Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Bioorg Med Chem Lett ; 98: 129546, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37944866

ABSTRACT

Epigenetic proteins containing YEATS domains (YD) are an emerging target class in drug discovery. Described herein are the discovery and characterization efforts associated with PFI-6, a new chemical probe for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). For hit identification, fragment-like mimetics of endogenous YD ligands (crotonylated histone-containing proteins), were synthesized via parallel medicinal chemistry (PMC) and screened for MLLT1 binding. Subsequent SAR studies led to iterative MLLT1/3 binding and selectivity improvements, culminating in the discovery of PFI-6. PFI-6 demonstrates good affinity and selectivity for MLLT1/3 vs. other human YD proteins (YEATS2/4) and engages MLLT3 in cells. Small-molecule X-ray co-crystal structures of two molecules, including PFI-6, bound to the YD of MLLT1/3 are also described. PFI-6 may be a useful tool molecule to better understand the biological effects associated with modulation of MLLT1/3.


Subject(s)
Histones , Nuclear Proteins , Humans , Nuclear Proteins/metabolism , Histones/metabolism , Protein Domains , Drug Discovery , Neoplasm Proteins/metabolism , Transcription Factors/metabolism
2.
Nat Chem Biol ; 15(8): 822-829, 2019 08.
Article in English | MEDLINE | ID: mdl-31285596

ABSTRACT

Here, we report the fragment-based discovery of BI-9321, a potent, selective and cellular active antagonist of the NSD3-PWWP1 domain. The human NSD3 protein is encoded by the WHSC1L1 gene located in the 8p11-p12 amplicon, frequently amplified in breast and squamous lung cancer. Recently, it was demonstrated that the PWWP1 domain of NSD3 is required for the viability of acute myeloid leukemia cells. To further elucidate the relevance of NSD3 in cancer biology, we developed a chemical probe, BI-9321, targeting the methyl-lysine binding site of the PWWP1 domain with sub-micromolar in vitro activity and cellular target engagement at 1 µM. As a single agent, BI-9321 downregulates Myc messenger RNA expression and reduces proliferation in MOLM-13 cells. This first-in-class chemical probe BI-9321, together with the negative control BI-9466, will greatly facilitate the elucidation of the underexplored biological function of PWWP domains.


Subject(s)
Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , CRISPR-Cas Systems , Cell Line , Cell Proliferation/drug effects , Cell Survival , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
3.
Mol Cell ; 52(1): 37-51, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24076217

ABSTRACT

The mechanisms that underlie and dictate the different biological outcomes of E2F-1 activity have yet to be elucidated. We describe the residue-specific methylation of E2F-1 by the asymmetric dimethylating protein arginine methyltransferase 1 (PRMT1) and symmetric dimethylating PRMT5 and relate the marks to different functional consequences of E2F-1 activity. Methylation by PRMT1 hinders methylation by PRMT5, which augments E2F-1-dependent apoptosis, whereas PRMT5-dependent methylation favors proliferation by antagonizing methylation by PRMT1. The ability of E2F-1 to prompt apoptosis in DNA damaged cells coincides with enhanced PRMT1 methylation. In contrast, cyclin A binding to E2F-1 impedes PRMT1 methylation and augments PRMT5 methylation, thus ensuring that E2F-1 is locked into its cell-cycle progression mode. The Tudor domain protein p100-TSN reads the symmetric methylation mark, and binding of p100-TSN downregulates E2F-1 apoptotic activity. Our results define an exquisite level of precision in the reader-writer interplay that governs the biological outcome of E2F-1 activity.


Subject(s)
Apoptosis , Cell Proliferation , E2F1 Transcription Factor/metabolism , Protein Processing, Post-Translational , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Amino Acid Motifs , Arginine , Cell Line, Tumor , Chromatin Assembly and Disassembly , Cyclin A/metabolism , DNA Damage , E2F1 Transcription Factor/genetics , Gene Expression Regulation , Humans , Methylation , Promoter Regions, Genetic , Protein Binding , Protein-Arginine N-Methyltransferases/genetics , RNA Interference , Repressor Proteins/genetics , Signal Transduction , Transcription, Genetic , Transfection
4.
J Am Chem Soc ; 141(22): 8951-8968, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31060360

ABSTRACT

Covalent probes can display unmatched potency, selectivity, and duration of action; however, their discovery is challenging. In principle, fragments that can irreversibly bind their target can overcome the low affinity that limits reversible fragment screening, but such electrophilic fragments were considered nonselective and were rarely screened. We hypothesized that mild electrophiles might overcome the selectivity challenge and constructed a library of 993 mildly electrophilic fragments. We characterized this library by a new high-throughput thiol-reactivity assay and screened them against 10 cysteine-containing proteins. Highly reactive and promiscuous fragments were rare and could be easily eliminated. In contrast, we found hits for most targets. Combining our approach with high-throughput crystallography allowed rapid progression to potent and selective probes for two enzymes, the deubiquitinase OTUB2 and the pyrophosphatase NUDT7. No inhibitors were previously known for either. This study highlights the potential of electrophile-fragment screening as a practical and efficient tool for covalent-ligand discovery.


Subject(s)
Drug Evaluation, Preclinical/methods , Electrons , HEK293 Cells , Humans , Ligands , Models, Molecular , Molecular Weight , Protein Conformation , Time Factors
5.
J Org Chem ; 84(17): 11068-11079, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31409063

ABSTRACT

The atom transfer radical addition reaction catalyzed by triphenylphosphine and copper(I) halide is described. The reaction proceeds under irradiation with 365 nm light using a light-emitting diode and was performed in regular glassware. The proposed mechanism involves the formation of quaternary phosphonium salt, which undergoes single electron reduction by copper(I) salt via photo-induced electron transfer. The method works well for terminal alkenes and activated organic halides such as esters of bromo- and iodoacetic acid and bromoacetonitrile. gem-Difluorinated styrenes, for which atom transfer reactions are rare, also proved to be good substrates for this phosphine-/copper-catalyzed protocol.

6.
Angew Chem Int Ed Engl ; 58(4): 1007-1012, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30589164

ABSTRACT

Bromodomain-containing proteins are epigenetic modulators involved in a wide range of cellular processes, from recruitment of transcription factors to pathological disruption of gene regulation and cancer development. Since the druggability of these acetyl-lysine reader domains was established, efforts were made to develop potent and selective inhibitors across the entire family. Here we report the development of a small molecule-based approach to covalently modify recombinant and endogenous bromodomain-containing proteins by targeting a conserved lysine and a tyrosine residue in the variable ZA or BC loops. Moreover, the addition of a reporter tag allowed in-gel visualization and pull-down of the desired bromodomains.


Subject(s)
Carbamates/chemistry , Histones/chemistry , Lysine/chemistry , Protein Domains , Pyridazines/chemistry , Triazoles/chemistry , Acetylation , Amino Acid Sequence , Binding Sites , Conserved Sequence , Molecular Docking Simulation , Protein Binding
7.
Angew Chem Int Ed Engl ; 58(2): 515-519, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30431220

ABSTRACT

Histone lysine demethylases (KDMs) are involved in the dynamic regulation of gene expression and they play a critical role in several biological processes. Achieving selectivity over the different KDMs has been a major challenge for KDM inhibitor development. Here we report potent and selective KDM5 covalent inhibitors designed to target cysteine residues only present in the KDM5 sub-family. The covalent binding to the targeted proteins was confirmed by MS and time-dependent inhibition. Additional competition assays show that compounds were non 2-OG competitive. Target engagement and ChIP-seq analysis showed that the compounds inhibited the KDM5 members in cells at nano- to micromolar levels and induce a global increase of the H3K4me3 mark at transcriptional start sites.

8.
Nat Chem Biol ; 12(7): 504-10, 2016 07.
Article in English | MEDLINE | ID: mdl-27159579

ABSTRACT

Bromodomain-containing proteins of the BET family recognize histone lysine acetylation and mediate transcriptional activation of target genes such as the MYC oncogene. Pharmacological inhibitors of BET domains promise therapeutic benefits in a variety of cancers. We performed a high-diversity chemical compound screen for agents capable of modulating BRD4-dependent heterochromatization of a generic reporter in human cells. In addition to known and new compounds targeting BRD4, we identified small molecules that mimic BRD4 inhibition without direct engagement. One such compound was a potent inhibitor of the second bromodomain of TAF1. Using this inhibitor, we discovered that TAF1 synergizes with BRD4 to control proliferation of cancer cells, making TAF1 an attractive epigenetic target in cancers driven by MYC.


Subject(s)
Chromatin/chemistry , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Small Molecule Libraries/pharmacology , TATA-Binding Protein Associated Factors/antagonists & inhibitors , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/antagonists & inhibitors , Transcription Factor TFIID/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Cell Cycle Proteins , Cell Line , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/genetics , Chromatin/metabolism , Histone Acetyltransferases/chemistry , Humans , Molecular Structure , Nuclear Proteins/chemistry , Protein Domains/drug effects , Small Molecule Libraries/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Transcription Factors/chemistry
9.
Org Biomol Chem ; 16(2): 336, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29256564

ABSTRACT

Correction for 'Synthesis of kinase inhibitors containing a pentafluorosulfanyl moiety' by Supojjanee Sansook et al., Org. Biomol. Chem., 2017, 15, 8655-8660.

10.
Bioorg Med Chem ; 26(11): 2965-2972, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29567296

ABSTRACT

The polyadenosine-diphosphate-ribose polymerase 14 (PARP14) has been implicated in DNA damage response pathways for homologous recombination. PARP14 contains three (ADP ribose binding) macrodomains (MD) whose exact contribution to overall PARP14 function in pathology remains unclear. A medium throughput screen led to the identification of N-(2(-9H-carbazol-1-yl)phenyl)acetamide (GeA-69, 1) as a novel allosteric PARP14 MD2 (second MD of PARP14) inhibitor. We herein report medicinal chemistry around this novel chemotype to afford a sub-micromolar PARP14 MD2 inhibitor. This chemical series provides a novel starting point for further development of PARP14 chemical probes.


Subject(s)
Cysteine Endopeptidases/chemistry , Drug Discovery , Poly(ADP-ribose) Polymerase Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerases/chemistry , Allosteric Regulation , Carbazoles/chemistry , Humans , Inhibitory Concentration 50 , Models, Biological , Molecular Docking Simulation , Molecular Structure , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/drug effects , Structure-Activity Relationship
11.
Nucleic Acids Res ; 44(9): e88, 2016 05 19.
Article in English | MEDLINE | ID: mdl-26893353

ABSTRACT

Epigenetic modifications of histone tails play an essential role in the regulation of eukaryotic transcription. Writer and eraser enzymes establish and maintain the epigenetic code by creating or removing posttranslational marks. Specific binding proteins, called readers, recognize the modifications and mediate epigenetic signalling. Here, we present a versatile assay platform for the investigation of the interaction between methyl lysine readers and their ligands. This can be utilized for the screening of small-molecule inhibitors of such protein-protein interactions and the detailed characterization of the inhibition. Our platform is constructed in a modular way consisting of orthogonal in vitro binding assays for ligand screening and verification of initial hits and biophysical, label-free techniques for further kinetic characterization of confirmed ligands. A stability assay for the investigation of target engagement in a cellular context complements the platform. We applied the complete evaluation chain to the Tudor domain containing protein Spindlin1 and established the in vitro test systems for the double Tudor domain of the histone demethylase JMJD2C. We finally conducted an exploratory screen for inhibitors of the interaction between Spindlin1 and H3K4me3 and identified A366 as the first nanomolar small-molecule ligand of a Tudor domain containing methyl lysine reader.


Subject(s)
Cell Cycle Proteins/metabolism , Epigenesis, Genetic/genetics , Histones/metabolism , Microtubule-Associated Proteins/metabolism , Phosphoproteins/metabolism , Protein Processing, Post-Translational/physiology , Cell Line, Tumor , HL-60 Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Ligands , Lysine/chemistry , Methylation , Protein Binding/physiology , Protein Domains
12.
Proc Natl Acad Sci U S A ; 112(34): 10768-73, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261308

ABSTRACT

Th17 responses are critical to a variety of human autoimmune diseases, and therapeutic targeting with monoclonal antibodies against IL-17 and IL-23 has shown considerable promise. Here, we report data to support selective bromodomain blockade of the transcriptional coactivators CBP (CREB binding protein) and p300 as an alternative approach to inhibit human Th17 responses. We show that CBP30 has marked molecular specificity for the bromodomains of CBP and p300, compared with 43 other bromodomains. In unbiased cellular testing on a diverse panel of cultured primary human cells, CBP30 reduced immune cell production of IL-17A and other proinflammatory cytokines. CBP30 also inhibited IL-17A secretion by Th17 cells from healthy donors and patients with ankylosing spondylitis and psoriatic arthritis. Transcriptional profiling of human T cells after CBP30 treatment showed a much more restricted effect on gene expression than that observed with the pan-BET (bromo and extraterminal domain protein family) bromodomain inhibitor JQ1. This selective targeting of the CBP/p300 bromodomain by CBP30 will potentially lead to fewer side effects than with the broadly acting epigenetic inhibitors currently in clinical trials.


Subject(s)
Benzimidazoles/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-17/metabolism , Isoxazoles/pharmacology , Th17 Cells/drug effects , p300-CBP Transcription Factors/antagonists & inhibitors , Adult , Aged , Arthritis, Psoriatic/metabolism , Arthritis, Psoriatic/pathology , Azepines/pharmacology , Benzimidazoles/chemistry , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Calorimetry , Cells, Cultured , Crystallography, X-Ray , Drug Evaluation, Preclinical , Female , Gene Expression Regulation/drug effects , Humans , Immunosuppressive Agents/chemistry , Interleukin-17/biosynthesis , Interleukin-17/genetics , Isoxazoles/chemistry , Kinetics , Male , Middle Aged , Models, Molecular , Molecular Structure , Protein Conformation , Protein Structure, Tertiary/drug effects , Recombinant Proteins/metabolism , Spondylitis, Ankylosing/metabolism , Spondylitis, Ankylosing/pathology , Structure-Activity Relationship , Th17 Cells/immunology , Triazoles/pharmacology
13.
Angew Chem Int Ed Engl ; 57(50): 16302-16307, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30288907

ABSTRACT

YEATS domain (YD) containing proteins are an emerging class of epigenetic targets in drug discovery. Dysregulation of these modified lysine-binding proteins has been linked to the onset and progression of cancers. We herein report the discovery and characterisation of the first small-molecule chemical probe, SGC-iMLLT, for the YD of MLLT1 (ENL/YEATS1) and MLLT3 (AF9/YEATS3). SGC-iMLLT is a potent and selective inhibitor of MLLT1/3-histone interactions. Excellent selectivity over other human YD proteins (YEATS2/4) and bromodomains was observed. Furthermore, our probe displays cellular target engagement of MLLT1 and MLLT3. The first small-molecule X-ray co-crystal structures with the MLLT1 YD are also reported. This first-in-class probe molecule can be used to understand MLLT1/3-associated biology and the therapeutic potential of small-molecule YD inhibitors.


Subject(s)
Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Small Molecule Libraries/chemistry , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Crystallography, X-Ray , Histones/metabolism , Humans , Molecular Docking Simulation , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Domains , Protein Interaction Maps/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism
14.
J Org Chem ; 82(6): 3270-3275, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28218543

ABSTRACT

A method for the synthesis of 3-hydroxy-4,4-difluoropyrrolidines from α,α-difluoro-ß-bromoketones is described. The reaction involves methylenation of the carbonyl group with tetrahydrothiophenium ylide followed by coupling with primary amines.

15.
J Org Chem ; 82(24): 12967-12974, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29172505

ABSTRACT

A method for the synthesis of diversely substituted 3-fluoropyridines from two ketone components is described. The reaction involves photoredox coupling of α,α-difluoro-ß-iodoketones with silyl enol ethers catalyzed by fac-Ir(ppy)3 under blue LED irradiation with subsequent one-pot condensation with ammonium acetate. Based on cyclic voltammetry studies, it was determined that α,α-difluoro-ß-iodoketones are reduced notably easier compared to 2,2,2-trifluoro-1-iodoethane, which may be ascribed to the influence of the carbonyl group.

16.
Org Biomol Chem ; 15(40): 8655-8660, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-28984325

ABSTRACT

A series of 3-methylidene-1H-indol-2(3H)-ones substituted with a 5- or 6-pentafluorosulfanyl group has been synthesized by a Knoevenagel condensation reaction of SF5-substituted oxindoles with a range of aldehydes. The resulting products were characterized by X-ray crystallography studies and were tested for biological activity versus a panel of cell lines and protein kinases. Some exhibited single digit nM activity.


Subject(s)
Fluorides/pharmacology , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Sulfur Compounds/pharmacology , Cell Line , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Fluorides/chemical synthesis , Fluorides/chemistry , Humans , Indoles/chemical synthesis , Indoles/chemistry , Models, Molecular , Molecular Structure , Oxindoles , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Sulfur Compounds/chemical synthesis , Sulfur Compounds/chemistry
17.
Bioorg Med Chem ; 25(9): 2657-2665, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28341403

ABSTRACT

The PIM family of serine/threonine kinases have become an attractive target for anti-cancer drug development, particularly for certain hematological malignancies. Here, we describe the discovery of a series of inhibitors of the PIM kinase family using a high throughput screening strategy. Through a combination of molecular modeling and optimization studies, the intrinsic potencies and molecular properties of this series of compounds was significantly improved. An excellent pan-PIM isoform inhibition profile was observed across the series, while optimized examples show good selectivity over other kinases. Two PIM-expressing leukemic cancer cell lines, MV4-11 and K562, were employed to evaluate the in vitro anti-proliferative effects of selected inhibitors. Encouraging activities were observed for many examples, with the best example (44) giving an IC50 of 0.75µM against the K562 cell line. These data provide a promising starting point for further development of this series as a new cancer therapy through PIM kinase inhibition.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Rhodanine/analogs & derivatives , Sulfonamides/pharmacology , Thiazolidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , K562 Cells , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Molecular Docking Simulation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Rhodanine/chemical synthesis , Rhodanine/pharmacokinetics , Rhodanine/pharmacology , Solubility , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Thiazolidines/chemical synthesis , Thiazolidines/pharmacokinetics
18.
Proc Natl Acad Sci U S A ; 111(31): 11341-6, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25049398

ABSTRACT

The retinoblastoma tumor suppressor protein pRb is a key regulator of cell cycle progression and mediator of the DNA damage response. Lysine methylation at K810, which occurs within a critical Cdk phosphorylation motif, holds pRb in the hypophosphorylated growth-suppressing state. We show here that methyl K810 is read by the tandem tudor domain containing tumor protein p53 binding protein 1 (53BP1). Structural elucidation of 53BP1 in complex with a methylated K810 pRb peptide emphasized the role of the 53BP1 tandem tudor domain in recognition of the methylated lysine and surrounding residues. Significantly, binding of 53BP1 to methyl K810 occurs on E2 promoter binding factor target genes and allows pRb activity to be effectively integrated with the DNA damage response. Our results widen the repertoire of cellular targets for 53BP1 and suggest a previously unidentified role for 53BP1 in regulating pRb tumor suppressor activity.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Retinoblastoma Protein/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cellular Senescence , Chromatin/metabolism , DNA Repair , Humans , Methylation , Mice , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Tertiary , Retinoblastoma Protein/chemistry , Tumor Suppressor p53-Binding Protein 1
19.
Angew Chem Int Ed Engl ; 56(49): 15555-15559, 2017 12 04.
Article in English | MEDLINE | ID: mdl-28976073

ABSTRACT

Histone lysine demethylases (KDMs) are of critical importance in the epigenetic regulation of gene expression, yet there are few selective, cell-permeable inhibitors or suitable tool compounds for these enzymes. We describe the discovery of a new class of inhibitor that is highly potent towards the histone lysine demethylases KDM2A/7A. A modular synthetic approach was used to explore the chemical space and accelerate the investigation of key structure-activity relationships, leading to the development of a small molecule with around 75-fold selectivity towards KDM2A/7A versus other KDMs, as well as cellular activity at low micromolar concentrations.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , F-Box Proteins/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , F-Box Proteins/metabolism , HeLa Cells , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Molecular Structure , Structure-Activity Relationship
20.
Angew Chem Int Ed Engl ; 56(3): 827-831, 2017 01 16.
Article in English | MEDLINE | ID: mdl-27966810

ABSTRACT

The p300/CBP-associated factor (PCAF) and related GCN5 bromodomain-containing lysine acetyl transferases are members of subfamily I of the bromodomain phylogenetic tree. Iterative cycles of rational inhibitor design and biophysical characterization led to the discovery of the triazolopthalazine-based L-45 (dubbed L-Moses) as the first potent, selective, and cell-active PCAF bromodomain (Brd) inhibitor. Synthesis from readily available (1R,2S)-(-)-norephedrine furnished L-45 in enantiopure form. L-45 was shown to disrupt PCAF-Brd histone H3.3 interaction in cells using a nanoBRET assay, and a co-crystal structure of L-45 with the homologous Brd PfGCN5 from Plasmodium falciparum rationalizes the high selectivity for PCAF and GCN5 bromodomains. Compound L-45 shows no observable cytotoxicity in peripheral blood mononuclear cells (PBMC), good cell-permeability, and metabolic stability in human and mouse liver microsomes, supporting its potential for in vivo use.


Subject(s)
Azo Compounds/pharmacology , Drug Discovery , Hydralazine/pharmacology , Molecular Probes/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Azo Compounds/chemical synthesis , Azo Compounds/chemistry , Dose-Response Relationship, Drug , Hydralazine/chemical synthesis , Hydralazine/chemistry , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL