinolines/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/enzymology , Male , Methylation , Mice, Inbred Strains , Models, Molecular , Molecular Structure , Protein Binding , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays , snRNP Core Proteins/metabolism
14.
Mol Cancer Ther ; 13(12): 3062-73, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25253781

ABSTRACT

The EZH2 methyltransferase silences gene expression through methylation of histone H3 on lysine 27 (H3K27). Recently, EZH2 mutations have been reported at Y641, A677, and A687 in non-Hodgkin lymphoma. Although the Y641F/N/S/H/C and A677G mutations exhibit clearly increased activity with substrates dimethylated at lysine 27 (H3K27me2), the A687V mutant has been shown to prefer a monomethylated lysine 27 (H3K27me1) with little gain of activity toward H3K27me2. Herein, we demonstrate that despite this unique substrate preference, A687V EZH2 still drives increased H3K27me3 when transiently expressed in cells. However, unlike the previously described mutants that dramatically deplete global H3K27me2 levels, A687V EZH2 retains normal levels of H3K27me2. Sequencing of B-cell-derived cancer cell lines identified an acute lymphoblastic leukemia cell line harboring this mutation. Similar to exogenous expression of A687V EZH2, this cell line exhibited elevated H3K27me3 while possessing H3K27me2 levels higher than Y641- or A677-mutant lines. Treatment of A687V EZH2-mutant cells with GSK126, a selective EZH2 inhibitor, was associated with a global decrease in H3K27me3, robust gene activation, caspase activation, and decreased proliferation. Structural modeling of the A687V EZH2 active site suggests that the increased catalytic activity with H3K27me1 may be due to a weakened interaction with an active site water molecule that must be displaced for dimethylation to occur. These findings suggest that A687V EZH2 likely increases global H3K27me3 indirectly through increased catalytic activity with H3K27me1 and cells harboring this mutation are highly dependent on EZH2 activity for their survival.


Subject(s)
Histones/metabolism , Mutation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cell Line, Tumor , Cluster Analysis , Enhancer of Zeste Homolog 2 Protein , Gene Expression , Gene Expression Profiling , Gene Silencing , Heterozygote , Humans , Lysine/metabolism , Methylation , Models, Molecular , Molecular Sequence Data , Polycomb Repressive Complex 2/chemistry , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Conformation , Sequence Alignment , Substrate Specificity , Transcriptional Activation
15.
ACS Chem Biol ; 9(3): 622-9, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24304166

ABSTRACT

EZH2/PRC2 catalyzes transcriptionally repressive methylation at lysine 27 of histone H3 and has been associated with numerous cancer types. Point mutations in EZH2 at Tyr641 and Ala677 identified in non-Hodgkin lymphomas alter substrate specificity and result in increased trimethylation at histone H3K27. Interestingly, EZH2/PRC2 is activated by binding H3K27me3 marks on histones, and this activation is proposed as a mechanism for self-propagation of gene silencing. Recent work has identified GSK126 as a potent, selective, SAM-competitive inhibitor of EZH2 capable of globally decreasing H3K27 trimethylation in cells. Here we show that activation of PRC2 by an H3 peptide trimethylated at K27 is primarily an effect on the rate-limiting step (kcat) with no effect on substrate binding (Km). Additionally, GSK126 is shown to have a significantly longer residence time of inhibition on the activated form of EZH2/PRC2 as compared to unactivated EZH2/PRC2. Overall inhibition constant (Ki*) values for GSK126 were determined to be as low as 93 pM and appear to be driven by slow dissociation of inhibitor from the activated enzyme. The data suggest that activation of EZH2 allows the enzyme to adopt a conformation that possesses greater affinity for GSK126. The long residence time of GSK126 may be beneficial in vivo and may result in durable target inhibition after drug systemic clearance.


Subject(s)
Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Pyridones/pharmacology , Allosteric Regulation , Allosteric Site , Binding, Competitive , Dose-Response Relationship, Drug , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Indoles/chemistry , Methylation , Nucleosomes/drug effects , Nucleosomes/enzymology , Point Mutation , Polycomb Repressive Complex 2/genetics , Protein Binding , Pyridones/chemistry , Structure-Activity Relationship , Substrate Specificity , Time Factors
16.
J Biomol Screen ; 17(10): 1279-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22904200

ABSTRACT

Histone methyltransferases (HMT) catalyze the methylation of histone tail lysines, resulting in changes in gene transcription. Misregulation of these enzymes has been associated with various forms of cancer, making this target class a potential new area for the development of novel chemotherapeutics. EZH2 is the catalytic component of the polycomb group repressive complex (PRC2), which selectively methylates histone H3 lysine 27 (H3K27). EZH2 is overexpressed in prostate, breast, bladder, brain, and other tumor types and is recognized as a molecular marker for cancer progression and aggressiveness. Several new reagents and assays were developed to aid in the identification of EZH2 inhibitors, and these were used to execute two high-throughput screening campaigns. Activity assays using either an H3K27 peptide or nucleosomes as substrates for methylation are described. The strategy to screen EZH2 with either a surrogate peptide or a natural substrate led to the identification of the same tractable series. Compounds from this series are reversible, are [(3)H]-S-adenosyl-L-methionine competitive, and display biochemical inhibition of H3K27 methylation.


Subject(s)
High-Throughput Screening Assays/methods , Nucleosomes/metabolism , Peptides/metabolism , Polycomb Repressive Complex 2/metabolism , Drug Screening Assays, Antitumor/methods , Enhancer of Zeste Homolog 2 Protein , Humans , Indicators and Reagents , Kinetics , Peptides/antagonists & inhibitors , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/chemistry , Reproducibility of Results
17.
Proc Natl Acad Sci U S A ; 109(8): 2989-94, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22323599

ABSTRACT

Trimethylation of histone H3 on lysine 27 (H3K27me3) is a repressive posttranslational modification mediated by the histone methyltransferase EZH2. EZH2 is a component of the polycomb repressive complex 2 and is overexpressed in many cancers. In B-cell lymphomas, its substrate preference is frequently altered through somatic mutation of the EZH2 Y641 residue. Herein, we identify mutation of EZH2 A677 to a glycine (A677G) among lymphoma cell lines and primary tumor specimens. Similar to Y641 mutant cell lines, an A677G mutant cell line revealed aberrantly elevated H3K27me3 and decreased monomethylated H3K27 (H3K27me1) and dimethylated H3K27 (H3K27me2). A677G EZH2 possessed catalytic activity with a substrate specificity that was distinct from those of both WT EZH2 and Y641 mutants. Whereas WT EZH2 displayed a preference for substrates with less methylation [unmethylated H3K27 (H3K27me0):me1:me2 k(cat)/K(m) ratio = 9:6:1] and Y641 mutants preferred substrates with greater methylation (H3K27me0:me1:me2 k(cat)/K(m) ratio = 1:2:13), the A677G EZH2 demonstrated nearly equal efficiency for all three substrates (H3K27me0:me1:me2 k(cat)/K(m) ratio = 1.1:0.6:1). When transiently expressed in cells, A677G EZH2, but not WT EZH2, increased global H3K27me3 and decreased H3K27me2. Structural modeling of WT and mutant EZH2 suggested that the A677G mutation acquires the ability to methylate H3K27me2 through enlargement of the lysine tunnel while preserving activity with H3K27me0/me1 substrates through retention of the Y641 residue that is crucial for orientation of these smaller substrates. This mutation highlights the interplay between Y641 and A677 residues in the substrate specificity of EZH2 and identifies another lymphoma patient population that harbors an activating mutation of EZH2.


Subject(s)
Alanine/genetics , DNA-Binding Proteins/genetics , Histones/metabolism , Lymphoma, B-Cell/enzymology , Lymphoma, B-Cell/genetics , Lysine/metabolism , Mutation/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Line, Tumor , DNA Mutational Analysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Glycine/genetics , Heterozygote , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Polycomb Repressive Complex 2 , Substrate Specificity , Transcription Factors/chemistry , Transcription Factors/metabolism
18.
Biochem J ; 436(2): 363-9, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21410436

ABSTRACT

The HIF (hypoxia-inducible factor) plays a central regulatory role in oxygen homoeostasis. HIF proteins are regulated by three Fe(II)- and α-KG (α-ketoglutarate)-dependent prolyl hydroxylase enzymes [PHD (prolyl hydroxylase domain) isoenzymes 1-3 or PHD1, PHD2 and PHD3] and one asparaginyl hydroxylase [FIH (factor inhibiting HIF)]. The prolyl hydroxylases control the abundance of HIF through oxygen-dependent hydroxylation of specific proline residues in HIF proteins, triggering subsequent ubiquitination and proteasomal degradation. FIH inhibits the HIF transcription activation through asparagine hydroxylation. Understanding the precise roles and regulation of these four Fe(II)- and α-KG-dependent hydroxylases is of great importance. In the present paper, we report the biochemical characterization of the first HIF protein substrates that contain the CODDD (C-terminal oxygen-dependent degradation domain), the NODDD (N-terminal oxygen-dependent degradation domain) and the CAD (C-terminal transactivation domain). Using LC-MS/MS (liquid chromatography-tandem MS) detection, we show that all three PHD isoenzymes have a strong preference for hydroxylation of the CODDD proline residue over the NODDD proline residue and the preference is observed for both HIF1α and HIF2α protein substrates. In addition, steady-state kinetic analyses show differential substrate selectivity for HIF and α-KG in reference to the three PHD isoforms and FIH.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Binding Sites , Humans , Hydroxylation , Isoenzymes/chemistry , Isoenzymes/metabolism , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/metabolism , Substrate Specificity
19.
Biochemistry ; 47(43): 11165-7, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18834144

ABSTRACT

Prolyl hydroxylase domain proteins (PHD isozymes 1-3) regulate levels of the alpha-subunit of the hypoxia inducible factor (HIF) through proline hydroxylation, earmarking HIFalpha for proteosome-mediated degradation. Under hypoxic conditions, HIF stabilization leads to enhanced transcription and regulation of a multitude of processes, including erythropoiesis. Herein, we examine the biochemical characterization of PHD2 variants, Arg371His and Pro317Arg, identified from patients with familial erythrocytosis. The variants display differential effects on catalytic rate and substrate binding, implying that partial inhibition or selective inhibition with regard to HIFalpha isoforms of PHD2 could result in the phenotype displayed by patients with familial erythrocytosis.


Subject(s)
Genetic Variation , Polycythemia/genetics , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/genetics , Amino Acid Motifs , Amino Acid Substitution , Arginine/metabolism , Binding Sites , Catalysis , Crystallography, X-Ray , Histidine/metabolism , Humans , Hydrogen Bonding , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Models, Molecular , Polycythemia/metabolism , Procollagen-Proline Dioxygenase/metabolism , Proline/chemistry , Proline/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity
20.
Anal Biochem ; 353(1): 69-74, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16643838

ABSTRACT

The Fe(II)/2-oxoglutarate-dependent dioxygenases are a catalytically diverse family of nonheme iron enzymes that oxidize their primary substrates while decomposing the 2-oxoglutarate cosubstrate to form succinate and CO(2). We report a generic assay for these enzymes that uses succinyl-coenzyme A synthetase, pyruvate kinase, and lactate dehydrogenase to couple the formation of the product succinate to the conversion of reduced nicotinamide adenine dinucleotide to nicotinamide adenine dinucleotide. We demonstrate the utility of this new method by measuring the kinetic parameters of two bacterial Fe(II)/2-oxoglutarate-dependent dioxygenases. Significantly, this method can be used to investigate both the productive turnover reactions and the nonproductive "uncoupled" decarboxylation reactions of this enzyme family, as demonstrated by using wild-type and variant forms of 2-oxoglutarate-dependent taurine dioxygenase. This assay is amenable to miniaturization and easily adapted to a format suitable for high-throughput screening; thus, it will be a valuable tool to study Fe(II)/2-oxoglutarate-dependent dioxygenases.


Subject(s)
Dioxygenases/metabolism , Iron/chemistry , Iron/metabolism , Ketoglutaric Acids/metabolism , Succinic Acid/analysis , Ferrous Compounds/chemistry , Kinetics , L-Lactate Dehydrogenase/metabolism , NAD/metabolism , Oxidation-Reduction , Pyruvate Kinase/metabolism , Spectrophotometry , Substrate Specificity , Succinate-CoA Ligases/metabolism , Succinic Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...