Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL