Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Am J Hum Genet ; 104(4): 709-720, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905399

ABSTRACT

The Mediator is an evolutionarily conserved, multi-subunit complex that regulates multiple steps of transcription. Mediator activity is regulated by the reversible association of a four-subunit module comprising CDK8 or CDK19 kinases, together with cyclin C, MED12 or MED12L, and MED13 or MED13L. Mutations in MED12, MED13, and MED13L were previously identified in syndromic developmental disorders with overlapping phenotypes. Here, we report CDK8 mutations (located at 13q12.13) that cause a phenotypically related disorder. Using whole-exome or whole-genome sequencing, and by international collaboration, we identified eight different heterozygous missense CDK8 substitutions, including 10 shown to have arisen de novo, in 12 unrelated subjects; a recurrent mutation, c.185C>T (p.Ser62Leu), was present in five individuals. All predicted substitutions localize to the ATP-binding pocket of the kinase domain. Affected individuals have overlapping phenotypes characterized by hypotonia, mild to moderate intellectual disability, behavioral disorders, and variable facial dysmorphism. Congenital heart disease occurred in six subjects; additional features present in multiple individuals included agenesis of the corpus callosum, ano-rectal malformations, seizures, and hearing or visual impairments. To evaluate the functional impact of the mutations, we measured phosphorylation at STAT1-Ser727, a known CDK8 substrate, in a CDK8 and CDK19 CRISPR double-knockout cell line transfected with wild-type (WT) or mutant CDK8 constructs. These experiments demonstrated a reduction in STAT1 phosphorylation by all mutants, in most cases to a similar extent as in a kinase-dead control. We conclude that missense mutations in CDK8 cause a developmental disorder that has phenotypic similarity to syndromes associated with mutations in other subunits of the Mediator kinase module, indicating probable overlap in pathogenic mechanisms.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Developmental Disabilities/genetics , Mediator Complex/genetics , Mutation, Missense , Brain/abnormalities , Child , Child, Preschool , Cyclin C/genetics , Cyclin-Dependent Kinases/genetics , Exome , Female , Heart Defects, Congenital/genetics , Heterozygote , Humans , Infant , Intellectual Disability/genetics , Male , Mutation , Phenotype , Phosphorylation , Syndrome
2.
Pediatr Blood Cancer ; 66(9): e27888, 2019 09.
Article in English | MEDLINE | ID: mdl-31207107

ABSTRACT

BACKGROUND: Ewing sarcoma and desmoplastic small round cell tumors (DSRCT) are rare and clinically aggressive sarcomas usually characterized by oncogenic fusion proteins involving EWS. Emerging studies of Ewing sarcoma have demonstrated EWS-FLI1-driven chromatin remodeling as a key aspect of tumorigenicity. In particular, the lysine-specific demethylase KDM1A/LSD1 is linked to transcriptional regulation of target genes orchestrated by the EWS portion of the fusion protein interacting with repressive chromatin-remodeling complexes. Consistent with this model, depletion of KDM1A supports it is a molecular therapeutic target in Ewing sarcoma cells, but effective drugs need to be identified. PROCEDURE: A comprehensive phenotypic analysis of the effects of catalytic KDM1A inhibitors ORY-1001 and GSK2879552, including clinically relevant doses, was carried out in 2D and 3D spheroid models of Ewing sarcoma and DSRCT. RESULTS: Catalytic inhibition of KDM1A did not affect cell viability in 2D and 3D assays and had no impact on invasion in a 3D assay. CONCLUSIONS: Overall, evidence presented here does not support inhibition of KDM1A catalytic demethylase activity as an effective therapeutic strategy for Ewing sarcoma or DSRCT. However, roles of KDM1A beyond its demethylase activity should be considered for these sarcomas.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Enzyme Inhibitors , Histone Demethylases/antagonists & inhibitors , Sarcoma, Ewing , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/enzymology , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Demethylases/metabolism , Humans , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/enzymology
3.
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.

4.
Br J Cancer ; 116(9): 1166-1176, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28334731

ABSTRACT

BACKGROUND: The main role of the cell cycle is to enable error-free DNA replication, chromosome segregation and cytokinesis. One of the best characterised checkpoint pathways is the spindle assembly checkpoint, which prevents anaphase onset until the appropriate attachment and tension across kinetochores is achieved. MPS1 kinase activity is essential for the activation of the spindle assembly checkpoint and has been shown to be deregulated in human tumours with chromosomal instability and aneuploidy. Therefore, MPS1 inhibition represents an attractive strategy to target cancers. METHODS: To evaluate CCT271850 cellular potency, two specific antibodies that recognise the activation sites of MPS1 were used and its antiproliferative activity was determined in 91 human cancer cell lines. DLD1 cells with induced GFP-MPS1 and HCT116 cells were used in in vivo studies to directly measure MPS1 inhibition and efficacy of CCT271850 treatment. RESULTS: CCT271850 selectively and potently inhibits MPS1 kinase activity in biochemical and cellular assays and in in vivo models. Mechanistically, tumour cells treated with CCT271850 acquire aberrant numbers of chromosomes and the majority of cells divide their chromosomes without proper alignment because of abrogation of the mitotic checkpoint, leading to cell death. We demonstrated a moderate level of efficacy of CCT271850 as a single agent in a human colorectal carcinoma xenograft model. CONCLUSIONS: CCT271850 is a potent, selective and orally bioavailable MPS1 kinase inhibitor. On the basis of in vivo pharmacodynamic vs efficacy relationships, we predict that more than 80% inhibition of MPS1 activity for at least 24 h is required to achieve tumour stasis or regression by CCT271850.


Subject(s)
Cell Cycle Proteins/genetics , Heterocyclic Compounds, 4 or More Rings/administration & dosage , M Phase Cell Cycle Checkpoints/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , HCT116 Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
5.
Nat Chem Biol ; 11(12): 973-980, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502155

ABSTRACT

There is unmet need for chemical tools to explore the role of the Mediator complex in human pathologies ranging from cancer to cardiovascular disease. Here we determine that CCT251545, a small-molecule inhibitor of the WNT pathway discovered through cell-based screening, is a potent and selective chemical probe for the human Mediator complex-associated protein kinases CDK8 and CDK19 with >100-fold selectivity over 291 other kinases. X-ray crystallography demonstrates a type 1 binding mode involving insertion of the CDK8 C terminus into the ligand binding site. In contrast to type II inhibitors of CDK8 and CDK19, CCT251545 displays potent cell-based activity. We show that CCT251545 and close analogs alter WNT pathway-regulated gene expression and other on-target effects of modulating CDK8 and CDK19, including expression of genes regulated by STAT1. Consistent with this, we find that phosphorylation of STAT1(SER727) is a biomarker of CDK8 kinase activity in vitro and in vivo. Finally, we demonstrate in vivo activity of CCT251545 in WNT-dependent tumors.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Molecular Probes/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Spiro Compounds/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/genetics , Humans , Models, Molecular , Molecular Probes/chemistry , Molecular Structure , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Spiro Compounds/chemistry
6.
Xenobiotica ; 47(9): 771-777, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27618572

ABSTRACT

1. We have previously described C8-substituted pyrido[3,4-d]pyrimidin-4(3H)-one derivatives as cell permeable inhibitors of the KDM4 and KDM5 subfamilies of JmjC histone lysine demethylases. 2. Although exemplar compound 1 exhibited moderate clearance in mouse liver microsomes, it was highly cleared in vivo due to metabolism by aldehyde oxidase (AO). Similar human and mouse AO-mediated metabolism was observed with the pyrido[3,4-d]pyrimidin-4(3H)-one scaffold and other C8-substituted derivatives. 3. We identified the C2-position as the oxidation site by LC-MS and 1H-NMR and showed that C2-substituted derivatives are no longer AO substrates. 4. In addition to the experimental data, these observations are supported by molecular modelling studies in the human AO protein crystal structure.


Subject(s)
Aldehyde Oxidase/antagonists & inhibitors , Pyrimidines/metabolism , Animals , Humans , Mice , Models, Molecular , Proton Magnetic Resonance Spectroscopy , Structure-Activity Relationship
7.
Angew Chem Int Ed Engl ; 56(38): 11409-11414, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28700101

ABSTRACT

The conformation of the activation loop (T-loop) of protein kinases underlies enzymatic activity and influences the binding of small-molecule inhibitors. By using single-molecule fluorescence spectroscopy, we have determined that phosphorylated Aurora A kinase is in dynamic equilibrium between a DFG-in-like active T-loop conformation and a DFG-out-like inactive conformation, and have measured the rate constants of interconversion. Addition of the Aurora A activating protein TPX2 shifts the equilibrium towards an active T-loop conformation whereas addition of the inhibitors MLN8054 and CD532 favors an inactive T-loop. We show that Aurora A binds TPX2 and MLN8054 simultaneously and provide a new model for kinase conformational behavior. Our approach will enable conformation-specific effects to be integrated into inhibitor discovery across the kinome, and we outline some immediate consequences for structure-based drug discovery.


Subject(s)
Aurora Kinase A/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Aurora Kinase A/metabolism , Fluorescence , Humans , Ligands , Models, Molecular , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry
8.
Bioorg Med Chem Lett ; 26(5): 1443-51, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26852363

ABSTRACT

Here we describe the discovery and optimization of 3-benzylindazoles as potent and selective inhibitors of CDK8, also modulating CDK19, discovered from a high-throughput screening (HTS) campaign sampling the Merck compound collection. The primary hits with strong HSP90 affinity were subsequently optimized to potent and selective CDK8 inhibitors which demonstrate inhibition of WNT pathway activity in cell-based assays. X-ray crystallographic data demonstrated that 3-benzylindazoles occupy the ATP binding site of CDK8 and adopt a Type I binding mode. Medicinal chemistry optimization successfully led to improved potency, physicochemical properties and oral pharmacokinetics. Modulation of phospho-STAT1, a pharmacodynamic biomarker of CDK8, was demonstrated in an APC-mutant SW620 human colorectal carcinoma xenograft model following oral administration.


Subject(s)
Colorectal Neoplasms/drug therapy , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Drug Discovery , HSP90 Heat-Shock Proteins/metabolism , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Colorectal Neoplasms/metabolism , Crystallography, X-Ray , Cyclin-Dependent Kinase 8/metabolism , Dose-Response Relationship, Drug , Humans , Indazoles/administration & dosage , Indazoles/chemistry , Mice , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Rats , Structure-Activity Relationship , Substrate Specificity
9.
Bioorg Med Chem Lett ; 25(19): 4203-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26296477

ABSTRACT

Introduction of a 1-benzyl-1H-pyrazol-4-yl moiety at C7 of the imidazo[4,5-b]pyridine scaffold provided 7a which inhibited a range of kinases including Aurora-A. Modification of the benzyl group in 7a, and subsequent co-crystallisation of the resulting analogues with Aurora-A indicated distinct differences in binding mode dependent upon the pyrazole N-substituent. Compounds 7a and 14d interact with the P-loop whereas 14a and 14b engage with Thr217 in the post-hinge region. These crystallographic insights provide options for the design of compounds interacting with the DFG motif or with Thr217.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Aurora Kinases/chemistry , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallization , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Pyrazoles/chemistry , Pyridines/chemistry , Structure-Activity Relationship
10.
J Chem Inf Model ; 55(6): 1169-80, 2015 Jun 22.
Article in English | MEDLINE | ID: mdl-26054755

ABSTRACT

We describe the development and application of an integrated, multiobjective optimization workflow (MOARF) for directed medicinal chemistry design. This workflow couples a rule-based molecular fragmentation scheme (SynDiR) with a pharmacophore fingerprint-based fragment replacement algorithm (RATS) to broaden the scope of reconnection options considered in the generation of potential solution structures. Solutions are ranked by a multiobjective scoring algorithm comprising ligand-based (shape similarity) biochemical activity predictions as well as physicochemical property calculations. Application of this iterative workflow to optimization of the CDK2 inhibitor Seliciclib (CYC202, R-roscovitine) generated solution molecules in desired physicochemical property space. Synthesis and experimental evaluation of optimal solution molecules demonstrates CDK2 biochemical activity and improved human metabolic stability.


Subject(s)
Algorithms , Computational Biology/methods , Drug Design , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Drug Stability , Humans , Ligands , Microsomes/metabolism , Oxidation-Reduction , Purines/chemistry , Purines/metabolism , Purines/pharmacology , Roscovitine
11.
J Med Chem ; 67(2): 1061-1078, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38198226

ABSTRACT

Hedgehog signaling is involved in embryonic development and cancer growth. Functional activity of secreted Hedgehog signaling proteins is dependent on N-terminal palmitoylation, making the palmitoyl transferase Hedgehog acyltransferase (HHAT), a potential drug target and a series of 4,5,6,7-tetrahydrothieno[3,2-c]pyridines have been identified as HHAT inhibitors. Based on structural data, we designed and synthesized 37 new analogues which we profiled alongside 13 previously reported analogues in enzymatic and cellular assays. Our results show that a central amide linkage, a secondary amine, and (R)-configuration at the 4-position of the core are three key factors for inhibitory potency. Several potent analogues with low- or sub-µM IC50 against purified HHAT also inhibit Sonic Hedgehog (SHH) palmitoylation in cells and suppress the SHH signaling pathway. This work identifies IMP-1575 as the most potent cell-active chemical probe for HHAT function, alongside an inactive control enantiomer, providing tool compounds for validation of HHAT as a target in cellular assays.


Subject(s)
Hedgehog Proteins , Hedgehog Proteins/metabolism , Pyridines/chemistry , Pyridines/pharmacology
12.
Org Biomol Chem ; 11(14): 2335-47, 2013 Apr 14.
Article in English | MEDLINE | ID: mdl-23429655

ABSTRACT

We show that N3-MEM-protected imidazo[4,5-b]pyridines undergo efficient C2-functionalisation via direct C-H arylation. Twenty-two substituted imidazo[4,5-b]pyridines are prepared and iterative, selective elaboration of functionalised imidazo[4,5-b]pyridines gives 2,7- and 2,6-disubstituted derivatives in good yields from common intermediates. Mechanistic observations are consistent with a concerted-metallation-deprotonation mechanism facilitated by coordination of copper(I)iodide to the imidazo[4,5-b]pyridine.


Subject(s)
Imidazoles/chemistry , Pyridines/chemistry , Copper/chemistry , Iodides/chemistry , Molecular Structure , Stereoisomerism
13.
J Chem Inf Model ; 53(5): 1100-12, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23672464

ABSTRACT

We describe and apply a scaffold-focused virtual screen based upon scaffold trees to the mitotic kinase TTK (MPS1). Using level 1 of the scaffold tree, we perform both 2D and 3D similarity searches between a query scaffold and a level 1 scaffold library derived from a 2 million compound library; 98 compounds from 27 unique top-ranked level 1 scaffolds are selected for biochemical screening. We show that this scaffold-focused virtual screen prospectively identifies eight confirmed active compounds that are structurally differentiated from the query compound. In comparison, 100 compounds were selected for biochemical screening using a virtual screen based upon whole molecule similarity resulting in 12 confirmed active compounds that are structurally similar to the query compound. We elucidated the binding mode for four of the eight confirmed scaffold hops to TTK by determining their protein-ligand crystal structures; each represents a ligand-efficient scaffold for inhibitor design.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , User-Computer Interface , Cell Cycle Proteins/chemistry , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Models, Molecular , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein-Tyrosine Kinases/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
16.
J Chem Inf Model ; 52(10): 2516-25, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23009689

ABSTRACT

We describe a computational method, plane of best fit (PBF), to quantify and characterize the 3D character of molecules. This method is rapid and amenable to analysis of large diverse data sets. We compare PBF with alternative literature methods used to assess 3D character and apply the method to diverse data sets of fragment-like, drug-like, and natural product compound libraries. We show that exemplar fragment libraries underexploit the potential of 3D character in fragment-like chemical space and that drug-like molecules in the libraries examined are predominantly 2D in character.


Subject(s)
Algorithms , Biological Products/chemistry , Small Molecule Libraries/chemistry , Databases, Chemical , Databases, Pharmaceutical , Drug Design , Humans , Models, Molecular , Molecular Conformation
17.
Bioorg Med Chem ; 20(22): 6630-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23058106

ABSTRACT

Two closely related binding modes have previously been proposed for the ATP-competitive benzimidazole class of checkpoint kinase 2 (CHK2) inhibitors; however, neither binding mode is entirely consistent with the reported SAR. Unconstrained rigid docking of benzimidazole ligands into representative CHK2 protein crystal structures reveals an alternative binding mode involving a water-mediated interaction with the hinge region; docking which incorporates protein side chain flexibility for selected residues in the ATP binding site resulted in a refinement of the water-mediated hinge binding mode that is consistent with observed SAR. The flexible docking results are in good agreement with the crystal structures of four exemplar benzimidazole ligands bound to CHK2 which unambiguously confirmed the binding mode of these inhibitors, including the water-mediated interaction with the hinge region, and which is significantly different from binding modes previously postulated in the literature.


Subject(s)
Benzimidazoles/metabolism , Protein Serine-Threonine Kinases/metabolism , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Binding Sites , Checkpoint Kinase 2 , Crystallography, X-Ray , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary , Structure-Activity Relationship
18.
Hum Mutat ; 32(1): E1928-38, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20949628

ABSTRACT

Transforming growth factor-ß (TGF-ß) signalling plays a key role in colorectal cancer (CRC). Bone morphogenetic protein-4 (BMP4) is a member of the TGF-ß family of signal transduction molecules. To examine if germline mutation in BMP4 causes CRC we analysed 504 genetically enriched CRC cases (by virtue of early-onset disease, family history of CRC) for mutations in the coding sequence of BMP4. We identified three pathogenic mutations, p.R286X (g.8330C>T), p.W325C (g.8449G>T) and p.C373S (g.8592G>C), amongst the CRC cases which were not observed in 524 healthy controls. p.R286X localizes to the N-terminal of the TGF-ß1 prodomain truncating the protein prior to the active domain. p.W325C and p.C373S mutations are predicted from protein homology modelling with BMP2 to impact deleteriously on BMP4 function. Segregation of p.C373S with adenoma and hyperplastic polyp in first-degree relatives of the case suggests germline mutations may confer a juvenile polyposis-type phenotype. These findings suggest mutation of BMP4is a cause of CRC and the value of protein-based modelling in the elucidation of rare disease-causing variants.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Colorectal Neoplasms/genetics , Germ-Line Mutation/genetics , Adult , Aged , Amino Acid Sequence , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , Sequence Alignment , Transforming Growth Factor beta/genetics
19.
J Biol Chem ; 285(50): 39348-58, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20880844

ABSTRACT

Cellular stress in early mitosis activates the antephase checkpoint, resulting in the decondensation of chromosomes and delayed mitotic progression. Checkpoint with forkhead-associated and RING domains (CHFR) is central to this checkpoint, and its activity is ablated in many tumors and cancer cell lines through promoter hypermethylation or mutation. The interaction between the PAR-binding zinc finger (PBZ) of CHFR and poly(ADP-ribose) (PAR) is crucial for a functional antephase checkpoint. We determined the crystal structure of the cysteine-rich region of human CHFR (amino acids 425-664) to 1.9 Å resolution, which revealed a multizinc binding domain of elaborate topology within which the PBZ is embedded. The PBZ of CHFR closely resembles the analogous motifs from aprataxin-like factor and CG1218-PA, which lie within unstructured regions of their respective proteins. Based on co-crystal structures of CHFR bound to several different PAR-like ligands (adenosine 5'-diphosphoribose, adenosine monophosphate, and P(1)P(2)-diadenosine 5'-pyrophosphate), we made a model of the CHFR-PAR interaction, which we validated using site-specific mutagenesis and surface plasmon resonance. The PBZ motif of CHFR recognizes two adenine-containing subunits of PAR and the phosphate backbone that connects them. More generally, PBZ motifs may recognize different numbers of PAR subunits as required to carry out their functions.


Subject(s)
Adenosine Diphosphate Ribose/chemistry , Cell Cycle Proteins/chemistry , Neoplasm Proteins/chemistry , Adenosine Diphosphate/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray/methods , Humans , Ligands , Mitosis , Molecular Sequence Data , Poly-ADP-Ribose Binding Proteins , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance , Temperature , Ubiquitin-Protein Ligases , Zinc/chemistry , Zinc Fingers
20.
Proteins ; 79(7): 2109-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21557316

ABSTRACT

Deregulation of protein kinases is associated with numerous diseases, making them important targets for drug discovery. The majority of drugs target the catalytic site of these proteins, but due to the high level of similarity within the ATP binding sites of protein kinases, it is often difficult to achieve the required pharmacological selectivity. In this study, we describe the identification and subsequent analysis of water patterns in the ATP binding sites of 171 protein kinase structures, comprising 19 different kinases from various branches of the kinome, and demonstrate that structurally similar binding sites often have significantly different water patterns. We show that the observed variations in water patterns of different, but structurally similar kinases can be exploited in the structure-based design of potent and selective kinase inhibitors.


Subject(s)
Protein Kinases/chemistry , Water/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding Sites , Cluster Analysis , Databases, Protein , Humans , Models, Molecular , Protein Conformation , Protein Kinases/metabolism , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL