Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Biochem J ; 440(1): 85-93, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21774789

ABSTRACT

We describe in the present paper mutations of the catalytic subunit α of PKA (protein kinase A) that introduce amino acid side chains into the ATP-binding site and progressively transform the pocket to mimic that of Aurora protein kinases. The resultant PKA variants are enzymatically active and exhibit high affinity for ATP site inhibitors that are specific for Aurora kinases. These features make the Aurora-chimaeric PKA a valuable tool for structure-based drug discovery tasks. Analysis of crystal structures of the chimaera reveal the roles for individual amino acid residues in the binding of a variety of inhibitors, offering key insights into selectivity mechanisms. Furthermore, the high affinity for Aurora kinase-specific inhibitors, combined with the favourable crystallizability properties of PKA, allow rapid determination of inhibitor complex structures at an atomic resolution. We demonstrate the utility of the Aurora-chimaeric PKA by measuring binding kinetics for three Aurora kinase-specific inhibitors, and present the X-ray structures of the chimaeric enzyme in complex with VX-680 (MK-0457) and JNJ-7706621 [Aurora kinase/CDK (cyclin-dependent kinase) inhibitor].


Subject(s)
Adenosine Triphosphate/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Amino Acid Sequence , Amino Acid Substitution , Aurora Kinases , Binding Sites , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Kinetics , Molecular Sequence Data , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins , Sequence Alignment , Triazoles/pharmacology
2.
J Mol Biol ; 403(1): 66-77, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20732331

ABSTRACT

Crystal structures of the catalytic subunit α of cAMP-dependent protein kinase (PKAc) with three adenosine analogue-oligoarginine conjugates (ARCs) are presented. The rationally designed ARCs include moieties that, in combination, target both the ATP- and the peptide-substrate-binding sites of PKAc, thereby taking advantage of high-affinity binding interactions offered by the ATP site while utilizing an additional mechanism for target specificity via binding to the peptide substrate site. The crystal structures demonstrate that, in accord with the previously reported bisubstrate character of ARCs, the inhibitors occupy both binding sites of PKAc. Further, they show new binding modes that may also apply to natural protein substrates of PKAc, which have not been revealed by previous crystallographic studies. The crystal structures described here contribute to the understanding of the substrate-binding patterns of PKAc and should also facilitate the design of inhibitors targeting PKAc and related protein kinases.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Adenosine/chemistry , Arginine/chemistry , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Models, Molecular , Molecular Structure , Protein Structure, Tertiary
3.
Proteomics ; 10(16): 2890-900, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20564261

ABSTRACT

An expression-uncoupled tandem affinity purification assay is introduced which differs from the standard TAP assay by uncoupling the expression of the TAP-bait protein from the target cells. Here, the TAP-tagged bait protein is expressed in Escherichia coli and purified. The two concatenated purification steps of the classical TAP are performed after addition of the purified bait to brain tissue homogenates, cell and nuclear extracts. Without prior genetic manipulation of the target, upscaling, free choice of cell compartments and avoidance of expression triggered heat shock responses could be achieved in one go. By the strategy of separating bait expression from the prey protein environment numerous established, mostly tissue-specific binding partners of the protein kinase A catalytic subunit Cbeta1 were identified, including interactions in binary, ternary and quaternary complexes. In addition, the previously unknown small molecule inhibitor-dependent interaction of Cbeta1 with the cell cycle and apoptosis regulatory protein-1 was verified. The uncoupled tandem affinity purification procedure presented here expands the application range of the in vivo TAP assay and may serve as a simple strategy for identifying cell- and tissue-specific protein complexes.


Subject(s)
Carrier Proteins/metabolism , Cloning, Molecular/methods , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Proteomics/methods , Affinity Labels , Animals , Apoptosis Regulatory Proteins , Brain Chemistry , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Cell Nucleus/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Humans , Mice , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reproducibility of Results
4.
Anal Bioanal Chem ; 395(6): 1713-20, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19590856

ABSTRACT

The catalytic subunit of recombinant wild-type cyclic adenosine monophosphate-dependent protein kinase A (PKA) has been analyzed by a combination of 1D gel electrophoresis, in-gel digestion by trypsin, chymotrypsin, or endoproteinase AspN, and nano-ultraperformance liquid chromatography--MS/MS. The MS/MS spectra were annotated by MASCOT and the annotations were manually controlled. Using Ga(III)-immobilized metal ion affinity chromatography (IMAC), in addition to the four established autophosphorylation sites of the catalytic subunit of recombinant PKA, pSer10, pSer139, pThr197, and pSer338, six new phosphorylated residues have been characterized--pSer14, pThr48, pSer53, pSer212, pSer259, and pSer325. The established phosphorylation sites all are part of a PKA consensus motif and were found to be almost completely modified. In contrast, the newly detected sites were only partially phosphorylated. For estimation of their degree of phosphorylation, a method based on signal intensity measurements was used. For this purpose, signal intensities of all phospho- and non-phosphopeptides containing a particular site were added for estimation of site-specific phosphorylation degrees. This addition was performed over all peptides observed in the different digestion experiments, including their different charge states. pThr48 and pSer259 are located within PKA consensus motifs and were observed to be phosphorylated at 20% and 24%, respectively. pSer14 and pSer53 are located within inverted PKA consensus motifs and were found to be phosphorylated around 10% and 15%, respectively. The sequence environments of pSer212 and pSer325 have no similarity to the PKA consensus motif at all and were observed to be phosphorylated at about 5% or lower. All newly observed phosphorylation sites are located at the surface of the native protein structure of the PKA catalytic subunit. The results add new information on the theme of site-specific (auto)phosphorylation by PKA.


Subject(s)
Chromatography, High Pressure Liquid/methods , Cyclic AMP-Dependent Protein Kinases/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Catalytic Domain , Molecular Sequence Data , Phosphorylation , Recombinant Proteins/chemistry
5.
J Med Chem ; 52(2): 308-21, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19143565

ABSTRACT

The crystal structure of a complex of the catalytic subunit (type alpha) of cAMP-dependent protein kinase (PKA C alpha) with ARC-type inhibitor (ARC-1034), the presumed lead scaffold of previously reported adenosine-oligo-arginine conjugate-based (ARC-type) inhibitors, was solved. Structural elements important for interaction with the kinase were established with specifically modified derivatives of the lead compound. On the basis of this knowledge, a new generation of inhibitors, conjugates of adenosine-4'-dehydroxymethyl-4'-carboxylic acid moiety and oligo(D-arginine), was developed with inhibitory constants well into the subnanomolar range. The structural determinants of selectivity of the new compounds were established in assays with ROCK-II and PKBgamma.


Subject(s)
Adenosine/analogs & derivatives , Basophils/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dipeptides/chemistry , Protein Kinase Inhibitors/chemistry , Adenosine/chemistry , Adenosine/pharmacology , Amino Acid Sequence , Animals , Catalytic Domain , Cattle , Crystallography , Cyclic AMP-Dependent Protein Kinases/chemistry , Dipeptides/pharmacology , Fluorescence Polarization , Models, Molecular , Molecular Sequence Data , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Sequence Homology, Amino Acid
6.
J Biol Chem ; 281(34): 24818-30, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16699172

ABSTRACT

Controlling aberrant kinase-mediated cellular signaling is a major strategy in cancer therapy; successful protein kinase inhibitors such as Tarceva and Gleevec verify this approach. Specificity of inhibitors for the targeted kinase(s), however, is a crucial factor for therapeutic success. Based on homology modeling, we previously identified four amino acids in the active site of Rho-kinase that likely determine inhibitor specificities observed for Rho-kinase relative to protein kinase A (PKA) (in PKA numbering: T183A, L49I, V123M, and E127D), and a fifth (Q181K) that played a surprising role in PKA-PKB hybrid proteins. We have systematically mutated these residues in PKA to their counterparts in Rho-kinase, individually and in combination. Using four Rho-kinase-specific, one PKA-specific, and one pan-kinase-specific inhibitor, we measured the inhibitor-binding properties of the mutated proteins and identify the roles of individual residues as specificity determinants. Two combined mutant proteins, containing the combination of mutations T183A and L49I, closely mimic Rho-kinase. Kinetic results corroborate the hypothesis that side-chain identities form the major determinants of selectivity. An unexpected result of the analysis is the consistent contribution of the individual mutations by simple factors. Crystal structures of the surrogate kinase inhibitor complexes provide a detailed basis for an understanding of these selectivity determinant residues. The ability to obtain kinetic and structural data from these PKA mutants, combined with their Rho-kinase-like selectivity profiles, make them valuable for use as surrogate kinases for structure-based inhibitor design.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Benzamides , Cattle , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Erlotinib Hydrochloride , Imatinib Mesylate , Models, Molecular , Mutagenesis, Site-Directed , Piperazines/chemistry , Piperazines/metabolism , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Quinazolines/chemistry , Quinazolines/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity , rho-Associated Kinases
7.
Biochim Biophys Acta ; 1754(1-2): 38-49, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16269279

ABSTRACT

Protein crystallography can be used throughout the drug discovery process to obtain diverse information critical for structure based drug design. At a minimum, a single target structure may be available. Optimally, and especially for protein kinases, a broad range of crystal structures should be obtained to characterize target flexibility, structure modulation via co-factor binding or posttranslational modification, ligand induced conformational changes, and off-target complex structures for selectivity optimization. The flexibility of the protein kinases is in contrast to the need for "crystallizable" constructs, that is, proteins that crystallize under varying conditions and in varying crystal packing arrangements. Strategies to produce crystallizable protein kinase constructs include truncation to the catalytic domain, co-crystallization with rigidifying ligands, crystallization of known rigid forms, and point mutation to improve homogeneity or mimic less crystallizable proteins. PKA, the prototypical serine/threonine protein kinase, and SRC, a tyrosine kinase and the first identified oncoprotein, provide multiple examples of these various approaches to protein kinase crystallography for drug design.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Drug Design , Protein Kinases/chemistry , src-Family Kinases/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Binding/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Structure-Activity Relationship , src-Family Kinases/metabolism
8.
J Med Chem ; 48(1): 163-70, 2005 Jan 13.
Article in English | MEDLINE | ID: mdl-15634010

ABSTRACT

Protein kinase B (PKB)-selective inhibitors were designed, synthesized, and cocrystallized using the AGC kinase family protein kinase A (PKA, often called cAMP-dependent protein kinase); PKA has been used as a surrogate for other members of this family and indeed for protein kinases in general. The high homology between PKA and PKB includes very similar ATP binding sites and hence similar binding pockets for inhibitors, with only few amino acids that differ between the two kinases. A series of these sites were mutated in PKA in order to improve the surrogate model for a design of PKB-selective inhibitors. Namely, the PKA to PKB exchanges F187L and Q84E enable the design of the selective inhibitors described herein which mimic ATP but extend further into a site not occupied by ATP. In this pocket, selectivity over PKA can be achieved by the introduction of bulkier substituents. Analysis of the cocrystal structures and binding studies were performed to rationalize the selectivity and improve the design.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Binding Sites , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Models, Molecular , Mutation , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Reproducibility of Results , Structure-Activity Relationship
9.
Oncol Res ; 14(6): 267-78, 2004.
Article in English | MEDLINE | ID: mdl-15206489

ABSTRACT

The AGC group of protein kinases comprises several targets for small molecule inhibitors of therapeutic significance. Crystal structure data facilitate the design or improvement of selective inhibitory molecules. Cross-selectivity of kinase inhibitors is often observed among closely related enzymes. Usually an obstacle for inhibitor design, cross-selectivity can be useful to obtain structural data from a related kinase, if not available from the original target. Protein kinase A (PKA), a representative of the AGC kinase group, has been cocrystallized with AGC group inhibitors from diverse chemical groups, thus providing structural information about binding modes, selectivity, and cross-selectivity. "Ersatz" kinases were created by mutating the inhibitor binding site of PKA to resemble other related kinases from the AGC group. The cocrystallization of these ersatz kinases with certain AGC group small molecule inhibitors elucidated some aspects of protein kinase inhibitor selectivity in this group of kinases.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Binding Sites , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , rho-Associated Kinases
10.
Biochemistry ; 43(24): 7743-9, 2004 Jun 22.
Article in English | MEDLINE | ID: mdl-15196017

ABSTRACT

Protein kinases comprise the major enzyme family critically involved in signal transduction pathways; posttranslational modifications affect their regulation and determine signaling states. The prototype protein kinase A (PKA) possesses an N-terminal alpha-helix (Helix A) that is atypical for kinases and is thus a major distinguishing feature of PKA. Its physiological function may involve myristoylation at the N-terminus and modulation via phosphorylation at serine 10. Here we describe an unusual structure of an unmyristoylated PKA, unphosphorylated at serine 10, with a completely ordered N-terminus. Using standard conditions (e.g., PKI 5-24, ATP site ligand, MEGA-8), a novel 2-fold phosphorylated PKA variant showed the ordered N-terminus in a new crystal packing arrangement. Thus, the critical factor for structuring the N-terminus is apparently the absence of phosphorylation of Ser10. The flexibility of the N-terminus, its myristoylation, and the conformational dependence on the phosphorylation state are consistent with a functional role for myristoylation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Myristic Acid/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Phosphorylation , Protein Folding , Serine/metabolism
11.
J Biol Chem ; 279(22): 23679-90, 2004 May 28.
Article in English | MEDLINE | ID: mdl-14996846

ABSTRACT

As the key mediators of eukaryotic signal transduction, the protein kinases often cause disease, and in particular cancer, when disregulated. Appropriately selective protein kinase inhibitors are sought after as research tools and as therapeutic drugs; several have already proven valuable in clinical use. The AGC subfamily protein kinase C (PKC) was identified early as a cause of cancer, leading to the discovery of a variety of PKC inhibitors. Despite its importance and early discovery, no crystal structure for PKC has yet been reported. Therefore, we have co-crystallized PKC inhibitor bisindolyl maleimide 2 (BIM2) with PKA variants to study its binding interactions. BIM2 co-crystallized as an asymmetric pair of kinase-inhibitor complexes. In this asymmetric unit, the two kinase domains have different lobe configurations, and two different inhibitor conformers bind in different orientations. One kinase molecule (A) is partially open with respect to the catalytic conformation, the other (B) represents the most open conformation of PKA reported so far. In monomer A, the BIM2 inhibitor binds tightly via an induced fit in the ATP pocket. The indole moieties are rotated out of the plane with respect to the chemically related but planar inhibitor staurosporine. In molecule B a different conformer of BIM2 binds in a reversed orientation relative to the equivalent maleimide atoms in molecule A. Also, a critical active site salt bridge is disrupted, usually indicating the induction of an inactive conformation. Molecular modeling of the clinical phase III PKC inhibitor LY333531 into the electron density of BIM2 reveals the probable binding mechanism and explains selectivity properties of the inhibitor.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Maleimides/pharmacology , Protein Kinase C/antagonists & inhibitors , Animals , Binding Sites , Cattle , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/metabolism , Indoles/chemistry , Indoles/metabolism , Maleimides/chemistry , Maleimides/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry
12.
Structure ; 11(12): 1595-607, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14656443

ABSTRACT

Protein kinases require strict inactivation to prevent spurious cellular signaling; overactivity can cause cancer or other diseases and necessitates selective inhibition for therapy. Rho-kinase is involved in such processes as tumor invasion, cell adhesion, smooth muscle contraction, and formation of focal adhesion fibers, as revealed using inhibitor Y-27632. Another Rho-kinase inhibitor, HA-1077 or Fasudil, is currently used in the treatment of cerebral vasospasm; the related nanomolar inhibitor H-1152P improves on its selectivity and potency. We have determined the crystal structures of HA-1077, H-1152P, and Y-27632 in complexes with protein kinase A (PKA) as a surrogate kinase to analyze Rho-kinase inhibitor binding properties. Features conserved between PKA and Rho-kinase are involved in the key binding interactions, while a combination of residues at the ATP binding pocket that are unique to Rho-kinase may explain the inhibitors' Rho-kinase selectivity. Further, a second H-1152P binding site potentially points toward PKA regulatory domain interaction modulators.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Cyclic AMP-Dependent Protein Kinases/chemistry , Enzyme Inhibitors/pharmacology , Pyridines/pharmacology , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cattle , Cell Adhesion , Crystallography, X-Ray , Electrons , Escherichia coli/metabolism , Glycine/chemistry , Kinetics , Models, Chemical , Models, Molecular , Molecular Sequence Data , Muscle Contraction , Muscle, Smooth/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid
13.
J Mol Biol ; 329(5): 1021-34, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12798691

ABSTRACT

The mutation of well behaved enzymes in order to simulate less manageable cognates is the obvious approach to study specific features of the recalcitrant target. Accordingly, the prototypical protein kinase PKA serves as a model for many kinases, including the closely related PKB, an AGC family protein kinase now implicated as oncogenic in several cancers. Two residues that differ between the alpha isoforms of PKA and PKB at the adenine-binding site generate differing shapes of the binding surface and are likely to play a role in ligand selectivity. As the corresponding mutations in PKA, V123A would enlarge the adenine pocket, while L173M would alter both the shape and its electronic character of the adenine-binding surface. We have determined the structures of the corresponding double mutant (PKAB2: PKAalpha V123A, L173M) in apo and MgATP-bound states, and observed structural alterations of a residue not previously involved in ATP-binding interactions: the side-chain of Q181, which in native PKA points away from the ATP-binding site, adopts in apo double mutant protein a new rotamer conformation, which places the polar groups at the hinge region in the ATP pocket. MgATP binding forces Q181 back to the position seen in native PKA. The crystal structure shows that ATP binding geometry is identical with that in native PKA but in this case was determined under conditions with only a single Mg ion ligand. Surface plasmon resonance spectroscopy studies show that significant energy is required for this ligand-induced transition. An additional PKA/PKB mutation, Q181K, corrects the defect, as shown both by the crystal structure of triple mutant PKAB3 (PKAalpha V123A, L173M, Q181K) and by surface plasmon resonance spectroscopy binding studies with ATP and three isoquinoline inhibitors. Thus, the triple mutant serves well as an easily crystallizable model for PKB inhibitor interactions. Further, the phenomenon of Q181 shows how crystallographic analysis should accompany mutant studies to monitor possible spurious structural effects.


Subject(s)
Adenosine Triphosphate/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Binding Sites , Biosensing Techniques , Crystallography, X-Ray , Cyclic AMP-Dependent Protein Kinases/chemistry , Magnesium/metabolism , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Mutation , Protein Conformation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Sequence Homology, Amino Acid , Sulfur/chemistry
14.
Pharmacol Ther ; 93(2-3): 99-111, 2002.
Article in English | MEDLINE | ID: mdl-12191603

ABSTRACT

Protein kinases catalyze the phosphotransfer reaction fundamental to most signaling and regulatory processes in the eukaryotic cell. Absolute control of individual protein kinase activity is, therefore, of utmost importance to signaling fidelity in the cell. Mechanisms for activity modulation, including complete and reversible inactivation, have been shown by crystal structures of many active and inactive protein kinases. The structures of inactivated kinases, compared with those of active and catalytically competent kinases such as the protein kinase A catalytic subunit, highlight recurring structural alterations among a set of elements of the catalytic kinase core. These 'activity modulation sites' apparently comprise the principal evolved mechanisms for control of enzyme activity in the catalytic domain. In combination, they enable diverse physiological regulatory mechanisms operative for most protein kinases. Identification and characterization of these sites should impact strategies for discovery and design of target-specific therapeutic drugs as the range of structural variations for specific kinases becomes known. The principle site, the ATP-binding pocket, is the target of many physiological regulators and also most experimental or therapeutic inhibitors, which typically block it in a competitive or allosteric fashion. Co-crystallization studies with protein kinase A and other kinases have revealed binding features of several classes of protein kinase inhibitors. Ligand-induced structural changes are common and tend to optimize buried surface areas. The ability to optimize binding energies arising from the hydrophobic effect creates a logarithmic dependence of binding energy on buried surface areas. Exceptions to this rule arise for specific inhibitor classes, and possibly also as artifacts of structure determination.


Subject(s)
Adenosine Triphosphate/metabolism , Molecular Conformation , Protein Kinases , Signal Transduction/physiology , Binding Sites , Protein Kinase Inhibitors , Protein Kinases/metabolism , Protein Kinases/physiology , Signal Transduction/drug effects , Structure-Activity Relationship
15.
Proteomics ; 2(7): 911-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12124936

ABSTRACT

Using the combination of in-gel elastase digestion, immobilized metal affinity chromatography and high resolution electrospray tandem mass spectrometry, the phosphorylation sites of two phosphoproteins were determined. Complete coverage of all phosphorylation sites (Ser10, Ser139, Thr197, Ser338) of the model phosphoprotein protein kinase A C(alpha)-subunit could be achieved by this strategy in the low picomole range. In addition, three previously unknown phosphorylation sites of the human transcription initiation factor TIF-IA (Ser44, Ser170, Ser172) were determined in this way. Both phosphoproteins could be identified in a protein database on the basis of their elastase generated phosphopeptides alone. The data of seven phosphopeptides were used for identification of protein kinase A, and those of two phosphopeptides for TIF-IA, respectively. The accurate mass data of the electrospray mass spectra recorded at high resolution are extremely useful for sequencing of the elastase generated phosphopeptides and for protein identification by database searching.


Subject(s)
Chromatography, Affinity/methods , Pancreatic Elastase/metabolism , Phosphoproteins/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Databases, Protein , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Molecular Sequence Data , Molecular Weight , Phosphoproteins/metabolism , Phosphorylation , Pol1 Transcription Initiation Complex Proteins , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Transcription Factors/chemistry , Transcription Factors/metabolism
16.
Biochemistry ; 41(19): 5968-77, 2002 May 14.
Article in English | MEDLINE | ID: mdl-11993991

ABSTRACT

Cell signaling pathways rely on phosphotransfer reactions that are catalyzed by protein kinases. The protein kinases themselves are typically regulated by phosphorylation and concurrent structural rearrangements, both near the catalytic site and elsewhere. Thus, physiological function requires posttranslational modification and deformable structures. A prototypical example is provided by cyclic AMP-dependent protein kinase (PKA). It is activated by phosphorylation, is inhomogeneously phosphorylated when expressed in bacteria, and exhibits a wide range of dynamic properties. Here we use (31)P nuclear magnetic resonance (NMR) spectroscopy to characterize the phosphorylation states and to estimate the flexibility of the phosphorylation sites of 2-, 3-, and 4-fold phosphorylated PKA. The phosphorylation sites Ser10, Ser139, Thr197, and Ser338 are assigned to individual NMR resonances, assisted by complexation with AMP-PNP and dephosphorylation with alkaline phosphatase. Rotational diffusion correlation times estimated from resonance line widths show progressively increasing flexibilities for phosphothreonine 197, phosphoserines 139 and 338, and disorder at phosphoserine 10, consistent with crystal structures of PKA. However, because the apparent rotational diffusion correlation time fitted for phosphothreonine 197 of the activation loop is longer than the overall PKA rotational diffusion time, microsecond to millisecond time scale conformational exchange effects involving motions of phosphothreonine 197 are probable. These may represent "open"-"closed" transitions of the uncomplexed protein in solution. These data represent direct measurements of flexibilities also associated with functional properties, such as ATP binding and membrane association, and illustrate the applicability of (31)P NMR for functional and dynamic characterization of protein kinase phosphorylation sites.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , Alkaline Phosphatase/metabolism , Animals , Binding Sites , Cyclic AMP-Dependent Protein Kinases/metabolism , In Vitro Techniques , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Phosphorus , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine/chemistry , Threonine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL