Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Sci Transl Med ; 14(675): eabp9159, 2022 12 14.
Article En | MEDLINE | ID: mdl-36516271

The epidermis is a barrier that prevents water loss while keeping harmful substances from penetrating the host. The impermeable cornified layer of the stratum corneum is maintained by balancing continuous turnover driven by epidermal basal cell proliferation, suprabasal cell differentiation, and corneal shedding. The epidermal desquamation process is tightly regulated by balance of the activities of serine proteases of the Kallikrein-related peptidases (KLK) family and their cognate inhibitor lymphoepithelial Kazal type-related inhibitor (LEKTI), which is encoded by the serine peptidase inhibitor Kazal type 5 gene. Imbalance of proteolytic activity caused by a deficiency of LEKTI leads to excessive desquamation due to increased activities of KLK5, KLK7, and KLK14 and results in Netherton syndrome (NS), a debilitating condition with an unmet clinical need. Increased activity of KLKs may also be pathological in other dermatoses such as atopic dermatitis (AD). Here, we describe the discovery of inhibitory antibodies against murine KLK5 and KLK7 that could compensate for the deficiency of LEKTI in NS. These antibodies are protective in mouse models of NS and AD and, when combined, promote improved skin barrier integrity and reduced inflammation. To translate these findings, we engineered a humanized bispecific antibody capable of potent inhibition of human KLK5 and KLK7. A crystal structure of KLK5 bound to the inhibitory Fab revealed that the antibody binds distal to its active site and uses a relatively unappreciated allosteric inhibition mechanism. Treatment with the bispecific anti-KLK5/7 antibody represents a promising therapy for clinical development in NS and other inflammatory dermatoses.


Dermatitis, Atopic , Netherton Syndrome , Skin Diseases , Mice , Humans , Animals , Netherton Syndrome/genetics , Netherton Syndrome/metabolism , Netherton Syndrome/pathology , Dermatitis, Atopic/pathology , Serine Peptidase Inhibitor Kazal-Type 5/metabolism , Epidermis/pathology , Skin Diseases/metabolism , Antibodies/metabolism , Kallikreins/metabolism
2.
Bioorg Med Chem ; 29: 115865, 2021 01 01.
Article En | MEDLINE | ID: mdl-33285410

Recent years have seen a resurgence in drug discovery efforts aimed at the identification of covalent inhibitors which has led to an explosion of literature reports in this area and most importantly new approved therapies. These reports and breakthroughs highlight the significant investments made across the industry in SAR campaigns to optimize inhibitors. The potency of covalent inhibitors is generally considered to be more accurately described by the time-independent kinetic parameter kinact/Ki rather than a by a simple IC50 since the latter is a time-dependent parameter. Enzyme substrate concentrations are an additional important factor to consider when attempting to translate parameters derived from enzymology experiments to phenotypic behavior in a physiologically relevant cell-based system. Theoretical and experimental investigations into the relationship between IC50, time, substrate concentration and Kinact/Ki provided us with an effective approach to provide meaningful data for SAR optimization. The data we generated for our JAK3 irreversible covalent inhibitor program using IC50 values provided by enzyme assays with long incubations (>1h) coupled with physiological substrate concentration provided the medicinal chemist with optimal information in a rapid and efficient manner. We further document the wide applicability of this method by applying it to other enzymes systems where we have run covalent inhibitor programs.


Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Janus Kinase 3/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemistry , Recombinant Proteins , Structure-Activity Relationship
3.
ACS Chem Biol ; 14(6): 1205-1216, 2019 06 21.
Article En | MEDLINE | ID: mdl-31059222

Access to cryptic binding pockets or allosteric sites on a kinase that present themselves when the enzyme is in a specific conformational state offers a paradigm shift in designing the next generation small molecule kinase inhibitors. The current work showcases an extensive and exhaustive array of in vitro biochemical and biophysical tools and techniques deployed along with structural biology efforts of inhibitor-bound kinase complexes to characterize and confirm the cryptic allosteric binding pocket and docking mode of the small molecule actives identified for hTrkA. Specifically, assays were designed and implemented to lock the kinase in a predominantly active or inactive conformation and the effect of the kinase inhibitor probed to understand the hTrkA binding and hTrkB selectivity. The current outcome suggests that inhibitors with a fast association rate take advantage of the inactive protein conformation and lock the kinase state by also exhibiting a slow off-rate. This in turn shifts the inactive/active state protein conformational equilibrium cycle, affecting the subsequent downstream signaling.


Protein Kinase Inhibitors/pharmacology , Receptor, trkA/antagonists & inhibitors , Allosteric Regulation , Animals , Computer Simulation , Humans , Ligands , Neurites , PC12 Cells , Protein Kinase Inhibitors/metabolism , Rats , Receptor, trkA/metabolism
4.
ACS Med Chem Lett ; 10(1): 80-85, 2019 Jan 10.
Article En | MEDLINE | ID: mdl-30655951

Potent covalent inhibitors of Bruton's tyrosine kinase (BTK) based on an aminopyrazole carboxamide scaffold have been identified. Compared to acrylamide-based covalent reactive groups leading to irreversible protein adducts, cyanamide-based reversible-covalent inhibitors provided the highest combined BTK potency and EGFR selectivity. The cyanamide covalent mechanism with BTK was confirmed through enzyme kinetic, NMR, MS, and X-ray crystallographic studies. The lead cyanamide-based inhibitors demonstrated excellent kinome selectivity and rat pharmacokinetic properties.

5.
Protein Pept Lett ; 19(5): 485-91, 2012 May.
Article En | MEDLINE | ID: mdl-22486643

The Janus kinase (JAK) family consists of four members: JAK-1, -2, -3 and tyrosine kinase 2 (TYK-2). Recent work suggests that cytokine signaling through TYK-2 may play a critical role in a number of inflammatory processes. We recently described the purification and characterization of phosphorylated isoforms of the TYK-2 kinase domain (TYK-2 KD) and its high resolution 3D structure in the presence of inhibitors. We now report the expression and a two-step purification procedure for the doubly tagged full-length construct, H6-FL-TYK-2-FLAG, and examine its properties compared to those of TYK-2 KD. In the presence of ATP and a peptide substrate, H6-FL-TYK-2-FLAG showed a marked lag in phosphopeptide product formation, while TYK-2 KD showed no such lag. This lag could be eliminated by ATP pretreatment, suggesting that the H6-FL-TYK-2-FLAG enzyme was activated by phosphorylation. The potencies of several nanomolar inhibitors were similar for TYK-2 KD and H6-FL-TYK-2-FLAG. However, these same inhibitors were about 1000 times less potent inhibiting the autophosphorylation of H6-FL-TYK-2-FLAG than they were inhibiting the phosphorylation of a peptide substrate modeled after the activation loop sequence of TYK-2. This intriguing result suggests that autophosphorylation and, thus, activation of H6-FL-TYK-2-FLAG is relatively insensitive to inhibition and that present inhibitors act to inhibit TYK-2 subsequent to activation. Inhibition of TYK-2 autophosphorylation may represent a new area of investigation for the JAK family.


TYK2 Kinase/antagonists & inhibitors , TYK2 Kinase/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Catalytic Domain , Electrophoresis, Polyacrylamide Gel , Histidine/chemistry , Humans , Kinetics , Oligopeptides/chemistry , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , TYK2 Kinase/chemistry , TYK2 Kinase/isolation & purification
6.
Biochem J ; 444(1): 79-88, 2012 May 15.
Article En | MEDLINE | ID: mdl-22397330

SphK (sphingosine kinase) is the major source of the bioactive lipid and GPCR (G-protein-coupled receptor) agonist S1P (sphingosine 1-phosphate). S1P promotes cell growth, survival and migration, and is a key regulator of lymphocyte trafficking. Inhibition of S1P signalling has been proposed as a strategy for treatment of inflammatory diseases and cancer. In the present paper we describe the discovery and characterization of PF-543, a novel cell-permeant inhibitor of SphK1. PF-543 inhibits SphK1 with a K(i) of 3.6 nM, is sphingosine-competitive and is more than 100-fold selective for SphK1 over the SphK2 isoform. In 1483 head and neck carcinoma cells, which are characterized by high levels of SphK1 expression and an unusually high rate of S1P production, PF-543 decreased the level of endogenous S1P 10-fold with a proportional increase in the level of sphingosine. In contrast with past reports that show that the growth of many cancer cell lines is SphK1-dependent, specific inhibition of SphK1 had no effect on the proliferation and survival of 1483 cells, despite a dramatic change in the cellular S1P/sphingosine ratio. PF-543 was effective as a potent inhibitor of S1P formation in whole blood, indicating that the SphK1 isoform of sphingosine kinase is the major source of S1P in human blood. PF-543 is the most potent inhibitor of SphK1 described to date and it will be useful for dissecting specific roles of SphK1-driven S1P signalling.


Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyrrolidines/pharmacology , Sphingosine/analogs & derivatives , Sulfones/pharmacology , Cell Line, Tumor , Cell Membrane Permeability , Humans , Lysophospholipids/blood , Methanol , Phosphorylation , Pyrrolidines/chemical synthesis , Pyrrolidines/metabolism , Sphingosine/blood , Sphingosine/metabolism , Substrate Specificity , Sulfones/chemical synthesis , Sulfones/metabolism
7.
Anal Biochem ; 421(2): 556-65, 2012 Feb 15.
Article En | MEDLINE | ID: mdl-22107886

Fatty acid amide hydrolase (FAAH) is one of the main enzymes responsible for the degradation of the endocannabinoid anandamide (N-arachidonoylethanolamine, AEA). FAAH inhibitors may be useful in treating many disorders involving inflammation and pain. Although brain FAAH may be the relevant target for inhibition, rat studies show a correlation between blood and brain FAAH inhibition, allowing blood FAAH activity to be used as a target biomarker. Building on experience with a rat leukocyte FAAH activity assay using [³H]AEA, we have developed a human leukocyte assay using stably labeled [²H4]AEA as substrate. The deuterium-labeled ethanolamine reaction product ([²H4]EA) was analyzed by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) in the positive electrospray ionization (ESI) mode. The response for [²H4]EA was linear from 10 nM to 10 µM, and the analysis time was less than 6 min/sample. Results using the [²H4]AEA and HPLC-MS/MS method agreed well with those obtained using the [³H]AEA radiometric assay. In addition to using a nonradioactive substrate, the HPLC-MS/MS method had increased sensitivity with lower background. Importantly, the assay preserved partial FAAH inhibition resulting from ex vivo treatment with a time-dependent irreversible inhibitor, suggesting its utility with clinical samples. The assay has been used to profile the successful inhibition of FAAH in recent clinical trials.


Amidohydrolases/blood , Chromatography, High Pressure Liquid/methods , Leukocytes/enzymology , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Amidohydrolases/antagonists & inhibitors , Biomarkers/blood , Humans
8.
9.
Biochim Biophys Acta ; 1804(9): 1817-31, 2010 Sep.
Article En | MEDLINE | ID: mdl-20580860

Because of their central role in programmed cell death, the caspases are attractive targets for developing new therapeutics against cancer and autoimmunity, myocardial infarction and ischemic damage, and neurodegenerative diseases. We chose to target caspase-3, an executioner caspase, and caspase-8, an initiator caspase, based on the vast amount of information linking their functions to diseases. Through a structure-based drug design approach, a number of novel beta-strand peptidomimetic compounds were synthesized. Kinetic studies of caspase-3 and caspase-8 inhibition were carried out with these urazole ring-containing irreversible peptidomimetics and a known irreversible caspase inhibitor, Z-VAD-fmk. Using a stopped-flow fluorescence assay, we were able to determine individual kinetic parameters of caspase-3 and caspase-8 inhibition by these inhibitors. Z-VAD-fmk and the peptidomimetic inhibitors inhibit caspase-3 and caspase-8 via a three-step kinetic mechanism. Inhibition of both caspase-3 and caspase-8 by Z-VAD-fmk and of caspase-3 by the peptidomimetic inhibitors proceeds via two rapid equilibrium steps followed by a relatively fast inactivation step. However, caspase-8 inhibition by the peptidomimetics goes through a rapid equilibrium step, a slow-binding reversible step, and an extremely slow inactivation step. The crystal structures of inhibitor complexes of caspases-3 and -8 validate the design of the inhibitors by illustrating in detail how they mimic peptide substrates. One of the caspase-8 structures also shows binding at a secondary, allosteric site, providing a possible route to the development of noncovalent small molecule modulators of caspase activity.


Caspase 3/chemistry , Caspase 8/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Caspase Inhibitors , Crystallization , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/chemical synthesis , Humans , Kinetics , Molecular Structure , Protein Conformation
10.
Biochem Biophys Res Commun ; 396(2): 543-8, 2010 May 28.
Article En | MEDLINE | ID: mdl-20435017

The Janus kinase family consists of four members: JAK-1, -2, -3 and TYK-2. While JAK-2 and JAK-3 have been well characterized biochemically, there is little data on TYK-2. Recent work suggests that TYK-2 may play a critical role in the development of a number of inflammatory processes. We have carried out a series of biochemical studies to better understand TYK-2 enzymology and its inhibition profile, in particular how the TYK-2 phosphorylated forms differ from each other and from the other JAK family members. We have expressed and purified milligram quantities of the TYK-2 kinase domain (KD) to high purity and developed a method to separate the non-, mono- (pY(1054)) and di-phosphorylated forms of the enzyme. Kinetic studies (k(cat(app))/K(m(app))) indicated that phosphorylation of the TYK-2-KD (pY(1054)) increased the catalytic efficiency 4.4-fold compared to its non-phosphorylated form, while further phosphorylation to generate the di-phosphorylated enzyme imparted no further increase in activity. These results are in contrast to those obtained with the JAK-2-KD and JAK-3-KD, where little or no increase in activity occurred upon mono-phosphorylation, while di-phosphorylation resulted in a 5.1-fold increase in activity for the JAK-2-KD. Moreover, ATP-competitive inhibitors demonstrated 10-30-fold shifts in potency (K(i(app))) as a result of the TYK-2-KD phosphorylation state, while the shifts for JAK-3-KD were only 2-3-fold and showed little or no change for JAK-2-KD. Thus, the phosphorlyation state imparted differential effects on both activity and inhibition within the JAK family of kinases.


Janus Kinase 2/biosynthesis , Janus Kinase 2/isolation & purification , Janus Kinase 3/biosynthesis , Janus Kinase 3/isolation & purification , TYK2 Kinase/biosynthesis , TYK2 Kinase/isolation & purification , Animals , Catalysis , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Mice , Phosphorylation , Protein Structure, Tertiary , TYK2 Kinase/antagonists & inhibitors
11.
Bioorg Med Chem Lett ; 20(2): 576-80, 2010 Jan 15.
Article En | MEDLINE | ID: mdl-20005097

Potent, highly selective and orally-bioavailable MMP-13 inhibitors have been identified based upon a (pyridin-4-yl)-2H-tetrazole scaffold. Co-crystal structure analysis revealed that the inhibitors bind at the S(1)(') active site pocket and are not ligands for the catalytic zinc atom. Compound 29b demonstrated reduction of cartilage degradation biomarker (TIINE) levels associated with cartilage protection in a preclinical rat osteoarthritis model.


Matrix Metalloproteinase Inhibitors , Osteoarthritis/drug therapy , Picolinic Acids/chemistry , Protease Inhibitors/chemistry , Tetrazoles/chemistry , Administration, Oral , Animals , Binding Sites , Cartilage/drug effects , Cartilage/metabolism , Catalytic Domain , Crystallography, X-Ray , Disease Models, Animal , Drug Discovery , Matrix Metalloproteinase 13/metabolism , Picolinic Acids/chemical synthesis , Picolinic Acids/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Rats , Tetrazoles/chemical synthesis , Tetrazoles/pharmacology , Zinc/chemistry
12.
J Biol Chem ; 284(36): 24185-91, 2009 Sep 04.
Article En | MEDLINE | ID: mdl-19586907

Several inhibitors of a series of cis-1(S)2(R)-amino-2-indanol-based compounds were reported to be selective for the aggrecanases, ADAMTS-4 and -5 over other metalloproteases. To understand the nature of this selectivity for aggrecanases, the inhibitors, along with the broad spectrum metalloprotease inhibitor marimastat, were independently bound to the catalytic domain of ADAMTS-5, and the corresponding crystal structures were determined. By comparing the structures, it was determined that the specificity of the relative inhibitors for ADAMTS-5 was not driven by a specific interaction, such as zinc chelation, hydrogen bonding, or charge interactions, but rather by subtle and indirect factors, such as water bridging, ring rigidity, pocket size, and shape, as well as protein conformation flexibility.


Endopeptidases/chemistry , Enzyme Inhibitors/chemistry , ADAM Proteins/chemistry , ADAMTS4 Protein , ADAMTS5 Protein , Animals , Cattle , Humans , Hydrogen Bonding , Procollagen N-Endopeptidase/chemistry , Protein Structure, Tertiary , Structural Homology, Protein , Zinc/chemistry
13.
Biochemistry ; 46(21): 6393-401, 2007 May 29.
Article En | MEDLINE | ID: mdl-17487981

ADAMTS-4 (aggrecanase-1) is implicated in the breakdown of articular cartilage and is an attractive target for therapeutic intervention in arthritis. Cleavage of the native substrate, aggrecan, occurs through exosite interactions and peptide sequence recognition. Although expected to be competitive with aggrecan, the hydroxamic acid, SC81956, demonstrated noncompetitive inhibition kinetics with a Ki of 23 nM. The IC50 of SC81956 did not change when aggrecan was varied from 12.8 to 200 nM (0.2-3.3 times the apparent aggrecan Km of 61 nM) but was shifted as expected for a competitive inhibitor when increasing levels of a low molecular weight peptide substrate were added to a fluorogenic peptide assay system. These observations are consistent with a model for aggrecan cleavage where substrate initially binds at an exosite, followed by binding of the appropriate peptide sequence at the active site. A peptide-competitive inhibitor could bind both free enzyme and initial substrate-enzyme exosite complex but would be excluded by the final Michaelis complex. Noncompetitive appearing kinetics for such inhibitors is predicted as long as the equilibrium between the two forms of enzyme-substrate complex significantly favors the initial exosite complex. In support, hydrolysis of a low molecular weight peptide substrate and its inhibition by SC81956 were unaffected by aggrecan concentrations substantially above the Km. These observations suggest that the apparent Km for aggrecan cleavage predominately reflects the exosite interaction. Consequently, the efficacy of active-site inhibitors of ADAMTS-4 will not be limited by competition with native substrate as predicted from the Km determined by traditional kinetic models.


ADAM Proteins/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Procollagen N-Endopeptidase/antagonists & inhibitors , ADAMTS4 Protein , Animals , Binding Sites , Binding, Competitive , Cartilage, Articular/metabolism , Cattle , Humans , Kinetics , Peptides/metabolism , Substrate Specificity
14.
J Biol Chem ; 279(17): 17554-61, 2004 Apr 23.
Article En | MEDLINE | ID: mdl-14715656

Osteoarthritis is characterized by the loss of aggrecan and collagen from the cartilage extracellular matrix. The proteinases responsible for the breakdown of cartilage aggrecan include ADAMTS-4 (aggrecanase 1) and ADAMTS-5 (aggrecanase 2). Post-translational inhibition of ADAMTS-4/-5 activity may be important for maintaining normal homeostasis of aggrecan metabolism, and thus, any disruption to this inhibition could lead to accelerated aggrecan breakdown. To date TIMP-3 (tissue inhibitor of matrix metalloproteinases-3) is the only endogenous inhibitor of ADAMTS-4/-5 that has been identified. In the present studies we identify alpha(2)-macroglobulin (alpha(2)M) as an additional endogenous inhibitor of ADAMTS-4 and ADAMTS-5. alpha(2)M inhibited the activity of both ADAMTS-4 and ADAMTS-5 in a concentration-dependent manner, demonstrating 1:1 stoichiometry with second-order rate constants on the order of 10(6) and 10(5) m(-1) s(-1), respectively. Inhibition of the aggrecanases was mediated by proteolysis of the bait region within alpha(2)M, resulting in physical entrapment of these proteinases. Both ADAMTS-4 and ADAMTS-5 cleaved alpha(2)M at Met(690)/Gly(691), representing a novel proteinase cleavage site within alpha(2)M and a novel site of cleavage for ADAMTS-4 and ADAMTS-5. Finally, the use of the anti-neoepitope antibodies to detect aggrecanase-generated alpha(2)M-fragments in synovial fluid was investigated and found to be uninformative.


Enzyme Inhibitors/chemistry , Metalloendopeptidases/metabolism , Procollagen N-Endopeptidase/metabolism , alpha-Macroglobulins/physiology , ADAM Proteins , ADAMTS4 Protein , ADAMTS5 Protein , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , Cattle , Cell Line , Dose-Response Relationship, Drug , Drosophila , Epitopes/chemistry , Glutamic Acid/chemistry , Humans , Kinetics , Metalloendopeptidases/antagonists & inhibitors , Methylamines/chemistry , Molecular Sequence Data , Nasal Septum/metabolism , Peptides/chemistry , Procollagen N-Endopeptidase/antagonists & inhibitors , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Synovial Fluid/metabolism , Time Factors , Tissue Inhibitor of Metalloproteinase-3/metabolism , alpha-Macroglobulins/metabolism
15.
Antiviral Res ; 55(2): 291-306, 2002 Aug.
Article En | MEDLINE | ID: mdl-12103430

Herpesviruses encode a protease that is essential for virus replication. The protease undergoes cleavage to a processed form during capsid maturation. A recombinant 75 kDa form of the protease from human cytomegalovirus was purified and compared with the recombinant 29 kDa processed form. Modification with an active site titrant suggested that most of each recombinant protease preparation was active (66 and 86%, respectively). Protease activity was compared using a low-molecular weight peptide substrate and the native substrate, capsid assembly protein. In addition, a cell-based assay for both enzymes was developed in which the target sequence of the protease has been fused inframe into the herpes simplex virus VP16 molecule. Cleavage of the fusion protein by the protease releases the carboxyl terminal transactivation domain, resulting in a decrease in the ability of the fusion molecule to transactivate a target promoter linked to a reporter gene in mammalian cells. Results suggest that the 75 kDa form of the enzyme is significantly less active than the 29 kDa form by all criteria.


Cytomegalovirus/enzymology , Endopeptidases/metabolism , Serine Endopeptidases/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Capsid , Chlorocebus aethiops , Cytomegalovirus/genetics , Endopeptidases/biosynthesis , Endopeptidases/genetics , Endopeptidases/isolation & purification , Escherichia coli , Herpes Simplex Virus Protein Vmw65/genetics , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Molecular Sequence Data , Molecular Weight , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/genetics , Substrate Specificity , Transcriptional Activation , Vero Cells , Viral Proteins/biosynthesis , Viral Proteins/isolation & purification , beta-Galactosidase/metabolism
16.
Biochim Biophys Acta ; 1598(1-2): 88-97, 2002 Jul 29.
Article En | MEDLINE | ID: mdl-12147348

The kinetic mechanism of mitogen-activated protein kinase activated protein kinase-2 (MAPKAPK2) was investigated using a peptide (LKRSLSEM) based on the phosphorylation site found in serum response factor (SRF). Initial velocity studies yielded a family of double-reciprocal lines that appear parallel and indicative of a ping-pong mechanism. The use of dead-end inhibition studies did not provide a definitive assignment of a reaction mechanism. However, product inhibition studies suggested that MAPKAPK2 follows an ordered bi-bi kinetic mechanism, where ATP must bind to the enzyme prior to the SRF-peptide and the phosphorylated product is released first, followed by ADP. In agreement with these latter results, surface plasmon resonance measurements demonstrate that the binding of the inhibitor peptide to MAPKAPK2 requires the presence of ATP. Furthermore, competitive inhibitors of ATP, adenosine 5'-(beta,gamma-imino)triphosphate (AMPPNP) and a staurosporine analog (K252a), can inhibit this ATP-dependent binding providing further evidence that the peptide substrate binds preferably to the E:ATP complex.


Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins , Kinetics , Peptide Fragments/chemistry , Peptide Mapping , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/isolation & purification , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity
...