Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
SLAS Discov ; 22(4): 418-424, 2017 04.
Article in English | MEDLINE | ID: mdl-28296537

ABSTRACT

The Hedgehog pathway is a key developmental signaling pathway but is also implicated in many types of cancer. The extracellular signaling protein Sonic hedgehog (Shh) requires dual lipidation for functional signaling, whereby N-terminal palmitoylation is performed by the enzyme Hedgehog acyltransferase (Hhat). Hhat is an attractive target for small-molecule inhibition to arrest Hedgehog signaling, and methods for assaying Hhat activity are central to understanding its function. However, all existing assays to quantify lipidation of peptides suffer limitations, such as safety hazards, high costs, extensive manual handling, restriction to stopped-assay measurements, or indirect assessment of lipidation. To address these limitations, we developed a microfluidic mobility shift assay (MSA) to analyze Shh palmitoylation. MSA allowed separation of fluorescently labeled Shh amine-substrate and palmitoylated Shh amide-product peptides based on differences in charge and hydrodynamic radius, coupled with online fluorescence intensity measurements for quantification. The MSA format was employed to study Hhat-catalyzed reactions, investigate Hhat kinetics, and determine small-molecule inhibitor IC50 values. Both real-time and stopped assays were performed, with the latter achieved via addition of excess unlabeled Shh peptide. The MSA format therefore allows direct and real-time fluorescence-based measurement of acylation and represents a powerful alternative technique in the study of N-lipidation.


Subject(s)
Acyltransferases/metabolism , Electrophoretic Mobility Shift Assay/methods , Hedgehog Proteins/metabolism , Microfluidics/methods , Protein Processing, Post-Translational , Acyltransferases/antagonists & inhibitors , Acyltransferases/genetics , Amino Acid Sequence , Electrophoretic Mobility Shift Assay/instrumentation , Enzyme Assays , Enzyme Inhibitors/pharmacology , HEK293 Cells , Hedgehog Proteins/genetics , Humans , Kinetics , Lipoylation/drug effects , Microfluidics/instrumentation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
2.
J Med Chem ; 59(11): 5221-37, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27167172

ABSTRACT

Multiparameter optimization of a series of 5-((4-aminopyridin-2-yl)amino)pyrazine-2-carbonitriles resulted in the identification of a potent and selective oral CHK1 preclinical development candidate with in vivo efficacy as a potentiator of deoxyribonucleic acid (DNA) damaging chemotherapy and as a single agent. Cellular mechanism of action assays were used to give an integrated assessment of compound selectivity during optimization resulting in a highly CHK1 selective adenosine triphosphate (ATP) competitive inhibitor. A single substituent vector directed away from the CHK1 kinase active site was unexpectedly found to drive the selective cellular efficacy of the compounds. Both CHK1 potency and off-target human ether-a-go-go-related gene (hERG) ion channel inhibition were dependent on lipophilicity and basicity in this series. Optimization of CHK1 cellular potency and in vivo pharmacokinetic-pharmacodynamic (PK-PD) properties gave a compound with low predicted doses and exposures in humans which mitigated the residual weak in vitro hERG inhibition.


Subject(s)
4-Aminopyridine/analogs & derivatives , Checkpoint Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , 4-Aminopyridine/chemical synthesis , 4-Aminopyridine/chemistry , 4-Aminopyridine/pharmacology , Checkpoint Kinase 1/metabolism , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazines/chemical synthesis , Pyrazines/chemistry , Structure-Activity Relationship
3.
J Med Chem ; 59(4): 1388-409, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26741168

ABSTRACT

We report the discovery of N-substituted 4-(pyridin-2-yl)thiazole-2-amine derivatives and their subsequent optimization, guided by structure-based design, to give 8-(1H-pyrazol-3-yl)pyrido[3,4-d]pyrimidin-4(3H)-ones, a series of potent JmjC histone N-methyl lysine demethylase (KDM) inhibitors which bind to Fe(II) in the active site. Substitution from C4 of the pyrazole moiety allows access to the histone peptide substrate binding site; incorporation of a conformationally constrained 4-phenylpiperidine linker gives derivatives such as 54j and 54k which demonstrate equipotent activity versus the KDM4 (JMJD2) and KDM5 (JARID1) subfamily demethylases, selectivity over representative exemplars of the KDM2, KDM3, and KDM6 subfamilies, cellular permeability in the Caco-2 assay, and, for 54k, inhibition of H3K9Me3 and H3K4Me3 demethylation in a cell-based assay.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Repressor Proteins/antagonists & inhibitors , Caco-2 Cells , Cell Membrane Permeability , Enzyme Inhibitors/pharmacokinetics , Humans , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Pyrimidinones/pharmacokinetics , Repressor Proteins/chemistry , Repressor Proteins/metabolism
4.
J Med Chem ; 56(22): 9122-35, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24195668

ABSTRACT

Aurora-A differs from Aurora-B/C at three positions in the ATP-binding pocket (L215, T217, and R220). Exploiting these differences, crystal structures of ligand-Aurora protein interactions formed the basis of a design principle for imidazo[4,5-b]pyridine-derived Aurora-A-selective inhibitors. Guided by a computational modeling approach, appropriate C7-imidazo[4,5-b]pyridine derivatization led to the discovery of highly selective inhibitors, such as compound 28c, of Aurora-A over Aurora-B. In HCT116 human colon carcinoma cells, 28c and 40f inhibited the Aurora-A L215R and R220K mutants with IC50 values similar to those seen for the Aurora-A wild type. However, the Aurora-A T217E mutant was significantly less sensitive to inhibition by 28c and 40f compared to the Aurora-A wild type, suggesting that the T217 residue plays a critical role in governing the observed isoform selectivity for Aurora-A inhibition. These compounds are useful small-molecule chemical tools to further explore the function of Aurora-A in cells.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Drug Design , Imidazoles/chemistry , Imidazoles/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Aurora Kinase A/chemistry , Aurora Kinase A/metabolism , Aurora Kinase B/chemistry , Aurora Kinase B/metabolism , Catalytic Domain , Drug Stability , HCT116 Cells , Humans , Imidazoles/metabolism , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Male , Mice , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Substrate Specificity
5.
Cancer Res ; 71(2): 463-72, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21239475

ABSTRACT

CHK2 is a checkpoint kinase involved in the ATM-mediated response to double-strand DNA breaks. Its potential as a drug target is still unclear, but inhibitors of CHK2 may increase the efficacy of genotoxic cancer therapies in a p53 mutant background by eliminating one of the checkpoints or DNA repair pathways contributing to cellular resistance. We report here the identification and characterization of a novel CHK2 kinase inhibitor, CCT241533. X-ray crystallography confirmed that CCT241533 bound to CHK2 in the ATP pocket. This compound inhibits CHK2 with an IC(50) of 3 nmol/L and shows minimal cross-reactivity against a panel of kinases at 1 µmol/L. CCT241533 blocked CHK2 activity in human tumor cell lines in response to DNA damage, as shown by inhibition of CHK2 autophosphorylation at S516, band shift mobility changes, and HDMX degradation. CCT241533 did not potentiate the cytotoxicity of a selection of genotoxic agents in several cell lines. However, this compound significantly potentiates the cytotoxicity of two structurally distinct PARP inhibitors. Clear induction of the pS516 CHK2 signal was seen with a PARP inhibitor alone, and this activation was abolished by CCT241533, implying that the potentiation of PARP inhibitor cell killing by CCT241533 was due to inhibition of CHK2. Consequently, our findings imply that CHK2 inhibitors may exert therapeutic activity in combination with PARP inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Bleomycin/administration & dosage , Cell Cycle/drug effects , Cell Line, Tumor , Checkpoint Kinase 2 , Crystallography, X-Ray , Drug Synergism , Enzyme Activation/drug effects , HCT116 Cells , HT29 Cells , HeLa Cells , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Models, Molecular , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Quinazolines/administration & dosage , Quinazolines/chemistry
6.
EMBO J ; 25(13): 3179-90, 2006 Jul 12.
Article in English | MEDLINE | ID: mdl-16794575

ABSTRACT

The protein kinase Chk2 (checkpoint kinase 2) is a major effector of the replication checkpoint. Chk2 activation is initiated by phosphorylation of Thr68, in the serine-glutamine/threonine-glutamine cluster domain (SCD), by ATM. The phosphorylated SCD-segment binds to the FHA domain of a second Chk2 molecule, promoting dimerisation of the protein and triggering phosphorylation of the activation segment/T-loop in the kinase domain. We have now determined the structure of the kinase domain of human Chk2 in complexes with ADP and a small-molecule inhibitor debromohymenialdisine. The structure reveals a remarkable dimeric arrangement in which T-loops are exchanged between protomers, to form an active kinase conformation in trans. Biochemical data suggest that this dimer is the biologically active state promoted by ATM-phosphorylation, and also suggests a mechanism for dimerisation-driven activation of Chk2 by trans-phosphorylation.


Subject(s)
DNA Damage , Models, Molecular , Protein Serine-Threonine Kinases/chemistry , Trans-Activators , Adenosine Diphosphate/chemistry , Ataxia Telangiectasia Mutated Proteins , Azepines/chemistry , Catalytic Domain , Cell Cycle Proteins/chemistry , Checkpoint Kinase 2 , DNA-Binding Proteins/chemistry , Dimerization , Enzyme Activation , Humans , Phosphorylation , Protein Conformation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Pyrroles/chemistry , Signal Transduction , Tumor Suppressor Proteins/chemistry
7.
Bioorg Med Chem Lett ; 15(14): 3338-43, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15955698

ABSTRACT

High-throughput screening identified the 3,4-diarylpyrazole CCT018159 as a novel and potent (7.1 microM) inhibitor of Hsp90 ATPase activity. Here, we describe the synthesis of CCT018159 and a number of close analogues together with data on their biochemical properties. Some initial structure-activity relationships are discussed, as well as the crystal structure of CCT018159 bound to Hsp90.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Enzyme Inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring , Pyrazoles , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Hydrogen Bonding , Models, Molecular , Molecular Conformation , Protein Conformation , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/classification , Pyrazoles/pharmacology , Structure-Activity Relationship , Yeasts/chemistry , Yeasts/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...