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.
Int J Mol Sci ; 22(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34576147

ABSTRACT

Drug-resistant Staphylococcus aureus is an imminent threat to public health, increasing the importance of drug discovery utilizing unexplored bacterial pathways and enzyme targets. De novo pyrimidine biosynthesis is a specialized, highly conserved pathway implicated in both the survival and virulence of several clinically relevant pathogens. Class I dihydroorotase (DHOase) is a separate and distinct enzyme present in gram positive bacteria (i.e., S. aureus, B. anthracis) that converts carbamoyl-aspartate (Ca-asp) to dihydroorotate (DHO)-an integral step in the de novo pyrimidine biosynthesis pathway. This study sets forth a high-throughput screening (HTS) of 3000 fragment compounds by a colorimetry-based enzymatic assay as a primary screen, identifying small molecule inhibitors of S. aureus DHOase (SaDHOase), followed by hit validation with a direct binding analysis using surface plasmon resonance (SPR). Competition SPR studies of six hit compounds and eight additional analogs with the substrate Ca-asp determined the best compound to be a competitive inhibitor with a KD value of 11 µM, which is 10-fold tighter than Ca-asp. Preliminary structure-activity relationship (SAR) provides the foundation for further structure-based antimicrobial inhibitor design against S. aureus.


Subject(s)
Dihydroorotase/antagonists & inhibitors , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Staphylococcus aureus/enzymology , Catalytic Domain , Dihydroorotase/chemistry , Dihydroorotase/isolation & purification , Dihydroorotase/metabolism , Enzyme Inhibitors/chemistry , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Staphylococcus aureus/drug effects , Structure-Activity Relationship
2.
Bioorg Med Chem ; 27(10): 1981-1989, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30940566

ABSTRACT

The development of new therapeutic agents against the coronavirus causing Middle East Respiratory Syndrome (MERS) is a continuing imperative. The initial MERS-CoV epidemic was contained entirely through public health measures, but episodic cases continue, as there are currently no therapeutic agents effective in the treatment of MERS-CoV, although multiple strategies have been proposed. In this study, we screened 30,000 compounds from three different compound libraries against one of the essential proteases, the papain-like protease (PLpro), using a fluorescence-based enzymatic assay followed by surface plasmon resonance (SPR) direct binding analysis for hit confirmation. Mode of inhibition assays and competition SPR studies revealed two compounds to be competitive inhibitors. To improve upon the inhibitory activity of the best hit compounds, a small fragment library consisting of 352 fragments was screened in the presence of each hit compound, resulting in one fragment that enhanced the IC50 value of the best hit compound by 3-fold. Molecular docking and MM/PBSA binding energy calculations were used to predict potential binding sites, providing insight for design and synthesis of next-generation compounds.


Subject(s)
Drug Design , Middle East Respiratory Syndrome Coronavirus/enzymology , Peptide Hydrolases/chemistry , Protease Inhibitors/chemistry , Small Molecule Libraries/chemistry , Viral Proteins/antagonists & inhibitors , Binding Sites , Electron Spin Resonance Spectroscopy , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Peptide Hydrolases/metabolism , Protease Inhibitors/metabolism , Protein Structure, Tertiary , Small Molecule Libraries/metabolism , Structure-Activity Relationship , Viral Proteins/metabolism
3.
Antiviral Res ; 139: 49-58, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28034741

ABSTRACT

Zika flavivirus infection during pregnancy appears to produce higher risk of microcephaly, and also causes multiple neurological problems such as Guillain-Barré syndrome. The Zika virus is now widespread in Central and South America, and is anticipated to become an increasing risk in the southern United States. With continuing global travel and the spread of the mosquito vector, the exposure is expected to accelerate, but there are no currently approved treatments against the Zika virus. The Zika NS2B/NS3 protease is an attractive drug target due to its essential role in viral replication. Our studies have identified several compounds with inhibitory activity (IC50) and binding affinity (KD) of ∼5-10 µM against the Zika NS2B-NS3 protease from testing 71 HCV NS3/NS4A inhibitors that were initially discovered by high-throughput screening of 40,967 compounds. Competition surface plasmon resonance studies and mechanism of inhibition analyses by enzyme kinetics subsequently determined the best compound to be a competitive inhibitor with a Ki value of 9.5 µM. We also determined the X-ray structure of the Zika NS2B-NS3 protease in a "pre-open conformation", a conformation never observed before for any flavivirus proteases. This provides the foundation for new structure-based inhibitor design.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Drug Discovery , Serine Proteases/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus/drug effects , Inhibitory Concentration 50 , Kinetics , Protein Conformation , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Surface Plasmon Resonance , Virus Replication/drug effects , Zika Virus/enzymology
4.
Bioorg Med Chem ; 24(19): 4536-4543, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27499369

ABSTRACT

Dihydroorotase (DHOase) is the third enzyme in the de novo pyrimidine synthesis pathway and is responsible for the reversible cyclization of carbamyl-aspartate (Ca-asp) to dihydroorotate (DHO). DHOase is further divided into two classes based on several structural characteristics, one of which is the length of the flexible catalytic loop that interacts with the substrate, Ca-asp, regulating the enzyme activity. Here, we present the crystal structure of Class I Bacillus anthracis DHOase with Ca-asp in the active site, which shows the peptide backbone of glycine in the shorter loop forming the necessary hydrogen bonds with the substrate, in place of the two threonines found in Class II DHOases. Despite the differences in the catalytic loop, the structure confirms that the key interactions between the substrate and active site residues are similar between Class I and Class II DHOase enzymes, which we further validated by mutagenesis studies. B. anthracis DHOase is also a potential antibacterial drug target. In order to identify prospective inhibitors, we performed high-throughput screening against several libraries using a colorimetric enzymatic assay and an orthogonal fluorescence thermal binding assay. Surface plasmon resonance was used for determining binding affinity (KD) and competition analysis with Ca-asp. Our results highlight that the primary difference between Class I and Class II DHOase is the catalytic loop. We also identify several compounds that can potentially be further optimized as potential B. anthracis inhibitors.


Subject(s)
Bacillus anthracis/enzymology , Dihydroorotase/antagonists & inhibitors , Dihydroorotase/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Anthrax/drug therapy , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/chemistry , Bacillus anthracis/metabolism , Crystallography, X-Ray , Dihydroorotase/metabolism , Humans , Models, Molecular , Protein Conformation
5.
Protein Expr Purif ; 125: 83-9, 2016 09.
Article in English | MEDLINE | ID: mdl-26344899

ABSTRACT

Quorum sensing (QS) has been recognized as an important biological phenomenon in which bacterial cells communicate and coordinate their gene expression and cellular processes with respect to population density. Bacillus anthracis is the etiological agent of fatal pulmonary anthrax infections, and the NprR/NprX QS system may be involved in its pathogenesis. NprR, renamed as aqsR for anthrax quorum sensing Regulator, is a transcriptional regulator that may control the expression of genes required for proliferation and survival. Currently, there is no protocol reported to over-express and purify B. anthracis AqsR. In this study, we describe cloning, purification, and confirmation of functional full-length B. anthracis AqsR protein. The AqsR gene was cloned into the pQE-30 vector with an HRV 3C protease recognition site between AqsR and the N-terminal His6-tag in order to yield near native AqsR after the His-tag cleavage, leaving only two additional amino acid residues at the N-terminus.


Subject(s)
Bacillus anthracis/metabolism , Bacterial Proteins , Gene Expression Regulation, Bacterial , Transcription Factors , Bacillus anthracis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Cell Line , Gene Expression , Humans , Quorum Sensing , Surface Plasmon Resonance , Tandem Mass Spectrometry , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/isolation & purification , Transcription Factors/metabolism
6.
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
7.
Anal Biochem ; 441(1): 87-94, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23769705

ABSTRACT

Dihydroorotase (DHOase) is the third enzyme in the de novo pyrimidine biosynthesis pathway and is a potential new antibacterial drug target. No target-based high-throughput screening (HTS) assay for this enzyme has been reported to date. Here, we optimized two colorimetric-based enzymatic assays that detect the ureido moiety of the DHOase substrate, carbamyl-aspartate (Ca-asp). Each assay was developed in a 40-µl assay volume using 384-well plates with a different color mix, diacetylmonoxime (DAMO)-thiosemicarbazide (TSC) or DAMO-antipyrine. The sensitivity and color interference of both color mixes were compared in the presence of common HTS buffer additives, including dimethyl sulfoxide, reducing agents, detergents, and bovine serum albumin. DAMO-TSC (Z'-factors 0.7-0.8) was determined to be superior to DAMO-antipyrine (Z'-factors 0.5-0.6) with significantly less variability within replicates. An HTS pilot screening with 29,552 compounds from four structurally diverse libraries confirmed the quality of our newly optimized colorimetric assay with DAMO-TSC. This robust method has no heating requirement, which was the main obstacle to applying previous assays to HTS. More important, this well-optimized HTS assay for DHOase, the first of its kind, should make it possible to screen large-scale compound libraries to develop new inhibitors against any enzymes that produce ureido functional groups.


Subject(s)
Aspartic Acid/analogs & derivatives , Colorimetry/methods , Dihydroorotase/analysis , Dihydroorotase/metabolism , Enzyme Assays/methods , High-Throughput Screening Assays/methods , Aspartic Acid/analysis , Aspartic Acid/chemistry , Bacillus anthracis/enzymology
8.
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
SELECTION OF CITATIONS
SEARCH DETAIL