Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 29(4): 555-571.e11, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34715055

ABSTRACT

Canonical targeting of Polycomb repressive complex 1 (PRC1) to repress developmental genes is mediated by cell-type-specific, paralogous chromobox (CBX) proteins (CBX2, 4, 6, 7, and 8). Based on their central role in silencing and their dysregulation associated with human disease including cancer, CBX proteins are attractive targets for small-molecule chemical probe development. Here, we have used a quantitative and target-specific cellular assay to discover a potent positive allosteric modulator (PAM) of CBX8. The PAM activity of UNC7040 antagonizes H3K27me3 binding by CBX8 while increasing interactions with nucleic acids. We show that treatment with UNC7040 leads to efficient and selective eviction of CBX8-containing PRC1 from chromatin, loss of silencing, and reduced proliferation across different cancer cell lines. Our discovery and characterization of UNC7040 not only reveals the most cellularly potent CBX8-specific chemical probe to date, but also corroborates a mechanism of Polycomb regulation by non-specific CBX nucleotide binding activity.


Subject(s)
Neoplasms , Polycomb Repressive Complex 1 , Cell Cycle Proteins/metabolism , Chromatin , Histones/metabolism , Humans , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Protein Binding
2.
Cell Chem Biol ; 26(10): 1365-1379.e22, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31422906

ABSTRACT

Polycomb-directed repression of gene expression is frequently misregulated in human diseases. A quantitative and target-specific cellular assay was utilized to discover the first potent positive allosteric modulator (PAM) peptidomimetic, UNC4976, of nucleic acid binding by CBX7, a chromodomain methyl-lysine reader of Polycomb repressive complex 1. The PAM activity of UNC4976 resulted in enhanced efficacy across three orthogonal cellular assays by simultaneously antagonizing H3K27me3-specific recruitment of CBX7 to target genes while increasing non-specific binding to DNA and RNA. PAM activity thereby reequilibrates PRC1 away from H3K27me3 target regions. Together, our discovery and characterization of UNC4976 not only revealed the most cellularly potent PRC1-specific chemical probe to date, but also uncovers a potential mechanism of Polycomb regulation with implications for non-histone lysine methylated interaction partners.


Subject(s)
Drug Discovery , Peptidomimetics/pharmacology , Polycomb Repressive Complex 1/metabolism , Allosteric Regulation/drug effects , Animals , HEK293 Cells , HeLa Cells , Humans , Mice , Peptidomimetics/chemistry
5.
ACS Chem Biol ; 11(9): 2475-83, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27356154

ABSTRACT

Efforts to develop strategies for small-molecule chemical probe discovery against the readers of the methyl-lysine (Kme) post-translational modification have been met with limited success. Targeted disruption of these protein-protein interactions via peptidomimetic inhibitor optimization is a promising alternative to small-molecule hit discovery; however, recognition of identical peptide motifs by multiple Kme reader proteins presents a unique challenge in the development of selective Kme reader chemical probes. These selectivity challenges are exemplified by the Polycomb repressive complex 1 (PRC1) chemical probe, UNC3866, which demonstrates submicromolar off-target affinity toward the non-PRC1 chromodomains CDYL2 and CDYL. Moreover, since peptidomimetics are challenging subjects for structure-activity relationship (SAR) studies, traditional optimization of UNC3866 would prove costly and time-consuming. Herein, we report a broadly applicable strategy for the affinity-based, target-class screening of chromodomains via the repurposing of UNC3866 in an efficient, combinatorial peptide library. A first-generation library yielded UNC4991, a UNC3866 analogue that exhibits a distinct selectivity profile while maintaining submicromolar affinity toward the CDYL chromodomains. Additionally, in vitro pull-down experiments from HeLa nuclear lysates further demonstrate the selectivity and utility of this compound for future elucidation of CDYL protein function.


Subject(s)
Molecular Probes/chemistry , Proteins/chemistry , Ligands , Protein Binding , Structure-Activity Relationship
6.
Nat Chem Biol ; 12(3): 180-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26807715

ABSTRACT

We report the design and characterization of UNC3866, a potent antagonist of the methyllysine (Kme) reading function of the Polycomb CBX and CDY families of chromodomains. Polycomb CBX proteins regulate gene expression by targeting Polycomb repressive complex 1 (PRC1) to sites of H3K27me3 via their chromodomains. UNC3866 binds the chromodomains of CBX4 and CBX7 most potently, with a K(d) of ∼100 nM for each, and is 6- to 18-fold selective as compared to seven other CBX and CDY chromodomains while being highly selective over >250 other protein targets. X-ray crystallography revealed that UNC3866's interactions with the CBX chromodomains closely mimic those of the methylated H3 tail. UNC4195, a biotinylated derivative of UNC3866, was used to demonstrate that UNC3866 engages intact PRC1 and that EED incorporation into PRC1 is isoform dependent in PC3 prostate cancer cells. Finally, UNC3866 inhibits PC3 cell proliferation, consistent with the known ability of CBX7 overexpression to confer a growth advantage, whereas UNC4219, a methylated negative control compound, has negligible effects.


Subject(s)
Oligopeptides/pharmacology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Animals , Biological Availability , Biotinylation , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Gene Expression Regulation/genetics , Humans , Isomerism , Ligases , Male , Methylation , Mice , Models, Molecular , Polycomb Repressive Complex 1/biosynthesis , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Substrate Specificity , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
7.
ACS Chem Biol ; 11(3): 722-8, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26317848

ABSTRACT

L3MBTL3 recognizes mono- and dimethylated lysine residues on histone tails. The recently reported X-ray cocrystal structures of the chemical probe UNC1215 and inhibitor UNC2533 bound to the methyl-lysine reading MBT domains of L3MBTL3 demonstrate a unique and flexible 2:2 dimer mode of recognition. In this study, we describe our in vitro analysis of L3MBTL3 dimerization via its MBT domains and additionally show that this dimerization occurs within a cellular context in the absence of small molecule ligands. Furthermore, mutations to the first and second MBT domains abrogated L3MBTL3 dimerization both in vitro and in cells. These observations are consistent with the hypothesis that L3MBTL3 engages methylated histone tails as a dimer while carrying out its normal function and provides an explanation for the presence of repeated MBT domains within L3MBTL3.


Subject(s)
DNA-Binding Proteins/metabolism , Benzamides , Biotin , Cell-Free System , DNA-Binding Proteins/genetics , HeLa Cells , Histones , Humans , Ligands , Molecular Structure , Mutation , Piperidines , Protein Domains , Protein Multimerization
8.
ACS Chem Biol ; 10(4): 1072-81, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25590533

ABSTRACT

Improving our understanding of the role of chromatin regulators in the initiation, development, and suppression of cancer and other devastating diseases is critical, as they are integral players in regulating DNA integrity and gene expression. Developing small molecule inhibitors for this target class with cellular activity is a crucial step toward elucidating their specific functions. We specifically targeted the DNA damage response protein, 53BP1, which uses its tandem tudor domain to recognize histone H4 dimethylated on lysine 20 (H4K20me2), a modification related to double-strand DNA breaks. Through a cross-screening approach, we identified UNC2170 (1) as a micromolar ligand of 53BP1, which demonstrates at least 17-fold selectivity for 53BP1 as compared to other methyl-lysine (Kme) binding proteins tested. Structural studies revealed that the tert-butyl amine of UNC2170 anchors the compound in the methyl-lysine (Kme) binding pocket of 53BP1, making it competitive with endogenous Kme substrates. X-ray crystallography also demonstrated that UNC2170 binds at the interface of two tudor domains of a 53BP1 dimer. Importantly, this compound functions as a 53BP1 antagonist in cellular lysates and shows cellular activity by suppressing class switch recombination, a process which requires a functional 53BP1 tudor domain. These results demonstrate that UNC2170 is a functionally active, fragment-like ligand for 53BP1.


Subject(s)
Benzamides/metabolism , Diamines/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Animals , B-Lymphocytes/drug effects , Benzamides/chemistry , Benzamides/pharmacology , Binding Sites , Chromatin/metabolism , Crystallography, X-Ray , Diamines/chemistry , Diamines/pharmacology , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Ligands , Magnetic Resonance Spectroscopy , Mice, Inbred C57BL , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Structure-Activity Relationship , Tumor Suppressor p53-Binding Protein 1
9.
PLoS One ; 9(12): e116101, 2014.
Article in English | MEDLINE | ID: mdl-25542006

ABSTRACT

Ror2 is a Wnt ligand receptor that is overexpressed in a variety of tumors including clear cell renal cell carcinoma (ccRCC). Here we demonstrate that expression of wild type Ror2 results in increased tumorigenic properties in in vitro cell culture and in vivo xenograft models. In addition, Ror2 expression produced positive changes in both cell migration and invasion, which were dependent on matrix metalloprotease 2 (MMP2) activity. Mutations in key regions of the kinase domain of Ror2 resulted in the abrogation of increased tumor growth, cell migration, and cell invasion observed with expression of wild-type Ror2. Finally, we examined Ror2 expression as a prognostic biomarker for ccRCC utilizing the TCGA ccRCC dataset. High expression of Ror2 showed a significant correlation with higher clinical stage, nuclear grade, and tumor stage. Furthermore, high expression of Ror2 in ccRCC patients correlated with significant lower overall survival, cancer specific survival, and recurrence free survival. Together, these findings suggest that Ror2 plays a central role in influencing the ccRCC phenotype, and can be considered as a negative prognostic biomarker and potential therapeutic target in this cancer.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Kidney/pathology , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Animals , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Kidney/metabolism , Kidney Neoplasms/diagnosis , Kidney Neoplasms/genetics , Mice, Nude , Mutation , Neoplasm Invasiveness/diagnosis , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prognosis , Receptor Tyrosine Kinase-like Orphan Receptors/analysis
10.
J Med Chem ; 57(15): 6822-33, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25032507

ABSTRACT

The lysine methyltransferase SETD8 is the only known methyltransferase that catalyzes monomethylation of histone H4 lysine 20 (H4K20). Monomethylation of H4K20 has been implicated in regulating diverse biological processes including the DNA damage response. In addition to H4K20, SETD8 monomethylates non-histone substrates including proliferating cell nuclear antigen (PCNA) and promotes carcinogenesis by deregulating PCNA expression. However, selective inhibitors of SETD8 are scarce. The only known selective inhibitor of SETD8 to date is nahuoic acid A, a marine natural product, which is competitive with the cofactor. Here, we report the discovery of the first substrate-competitive inhibitor of SETD8, UNC0379 (1). This small-molecule inhibitor is active in multiple biochemical assays. Its affinity to SETD8 was confirmed by ITC (isothermal titration calorimetry) and SPR (surface plasmon resonance) studies. Importantly, compound 1 is selective for SETD8 over 15 other methyltransferases. We also describe structure-activity relationships (SAR) of this series.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Pyrrolidines/chemistry , Quinazolines/chemistry , Calorimetry , Histone-Lysine N-Methyltransferase/chemistry , Pyrrolidines/chemical synthesis , Quinazolines/chemical synthesis , Structure-Activity Relationship , Surface Plasmon Resonance
11.
J Med Chem ; 56(18): 7358-71, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24040942

ABSTRACT

Lysine methylation is a key epigenetic mark, the dysregulation of which is linked to many diseases. Small-molecule antagonism of methyl-lysine (Kme) binding proteins that recognize such epigenetic marks can improve our understanding of these regulatory mechanisms and potentially validate Kme binding proteins as drug-discovery targets. We previously reported the discovery of 1 (UNC1215), the first potent and selective small-molecule chemical probe of a methyl-lysine reader protein, L3MBTL3, which antagonizes the mono- and dimethyl-lysine reading function of L3MBTL3. The design, synthesis, and structure-activity relationship studies that led to the discovery of 1 are described herein. These efforts established the requirements for potent L3MBTL3 binding and enabled the design of novel antagonists, such as compound 2 (UNC1679), that maintain in vitro and cellular potency with improved selectivity against other MBT-containing proteins. The antagonists described were also found to effectively interact with unlabeled endogenous L3MBTL3 in cells.


Subject(s)
DNA-Binding Proteins/metabolism , Lysine/metabolism , Small Molecule Libraries/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , Drug Design , HEK293 Cells , Humans , Inhibitory Concentration 50 , Ligands , Models, Molecular , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Substrate Specificity
12.
ACS Chem Biol ; 8(6): 1324-34, 2013.
Article in English | MEDLINE | ID: mdl-23614352

ABSTRACT

EZH2 or EZH1 is the catalytic subunit of the polycomb repressive complex 2 that catalyzes methylation of histone H3 lysine 27 (H3K27). The trimethylation of H3K27 (H3K27me3) is a transcriptionally repressive post-translational modification. Overexpression of EZH2 and hypertrimethylation of H3K27 have been implicated in a number of cancers. Several selective inhibitors of EZH2 have been reported recently. Herein we disclose UNC1999, the first orally bioavailable inhibitor that has high in vitro potency for wild-type and mutant EZH2 as well as EZH1, a closely related H3K27 methyltransferase that shares 96% sequence identity with EZH2 in their respective catalytic domains. UNC1999 was highly selective for EZH2 and EZH1 over a broad range of epigenetic and non-epigenetic targets, competitive with the cofactor SAM and non-competitive with the peptide substrate. This inhibitor potently reduced H3K27me3 levels in cells and selectively killed diffused large B cell lymphoma cell lines harboring the EZH2(Y641N) mutant. Importantly, UNC1999 was orally bioavailable in mice, making this inhibitor a valuable tool for investigating the role of EZH2 and EZH1 in chronic animal studies. We also designed and synthesized UNC2400, a close analogue of UNC1999 with potency >1,000-fold lower than that of UNC1999 as a negative control for cell-based studies. Finally, we created a biotin-tagged UNC1999 (UNC2399), which enriched EZH2 in pull-down studies, and a UNC1999-dye conjugate (UNC2239) for co-localization studies with EZH2 in live cells. Taken together, these compounds represent a set of useful tools for the biomedical community to investigate the role of EZH2 and EZH1 in health and disease.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Administration, Oral , Animals , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Histones/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/enzymology , Male , Methylation/drug effects , Mice , Polycomb Repressive Complex 2/metabolism
13.
Nat Chem Biol ; 9(3): 184-91, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23292653

ABSTRACT

We describe the discovery of UNC1215, a potent and selective chemical probe for the methyllysine (Kme) reading function of L3MBTL3, a member of the malignant brain tumor (MBT) family of chromatin-interacting transcriptional repressors. UNC1215 binds L3MBTL3 with a K(d) of 120 nM, competitively displacing mono- or dimethyllysine-containing peptides, and is greater than 50-fold more potent toward L3MBTL3 than other members of the MBT family while also demonstrating selectivity against more than 200 other reader domains examined. X-ray crystallography identified a unique 2:2 polyvalent mode of interaction between UNC1215 and L3MBTL3. In cells, UNC1215 is nontoxic and directly binds L3MBTL3 via the Kme-binding pocket of the MBT domains. UNC1215 increases the cellular mobility of GFP-L3MBTL3 fusion proteins, and point mutants that disrupt the Kme-binding function of GFP-L3MBTL3 phenocopy the effects of UNC1215 on localization. Finally, UNC1215 was used to reveal a new Kme-dependent interaction of L3MBTL3 with BCLAF1, a protein implicated in DNA damage repair and apoptosis.


Subject(s)
Benzamides/pharmacology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Drug Discovery , Lysine/analogs & derivatives , Molecular Probes/pharmacology , Piperidines/pharmacology , Benzamides/chemistry , Benzamides/metabolism , Binding, Competitive/drug effects , Crystallography, X-Ray , DNA-Binding Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Lysine/antagonists & inhibitors , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Molecular Probes/chemistry , Molecular Probes/metabolism , Molecular Structure , Piperidines/chemistry , Piperidines/metabolism , Protein Structure, Tertiary , Repressor Proteins/metabolism , Structure-Activity Relationship , Tumor Suppressor Proteins/metabolism
14.
Medchemcomm ; 4(11): 1501-1507, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24466405

ABSTRACT

We recently reported the discovery of UNC1215, a potent and selective chemical probe for the L3MBTL3 methyllysine reader domain. In this article, we describe the development of structure-activity relationships (SAR) of a second series of potent L3MBTL3 antagonists which evolved from the structure of the chemical probe UNC1215. These compounds are selective for L3MBTL3 against a panel of methyllysine reader proteins, particularly the related MBT family proteins, L3MBTL1 and MBTD1. A co-crystal structure of L3MBTL3 and one of the most potent compounds suggests that the L3MBTL3 dimer rotates about the dimer interface to accommodate ligand binding.

15.
Nat Chem Biol ; 7(8): 566-74, 2011 Jul 10.
Article in English | MEDLINE | ID: mdl-21743462

ABSTRACT

Protein lysine methyltransferases G9a and GLP modulate the transcriptional repression of a variety of genes via dimethylation of Lys9 on histone H3 (H3K9me2) as well as dimethylation of non-histone targets. Here we report the discovery of UNC0638, an inhibitor of G9a and GLP with excellent potency and selectivity over a wide range of epigenetic and non-epigenetic targets. UNC0638 treatment of a variety of cell lines resulted in lower global H3K9me2 levels, equivalent to levels observed for small hairpin RNA knockdown of G9a and GLP with the functional potency of UNC0638 being well separated from its toxicity. UNC0638 markedly reduced the clonogenicity of MCF7 cells, reduced the abundance of H3K9me2 marks at promoters of known G9a-regulated endogenous genes and disproportionately affected several genomic loci encoding microRNAs. In mouse embryonic stem cells, UNC0638 reactivated G9a-silenced genes and a retroviral reporter gene in a concentration-dependent manner without promoting differentiation.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/metabolism , Quinazolines/pharmacology , Animals , Cell Line , Gene Silencing , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Molecular Structure
16.
J Med Chem ; 54(7): 2504-11, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21417280

ABSTRACT

Proteins which bind methylated lysines ("readers" of the histone code) are important components in the epigenetic regulation of gene expression and can also modulate other proteins that contain methyl-lysine such as p53 and Rb. Recognition of methyl-lysine marks by MBT domains leads to compaction of chromatin and a repressed transcriptional state. Antagonists of MBT domains would serve as probes to interrogate the functional role of these proteins and initiate the chemical biology of methyl-lysine readers as a target class. Small-molecule MBT antagonists were designed based on the structure of histone peptide-MBT complexes and their interaction with MBT domains determined using a chemiluminescent assay and ITC. The ligands discovered antagonize native histone peptide binding, exhibiting 5-fold stronger binding affinity to L3MBTL1 than its preferred histone peptide. The first cocrystal structure of a small molecule bound to L3MBTL1 was determined and provides new insights into binding requirements for further ligand design.


Subject(s)
Lysine/metabolism , Nuclear Proteins/metabolism , Small Molecule Libraries/metabolism , Binding Sites , Calorimetry , Drug Discovery , High-Throughput Screening Assays , Humans , Ligands , Luminescent Measurements , Methylation , Models, Molecular , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Peptidomimetics/chemistry , Peptidomimetics/metabolism , Peptidomimetics/pharmacology , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
17.
J Am Chem Soc ; 133(14): 5357-62, 2011 Apr 13.
Article in English | MEDLINE | ID: mdl-21428286

ABSTRACT

Histone lysine methylation (Kme) encodes essential information modulating many biological processes including gene expression and transcriptional regulation. However, the atomic-level recognition mechanisms of methylated histones by their respective adaptor proteins are still elusive. For instance, it is unclear how L3MBTL1, a methyl-lysine histone code reader, recognizes equally well both mono- and dimethyl marks but ignores unmodified and trimethylated lysine residues. We made use of molecular dynamics (MD) and free energy perturbation (FEP) techniques in order to investigate the energetics and dynamics of the methyl-lysine recognition. Isothermal titration calorimetry (ITC) was employed to experimentally validate the computational findings. Both computational and experimental methods were applied to a set of designed "biophysical" probes that mimic the shape of a single lysine residue and reproduce the binding affinities of cognate histone peptides. Our results suggest that, besides forming favorable interactions, the L3MBTL1 binding pocket energetically penalizes both methylation states and has most probably evolved as a "compromise" that nonoptimally fits to both mono- and dimethyl-lysine marks.


Subject(s)
Lysine/chemistry , Lysine/metabolism , Molecular Dynamics Simulation , Molecular Probes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Binding Sites , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Histones/chemistry , Histones/metabolism , Methylation , Molecular Probes/chemistry , Protein Conformation , Protein Structure, Tertiary , Thermodynamics
18.
Chem Biol ; 17(7): 695-704, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20659682

ABSTRACT

The discovery of small molecules targeting the >80 enzymes that add (methyltransferases) or remove (demethylases) methyl marks from lysine and arginine residues, most notably present in histone tails, may yield unprecedented chemotherapeutic agents and facilitate regenerative medicine. To better enable chemical exploration of these proteins, we have developed a highly quantitative microfluidic capillary electrophoresis assay to enable full mechanistic studies of these enzymes and the kinetics of their inhibition. This technology separates small biomolecules, i.e., peptides, based on their charge-to-mass ratio. Methylation, however, does not alter the charge of peptide substrates. To overcome this limitation, we have employed a methylation-sensitive endoproteinase strategy to separate methylated from unmethylated peptides. The assay was validated on a lysine methyltransferase (G9a) and a lysine demethylase (LSD1) and was employed to investigate the inhibition of G9a by small molecules.


Subject(s)
Electrophoresis, Capillary/instrumentation , Enzyme Assays/methods , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Microfluidic Analytical Techniques , Amino Acid Sequence , Binding, Competitive , Drug Evaluation, Preclinical , Enzyme Assays/instrumentation , Histone Demethylases/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Kinetics , Methylation , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , S-Adenosylmethionine/metabolism , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacology
19.
J Med Chem ; 53(15): 5844-57, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20614940

ABSTRACT

Protein lysine methyltransferase G9a, which catalyzes methylation of lysine 9 of histone H3 (H3K9) and lysine 373 (K373) of p53, is overexpressed in human cancers. Genetic knockdown of G9a inhibits cancer cell growth, and the dimethylation of p53 K373 results in the inactivation of p53. Initial SAR exploration of the 2,4-diamino-6,7-dimethoxyquinazoline template represented by 3a (BIX01294), a selective small molecule inhibitor of G9a and GLP, led to the discovery of 10 (UNC0224) as a potent G9a inhibitor with excellent selectivity. A high resolution X-ray crystal structure of the G9a-10 complex, the first cocrystal structure of G9a with a small molecule inhibitor, was obtained. On the basis of the structural insights revealed by this cocrystal structure, optimization of the 7-dimethylaminopropoxy side chain of 10 resulted in the discovery of 29 (UNC0321) (Morrison K(i) = 63 pM), which is the first G9a inhibitor with picomolar potency and the most potent G9a inhibitor to date.


Subject(s)
Azepines/chemical synthesis , Histocompatibility Antigens/chemistry , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Models, Molecular , Quinazolines/chemical synthesis , Azepines/chemistry , Crystallography, X-Ray , Drug Design , Humans , Protein Binding , Protein Conformation , Quinazolines/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL