Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Bioorg Med Chem ; 24(4): 596-605, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26740153

ABSTRACT

The de novo purine biosynthesis pathway is an attractive target for antibacterial drug design, and PurE from this pathway has been identified to be crucial for Bacillus anthracis survival in serum. In this study we adopted a fragment-based hit discovery approach, using three screening methods-saturation transfer difference nucleus magnetic resonance (STD-NMR), water-ligand observed via gradient spectroscopy (WaterLOGSY) NMR, and surface plasmon resonance (SPR), against B. anthracis PurE (BaPurE) to identify active site binding fragments by initially testing 352 compounds in a Zenobia fragment library. Competition STD NMR with the BaPurE product effectively eliminated non-active site binding hits from the primary hits, selecting active site binders only. Binding affinities (dissociation constant, KD) of these compounds varied between 234 and 301µM. Based on test results from the Zenobia compounds, we subsequently developed and applied a streamlined fragment screening strategy to screen a much larger library consisting of 3000 computationally pre-selected fragments. Thirteen final fragment hits were confirmed to exhibit binding affinities varying from 14µM to 700µM, which were categorized into five different basic scaffolds. All thirteen fragment hits have ligand efficiencies higher than 0.30. We demonstrated that at least two fragments from two different scaffolds exhibit inhibitory activity against the BaPurE enzyme.


Subject(s)
Bacillus anthracis/enzymology , Enzyme Inhibitors/pharmacology , Intramolecular Transferases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Catalytic Domain/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Intramolecular Transferases/isolation & purification , Intramolecular Transferases/metabolism , Magnetic Resonance Spectroscopy , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance
2.
ACS Chem Biol ; 10(6): 1456-65, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25746232

ABSTRACT

The Middle East Respiratory Syndrome coronavirus (MERS-CoV) papain-like protease (PLpro) blocking loop 2 (BL2) structure differs significantly from that of SARS-CoV PLpro, where it has been proven to play a crucial role in SARS-CoV PLpro inhibitor binding. Four SARS-CoV PLpro lead inhibitors were tested against MERS-CoV PLpro, none of which were effective against MERS-CoV PLpro. Structure and sequence alignments revealed that two residues, Y269 and Q270, responsible for inhibitor binding to SARS-CoV PLpro, were replaced by T274 and A275 in MERS-CoV PLpro, making critical binding interactions difficult to form for similar types of inhibitors. High-throughput screening (HTS) of 25 000 compounds against both PLpro enzymes identified a small fragment-like noncovalent dual inhibitor. Mode of inhibition studies by enzyme kinetics and competition surface plasmon resonance (SPR) analyses suggested that this compound acts as a competitive inhibitor with an IC50 of 6 µM against MERS-CoV PLpro, indicating that it binds to the active site, whereas it acts as an allosteric inhibitor against SARS-CoV PLpro with an IC50 of 11 µM. These results raised the possibility that inhibitor recognition specificity of MERS-CoV PLpro may differ from that of SARS-CoV PLpro. In addition, inhibitory activity of this compound was selective for SARS-CoV and MERS-CoV PLpro enzymes over two human homologues, the ubiquitin C-terminal hydrolases 1 and 3 (hUCH-L1 and hUCH-L3).


Subject(s)
Antiviral Agents/chemistry , Middle East Respiratory Syndrome Coronavirus/chemistry , Protease Inhibitors/chemistry , Severe acute respiratory syndrome-related coronavirus/chemistry , Viral Proteins/antagonists & inhibitors , Allosteric Regulation , Amino Acid Sequence , Catalytic Domain , Coronavirus 3C Proteases , Cysteine Endopeptidases/chemistry , Endopeptidases/chemistry , High-Throughput Screening Assays , Humans , Kinetics , Middle East Respiratory Syndrome Coronavirus/enzymology , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Severe acute respiratory syndrome-related coronavirus/enzymology , Species Specificity , Surface Plasmon Resonance , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/chemistry , Viral Proteins/chemistry
3.
Bioorg Med Chem ; 22(1): 167-77, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24332657

ABSTRACT

We have used a combination of virtual screening (VS) and high-throughput screening (HTS) techniques to identify novel, non-peptidic small molecule inhibitors against human SARS-CoV 3CLpro. A structure-based VS approach integrating docking and pharmacophore based methods was employed to computationally screen 621,000 compounds from the ZINC library. The screening protocol was validated using known 3CLpro inhibitors and was optimized for speed, improved selectivity, and for accommodating receptor flexibility. Subsequently, a fluorescence-based enzymatic HTS assay was developed and optimized to experimentally screen approximately 41,000 compounds from four structurally diverse libraries chosen mainly based on the VS results. False positives from initial HTS hits were eliminated by a secondary orthogonal binding analysis using surface plasmon resonance (SPR). The campaign identified a reversible small molecule inhibitor exhibiting mixed-type inhibition with a K(i) value of 11.1 µM. Together, these results validate our protocols as suitable approaches to screen virtual and chemical libraries, and the newly identified compound reported in our study represents a promising structural scaffold to pursue for further SARS-CoV 3CLpro inhibitor development.


Subject(s)
Severe acute respiratory syndrome-related coronavirus/enzymology , High-Throughput Screening Assays , Humans , Models, Molecular , Molecular Structure , Severe acute respiratory syndrome-related coronavirus/metabolism , Tissue Engineering , Tissue Scaffolds
4.
PLoS One ; 8(10): e75144, 2013.
Article in English | MEDLINE | ID: mdl-24130685

ABSTRACT

Development of drug-resistant mutations has been a major problem with all currently developed Hepatitis C Virus (HCV) NS3/4A inhibitors, including the two FDA approved drugs, significantly reducing the efficacy of these inhibitors. The high incidence of drug-resistance mutations and the limited utility of these inhibitors against only genotype 1 highlight the need for novel, broad-spectrum HCV therapies. Here we used high-throughput screening (HTS) to identify low molecular weight inhibitors against NS3/4A from multiple genotypes. A total of 40,967 compounds from four structurally diverse molecular libraries were screened by HTS using fluorescence-based enzymatic assays, followed by an orthogonal binding analysis using surface plasmon resonance (SPR) to eliminate false positives. A novel small molecule compound was identified with an IC50 value of 2.2 µM against the NS3/4A from genotype 1b. Mode of inhibition analysis subsequently confirmed this compound to be a competitive inhibitor with respect to the substrate, indicating direct binding to the protease active site, rather than to the allosteric binding pocket that was discovered to be the binding site of a few recently discovered small molecule inhibitors. This newly discovered inhibitor also showed promising inhibitory activity against the NS3/4As from three other HCV genotypes, as well as five common drug-resistant mutants of genotype 1b NS3/4A. The inhibitor was selective for NS3 from multiple HCV genotypes over two human serine proteases, and a whole cell lysate assay confirmed inhibitory activity in the cellular environment. This compound provides a lead for further development of potentially broader spectrum inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/enzymology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acid Sequence , Drug Evaluation, Preclinical/methods , Hepacivirus/drug effects , Molecular Sequence Data , Sequence Homology, Amino Acid
5.
ChemMedChem ; 8(8): 1361-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23788528

ABSTRACT

We previously developed two potent chemical classes that inhibit the essential papain-like protease (PLpro) of severe acute respiratory syndrome coronavirus. In this study, we applied a novel approach to identify small fragments that act synergistically with these inhibitors. A fragment library was screened in combination with four previously developed lead inhibitors by fluorescence-based enzymatic assays. Several fragment compounds synergistically enhanced the inhibitory activity of the lead inhibitors by approximately an order of magnitude. Surface plasmon resonance measurements showed that three fragments bind specifically to the PLpro enzyme. Mode of inhibition, computational solvent mapping, and molecular docking studies suggest that these fragments bind adjacent to the binding site of the lead inhibitors and further stabilize the inhibitor-bound state. We propose potential next-generation compounds based on a computational fragment-merging approach. This approach provides an alternative strategy for lead optimization for cases in which direct co-crystallization is difficult.


Subject(s)
Antiviral Agents/chemistry , Drug Design , Protease Inhibitors/chemistry , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/antagonists & inhibitors , Antiviral Agents/metabolism , Binding Sites , Coronavirus 3C Proteases , Cysteine Endopeptidases/metabolism , Drug Synergism , Humans , Kinetics , Molecular Docking Simulation , Protease Inhibitors/metabolism , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Surface Plasmon Resonance , Viral Proteins/metabolism
6.
J Chem Inf Model ; 53(3): 560-72, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23432621

ABSTRACT

We have developed a rigorous computational screening protocol to identify novel fragment-like inhibitors of N(5)-CAIR mutase (PurE), a key enzyme involved in de novo purine synthesis that represents a novel target for the design of antibacterial agents. This computational screening protocol utilizes molecular docking, graphics processing unit (GPU)-accelerated molecular dynamics, and Molecular Mechanics/Poisson-Boltzmann Surface Area (MM/PBSA) free energy estimations to investigate the binding modes and energies of fragments in the active sites of PurE. PurE is a functional octamer comprised of identical subunits. The octameric structure, with its eight active sites, provided a distinct advantage in these studies because, for a given simulation length, we were able to place eight separate fragment compounds in the active sites to increase the throughput of the MM/PBSA analysis. To validate this protocol, we have screened an in-house fragment library consisting of 352 compounds. The theoretical results were then compared with the results of two experimental fragment screens, Nuclear Magnetic Resonance (NMR) and Surface Plasmon Resonance (SPR) binding analyses. In these validation studies, the protocol was able to effectively identify the competitive binders that had been independently identified by experimental testing, suggesting the potential utility of this method for the identification of novel fragments for future development as PurE inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Discovery/methods , Drug Resistance, Multiple, Bacterial/genetics , High-Throughput Screening Assays/methods , Algorithms , Anti-Bacterial Agents/chemistry , Binding, Competitive , Computational Biology , Crystallography, X-Ray , Escherichia coli/drug effects , Escherichia coli/genetics , Intramolecular Transferases/drug effects , Intramolecular Transferases/genetics , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Library , Protein Binding , ROC Curve , Small Molecule Libraries , User-Computer Interface
7.
Protein Expr Purif ; 88(1): 98-106, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23246866

ABSTRACT

Staphylococcus aureus is a pathogenic bacterium that causes a variety of mild to lethal human diseases. The rapid spread of multidrug-resistant strains makes the discovery of new antimicrobial agents critical. Dihydroorotase (PyrC), the third enzyme in the bacterial pyrimidine biosynthesis pathway, is structurally and mechanistically distinct from its mammalian counterpart. It has been confirmed to be essential in S. aureus making it an attractive antibacterial drug target. No protocol to express and purify S. aureus PyrC (SaPyrC) has been reported. To obtain the SaPyrC enzyme and overcome anticipated solubility problems, the SaPyrC gene was cloned into the pET-SUMO vector. The N-terminal His-SUMO fused SaPyrC was expressed in Escherichia coli BL21 (DE3) with an HRV 3C protease recognition site inserted between the SUMO tag and SaPyrC to allow for improved cleavage by HRV protease. Purification of cleaved protein using HisTrap affinity and gel filtration columns resulted in native SaPyrC with estimated 95% purity and 40% yield. Both His-SUMO tagged and native SaPyrC form dimers, and enzyme characterization studies have shown that the His-SUMO tag affects enzyme activity slightly. Forward and reverse kinetic rate constants for both tagged and native SaPyrC were determined, and pH profiling studies revealed the optimal pH values for forward and reverse reactions.


Subject(s)
Dihydroorotase/genetics , Dihydroorotase/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Staphylococcus aureus/enzymology , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/metabolism , Cloning, Molecular , Dihydroorotase/biosynthesis , Drug Resistance, Multiple, Bacterial , Escherichia coli/genetics , Gene Expression , Genetic Vectors , Humans , Recombinant Fusion Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/metabolism , Staphylococcal Infections/enzymology , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology
8.
J Chem Inf Model ; 52(8): 2245-56, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22697413

ABSTRACT

Drug discovery and design for inhibition of the Hepatitis C Virus (HCV) NS3/4A serine protease is a major challenge. The broad, shallow, and generally featureless nature of the active site makes it a difficult target for "hit" selection especially using standard docking programs. There are several macrocyclic NS3/4A protease inhibitors that have been approved or are in clinical trials to treat chronic HCV (alone or as combination therapy), but most of the current therapies for HCV infection have untoward side effects, indicating a continuing medical need for the discovery of novel therapeutics with improved efficacy. In this study, we designed and implemented a two-tiered and progressive docking regime that successfully identified five non-macrocyclic small molecules that show inhibitory activity in the low micromolar range. Of these, four compounds show varying inhibition against HCV subgenotypes 1b, 1a, 2a, and 4d. The top inhibitor (3) has an IC(50) value of 15 µM against both subgenotypes 1b and 2a of the NS3/4A protease enzyme. Another inhibitor, 1, inhibits all four subgenotypes with moderate activity, showing highest activity for genotype 2a (24 µM). The five inhibitors presented in this study could be valuable candidates for future hit to lead optimization. Additionally, enzyme-inhibitor interaction models presented herein provide key information regarding structural differences between the active sites of the NS3/4A protease of the HCV subgenotype 1a and 1b that might explain the variable inhibitory activity between subgenotypes of the small molecule inhibitors identified here.


Subject(s)
Computational Biology/methods , Drug Evaluation, Preclinical/methods , Hepacivirus/enzymology , Protease Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Catalytic Domain , Inhibitory Concentration 50 , Molecular Docking Simulation , Protease Inhibitors/metabolism , Small Molecule Libraries/metabolism , User-Computer Interface , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...