Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 11(4): 497-505, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32292556

ABSTRACT

A combination of focused library and virtual screening, hit expansion, and rational design has resulted in the development of a series of inhibitors of RETV804M kinase, the anticipated drug-resistant mutant of RET kinase. These agents do not inhibit the wild type (wt) isoforms of RET or KDR and therefore offer a potential adjunct to RET inhibitors currently undergoing clinical evaluation.

2.
Essays Biochem ; 61(5): 495-503, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118096

ABSTRACT

The ongoing explosion in genomics data has long since outpaced the capacity of conventional biochemical methodology to verify the large number of hypotheses that emerge from the analysis of such data. In contrast, it is still a gold-standard for early phenotypic validation towards small-molecule drug discovery to use probe molecules (or tool compounds), notwithstanding the difficulty and cost of generating them. Rational structure-based approaches to ligand discovery have long promised the efficiencies needed to close this divergence; in practice, however, this promise remains largely unfulfilled, for a host of well-rehearsed reasons and despite the huge technical advances spearheaded by the structural genomics initiatives of the noughties. Therefore the current, fourth funding phase of the Structural Genomics Consortium (SGC), building on its extensive experience in structural biology of novel targets and design of protein inhibitors, seeks to redefine what it means to do structural biology for drug discovery. We developed the concept of a Target Enabling Package (TEP) that provides, through reagents, assays and data, the missing link between genetic disease linkage and the development of usefully potent compounds. There are multiple prongs to the ambition: rigorously assessing targets' genetic disease linkages through crowdsourcing to a network of collaborating experts; establishing a systematic approach to generate the protocols and data that comprise each target's TEP; developing new, X-ray-based fragment technologies for generating high quality chemical matter quickly and cheaply; and exploiting a stringently open access model to build multidisciplinary partnerships throughout academia and industry. By learning how to scale these approaches, the SGC aims to make structures finally serve genomics, as originally intended, and demonstrate how 3D structures systematically allow new modes of druggability to be discovered for whole classes of targets.


Subject(s)
Drug Design , Drug Discovery/methods , Drugs, Investigational/chemistry , Proteins/chemistry , Small Molecule Libraries/chemistry , Binding Sites , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Drugs, Investigational/chemical synthesis , Genomics/methods , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Proteins/agonists , Proteins/antagonists & inhibitors , Proteins/metabolism , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship
4.
Eur J Med Chem ; 126: 1118-1128, 2017 Jan 27.
Article in English | MEDLINE | ID: mdl-28039837

ABSTRACT

We present comprehensive testing of solvent representation in quantum mechanics (QM)-based scoring of protein-ligand affinities. To this aim, we prepared 21 new inhibitors of cyclin-dependent kinase 2 (CDK2) with the pyrazolo[1,5-a]pyrimidine core, whose activities spanned three orders of magnitude. The crystal structure of a potent inhibitor bound to the active CDK2/cyclin A complex revealed that the biphenyl substituent at position 5 of the pyrazolo[1,5-a]pyrimidine scaffold was located in a previously unexplored pocket and that six water molecules resided in the active site. Using molecular dynamics, protein-ligand interactions and active-site water H-bond networks as well as thermodynamics were probed. Thereafter, all the inhibitors were scored by the QM approach utilizing the COSMO implicit solvent model. Such a standard treatment failed to produce a correlation with the experiment (R2 = 0.49). However, the addition of the active-site waters resulted in significant improvement (R2 = 0.68). The activities of the compounds could thus be interpreted by taking into account their specific noncovalent interactions with CDK2 and the active-site waters. In summary, using a combination of several experimental and theoretical approaches we demonstrate that the inclusion of explicit solvent effects enhance QM/COSMO scoring to produce a reliable structure-activity relationship with physical insights. More generally, this approach is envisioned to contribute to increased accuracy of the computational design of novel inhibitors.


Subject(s)
Catalytic Domain , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quantum Theory , Solvents/chemistry , Water/chemistry , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Drug Design , Humans , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/metabolism , Structure-Activity Relationship
5.
PLoS One ; 9(8): e105688, 2014.
Article in English | MEDLINE | ID: mdl-25144743

ABSTRACT

The Cop9 signalosome complex (CSN) regulates the functional cycle of the major E3 ubiquitin ligase family, the cullin RING E3 ubiquitin ligases (CRLs). Activated CRLs are covalently modified by the ubiquitin-like protein Nedd8 (neural precursor cell expressed developmentally down-regulated protein 8). CSN serves an essential role in myriad cellular processes by reversing this modification through the isopeptidase activity of its CSN5 subunit. CSN5 alone is inactive due to an auto-inhibited conformation of its catalytic domain. Here we report the molecular basis of CSN5 catalytic domain activation and unravel a molecular hierarchy in CSN deneddylation activity. The association of CSN5 and CSN6 MPN (for Mpr1/Pad1 N-terminal) domains activates its isopeptidase activity. The CSN5/CSN6 module, however, is inefficient in CRL deneddylation, indicating a requirement of further elements in this reaction such as other CSN subunits. A hybrid molecular model of CSN5/CSN6 provides a structural framework to explain these functional observations. Docking this model into a published CSN electron density map and using distance constraints obtained from cross-linking coupled to mass-spectrometry, we find that the C-termini of the CSN subunits could form a helical bundle in the centre of the structure. They likely play a key scaffolding role in the spatial organization of CSN and precise positioning of the dimeric MPN catalytic core.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Multiprotein Complexes/chemistry , Peptide Hydrolases/chemistry , Protein Multimerization , Adaptor Proteins, Signal Transducing/metabolism , COP9 Signalosome Complex , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Multiprotein Complexes/metabolism , NEDD8 Protein , Peptide Hydrolases/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Subunits , Ubiquitins/metabolism
6.
ACS Chem Biol ; 9(6): 1251-6, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24669831

ABSTRACT

We have used a chemically diverse panel of kinase inhibitors to assess the chemical similarity of the ATP-binding sites of cyclin-dependent kinase (CDK) subfamily members in a range of activation states. Using this approach, we find that different activation states of a particular CDK may differ from each other as much as different CDKs in the same activation state. We also find that inhibitors discriminate more effectively among CDK family members in their monomeric state than in their cyclin-bound state, providing direct evidence for the belief that selective binding to inactive kinase states might be more readily achieved than selective binding to active states.


Subject(s)
Adenosine Triphosphate/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Enzyme Inhibitors/metabolism , Amino Acid Sequence , Binding Sites , Enzyme Inhibitors/chemistry , Humans , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Signal Transduction
7.
Curr Protein Pept Sci ; 15(5): 504-17, 2014.
Article in English | MEDLINE | ID: mdl-24555901

ABSTRACT

MPN (Mpr1/Pad1 N-terminal) domain-containing proteins are present throughout all domains of life. In eukaryotes, MPN domain-containing proteins are commonly found in association with other molecules in large protein complexes, where examples comprise; the 26S proteasome and the COP9 (Constitutive photomorphogenesis 9) signalosome complexes, including the MPN subunits, POH1 and Mov34, CSN5 and CSN6, respectively. Examples of MPN domaincontaining proteins that are not incorporated in a large multi-protein complex have also been reported and include AMSH (for associated molecule with the SH3 domain of STAM) and the AMSH-Like Protein (AMSH-LP). Within the MPN domain super-family, two main subclasses have been characterised: the MPN⁺ and MPN⁻ domain-containing proteins. MPN⁺ domain-containing proteins are classified as metalloenzymes responsible for isopeptidase activity. These proteins display a JAMM (JAB1-MPN-MOV34) metalloisopeptidase motif, typically consisting of a canonical sequence (E-x[2]-H-S/T-Hx[7]-S-x[2]-D) and coordinating a zinc ion. The JAMM motif specifies a catalytic centre essential for selective hydrolysis of linkages, contained between ubiquitin/ubiquitin-like proteins and target proteins or between ubiquitin monomers within a polymeric chain. The MPN⁻ family classifies proteins, which lack the key residues present in the typical JAMM motif. These MPN⁻ proteins are void of catalytic activity, but recent studies have proposed a role in mediating protein-protein interactions, in acting as a scaffold or in activity regulation. In light of recent structural and functional studies, a more detailed understanding of these proteins has been gained and is given in the present review.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Animals , Humans , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary
8.
J Med Chem ; 57(1): 56-70, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24304238

ABSTRACT

Evaluation of the effects of purine C-8 substitution within a series of CDK1/2-selective O(6)-cyclohexylmethylguanine derivatives revealed that potency decreases initially with increasing size of the alkyl substituent. Structural analysis showed that C-8 substitution is poorly tolerated, and to avoid unacceptable steric interactions, these compounds adopt novel binding modes. Thus, 2-amino-6-cyclohexylmethoxy-8-isopropyl-9H-purine adopts a "reverse" binding mode where the purine backbone has flipped 180°. This provided a novel lead chemotype from which we have designed more potent CDK2 inhibitors using, in the first instance, quantum mechanical energy calculations. Introduction of an ortho-tolyl or ortho-chlorophenyl group at the purine C-8 position restored the potency of these "reverse" binding mode inhibitors to that of the parent 2-amino-6-cyclohexylmethoxy-9H-purine. By contrast, the corresponding 8-(2-methyl-3-sulfamoylphenyl)-purine derivative exhibited submicromolar CDK2-inhibitory activity by virtue of engineered additional interactions with Asp86 and Lys89 in the reversed binding mode, as confirmed by X-ray crystallography.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Binding Sites , Crystallography, X-Ray , Drug Design , Models, Molecular , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
9.
Proc Natl Acad Sci U S A ; 110(4): 1273-8, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23288897

ABSTRACT

The COP9 (Constitutive photomorphogenesis 9) signalosome (CSN), a large multiprotein complex that resembles the 19S lid of the 26S proteasome, plays a central role in the regulation of the E3-cullin RING ubiquitin ligases (CRLs). The catalytic activity of the CSN complex, carried by subunit 5 (CSN5/Jab1), resides in the deneddylation of the CRLs that is the hydrolysis of the cullin-neural precursor cell expressed developmentally downregulated gene 8 (Nedd8)isopeptide bond. Whereas CSN-dependent CSN5 displays isopeptidase activity, it is intrinsically inactive in other physiologically relevant forms. Here we analyze the crystal structure of CSN5 in its catalytically inactive form to illuminate the molecular basis for its activation state. We show that CSN5 presents a catalytic domain that brings essential elements to understand its activity control. Although the CSN5 active site is catalytically competent and compatible with di-isopeptide binding, the Ins-1 segment obstructs access to its substrate-binding site, and structural rearrangements are necessary for the Nedd8-binding pocket formation. Detailed study of CSN5 by molecular dynamics unveils signs of flexibility and plasticity of the Ins-1 segment. These analyses led to the identification of a molecular trigger implicated in the active/inactive switch that is sufficient to impose on CSN5 an active isopeptidase state. We show that a single mutation in the Ins-1 segment restores biologically relevant deneddylase activity. This study presents detailed insights into CSN5 regulation. Additionally, a dynamic monomer-dimer equilibrium exists both in vitro and in vivo and may be functionally relevant.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/chemistry , Peptide Hydrolases/metabolism , Amino Acid Sequence , Arginine/chemistry , COP9 Signalosome Complex , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , NEDD8 Protein , Peptide Hydrolases/genetics , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Subunits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Ubiquitins/metabolism , Zinc/metabolism
10.
Chem Biol ; 19(8): 1028-40, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22921070

ABSTRACT

Cdk2 promotes DNA replication and is a promising cancer therapeutic target, but its functions appear redundant with Cdk1, an essential Cdk affected by most Cdk2 inhibitors. Here, we present an integrated multidisciplinary approach to address Cdk redundancy. Mathematical modeling of enzymology data predicted conditions allowing selective chemical Cdk2 inhibition. Together with experiments in Xenopus egg extracts, this supports a rate-limiting role for Cdk2 in DNA replication. To confirm this we designed inhibitor-resistant (ir)-Cdk2 mutants using a novel bioinformatics approach. Bypassing inhibition with ir-Cdk2 or with Cdk1 shows that Cdk2 is rate-limiting for replication in this system because Cdk1 is insufficiently active. Additionally, crystal structures and kinetics reveal alternative binding modes of Cdk1-selective and Cdk2-selective inhibitors and mechanisms of Cdk2 inhibitor resistance. Our approach thus provides insight into structure, functions, and biochemistry of a cyclin-dependent kinase.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Amino Acid Sequence , Animals , Binding Sites , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , DNA Replication/drug effects , Humans , Interphase , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Ovum/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Xenopus/growth & development , Xenopus/metabolism
11.
Biochim Biophys Acta ; 1804(3): 511-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19822225

ABSTRACT

The cyclin-dependent kinases (CDKs) have been intensely studied because of their involvement in regulating essential cellular activities that include proliferation and transcription. A series of CDK2-containing structures have informed a general model for the molecular details of CDK activation and regulation. Recent structural studies of other members of the CDK family have lead to a re-appraisal of this model. In this review, we describe alternative CDK-cyclin assemblies taking the recently characterised CDK/cyclin complexes, CDK9/cyclinT1 and CDK4/cyclinD as examples. The differential effects of CDK phosphorylation on CDK activation state and substrate specificity are examined in the light of recent data on CDK2/cyclinA, CDK9/cyclinT, CDK4/cyclinD and Pho85/Pho80. We also present an overview of factors that affect CDK substrate specificity, and, in particular, the contributions that are made by the cyclin subunit. Finally, we review recent results that have helped to unravel the molecular mechanisms underlying the conflicting roles of the Cip/Kip CDK inhibitor family in CDK regulation.


Subject(s)
Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Cyclins/chemistry , Cyclins/metabolism , Models, Molecular , Animals , Humans , Protein Structure, Tertiary/physiology , Substrate Specificity/physiology
12.
Mol Cancer Ther ; 7(9): 2713-24, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790752

ABSTRACT

Cyclin-dependent kinases (CDKs) and their regulators show frequent abnormalities in tumors. Ten low molecular weight pharmacologic inhibitors of CDKs are currently in clinical trials against various cancers, including the 2,6,9-trisubstituted purine (R)-roscovitine (CYC202/Seliciclib). We here report the characterization of N-&-N1, a bioisoster of roscovitine displaying improved antitumoral properties. N-&-N1 shows exquisite selectivity for CDKs, with 2- to 3-fold enhanced potency compared with (R)-roscovitine. Inhibition of retinoblastoma protein phosphorylation and RNA polymerase II Ser2 phosphorylation in neuroblastoma SH-SY5Y cells exposed to N-&-N1 indicates that N-&-N1 is able to inhibit CDKs in a cellular context. N-&-N1 also down-regulates the expression of RNA polymerase. Cocrystal structures of N-&-N1 and (R)-roscovitine in complex with CDK2/cyclin A reveal that both inhibitors adopt similar binding modes. A competitive assay shows that, compared with (R)-roscovitine, N-&-N1 has reduced affinity for Erk2 and pyridoxal kinase. N-&-N1 triggers cell death in a panel of diverse cell lines. Cell death is accompanied by events characteristic of apoptosis: cytochrome c release, activation of effector caspases, and poly(ADP-ribose) polymerase cleavage. Induction of p53 and p21CIP1 and down-regulation of the Mcl-1 antiapoptotic factor were also observed. Studies in mice show that N-&-N1 has pharmacokinetics properties similar to those of (R)-roscovitine. Altogether, these results show that analogues of (R)-roscovitine can be designed with improved antitumor potential.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cell Death/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Models, Biological , Models, Molecular , Neoplasm Proteins/metabolism , Protein Kinase Inhibitors/pharmacokinetics , Purines/chemistry , Roscovitine , Swine , Tissue Extracts/metabolism , Xenograft Model Antitumor Assays
13.
Structure ; 16(2): 228-38, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18275814

ABSTRACT

Malaria is a major threat to world health. The identification of parasite targets for drug development is a priority and parasitic protein kinases suggest themselves as suitable targets as many display profound structural and functional divergences from their host counterparts. In this paper, we describe the structure of the orphan protein kinase, Plasmodium falciparum protein kinase 7 (PFPK7). Several Plasmodium protein kinases contain extensive insertions, and the structure of PFPK7 reveals how these may be accommodated as excursions from the canonical eukaryotic protein kinase fold. The constitutively active conformation of PFPK7 is stabilized by a structural motif in which the role of the conserved phosphorylated residue that assists in structuring the activation loop of many protein kinases is played by an arginine residue. We identify two series of PFPK7 ATP-competitive inhibitors and suggest further developments for the design of selective and potent PFPK7 lead compounds as potential antimalarials.


Subject(s)
Antimalarials/chemistry , Plasmodium falciparum/enzymology , Protein Kinases/chemistry , Protozoan Proteins/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Crystallography, X-Ray , Drug Design , Enzyme Activation , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Sequence Data , Peptides/metabolism , Phosphorylation , Protein Kinases/metabolism , Protozoan Proteins/metabolism
14.
J Med Chem ; 51(4): 737-51, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18232649

ABSTRACT

We report the synthesis and biological characterization of 3-(pyrimidin-4-yl)-7-azaindoles (meriolins), a chemical hybrid between the natural products meridianins and variolins, derived from marine organisms. Meriolins display potent inhibitory activities toward cyclin-dependent kinases (CDKs) and, to a lesser extent, other kinases (GSK-3, DYRK1A). The crystal structures of 1e (meriolin 5) and variolin B (Bettayeb, K.; Tirado, O. M.; Marionneau-Lambert, S.; Ferandin, Y.; Lozach, O.; Morris, J.; Mateo-Lozano, S.; Drückes, P.; Schächtele, C.; Kubbutat, M.; Liger, F.; Marquet, B.; Joseph, B.; Echalier, A.; Endicott, J.; Notario, V.; Meijer, L. Cancer Res. 2007, 67, 8325-8334) in complex with CDK2/cyclin A reveal that the two inhibitors are orientated in very different ways inside the ATP-binding pocket of the kinase. A structure-activity relationship provides further insight into the molecular mechanism of action of this family of kinase inhibitors. Meriolins are also potent antiproliferative and proapoptotic agents in cells cultured either as monolayers or in spheroids. Proapoptotic efficacy of meriolins correlates best with their CDK2 and CDK9 inhibitory activity. Meriolins thus constitute a promising class of pharmacological agents to be further evaluated against the numerous human diseases that imply abnormal regulation of CDKs including cancers, neurodegenerative disorders, and polycystic kidney disease.


Subject(s)
Aza Compounds/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Cyclin A/chemistry , Cyclin-Dependent Kinase 2/chemistry , Indoles/chemical synthesis , Pyrimidines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Aza Compounds/chemistry , Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Crystallization , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/chemistry , Drug Screening Assays, Antitumor , Humans , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Pyrimidines/chemistry , Pyrimidines/pharmacology , Spheroids, Cellular/drug effects , Structure-Activity Relationship
15.
Biochemistry ; 47(7): 1947-56, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18217775

ABSTRACT

A recombinant form of the prototypic diheme bacterial cytochrome c peroxidase (BCCP) from Pseudomonas aeruginosa (PsaCCP) has been expressed in Escherichia coli and purified to homogeneity. This material was used to carry out the first integrated biochemical, spectroscopic and structural investigation of the factors leading to reductive activation of this class of enzymes. A single, tightly bound, Ca2+ ion (K = 3 x 10(10) M-1) found at the domain interface of both the fully oxidized and mixed-valence forms of the enzyme is absolutely required for catalytic activity. Reduction of the electron-transferring (high-potential) heme in the presence of Ca2+ ions triggers substantial structural rearrangements around the active-site (low-potential) heme to allow substrate binding and catalysis. The enzyme also forms a mixed-valence state in the absence of Ca2+ ions, but a combination of electronic absorption, and EPR spectroscopies suggests that under these circumstances the low potential heme remains six-coordinate, unable to bind substrate and therefore catalytically inactive. Our observations strongly suggest that the two mixed-valence forms of native PsaCCP reported previously by Foote and colleagues (Foote, N., Peterson, J., Gadsby, P., Greenwood, C., and Thomson, A. (1985) Biochem. J. 230, 227-237) correspond to the Ca2+-loaded and -depleted forms of the enzyme.


Subject(s)
Cytochrome-c Peroxidase/chemistry , Pseudomonas aeruginosa/enzymology , Base Sequence , Catalysis , Cytochrome-c Peroxidase/metabolism , DNA Primers , Mass Spectrometry , Oxidation-Reduction , Protein Conformation , Spectrophotometry, Ultraviolet
16.
Cancer Res ; 67(17): 8325-34, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804748

ABSTRACT

Protein kinases represent promising anticancer drug targets. We describe here the meriolins, a new family of inhibitors of cyclin-dependent kinases (CDK). Meriolins represent a chemical structural hybrid between meridianins and variolins, two families of kinase inhibitors extracted from various marine invertebrates. Variolin B is currently in preclinical evaluation as an antitumor agent. A selectivity study done on 32 kinases showed that, compared with variolin B, meriolins display enhanced specificity toward CDKs, with marked potency on CDK2 and CDK9. The structures of pCDK2/cyclin A/variolin B and pCDK2/cyclin A/meriolin 3 complexes reveal that the two inhibitors bind within the ATP binding site of the kinase, but in different orientations. Meriolins display better antiproliferative and proapoptotic properties in human tumor cell cultures than their parent molecules, meridianins and variolins. Phosphorylation at CDK1, CDK4, and CDK9 sites on, respectively, protein phosphatase 1alpha, retinoblastoma protein, and RNA polymerase II is inhibited in neuroblastoma SH-SY5Y cells exposed to meriolins. Apoptosis triggered by meriolins is accompanied by rapid Mcl-1 down-regulation, cytochrome c release, and activation of caspases. Meriolin 3 potently inhibits tumor growth in two mouse xenograft cancer models, namely, Ewing's sarcoma and LS174T colorectal carcinoma. Meriolins thus constitute a new CDK inhibitory scaffold, with promising antitumor activity, derived from molecules initially isolated from marine organisms.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Aza Compounds/chemistry , Aza Compounds/metabolism , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cells, Cultured , Crystallography, X-Ray , Cyclin A/chemistry , Cyclin A/metabolism , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/chemistry , Cyclin-Dependent Kinases/metabolism , Drug Evaluation, Preclinical , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Models, Molecular , Protein Binding , Pyrimidines/chemistry , Pyrimidines/metabolism , Substrate Specificity , Xenograft Model Antitumor Assays
17.
J Am Chem Soc ; 128(18): 6012-3, 2006 May 10.
Article in English | MEDLINE | ID: mdl-16669651

ABSTRACT

beta-Piperidinoethylsulfides are oxidized by m-chloroperbenzoic acid to intermediates containing both N-oxide and sulfone functions. These undergo a Cope-type elimination to a vinylsulfone that can be captured by amines to afford beta-aminoethylsulfones. When a beta-aminoethylsulfone group is linked to the 4-position of a phenyl group attached at N-2 of O6-cyclohexylmethylguanine, the resulting derivatives are inhibitors of the cyclin-dependent kinase CDK2. One of the most potent inhibitors (IC50 = 45 nM) contained a N-3-hydroxypropyl group on the aminoethylsulfonyl substituent. The crystal structure of this inhibitor bound to CDK2/cyclin A was determined and shows an unusual network of hydrogen bonds. The synthetic methodology developed can be utilized in multiple-parallel format and has numerous potential applications in medicinal chemistry.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfones/chemistry , Sulfones/pharmacology , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/chemistry , Drug Design , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfones/chemical synthesis
18.
Structure ; 14(1): 107-17, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16407070

ABSTRACT

Bacterial cytochrome c peroxidases contain an electron transferring (E) heme domain and a peroxidatic (P) heme domain. All but one of these enzymes are isolated in an inactive oxidized state and require reduction of the E heme by a small redox donor protein in order to activate the P heme. Here we present the structures of the inactive oxidized and active mixed valence enzyme from Paracoccus pantotrophus. Chain flexibility in the former, as expressed by the crystallographic temperature factors, is strikingly distributed in certain loop regions, and these coincide with the regions of conformational change that occur in forming the active mixed valence enzyme. On the basis of these changes, we postulate a series of events that occur to link the trigger of the electron entering the E heme from either pseudoazurin or cytochrome c(550) and the dissociation of a coordinating histidine at the P heme, which allows substrate access.


Subject(s)
Cytochrome-c Peroxidase/chemistry , Cytochrome-c Peroxidase/physiology , Heme/analogs & derivatives , Oxidoreductases/metabolism , Paracoccus pantotrophus/enzymology , Amino Acid Sequence , Binding Sites , Calcium/metabolism , Catalysis , Crystallography, X-Ray , Enzyme Activation/physiology , Heme/chemistry , Heme/metabolism , Histidine/genetics , Histidine/metabolism , Molecular Sequence Data , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/physiology , Protein Conformation , Protein Structure, Tertiary , Pseudomonas/enzymology , Sequence Alignment
19.
J Inorg Biochem ; 100(4): 551-67, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16434100

ABSTRACT

The bacterial cytochrome c peroxidases contain an electron-transferring haem c (E) and a peroxidatic haem c (P). Many are isolated in an inactive oxidised state. Reduction of the E haem promotes Ca(2+)-dependent spin state and coordination changes at the P haem rendering it accessible to ligand. Recent crystallographic work on the oxidised and mixed valence enzymes has suggested a mechanism by which an electron entering the E haem remotely triggers this activation of the P haem. Binding of hydrogen peroxide at the activated P haem leads to an intermediate catalytic form containing two oxidising equivalents, one of which is a ferryl oxene. This form of the enzyme is then reduced by two single electron transfers to the E haem delivered by small redox proteins such as cytochromes or cupredoxins. The binding of these small redox proteins is dominated by global electrostatic forces but the interfaces of the electron transfer complexes that are formed are largely hydrophobic and relatively non-specific. These features allow very high electron transfer rates in the steady state.


Subject(s)
Bacterial Proteins/chemistry , Cytochrome-c Peroxidase/chemistry , Heme/chemistry , Amino Acid Sequence , Bacterial Proteins/metabolism , Crystallography, X-Ray , Cytochrome-c Peroxidase/metabolism , Electron Transport , Enzyme Activation , Heme/metabolism , Iron/chemistry , Iron/metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Oxidation-Reduction , Protein Conformation , Structure-Activity Relationship
20.
Biochemistry ; 44(6): 1755-67, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15697201

ABSTRACT

Peroxiredoxins (Prxs) constitute a family of thiol peroxidases that reduce hydrogen peroxide, peroxinitrite, and hydroperoxides using a strictly conserved cysteine. Very abundant in all organisms, Prxs are produced as diverse isoforms characterized by different catalytic mechanisms and various thiol-containing reducing agents. The oligomeric state of Prxs and the link with their functionality is a subject of intensive research. We present here a combined X-ray and nuclear magnetic resonance (NMR) study of a plant Prx that belongs to the D-Prx (type II) subfamily. The Populus trichocarpa Prx is the first Prx shown to be regenerated in vitro by both the glutaredoxin and thioredoxin systems. The crystal structure and solution NMR provide evidence that the reduced protein is a specific noncovalent homodimer both in the crystal and in solution. The dimer interface is roughly perpendicular to the plane of the central beta sheet and differs from the interface of A- and B-Prx dimers, where proteins associate in the plane parallel to the beta sheet. The homodimer interface involves residues strongly conserved in the D (type II) Prxs, suggesting that all Prxs of this family can homodimerize. The study provides a new insight into the Prx oligomerism and the basis for protein-protein and enzyme-substrate interaction studies by NMR.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Oxidoreductases/chemistry , Peroxidases/chemistry , Plant Proteins/chemistry , Populus/enzymology , Thermodynamics , Thioredoxins/chemistry , Amino Acid Sequence , Binding Sites , Consensus Sequence , Crystallization , Crystallography, X-Ray , Dimerization , Glutaredoxins , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular/methods , Oxidoreductases/metabolism , Peroxidases/metabolism , Peroxiredoxins , Plant Proteins/metabolism , Solutions , Surface Properties , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...