Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(10): 105257, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37716702

ABSTRACT

RNA methylation is a ubiquitous post-transcriptional modification found in diverse RNA classes and is a critical regulator of gene expression. In this study, we used Zika virus RNA methyltransferase (MTase) to develop a highly sensitive microplate assay that uses a biotinylated RNA substrate and radiolabeled AdoMet coenzyme. The assay is fast, highly reproducible, exhibits linear progress-curve kinetics under multiple turnover conditions, has high sensitivity in competitive inhibition assays, and significantly lower background levels compared with the currently used method. Using our newly developed microplate assay, we observed no significant difference in the catalytic constants of the full-length nonstructural protein 5 enzyme and the truncated MTase domain. These data suggest that, unlike the Zika virus RNA-dependent RNA polymerase activity, the MTase activity is unaffected by RNA-dependent RNA polymerase-MTase interdomain interaction. Given its quantitative nature and accuracy, this method can be used to characterize various RNA MTases, and, therefore, significantly contribute to the field of epitranscriptomics and drug development against infectious diseases.


Subject(s)
Biological Assay , Methyltransferases , Drug Development , Methyltransferases/metabolism , RNA , RNA-Dependent RNA Polymerase/metabolism , Zika Virus/enzymology , Gene Expression Profiling , Epigenesis, Genetic , Biological Assay/methods , Biotinylation , Protein Structure, Tertiary
2.
Nature ; 558(7711): 610-614, 2018 06.
Article in English | MEDLINE | ID: mdl-29925952

ABSTRACT

Viral infections continue to represent major challenges to public health, and an enhanced mechanistic understanding of the processes that contribute to viral life cycles is necessary for the development of new therapeutic strategies 1 . Viperin, a member of the radical S-adenosyl-L-methionine (SAM) superfamily of enzymes, is an interferon-inducible protein implicated in the inhibition of replication of a broad range of RNA and DNA viruses, including dengue virus, West Nile virus, hepatitis C virus, influenza A virus, rabies virus 2 and HIV3,4. Viperin has been suggested to elicit these broad antiviral activities through interactions with a large number of functionally unrelated host and viral proteins3,4. Here we demonstrate that viperin catalyses the conversion of cytidine triphosphate (CTP) to 3'-deoxy-3',4'-didehydro-CTP (ddhCTP), a previously undescribed biologically relevant molecule, via a SAM-dependent radical mechanism. We show that mammalian cells expressing viperin and macrophages stimulated with IFNα produce substantial quantities of ddhCTP. We also establish that ddhCTP acts as a chain terminator for the RNA-dependent RNA polymerases from multiple members of the Flavivirus genus, and show that ddhCTP directly inhibits replication of Zika virus in vivo. These findings suggest a partially unifying mechanism for the broad antiviral effects of viperin that is based on the intrinsic enzymatic properties of the protein and involves the generation of a naturally occurring replication-chain terminator encoded by mammalian genomes.


Subject(s)
Antiviral Agents/metabolism , Cytidine Triphosphate/metabolism , Genome, Human/genetics , Proteins/genetics , Proteins/metabolism , Transcription Termination, Genetic , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Cytidine Triphosphate/biosynthesis , Cytidine Triphosphate/chemistry , HEK293 Cells , Humans , Oxidoreductases Acting on CH-CH Group Donors , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Ribonucleotides , Substrate Specificity , Vero Cells , Zika Virus/enzymology , Zika Virus/metabolism
3.
Arch Pharm (Weinheim) ; 357(9): e2400250, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38809037

ABSTRACT

Three new series of macrocyclic active site-directed inhibitors of the Zika virus (ZIKV) NS2B-NS3 protease were synthesized. First, attempts were made to replace the basic P3 lysine residue of our previously described inhibitors with uncharged and more hydrophobic residues. This provided numerous compounds with inhibition constants between 30 and 50 nM. A stronger reduction of the inhibitory potency was observed when the P2 lysine was replaced by neutral residues, all of these inhibitors possess Ki values >1 µM. However, it is possible to replace the P2 lysine with the less basic 3-aminomethylphenylalanine, which provides a similarly potent inhibitor of the ZIKV protease (Ki = 2.69 nM). Crystal structure investigations showed that the P2 benzylamine structure forms comparable interactions with the protease as lysine. Twelve additional structures of these inhibitors in complex with the protease were determined, which explain many, but not all, SAR data obtained in this study. All individual modifications in the P2 or P3 position resulted in inhibitors with low antiviral efficacy in cell culture. Therefore, a third inhibitor series with combined modifications was synthesized; all of them contain a more hydrophobic d-cyclohexylalanine in the linker segment. At a concentration of 40 µM, two of these compounds possess similar antiviral potency as ribavirin at 100 µM. Due to their reliable crystallization in complex with the ZIKV protease, these cyclic compounds are very well suited for a rational structure-based development of improved inhibitors.


Subject(s)
Antiviral Agents , Macrocyclic Compounds , Zika Virus , Zika Virus/enzymology , Zika Virus/drug effects , Structure-Activity Relationship , Macrocyclic Compounds/pharmacology , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Molecular Structure , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Dose-Response Relationship, Drug , Serine Endopeptidases/metabolism , Humans , Viral Protease Inhibitors/pharmacology , Viral Protease Inhibitors/chemical synthesis , Viral Protease Inhibitors/chemistry , Crystallography, X-Ray , Viral Proteases , Nucleoside-Triphosphatase , DEAD-box RNA Helicases
4.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673962

ABSTRACT

In the global pandemic scenario, dengue and zika viruses (DENV and ZIKV, respectively), both mosquito-borne members of the flaviviridae family, represent a serious health problem, and considering the absence of specific antiviral drugs and available vaccines, there is a dire need to identify new targets to treat these types of viral infections. Within this drug discovery process, the protease NS2B/NS3 is considered the primary target for the development of novel anti-flavivirus drugs. The NS2B/NS3 is a serine protease that has a dual function both in the viral replication process and in the elusion of the innate immunity. To date, two main classes of NS2B/NS3 of DENV and ZIKV protease inhibitors have been discovered: those that bind to the orthosteric site and those that act at the allosteric site. Therefore, this perspective article aims to discuss the main features of the use of the most potent NS2B/NS3 inhibitors and their impact at the social level.


Subject(s)
Antiviral Agents , Dengue , Protease Inhibitors , Zika Virus Infection , Animals , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , DEAD-box RNA Helicases , Dengue/drug therapy , Dengue/virology , Dengue Virus/drug effects , Nucleoside-Triphosphatase , Protease Inhibitors/therapeutic use , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Serine Endopeptidases/metabolism , Serine Endopeptidases/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , Viral Proteases , Zika Virus/drug effects , Zika Virus/enzymology , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
5.
J Cell Biochem ; 124(1): 127-145, 2023 01.
Article in English | MEDLINE | ID: mdl-36502494

ABSTRACT

Numerous pathogens affecting human is present in the flavivirus family namely west nile, dengue, yellow fever, and zika which involves in development of global burden and distressing the environment economically. Till date, no approved drugs are available for targeting these viruses. The threat which urged the identification of small molecules for the inhibition of these viruses is the spreading of serious viral diseases. The recent outbreak of zika and dengue infections postured a solemn risk to worldwide public well-being. RNA-dependent RNA polymerase (RdRp) is the supreme adaptable enzymes of all the RNA viruses which is responsible for the replication and transcription of genome among the structural and nonstructural proteins of flaviviruses. It is understood that the RdRp of the flaviviruses are similar stating that the japanese encephalitis and west nile shares 70% identity with zika whereas the dengue serotype 2 and 3 shares the identity of 76% and 81%, respectively. In this study, we investigated the binding site of four flaviviral RdRp and provided insights into various interaction of the molecules using the computational approach. Our study helps in recognizing the potent compounds that could inhibit the viral protein as a common inhibitor. Additionally, with the conformational stability analysis, we proposed the possible mechanism of inhibition of the identified common small molecule toward RdRp of flavivirus. Finally, this study could be an initiative for the identification of common inhibitors and can be explored further for understanding the mechanism of action through in vitro studies for the study on efficacy.


Subject(s)
Drug Repositioning , Flavivirus , RNA-Dependent RNA Polymerase , Humans , Dengue/drug therapy , Flavivirus/drug effects , Flavivirus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Zika Virus/drug effects , Zika Virus/enzymology , Zika Virus Infection/drug therapy
6.
J Med Virol ; 94(2): 442-453, 2022 02.
Article in English | MEDLINE | ID: mdl-34636434

ABSTRACT

Zika virus (ZIKV) infections are associated with severe neurological complications and are a global public health concern. There are no approved vaccines or antiviral drugs to inhibit ZIKV replication. NS2B-NS3 protease (NS2B-NS3 pro), which is essential for viral replication, is a promising molecular target for anti-ZIKV drugs. We conducted a systematic review to identify compounds with promising effects against ZIKV; we discussed their pharmacodynamic and pharmacophoric characteristics. The online search, performed using the PubMed/MEDLINE and SCOPUS databases, yielded 56 articles; seven relevant studies that reported nine promising compounds with inhibitory activity against ZIKV NS2B-NS3 pro were selected. Of these, five (niclosamide, nitazoxanide, bromocriptine, temoporfin, and novobiocin) are currently available on the market and have been tested for off-label use against ZIKV. The 50% inhibitory concentration values of these compounds for the inhibition of NS2B-NS3 pro ranged at 0.38-21.6 µM; most compounds exhibited noncompetitive inhibition (66%). All compounds that could inhibit the NS2B-NS3 pro complex showed potent in vitro anti-ZIKV activity with a 50% effective concentration ranging 0.024-50 µM. The 50% cytotoxic concentration of the compounds assayed using A549, Vero, and WRL-69 cell lines ranged at 0.6-1388.02 µM and the selectivity index was 3.07-1698. This review summarizes the most promising antiviral agents against ZIKV that have inhibitory activity against viral proteases.


Subject(s)
Antiviral Agents/pharmacology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Animals , Antiviral Agents/chemistry , Humans , Molecular Targeted Therapy , Protease Inhibitors/chemistry , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Zika Virus/enzymology , Zika Virus Infection/virology
7.
J Chem Inf Model ; 62(24): 6825-6843, 2022 12 26.
Article in English | MEDLINE | ID: mdl-36239304

ABSTRACT

The Zika virus (ZIKV) is a neurotropic arbovirus considered a global threat to public health. Although there have been several efforts in drug discovery projects for ZIKV in recent years, there are still no antiviral drugs approved to date. Here, we describe the results of a global collaborative crowdsourced open science project, the OpenZika project, from IBM's World Community Grid (WCG), which integrates different computational and experimental strategies for advancing a drug candidate for ZIKV. Initially, molecular docking protocols were developed to identify potential inhibitors of ZIKV NS5 RNA-dependent RNA polymerase (NS5 RdRp), NS3 protease (NS2B-NS3pro), and NS3 helicase (NS3hel). Then, a machine learning (ML) model was built to distinguish active vs inactive compounds for the cytoprotective effect against ZIKV infection. We performed three independent target-based virtual screening campaigns (NS5 RdRp, NS2B-NS3pro, and NS3hel), followed by predictions by the ML model and other filters, and prioritized a total of 61 compounds for further testing in enzymatic and phenotypic assays. This yielded five non-nucleoside compounds which showed inhibitory activity against ZIKV NS5 RdRp in enzymatic assays (IC50 range from 0.61 to 17 µM). Two compounds thermally destabilized NS3hel and showed binding affinity in the micromolar range (Kd range from 9 to 35 µM). Moreover, the compounds LabMol-301 inhibited both NS5 RdRp and NS2B-NS3pro (IC50 of 0.8 and 7.4 µM, respectively) and LabMol-212 thermally destabilized the ZIKV NS3hel (Kd of 35 µM). Both also protected cells from death induced by ZIKV infection in in vitro cell-based assays. However, while eight compounds (including LabMol-301 and LabMol-212) showed a cytoprotective effect and prevented ZIKV-induced cell death, agreeing with our ML model for prediction of this cytoprotective effect, no compound showed a direct antiviral effect against ZIKV. Thus, the new scaffolds discovered here are promising hits for future structural optimization and for advancing the discovery of further drug candidates for ZIKV. Furthermore, this work has demonstrated the importance of the integration of computational and experimental approaches, as well as the potential of large-scale collaborative networks to advance drug discovery projects for neglected diseases and emerging viruses, despite the lack of available direct antiviral activity and cytoprotective effect data, that reflects on the assertiveness of the computational predictions. The importance of these efforts rests with the need to be prepared for future viral epidemic and pandemic outbreaks.


Subject(s)
Antiviral Agents , Protease Inhibitors , Zika Virus , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Molecular Docking Simulation , Peptide Hydrolases , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/chemistry , Zika Virus/drug effects , Zika Virus/enzymology , Zika Virus Infection/drug therapy
8.
Bioorg Med Chem ; 57: 116631, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35123179

ABSTRACT

Zika virus (ZIKV) is a member of the Flaviviridae family that can cause neurological disorders and congenital malformations. The NS2B-NS3 viral serine protease is an attractive target for the development of new antiviral agents against ZIKV. We report here a SAR study on a series of substrate-like linear tripeptides that inhibit in a non-covalent manner the NS2B-NS3 protease. Optimization of the residues at positions P1, P2, P3 and of the N-terminal and C-terminal portions of the tripeptide allowed the identification of inhibitors with sub-micromolar potency with phenylglycine as arginine-mimicking group and benzylamide as C-terminal fragment. Further SAR exploration and application of these structural changes to a series of peptides having a 4-substituted phenylglycine residue at the P1 position led to potent compounds showing double digit nanomolar inhibition of the Zika protease (IC50 = 30 nM) with high selectivity against trypsin-like proteases and the proteases of other flavivirus, such as Dengue 2 virus (DEN2V) and West Nile virus (WNV).


Subject(s)
Antiviral Agents/pharmacology , Peptides/pharmacology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dengue Virus/drug effects , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , RNA Helicases/antagonists & inhibitors , RNA Helicases/metabolism , Serine Endopeptidases/metabolism , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism , West Nile virus/drug effects , Zika Virus/enzymology
9.
Nucleic Acids Res ; 48(9): 5081-5093, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32313955

ABSTRACT

Flaviviruses, including dengue virus and Zika virus, contain a single-stranded positive sense RNA genome that encodes viral proteins essential for replication and also serves as the template for new genome synthesis. As these processes move in opposite directions along the genome, translation must be inhibited at a defined point following infection to clear the template of ribosomes to allow efficient replication. Here, we demonstrate in vitro and in cell-based assays that the viral RNA polymerase, NS5, inhibits translation of the viral genome. By reconstituting translation in vitro using highly purified components, we show that this translation block occurs at the initiation stage and that translation inhibition depends on NS5-RNA interaction, primarily through association with the 5' replication promoter region. This work supports a model whereby expression of a viral protein signals successful translation of the infecting genome, prompting a switch to a ribosome depleted replication-competent form.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Genome, Viral , Protein Biosynthesis , RNA, Viral/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Chlorocebus aethiops , Dengue Virus/enzymology , Peptide Chain Initiation, Translational , RNA, Viral/chemistry , Vero Cells , Virus Replication , Zika Virus/enzymology , Zika Virus/physiology
10.
Angew Chem Int Ed Engl ; 61(4): e202113857, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34825756

ABSTRACT

Constrained peptides are promising next-generation therapeutics. We report here a fundamentally new strategy for the facile generation of bicyclic peptides using linear precursor peptides with three cysteine residues and a non-toxic trivalent bismuth(III) salt. Peptide-bismuth bicycles form instantaneously at physiological pH, are stable in aqueous solution for many weeks, and much more resistant to proteolysis than their linear precursors. The strategy allows the in situ generation of bicyclic ligands for biochemical screening assays. We demonstrate this for two screening campaigns targeting the proteases from Zika and West Nile viruses, revealing a new lead compound that displayed inhibition constants of 23 and 150 nM, respectively. Bicyclic peptides are up to 130 times more active and 19 times more proteolytically stable than their linear analogs without bismuth.


Subject(s)
Bismuth/pharmacology , Peptide Hydrolases/metabolism , Peptides, Cyclic/pharmacology , Protease Inhibitors/pharmacology , Bismuth/chemistry , Dose-Response Relationship, Drug , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protein Conformation , West Nile virus/enzymology , Zika Virus/enzymology
11.
Biochem Biophys Res Commun ; 534: 666-671, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33208231

ABSTRACT

The association of Zika virus (ZIKV) infection with a congenital malformation in fetuses, neurological, and other systemic complications in adults have brought significant global health emergency. ZIKV targets nerve cells in the brain and causes cell death, such as pyroptosis, leading to neuroinflammation. Here we described a novel mechanism of pyroptosis caused by ZIKV protease. We found that ZIKV protease directly cleaved the GSDMD into N-terminal fragment (1-249) leading to pyroptosis in a caspase-independent manner, suggesting a direct mechanism of ZIKV-induced cell death and subsequent inflammation. Our findings might shed new light to explore the pathogenesis of ZIKV infections where ZIKV protease might be a suitable target for the development of antiviral agents.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Peptide Hydrolases/metabolism , Phosphate-Binding Proteins/metabolism , Pyroptosis/physiology , Viral Proteins/metabolism , Zika Virus/enzymology , Zika Virus/pathogenicity , Binding Sites , Caspases/metabolism , Cell Line , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Biological , Neurons/metabolism , Neurons/pathology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphate-Binding Proteins/chemistry , Proteolysis , Substrate Specificity , Zika Virus Infection/etiology , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
12.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32796069

ABSTRACT

Zika virus (ZIKV) remains a potentially significant public health concern because it can cause teratogenic effects, such as microcephaly in newborns and neurological disease, like Guillain-Barré syndrome. Together with efforts to develop a vaccine, the discovery of antiviral molecules is important to control ZIKV infections and to prevent its most severe symptoms. Here, we report the development of small nonnucleoside inhibitors (NNIs) of ZIKV RNA-dependent RNA polymerase (RdRp) activity. These NNIs target an allosteric pocket (N pocket) located next to a putative hinge region between the thumb and the palm subdomains that was originally described for dengue virus (DENV) RdRp. We first tested the activity of DENV RdRp N-pocket inhibitors against ZIKV RdRp, introduced chemical modifications into these molecules, and assessed their potency using both enzymatic and cell-based assays. The most potent compound had a 50% inhibitory concentration value of 7.3 µM and inhibited ZIKV replication in a cell-based assay with a 50% effective concentration value of 24.3 µM. Importantly, we report four high-resolution crystal structures detailing how these NNIs insert into the N pocket of ZIKV RdRp. Our observations point to subtle differences in the size, shape, chemical environment, and hydration of the N pocket from ZIKV RdRp from those of the N pocket from DENV RdRp that are crucial for the design of improved antiviral inhibitors with activity against ZIKV.IMPORTANCE Zika virus belongs to the Flavivirus genus, which comprises several important human pathogens. There is currently neither an approved vaccine nor antiviral drugs available to prevent infection by ZIKV. The nonstructural protein 5 (NS5) polymerase, which is responsible for replicating the viral RNA genome, represents one of the most promising targets for antiviral drug development. Starting from compounds recently developed against dengue virus NS5, we designed and synthesized inhibitors targeting Zika virus NS5. We show that these novel compounds inhibit viral replication by targeting the polymerase activity. High-resolution X-ray crystallographic structures of protein-inhibitor complexes demonstrated specific binding to an allosteric site within the polymerase, called the N pocket. This work paves the way for the future structure-based design of potent compounds specifically targeting ZIKV RNA polymerase activity.


Subject(s)
Antiviral Agents/chemical synthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Sulfones/chemical synthesis , Thiophenes/chemical synthesis , Viral Proteins/antagonists & inhibitors , Allosteric Regulation , Allosteric Site/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cricetulus , Drug Design , Gene Expression , Hepatocytes , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity , Sulfones/pharmacology , Thiophenes/pharmacology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus/enzymology , Zika Virus/genetics , Zika Virus/isolation & purification , Zika Virus Infection/virology
13.
PLoS Pathog ; 15(4): e1007656, 2019 04.
Article in English | MEDLINE | ID: mdl-30951555

ABSTRACT

Zika virus (ZIKV), a member of the Flaviviridae family, has emerged as a major public health threat, since ZIKV infection has been connected to microcephaly and other neurological disorders. Flavivirus genome replication is driven by NS5, an RNA-dependent RNA polymerase (RdRP) that also contains a N-terminal methyltransferase domain essential for viral mRNA capping. Given its crucial roles, ZIKV NS5 has become an attractive antiviral target. Here, we have used integrated structural biology approaches to characterize the supramolecular arrangement of the full-length ZIKV NS5, highlighting the assembly and interfaces between NS5 monomers within a dimeric structure, as well as the dimer-dimer interactions to form higher order fibril-like structures. The relative orientation of each monomer within the dimer provides a model to explain the coordination between MTase and RdRP domains across neighboring NS5 molecules and mutational studies underscore the crucial role of the MTase residues Y25, K28 and K29 in NS5 dimerization. The basic residue K28 also participates in GTP binding and competition experiments indicate that NS5 dimerization is disrupted at high GTP concentrations. This competition represents a first glimpse at a molecular level explaining how dimerization might regulate the capping process.


Subject(s)
Protein Conformation , Protein Multimerization , RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/chemistry , Zika Virus/enzymology , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism
14.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800884

ABSTRACT

The nucleotide analog sofosbuvir, licensed for the treatment of hepatitis C, recently revealed activity against the Zika virus (ZIKV) in vitro and in animal models. However, the ZIKV genetic barrier to sofosbuvir has not yet been characterized. In this study, in vitro selection experiments were performed in infected human hepatoma cell lines. Increasing drug pressure significantly delayed viral breakthrough (p = 0.029). A double mutant in the NS5 gene (V360L/V607I) emerged in 3 independent experiments at 40-80 µM sofosbuvir resulting in a 3.9 ± 0.9-fold half- maximal inhibitory concentration (IC50) shift with respect to the wild type (WT) virus. A triple mutant (C269Y/V360L/V607I), detected in one experiment at 80 µM, conferred a 6.8-fold IC50 shift with respect to the WT. Molecular dynamics simulations confirmed that the double mutant V360L/V607I impacts the binding mode of sofosbuvir, supporting its role in sofosbuvir resistance. Due to the distance from the catalytic site and to the lack of reliable structural data, the contribution of C269Y was not investigated in silico. By a combination of sequence analysis, phenotypic susceptibility testing, and molecular modeling, we characterized a double ZIKV NS5 mutant with decreased sofosbuvir susceptibility. These data add important information to the profile of sofosbuvir as a possible lead for anti-ZIKV drug development.


Subject(s)
Amino Acid Substitution , Antiviral Agents/pharmacology , Point Mutation , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Sofosbuvir/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus/drug effects , Animals , Antiviral Agents/therapeutic use , Binding Sites , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chlorocebus aethiops , Humans , Inhibitory Concentration 50 , Liver Neoplasms/pathology , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Selection, Genetic , Sofosbuvir/therapeutic use , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Zika Virus/enzymology , Zika Virus/genetics
15.
J Med Virol ; 92(1): 11-16, 2020 01.
Article in English | MEDLINE | ID: mdl-31436327

ABSTRACT

The Zika virus (ZIKV) outbreak, which started in the year 2015, is considered the fastest and most widely spread outbreak reported for this flavivirus. The polymerase domain of the NS5 protein has been targeted in other viral infections and is recognized as a suitable target in ZIKV infection. Different novel modified compounds against ZIKV NS5 have been tested in silico. A few structures have been solved for ZIKV polymerase and deposited in the protein data bank website. Two of these solved structures (with a resolution of less than 1.9 A) are used in this study to test the binding of 74 novel compounds in silico. Molecular docking is used to quantify the binding affinities of ZIKV polymerase and compare it to the hepatitis C virus NS5B. A total of 19 novel compounds revealed results that are either similar to or better than the physiological molecule, guanosine triphosphate. Water molecules are found to facilitate the binding of the compounds to ZIKV RNA-dependent RNA polymerase (RdRp) structures. The presented 19 novel compounds represent good binders to ZIKV RdRp and could be suitable candidates for developing a new and effective anti-ZIKV polymerase nucleotide inhibitor.


Subject(s)
Antiviral Agents/pharmacology , Guanosine/analogs & derivatives , Guanosine/pharmacology , Zika Virus/drug effects , Zika Virus/enzymology , Drug Discovery , Molecular Docking Simulation , Protein Binding , RNA-Dependent RNA Polymerase/antagonists & inhibitors
16.
Arch Biochem Biophys ; 695: 108631, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33080173

ABSTRACT

Among Flaviviridae, in West Nile virus (WNV) and Hepatitis C virus (HCV), the non-structural protein NS4A modulates the NTPase activity of viral helicases during nucleic acid unwinding through its N-terminal disordered residues (1-50). In HCV, the acidic NS4A also serves as a cofactor for regulating the NS3 protease activity. However, in case of Zika virus (ZIKV), the role of NS4A and its impact on activities of NS3 helicase and protease is not known. In order to elucidate the role of NS4A, we checked the NTPase activity of NS3 helicase and protease activity of NS3 protease in presence of NS4A N-terminal region (residues 1-48) peptide. Our enzyme kinetics results together with binding experiment clearly demonstrate that NS3 helicase in presence of NS4A peptide increased the rate of ATP hydrolysis whereas the protease activity of NS3 protease was not affected. Therefore, like WNV and HCV, our results establish a role of ZIKV NS4A being a cofactor for modulating the NTPase activity of ZIKV NS3 helicase.


Subject(s)
Nucleoside-Triphosphatase/chemistry , RNA Helicases/chemistry , Serine Endopeptidases/chemistry , Viral Proteins/chemistry , Zika Virus/enzymology , Coenzymes , Nucleoside-Triphosphatase/genetics , Protein Domains , RNA Helicases/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Zika Virus/genetics
17.
Bioorg Med Chem Lett ; 30(5): 126965, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31980339

ABSTRACT

In recent years, the Zika virus has emerged from a neglected flavivirus to a health-threatening pathogen that causes epidemic outbreaks associated with neurological disorders and congenital malformations. In addition to vaccine development, the discovery of specific antiviral agents has been pursued intensely. The Zika virus protease NS2B-NS3 catalyses the processing of the viral precursor polyprotein as an essential step during viral replication. Since the epidemic Zika virus outbreak in the Americas, several inhibitors of this protease have been reported. Substrate-derived peptides revealed important structural information about the active site, whilst more drug-like small molecules have been discovered as allosteric inhibitors.


Subject(s)
Antiviral Agents/therapeutic use , Serine Proteinase Inhibitors/therapeutic use , Viral Proteins/antagonists & inhibitors , Zika Virus Infection/drug therapy , Zika Virus/enzymology , Animals , Drug Discovery , Drug Repositioning , Humans , Serine Endopeptidases
18.
J Chem Inf Model ; 60(2): 731-737, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31850756

ABSTRACT

The NS2B-NS3 protease has been identified as an attractive target for drug development against Zika virus (ZIKV) and combined drug repurposing and structure-based virtual screening has improved the development of antiviral drugs. In this study, we performed a structure-based virtual screening of 1861 Food and Administration (FDA) approved drugs available in DrugBank by the selection and docking validation of crystal structure of ZIKV NS2B-NS3 protease (PDB ID 5H4I ) using Glide and DOCK 6 software. The antihistaminic chlorcyclizine (Grid score -24.8 kcal/mol) exhibited the most promising interaction with NS2B-NS3 protease in comparison to crystallography ligand (Grid score -15.6 kcal/mol) by interaction to Tyr161 by hydrophobic interactions in the binding site of NS2B-NS3 which is recognized as an important amino acid in substrate molecular recognition. Cytotoxicity and global antiviral activity assay in Vero cells by MTT method showed that chlorcyclizine reduced the ZIKV induced cytopathic effect (EC50 of 69.0 ± 7.3 µM and SI = 1.9), and explicit molecular dynamics simulations implemented on a NAMD program indicated great stability of chlorcyclizine in protease binding site, suggesting the repurposing of chlorcyclizine as a promising finding in anti-ZIKV drug development.


Subject(s)
Drug Repositioning , Molecular Dynamics Simulation , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Viral Proteins/antagonists & inhibitors , Zika Virus/enzymology , Animals , Chlorocebus aethiops , Protein Conformation , Serine Endopeptidases/chemistry , Serine Proteinase Inhibitors/toxicity , Vero Cells , Viral Proteins/chemistry
19.
J Chem Inf Model ; 60(2): 562-568, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31985225

ABSTRACT

The NS5 methyltransferase (MTase) has been reported as an attractive molecular target for antivirals discovery against the Zika virus (ZIKV). Here, we report structure-based virtual screening of 42 390 structures from the Development Therapeutics Program (DTP) AIDS Antiviral Screen Database. Among the docked compounds, ZINC1652386 stood out due to its high affinity for MTase in comparison to the cocrystallized ligand MS2042, which interacts with the Asp146 residue in the MTase binding site by hydrogen bonding. Subsequent molecular dynamics simulations predicted that this compound forms a stable complex with MTase within 50 ns. Thus, ZINC1652386 may represent a promising ZIKV methyltransferase inhibitor.


Subject(s)
Antiviral Agents/pharmacology , Methyltransferases/antagonists & inhibitors , Molecular Dynamics Simulation , Zika Virus/drug effects , Zika Virus/enzymology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Binding Sites , Databases, Pharmaceutical , Drug Evaluation, Preclinical , Hydrogen Bonding , Methyltransferases/chemistry , Methyltransferases/metabolism , Molecular Docking Simulation , Protein Conformation , User-Computer Interface
20.
J Am Chem Soc ; 141(17): 6832-6836, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31017399

ABSTRACT

Flaviviruses, including dengue, West Nile and recently emerged Zika virus, are important human pathogens, but there are no drugs to prevent or treat these viral infections. The highly conserved Flavivirus NS2B-NS3 protease is essential for viral replication and therefore a drug target. Compound screening followed by medicinal chemistry yielded a series of drug-like, broadly active inhibitors of Flavivirus proteases with IC50 as low as 120 nM. The inhibitor exhibited significant antiviral activities in cells (EC68: 300-600 nM) and in a mouse model of Zika virus infection. X-ray studies reveal that the inhibitors bind to an allosteric, mostly hydrophobic pocket of dengue NS3 and hold the protease in an open, catalytically inactive conformation. The inhibitors and their binding structures would be useful for rational drug development targeting Zika, dengue and other Flaviviruses.


Subject(s)
Antiviral Agents/therapeutic use , Protease Inhibitors/therapeutic use , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus Infection/drug therapy , Allosteric Site , Aminopyridines/chemical synthesis , Aminopyridines/metabolism , Aminopyridines/therapeutic use , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Cell Line, Tumor , Chlorocebus aethiops , Crystallography, X-Ray , Dengue Virus/enzymology , Drug Discovery , Female , Humans , Male , Mice, Inbred C57BL , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Pyrazines/chemical synthesis , Pyrazines/metabolism , Pyrazines/therapeutic use , Serine Endopeptidases/chemistry , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , West Nile virus/enzymology , Zika Virus/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL