Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Chemistry ; 29(40): e202301621, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37285583

ABSTRACT

Invited for the cover of this issue is the group of Michael Ashley Spies at the University of Iowa. The image depicts how mapping allosteric structure-activity relationships reveals the nexus between the active site and the remote allosteric pocket. Read the full text of the article at 10.1002/chem.202300872.

2.
Chemistry ; 29(40): e202300872, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37005499

ABSTRACT

Caspase-7 (C7), a cysteine protease involved in apoptosis, is a valuable drug target for its role in human diseases (e. g., Parkinson's, Alzheimer's, sepsis). The C7 allosteric site has great potential for small-molecule targeting, but numerous drug discovery efforts have identified precious few allosteric inhibitors. Here we present the first selective, drug-like inhibitor of C7 along with several other improved inhibitors based on our previous fragment hit. We also provide a rational basis for the impact of allosteric binding on the C7 catalytic cycle by using an integrated approach including X-ray crystallography, stopped-flow kinetics, and molecular dynamics simulations. Our findings suggest allosteric binding disrupts C7 pre-acylation by neutralization of the catalytic dyad, displacement of substrate from the oxyanion hole, and altered dynamics of substrate binding loops. This work advances drug targeting efforts and bolsters our understanding of allosteric structure-activity relationships (ASARs).


Subject(s)
Molecular Dynamics Simulation , Humans , Caspase 7/metabolism , Allosteric Regulation , Protein Conformation , Allosteric Site , Crystallography, X-Ray
3.
ChemMedChem ; 15(4): 376-384, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31876113

ABSTRACT

Glutamate racemases (GR) are members of the family of bacterial enzymes known as cofactor-independent racemases and epimerases and catalyze the stereoinversion of glutamate. D-amino acids are universally important for the proper construction of viable bacterial cell walls, and thus have been repeatedly validated as attractive targets for novel antimicrobial drug design. Significant aspects of the mechanism of this challenging stereoinversion remain unknown. The current study employs a combination of MD and QM/MM computational approaches to show that the GR from H. pylori must proceed via a pre-activation step, which is dependent on the enzyme's flexibility. This mechanism is starkly different from previously proposed mechanisms. These findings have immediate pharmaceutical relevance, as the H. pylori GR enzyme is a very attractive allosteric drug target. The results presented in this study offer a distinctly novel understanding of how AstraZeneca's lead series of inhibitors cripple the H. pylori GR's native motions, via prevention of this critical chemical pre-activation step. Our experimental studies, using SPR, fluorescence and NMR WaterLOGSY, show that H. pylori GR is not inhibited by the uncompetitive mechanism originally put forward by Lundqvist et al.. The current study supports a deep connection between native enzyme motions and chemical reactivity, which has strong relevance to the field of allosteric drug discovery.


Subject(s)
Amino Acid Isomerases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Helicobacter pylori/drug effects , Molecular Dynamics Simulation , Allosteric Regulation/drug effects , Amino Acid Isomerases/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Helicobacter pylori/enzymology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship
4.
ChemMedChem ; 13(23): 2514-2521, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30264520

ABSTRACT

The application of covalent inhibitors has experienced a renaissance within drug discovery programs in the last decade. To leverage the superior potency and drug target residence time of covalent inhibitors, there have been extensive efforts to develop highly specific covalent modifications to decrease off-target liabilities. Herein, we present a series of covalent inhibitors of an antimicrobial drug target, glutamate racemase, discovered through structure-based virtual screening. A combination of enzyme kinetics, mass spectrometry, and surface-plasmon resonance experiments details a highly specific 1,4-conjugate addition of a small-molecule inhibitor with a catalytic cysteine of glutamate racemase. Molecular dynamics simulations and quantum mechanics-molecular mechanics geometry optimizations reveal the chemistry of the conjugate addition. Two compounds from this series of inhibitors display antimicrobial potency similar to ß-lactam antibiotics, with significant activity against methicillin-resistant S. aureus strains. This study elucidates a detailed chemical rationale for covalent inhibition and provides a platform for the development of antimicrobials with a novel mechanism of action against a target in the cell wall biosynthesis pathway.


Subject(s)
Amino Acid Isomerases/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Amino Acid Isomerases/metabolism , Bacillus subtilis/drug effects , Bacillus subtilis/enzymology , Bacteria/drug effects , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Drug Discovery , Humans , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
Angew Chem Int Ed Engl ; 56(46): 14443-14447, 2017 11 13.
Article in English | MEDLINE | ID: mdl-28940929

ABSTRACT

The caspase family of cysteine proteases are highly sought-after drug targets owing to their essential roles in apoptosis, proliferation, and inflammation pathways. High-throughput screening efforts to discover inhibitors have gained little traction. Fragment-based screening has emerged as a powerful approach for the discovery of innovative drug leads. This method has become a central facet of drug discovery campaigns in the pharmaceutical industry and academia. A fragment-based drug discovery campaign against human caspase-7 resulted in the discovery of a novel series of allosteric inhibitors. An X-ray crystal structure of caspase-7 bound to a fragment hit and a thorough kinetic characterization of a zymogenic form of the enzyme were used to investigate the allosteric mechanism of inhibition. This work further advances our understanding of the mechanisms of allosteric control of this class of pharmaceutically relevant enzymes, and provides a new path forward for drug discovery efforts.


Subject(s)
Caspase 7/metabolism , Drug Discovery/methods , Allosteric Regulation/drug effects , Apoptosis , Caspase 7/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Isotopes/chemistry , Kinetics , Solvents/chemistry , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...