Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
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
2.
J Med Chem ; 61(23): 10665-10699, 2018 12 13.
Article En | MEDLINE | ID: mdl-30423248

Ongoing interest in the discovery of selective JAK3 inhibitors led us to design novel covalent inhibitors that engage the JAK3 residue Cys909 by cyanamide, a structurally and mechanistically differentiated electrophile from other cysteine reacting groups previously incorporated in JAK3 covalent inhibitors. Through crystallography, kinetic, and computational studies, interaction of cyanamide 12 with Cys909 was optimized leading to potent and selective JAK3 inhibitors as exemplified by 32. In relevant cell-based assays and in agreement with previous results from this group, 32 demonstrated that selective inhibition of JAK3 is sufficient to drive JAK1/JAK3-mediated cellular responses. The contribution from extrahepatic processes to the clearance of cyanamide-based covalent inhibitors was also characterized using metabolic and pharmacokinetic data for 12. This work also gave key insights into a productive approach to decrease glutathione/glutathione S-transferase-mediated clearance, a challenge typically encountered during the discovery of covalent kinase inhibitors.


Cyanamide/chemistry , Cyanamide/pharmacology , Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Cyanamide/pharmacokinetics , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Janus Kinase 3/chemistry , Male , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/pharmacokinetics , Rats , Tissue Distribution
3.
J Med Chem ; 60(5): 1971-1993, 2017 03 09.
Article En | MEDLINE | ID: mdl-28139931

Significant work has been dedicated to the discovery of JAK kinase inhibitors resulting in several compounds entering clinical development and two FDA approved NMEs. However, despite significant effort during the past 2 decades, identification of highly selective JAK3 inhibitors has eluded the scientific community. A significant effort within our research organization has resulted in the identification of the first orally active JAK3 specific inhibitor, which achieves JAK isoform specificity through covalent interaction with a unique JAK3 residue Cys-909. The relatively rapid resynthesis rate of the JAK3 enzyme presented a unique challenge in the design of covalent inhibitors with appropriate pharmacodynamics properties coupled with limited unwanted off-target reactivity. This effort resulted in the identification of 11 (PF-06651600), a potent and low clearance compound with demonstrated in vivo efficacy. The favorable efficacy and safety profile of this JAK3-specific inhibitor 11 led to its evaluation in several human clinical studies.


Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry , Pyrroles/chemistry , Signal Transduction/drug effects , Administration, Oral , Drug Design , Humans , Janus Kinase 3/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology
4.
ACS Chem Biol ; 11(12): 3442-3451, 2016 12 16.
Article En | MEDLINE | ID: mdl-27791347

PF-06651600, a newly discovered potent JAK3-selective inhibitor, is highly efficacious at inhibiting γc cytokine signaling, which is dependent on both JAK1 and JAK3. PF-06651600 allowed the comparison of JAK3-selective inhibition to pan-JAK or JAK1-selective inhibition, in relevant immune cells to a level that could not be achieved previously without such potency and selectivity. In vitro, PF-06651600 inhibits Th1 and Th17 cell differentiation and function, and in vivo it reduces disease pathology in rat adjuvant-induced arthritis as well as in mouse experimental autoimmune encephalomyelitis models. Importantly, by sparing JAK1 function, PF-06651600 selectively targets γc cytokine pathways while preserving JAK1-dependent anti-inflammatory signaling such as the IL-10 suppressive functions following LPS treatment in macrophages and the suppression of TNFα and IL-1ß production in IL-27-primed macrophages. Thus, JAK3-selective inhibition differentiates from pan-JAK or JAK1 inhibition in various immune cellular responses, which could potentially translate to advantageous clinical outcomes in inflammatory and autoimmune diseases.


Arthritis, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Janus Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Animals , Arthritis, Experimental/immunology , Disease Models, Animal , Drug Discovery , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Interleukin-10/immunology , Interleukin-1beta/immunology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 3/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Models, Molecular , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Rats , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/drug effects , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/immunology
5.
Sci Rep ; 6: 30859, 2016 08 16.
Article En | MEDLINE | ID: mdl-27527709

Interleukin-17A (IL-17A) is a principal driver of multiple inflammatory and immune disorders. Antibodies that neutralize IL-17A or its receptor (IL-17RA) deliver efficacy in autoimmune diseases, but no small-molecule IL-17A antagonists have yet progressed into clinical trials. Investigation of a series of linear peptide ligands to IL-17A and characterization of their binding site has enabled the design of novel macrocyclic ligands that are themselves potent IL-17A antagonists.


Interleukin-17/antagonists & inhibitors , Interleukin-17/chemistry , Peptides, Cyclic/pharmacology , Small Molecule Libraries/pharmacology , Algorithms , Binding Sites , Cells, Cultured , Drug Design , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/metabolism , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Molecular Dynamics Simulation , Peptides, Cyclic/chemistry , Protein Binding , Small Molecule Libraries/chemistry , Structure-Activity Relationship
6.
Biochemistry ; 50(45): 9865-75, 2011 Nov 15.
Article En | MEDLINE | ID: mdl-22010960

It has been inferred from structural and computational studies that the mechanism of DNA polymerases involves subtle but important discrete steps that occur between binding and recognition of the correct dNTP and chemical catalysis. These steps potentially include local conformational changes involving active site residues, reorganization of Mg(2+)-coordinating ligands, and proton transfer. Here we address this broad issue by conducting extensive transient state kinetic analyses of DNA polymerase ß (Pol ß). We also performed kinetic simulations to evaluate alternative kinetic models. These studies provide some support for two-step subdomain closing and define constraints under which a kinetically significant prechemistry step can occur. To experimentally identify additional microscopic steps, we developed a stopped flow absorbance assay to measure proton formation that occurs during catalysis. These studies provide direct evidence that formation of the enzyme-bound 3'-O(-) nucleophile is rate determining for chemistry. We additionally show that at low pH the chemical step is rate limiting for catalysis, but at high pH, a postchemistry conformational step is rate limiting due to a pH-dependent increase in the rate of nucleotidyl transfer. Finally, we performed exhaustive analyses of [Mg(2+)] and pH effects. In contrast to published studies, the results suggest an irregular pH dependence of k(pol), which is consistent with general base catalysis involving cooperativity between two or more protonic residues. Overall, the results represent significant advancement in the kinetic mechanism of Pol ß and also reconcile some computational and experimental findings.


DNA Polymerase beta/chemistry , DNA Polymerase beta/metabolism , Animals , Base Sequence , Catalytic Domain , DNA/genetics , DNA/metabolism , Deoxyadenine Nucleotides/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Kinetics , Ligands , Magnesium/metabolism , Models, Molecular , Protein Conformation , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
7.
Biochem J ; 420(2): 229-38, 2009 May 13.
Article En | MEDLINE | ID: mdl-19281452

PAP (polyadenylate polymerase) is the template-independent RNA polymerase responsible for synthesis of the 3' poly(A) tails of mRNA. To investigate the role of proton transfer in the catalytic mechanism of PAP, the pH dependence of the steady-state kinetic parameters of yeast PAP were determined for the forward (adenyl transfer) and reverse (pyrophosphorolysis) reactions. The results indicate that productive formation of an enzyme-RNA-MgATP complex is pH independent over a broad pH range, but that formation of an active enzyme-RNA-MgPPi complex is strongly pH dependent, consistent with the production of a proton on the enzyme in the forward reaction. The pH dependence of the maximum velocity of the forward reaction suggests two protonic species are involved in enzyme catalysis. Optimal enzyme activity requires one species to be protonated and the other deprotonated. The deuterium solvent isotope effect on Vmax is also consistent with proton transfer involved in catalysis of a rate-determining step. Finally, pKa calculations of PAP were performed by the MCCE (multiconformational continuum electrostatic) method. Together, the data support that the protonation of residues Lys215 and Tyr224 exhibit co-operativity that is important for MgATP2- and MgPPi2- binding/dissociation, and suggest these residues function in electrostatic, but not in general acid, catalysis.


Fungal Proteins/metabolism , Polynucleotide Adenylyltransferase/metabolism , Protons , Yeasts/enzymology , Adenosine Triphosphate/metabolism , Catalytic Domain , Fungal Proteins/chemistry , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Polynucleotide Adenylyltransferase/chemistry , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , Substrate Specificity
8.
Biochemistry ; 47(26): 6859-69, 2008 Jul 01.
Article En | MEDLINE | ID: mdl-18537269

In yeast, the mRNA processing enzyme poly(A) polymerase is tethered to the much larger 3'-end processing complex via Fip1, a 36 kDa protein of unknown structure. We report the 2.6 A crystal structure of yeast poly(A) polymerase in complex with a peptide containing residues 80-105 of Fip1. The Fip1 peptide binds to the outside surface of the C-terminal domain of the polymerase. On the basis of this structure, we designed a mutant of the polymerase (V498Y, C485R) that is lethal to yeast. The mutant is unable to bind Fip1 but retains full polymerase activity. Fip1 is found in all eukaryotes and serves to connect poly(A) polymerase to pre-mRNA processing complexes in yeast, plants, and mammals. However, the Fip1 sequence is highly divergent, and residues on both Pap1 and Fip1 at the observed interaction surface are poorly conserved. Herein we demonstrate using analytical ultracentrifugation, circular dichroism, proteolytic studies, and other techniques that, in the absence of Pap1, Fip1 is largely, if not completely, unfolded. We speculate that flexibility may be important for Fip1's function as a molecular scaffold.


Peptides/chemistry , Peptides/metabolism , Polynucleotide Adenylyltransferase/chemistry , Polynucleotide Adenylyltransferase/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , mRNA Cleavage and Polyadenylation Factors/chemistry , mRNA Cleavage and Polyadenylation Factors/metabolism , Amino Acid Sequence , Biophysical Phenomena , Biophysics , Conserved Sequence , Crystallography, X-Ray , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Pancreatitis-Associated Proteins , Polynucleotide Adenylyltransferase/genetics , Protein Binding , Protein Structure, Quaternary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , mRNA Cleavage and Polyadenylation Factors/genetics
9.
Structure ; 15(9): 1117-31, 2007 Sep.
Article En | MEDLINE | ID: mdl-17850751

We report the 1.8 A structure of yeast poly(A) polymerase (PAP) trapped in complex with ATP and a five residue poly(A) by mutation of the catalytically required aspartic acid 154 to alanine. The enzyme has undergone significant domain movement and reveals a closed conformation with extensive interactions between the substrates and all three polymerase domains. Both substrates and 31 buried water molecules are enclosed within a central cavity that is open at both ends. Four PAP mutants were subjected to detailed kinetic analysis, and studies of the adenylyltransfer (forward), pyrophosphorolysis (reverse), and nucleotidyltransfer reaction utilizing CTP for the mutants are presented. The results support a model in which binding of both poly(A) and the correct nucleotide, MgATP, induces a conformational change, resulting in formation of a stable, closed enzyme state. Thermodynamic considerations of the data are discussed as they pertain to domain closure, substrate specificity, and catalytic strategies utilized by PAP.


Adenosine Triphosphate/metabolism , Polynucleotide Adenylyltransferase/metabolism , RNA/metabolism , Catalysis , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Polynucleotide Adenylyltransferase/chemistry , Polynucleotide Adenylyltransferase/genetics , Protein Conformation , RNA/chemistry
10.
J Mol Biol ; 366(5): 1401-15, 2007 Mar 09.
Article En | MEDLINE | ID: mdl-17223131

Polyadenylate polymerase (PAP) catalyzes the synthesis of poly(A) tails on the 3'-end of pre-mRNA. PAP is composed of three domains: an N-terminal nucleotide-binding domain (homologous to the palm domain of DNA and RNA polymerases), a middle domain (containing other conserved, catalytically important residues), and a unique C-terminal domain (involved in protein-protein interactions required for 3'-end formation). Previous X-ray crystallographic studies have shown that the domains are arranged in a V-shape such that they form a central cleft with the active site located at the base of the cleft at the interface between the N-terminal and middle domains. In the previous studies, the nucleotides were bound directly to the N-terminal domain and exhibited a conspicuous lack of adenine-specific interactions that would constitute nucleotide recognition. Furthermore, it was postulated that base-specific contacts with residues in the middle domain could occur either as a result of a change in the conformation of the nucleotide or domain movement. To address these issues and to better characterize the structural basis of substrate recognition and catalysis, we report two new crystal structures of yeast PAP. A comparison of these structures reveals that the N-terminal and C-terminal domains of PAP move independently as rigid bodies along two well defined axes of rotation. Modeling of the nucleotide into the most closed state allows us to deduce specific nucleotide interactions involving residues in the middle domain (K215, Y224 and N226) that are proposed to be involved in substrate binding and specificity. To further investigate the nature of PAP domain flexibility, 2-aminopurine labeled molecular probes were employed in steady state fluorescence and acrylamide quenching experiments. The results suggest that the closed domain conformation is stabilized upon recognition of the correct subtrate, MgATP, in an enzyme-substrate ternary complex. The implications of these results on the enzyme mechanism of PAP and the possible role for domain motion in an induced fit mechanism are discussed.


Crystallography, X-Ray , Polynucleotide Adenylyltransferase/chemistry , Saccharomyces cerevisiae/enzymology , Spectrometry, Fluorescence , Binding Sites , Models, Molecular , Nucleotides/chemistry , Nucleotides/metabolism , Polynucleotide Adenylyltransferase/genetics , Polynucleotide Adenylyltransferase/metabolism , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Rotation , Spectrum Analysis, Raman , Substrate Specificity
11.
Biochemistry ; 44(21): 7777-86, 2005 May 31.
Article En | MEDLINE | ID: mdl-15909992

Polyadenylate polymerase (PAP) catalyzes the synthesis of 3'-polyadenylate tails onto mRNA. A comprehensive steady-state kinetic analysis of PAP was conducted which included initial velocity studies of the forward and reverse reactions, inhibition studies, and the use of alternative substrates. The reaction (A(n) + ATP <--> A(n+1) + PP(i)) is adequately described by a rapid equilibrium random mechanism. Several thermodynamic parameters for the reaction were determined or calculated, including the overall equilibrium constant (K(eq) = 84) and the apparent equilibrium constant of the internal step (K(int) = 4) which involves the rate-determining interconversion of central complexes. A large (100-fold) difference in Vmax accounts for nucleotide specificity (ATP vs CTP), despite an only 3-fold difference in Km. Comparison of the sulfur elemental effect on Vmax for ATP and CTP suggests that the chemical step is rate-determining for both reactions. Comparison of the sulfur elemental effect on Vmax/Km revealed differences in the mechanism by which either nucleotide is incorporated. Consistent with these data, an induced fit mechanism for nucleotide specificity is proposed whereby PAP couples a uniform binding mechanism, which selects for ATP, with a ground-state destabilization mechanism, which serves to accelerate the velocity for the correct substrate.


Polynucleotide Adenylyltransferase/chemistry , Polynucleotide Adenylyltransferase/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding, Competitive , Cytidine Triphosphate/chemistry , Diphosphates/chemistry , Diphosphates/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Stability , Kinetics , Magnesium Compounds/chemistry , Magnesium Compounds/metabolism , Models, Chemical , Poly A/chemistry , Poly A/metabolism , Polyadenylation , Polynucleotide Adenylyltransferase/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Substrate Specificity , Sulfur/chemistry , Thermodynamics
12.
Biochemistry ; 42(51): 15189-96, 2003 Dec 30.
Article En | MEDLINE | ID: mdl-14690429

The PLP-dependent, biosynthetic arginine decarboxylase (ADC) of Yersinia pestis was investigated using steady-state kinetics employing structural analogues of arginine as both alternative substrates and competitive inhibitors. The inhibitor analysis indicates that binding of the carboxyl and guanidinium groups of the substrate, l-arginine, provides essentially all of the free energy change realized upon substrate binding in the ground state. Furthermore, recognition of the guanidinium group is primarily responsible for substrate specificity. Comparison of the steady-state parameters for a series of alternative substrates that contained chemically modified guanidinium moieties provides evidence of a role for induced fit in ADC catalysis. ADC was also characterized by UV/vis and fluorescence spectrophotometry in the presence or absence of a number of arginine analogues. The enzyme complexes formed served as models for the adsorption complex and the external aldimine complex of the enzyme with the substrate.


Arginine/analogs & derivatives , Arginine/chemistry , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/biosynthesis , Enzyme Inhibitors/chemistry , Yersinia pestis/enzymology , Amino Acid Sequence , Canavanine/chemistry , Carboxy-Lyases/chemistry , Catalysis , Kinetics , Molecular Sequence Data , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Substrate Specificity , omega-N-Methylarginine/chemistry
13.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 4): 721-3, 2003 Apr.
Article En | MEDLINE | ID: mdl-12657792

The alpha(2) epsilon (2) subcomponent (218.6 kDa) of the 1.99 MDa acetyl-CoA decarbonylase/synthase (ACDS) multienzyme complex is an Ni/Fe-S enzyme that catalyzes reversible CO(2)/CO reduction in the context of acetyl-CoA synthesis. The ACDS complex is required for methanogenesis from acetate in methanogenic archaea. The alpha(2)epsilon(2) subcomponent from Methanosarcina thermophila, grown on acetate, was purified and crystallized. The crystals were mounted in a capillary and diffracted to 4.0 A resolution at room temperature. Different flash-cooling approaches were attempted, all of which resulted in poor diffraction.


Aldehyde Oxidoreductases/chemistry , Methanosarcina/enzymology , Multienzyme Complexes/chemistry , Acetyl Coenzyme A/biosynthesis , Crystallization , Crystallography, X-Ray , Protein Subunits/chemistry , X-Ray Diffraction
14.
Microbiology (Reading) ; 145 ( Pt 5): 1181-1190, 1999 May.
Article En | MEDLINE | ID: mdl-10376834

A siderophore-dependent iron transport system of the pathogenic yersiniae plays a role in the pathogenesis of these organisms. The structure of the yersiniabactin (Ybt) siderophore produced by Yersinia enterocolitica has been elucidated. This paper reports the purification of Ybt from Yersinia pestis and demonstrates that it has the same structure as Ybt from Y. enterocolitica. Purified Ybt had a formation constant for Fe3+ of approximately 4x10(-36). Addition of purified Ybt from Y. pestis enhanced iron uptake by a siderophore-negative (irp2) strain of Y. pestis. Maximal expression of the Ybt outer-membrane receptor, Psn, in this strain was dependent upon exogenously supplied Ybt. Regulation of Psn expression by Ybt occurred at the transcriptional level. Y. pestis DNA was used to construct irp2 and psn mutations in Yersinia pseudotuberculosis. The irp2 mutant strain no longer synthesized Ybt and the psn mutant strain could not use exogenously supplied Ybt. As in Y. pestis, Ybt was required for maximal expression of Psn. Regulation by Ybt occurred at the transcriptional level. In contrast to Y. pestis, in which a psn mutation does not repress synthesis of Ybt siderophore or expression of the iron-regulated HMWP1 and HMWP2 proteins, the same mutation in Y. pseudotuberculosis partially repressed these products.


Phenols , Siderophores/chemistry , Siderophores/isolation & purification , Thiazoles , Yersinia pestis/metabolism , Bacterial Outer Membrane Proteins , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Bacterial , Iron/metabolism , Iron-Binding Proteins , Mutation , Periplasmic Binding Proteins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Siderophores/metabolism , Transcription, Genetic , Yersinia enterocolitica/genetics , Yersinia enterocolitica/growth & development , Yersinia enterocolitica/metabolism , Yersinia pestis/chemistry , Yersinia pestis/genetics , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/growth & development , Yersinia pseudotuberculosis/metabolism
...