Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5179, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898037

ABSTRACT

Viral genetic diversity presents significant challenges in developing antivirals with broad-spectrum activity and high barriers to resistance. Here we report development of proteolysis targeting chimeras (PROTACs) targeting the dengue virus envelope (E) protein through coupling of known E fusion inhibitors to ligands of the CRL4CRBN E3 ubiquitin ligase. The resulting small molecules block viral entry through inhibition of E-mediated membrane fusion and interfere with viral particle production by depleting intracellular E in infected Huh 7.5 cells. This activity is retained in the presence of point mutations previously shown to confer partial resistance to the parental inhibitors due to decreased inhibitor-binding. The E PROTACs also exhibit broadened spectrum of activity compared to the parental E inhibitors against a panel of mosquito-borne flaviviruses. These findings encourage further exploration of targeted protein degradation as a differentiated and potentially advantageous modality for development of broad-spectrum direct-acting antivirals.


Subject(s)
Antiviral Agents , Dengue Virus , Flavivirus , Proteolysis , Virus Internalization , Humans , Proteolysis/drug effects , Animals , Antiviral Agents/pharmacology , Flavivirus/drug effects , Flavivirus/genetics , Flavivirus/metabolism , Virus Internalization/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Dengue Virus/genetics , Culicidae/virology , Ubiquitin-Protein Ligases/metabolism , Viral Envelope Proteins/metabolism , Cell Line
2.
bioRxiv ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38854003

ABSTRACT

Targeted protein degradation has been widely adopted as a new approach to eliminate both established and previously recalcitrant therapeutic targets. Here we report the development of small molecule degraders of the envelope (E) protein of dengue virus. We developed two classes of bivalent E-degraders, linking two previously reported E-binding small molecules, GNF-2 and CVM-2-12-2, to a glutarimide-based recruiter of the CRL4CRBN ligase to effect proteosome-mediated degradation of the E protein. ZXH-2-107 (based on GNF-2) is an E degrader with ABL inhibition while ZXH-8-004 (based on CVM-2-12-2) is a selective and potent E-degrader. These two compounds provide proof-of-concept that difficult-to-drug targets such as a viral envelope protein can be effectively eliminated using a bivalent degrader and provide starting points for the future development of a new class antiviral drugs.

3.
Antiviral Res ; 210: 105480, 2023 02.
Article in English | MEDLINE | ID: mdl-36567024

ABSTRACT

Targeted protein degradation (TPD) has emerged as a new modality in drug discovery. In this approach, small molecules are used to drive degradation of the target protein of interest. Whereas most direct-acting antivirals (DAAs) inhibit or derange the activity of their viral protein targets and have occupancy-driven pharmacology, small molecules with a TPD-based mechanism have event-driven pharmacology exerted through their ability to induce target degradation. These contrasting mechanisms can result in significant differences in drug efficacy and pharmacodynamics that may be useful in the development of new classes of antivirals. While now being widely pursued in cancer biology and autoimmune disease, TPD has not yet been widely applied as an antiviral strategy. Here, we briefly review TPD pharmacology along with the current status of tools available for developing small molecules that achieve antiviral activity through a TPD mechanism. We also highlight aspects of TPD that may be especially useful in the development of antivirals and that we hope will motivate pursuit of TPD-based antivirals by the antivirals research community.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Proteolysis , Hepatitis C, Chronic/drug therapy , Drug Discovery , Proteins
4.
J Med Virol ; 95(1): e28157, 2023 01.
Article in English | MEDLINE | ID: mdl-36117402

ABSTRACT

Coronavirus disease 2019 (COVID-19) remains a major public health concern, and vaccine unavailability, hesitancy, or failure underscore the need for discovery of efficacious antiviral drug therapies. Numerous approved drugs target protein kinases associated with viral life cycle and symptoms of infection. Repurposing of kinase inhibitors is appealing as they have been vetted for safety and are more accessible for COVID-19 treatment. However, an understanding of drug mechanism is needed to improve our understanding of the factors involved in pathogenesis. We tested the in vitro activity of three kinase inhibitors against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), including inhibitors of AXL kinase, a host cell factor that contributes to successful SARS-CoV-2 infection. Using multiple cell-based assays and approaches, gilteritinib, nintedanib, and imatinib were thoroughly evaluated for activity against SARS-CoV-2 variants. Each drug exhibited antiviral activity, but with stark differences in potency, suggesting differences in host dependency for kinase targets. Importantly, for gilteritinib, the amount of compound needed to achieve 90% infection inhibition, at least in part involving blockade of spike protein-mediated viral entry and at concentrations not inducing phospholipidosis (PLD), approached a clinically achievable concentration. Knockout of AXL, a target of gilteritinib and nintedanib, impaired SARS-CoV-2 variant infectivity, supporting a role for AXL in SARS-CoV-2 infection and supporting further investigation of drug-mediated AXL inhibition as a COVID-19 treatment. This study supports further evaluation of AXL-targeting kinase inhibitors as potential antiviral agents and treatments for COVID-19. Additional mechanistic studies are needed to determine underlying differences in virus response.


Subject(s)
COVID-19 , Humans , SARS-CoV-2/metabolism , COVID-19 Drug Treatment , Drug Repositioning , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Spike Glycoprotein, Coronavirus/metabolism
5.
Annu Rev Virol ; 8(1): 459-489, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34197186

ABSTRACT

Viral fusion glycoproteins catalyze membrane fusion during viral entry. Unlike most enzymes, however, they lack a conventional active site in which formation or scission of a specific covalent bond is catalyzed. Instead, they drive the membrane fusion reaction by cojoining highly regulated changes in conformation to membrane deformation. Despite the challenges in applying inhibitor design approaches to these proteins, recent advances in knowledge of the structures and mechanisms of viral fusogens have enabled the development of small-molecule inhibitors of both class I and class II viral fusion proteins. Here, we review well-validated inhibitors, including their discovery, targets, and mechanism(s) of action, while highlighting mechanistic similarities and differences. Together, these examples make a compelling case for small-molecule inhibitors as tools for probing the mechanisms of viral glycoprotein-mediated fusion and for viral glycoproteins as druggable targets.


Subject(s)
Viral Fusion Proteins , Virus Internalization , Membrane Fusion , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/chemistry
7.
Pharm Res ; 37(9): 167, 2020 Aug 10.
Article in English | MEDLINE | ID: mdl-32778962

ABSTRACT

The outbreak of COVID-19, the pandemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spurred an intense search for treatments by the scientific community. In the absence of a vaccine, the goal is to target the viral life cycle and alleviate the lung-damaging symptoms of infection, which can be life-threatening. There are numerous protein kinases associated with these processes that can be inhibited by FDA-approved drugs, the repurposing of which presents an alluring option as they have been thoroughly vetted for safety and are more readily available for treatment of patients and testing in clinical trials. Here, we characterize more than 30 approved kinase inhibitors in terms of their antiviral potential, due to their measured potency against key kinases required for viral entry, metabolism, or reproduction. We also highlight inhibitors with potential to reverse pulmonary insufficiency because of their anti-inflammatory activity, cytokine suppression, or antifibrotic activity. Certain agents are projected to be dual-purpose drugs in terms of antiviral activity and alleviation of disease symptoms, however drug combination is also an option for inhibitors with optimal pharmacokinetic properties that allow safe and efficacious co-administration with other drugs, such as antiviral agents, IL-6 blocking agents, or other kinase inhibitors.


Subject(s)
Antiviral Agents/therapeutic use , Coronavirus Infections/drug therapy , Drug Repositioning , Pneumonia, Viral/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , COVID-19 , Humans , Pandemics
8.
Can J Physiol Pharmacol ; 98(8): 483-489, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32640179

ABSTRACT

In response to the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), researchers are expeditiously searching for antiviral treatments able to alleviate the symptoms of infection, which can be life-threatening. Here, we provide a general overview of what is currently known about the structure and characteristic features of SARS-CoV-2, some of which could potentially be exploited for the purposes of antiviral therapy and vaccine development. This minireview also covers selected and noteworthy antiviral agents/supportive therapy out of hundreds of drugs that are being repurposed or tested as potential treatments for COVID-19, the disease caused by SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus , Coronavirus Infections , Pandemics , Pneumonia, Viral , Therapies, Investigational/methods , Betacoronavirus/isolation & purification , Betacoronavirus/pathogenicity , Betacoronavirus/physiology , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Coronavirus Infections/therapy , Humans , Pneumonia, Viral/epidemiology , Pneumonia, Viral/therapy , SARS-CoV-2 , Treatment Outcome , COVID-19 Drug Treatment
9.
J Biol Chem ; 295(35): 12426-12436, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32641492

ABSTRACT

Many RNA viruses create specialized membranes for genome replication by manipulating host lipid metabolism and trafficking, but in most cases, we do not know the molecular mechanisms responsible or how specific lipids may impact the associated membrane and viral process. For example, hepatitis C virus (HCV) causes a specific, large-fold increase in the steady-state abundance of intracellular desmosterol, an immediate precursor of cholesterol, resulting in increased fluidity of the membrane where HCV RNA replication occurs. Here, we establish the mechanism responsible for HCV's effect on intracellular desmosterol, whereby the HCV NS3-4A protease controls activity of 24-dehydrocholesterol reductase (DHCR24), the enzyme that catalyzes conversion of desmosterol to cholesterol. Our cumulative evidence for the proposed mechanism includes immunofluorescence microscopy experiments showing co-occurrence of DHCR24 and HCV NS3-4A protease; formation of an additional, faster-migrating DHCR24 species (DHCR24*) in cells harboring a HCV subgenomic replicon RNA or ectopically expressing NS3-4A; and biochemical evidence that NS3-4A cleaves DHCR24 to produce DHCR24* in vitro and in vivo We further demonstrate that NS3-4A cleaves DHCR24 between residues Cys91 and Thr92 and show that this reduces the intracellular conversion of desmosterol to cholesterol. Together, these studies demonstrate that NS3-4A directly cleaves DHCR24 and that this results in the enrichment of desmosterol in the membranes where NS3-4A and DHCR24 co-occur. Overall, this suggests a model in which HCV directly regulates the lipid environment for RNA replication through direct effects on the host lipid metabolism.


Subject(s)
Hepacivirus/enzymology , Lipid Metabolism , Membrane Lipids/metabolism , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Proteolysis , RNA, Viral/biosynthesis , Serine Proteases/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line, Tumor , Hepacivirus/genetics , Humans , Membrane Lipids/genetics , Nerve Tissue Proteins/genetics , Oxidoreductases Acting on CH-CH Group Donors/genetics , RNA, Viral/genetics , Serine Proteases/genetics , Viral Nonstructural Proteins/genetics
10.
mBio ; 11(2)2020 04 14.
Article in English | MEDLINE | ID: mdl-32291299

ABSTRACT

While the basic mechanisms of flavivirus entry and fusion are understood, little is known about the postfusion events that precede RNA replication, such as nucleocapsid disassembly. We describe here a sensitive, conditionally replication-defective yellow fever virus (YFV) entry reporter, YFVΔSK/Nluc, to quantitively monitor the translation of incoming, virus particle-delivered genomes. We validated that YFVΔSK/Nluc gene expression can be neutralized by YFV-specific antisera and requires known flavivirus entry pathways and cellular factors, including clathrin- and dynamin-mediated endocytosis, endosomal acidification, YFV E glycoprotein-mediated fusion, and cellular LY6E and RPLP1 expression. The initial round of YFV translation was shown to require cellular ubiquitylation, consistent with recent findings that dengue virus capsid protein must be ubiquitylated in order for nucleocapsid uncoating to occur. Importantly, translation of incoming YFV genomes also required valosin-containing protein (VCP)/p97, a cellular ATPase that unfolds and extracts ubiquitylated client proteins from large complexes. RNA transfection and washout experiments showed that VCP/p97 functions at a postfusion, pretranslation step in YFV entry. Finally, VCP/p97 activity was required by other flaviviruses in mammalian cells and by YFV in mosquito cells. Together, these data support a critical role for VCP/p97 in the disassembly of incoming flavivirus nucleocapsids during a postfusion step in virus entry.IMPORTANCE Flaviviruses are an important group of RNA viruses that cause significant human disease. The mechanisms by which flavivirus nucleocapsids are disassembled during virus entry remain unclear. Here, we used a yellow fever virus entry reporter, which expresses a sensitive reporter enzyme but does not replicate, to show that nucleocapsid disassembly requires the cellular protein-disaggregating enzyme valosin-containing protein, also known as p97.


Subject(s)
Valosin Containing Protein/metabolism , Virus Uncoating , Yellow fever virus/genetics , Yellow fever virus/physiology , Animals , Cell Line , Cricetinae , Female , Genes, Reporter , Genome, Viral , HEK293 Cells , HeLa Cells , Humans , Kidney/cytology , Mice , Mice, Inbred C57BL , Valosin Containing Protein/genetics , Virus Internalization , Virus Replication
11.
J Biol Chem ; 295(6): 1694-1703, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31914414

ABSTRACT

Small-molecule inhibitors of translation are critical tools to study the molecular mechanisms of protein synthesis. In this study, we sought to characterize how QL47, a host-targeted, small-molecule antiviral agent, inhibits steady-state viral protein expression. We demonstrate that this small molecule broadly inhibits both viral and host protein synthesis and targets a translation step specific to eukaryotic cells. We show that QL47 inhibits protein neosynthesis initiated by both canonical cap-driven and noncanonical initiation strategies, most likely by targeting an early step in translation elongation. Our findings thus establish QL47 as a new small-molecule inhibitor that can be utilized to probe the eukaryotic translation machinery and that can be further developed as a new therapeutic agent.


Subject(s)
Antiviral Agents/pharmacology , Protein Biosynthesis/drug effects , Small Molecule Libraries/pharmacology , Antiviral Agents/chemistry , Cell Line , HEK293 Cells , Humans , Proteins/metabolism , Small Molecule Libraries/chemistry , Viral Proteins/metabolism , Virus Diseases/drug therapy , Virus Diseases/metabolism , Viruses/drug effects , Viruses/metabolism
13.
Nat Commun ; 10(1): 3468, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31371704

ABSTRACT

Targeted protein degradation is a promising drug development paradigm. Here we leverage this strategy to develop a new class of small molecule antivirals that induce proteasomal degradation of viral proteins. Telaprevir, a reversible-covalent inhibitor that binds to the hepatitis C virus (HCV) protease active site is conjugated to ligands that recruit the CRL4CRBN ligase complex, yielding compounds that can both inhibit and induce the degradation of the HCV NS3/4A protease. An optimized degrader, DGY-08-097, potently inhibits HCV in a cellular infection model, and we demonstrate that protein degradation contributes to its antiviral activity. Finally, we show that this new class of antiviral agents can overcome viral variants that confer resistance to traditional enzymatic inhibitors such as telaprevir. Overall, our work provides proof-of-concept that targeted protein degradation may provide a new paradigm for the development of antivirals with superior resistance profiles.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/chemistry , Cell Line, Tumor , Drug Design , Drug Resistance, Viral/genetics , Gene Knockdown Techniques , HEK293 Cells , Hepacivirus/drug effects , Hepacivirus/metabolism , Hepatitis C/drug therapy , Hepatitis C/genetics , Hepatitis C/virology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/pharmacology , Proof of Concept Study , Protease Inhibitors/chemistry , Proteolysis/drug effects , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/metabolism
14.
PLoS Pathog ; 15(1): e1007553, 2019 01.
Article in English | MEDLINE | ID: mdl-30703168

ABSTRACT

[This corrects the article DOI: 10.1371/journal.ppat.1002627.].

15.
Antiviral Res ; 164: 147-153, 2019 04.
Article in English | MEDLINE | ID: mdl-30771406

ABSTRACT

The recent emergence of Zika virus, a mosquito-borne flavivirus, in the Americas has shed light on the severe neurological diseases associated with infection, notably congenital microcephaly in newborns and Guillain-Barré syndrome in adults. Despite the recent focus on Zika virus, there are currently no approved vaccines or antiviral therapies available to treat or prevent infection. In this study we established a competitive amplified luminescent proximity homogeneous assay (ALPHAscreen) to identify small molecule inhibitors targeting the envelope protein of Zika virus (Zika E). We utilized this assay to screen two libraries of nearly 27,000 compounds and identified seven novel inhibitors of Zika E. Characterization of these primary screening leads demonstrated that inhibition of Zika virus occurs at non-cytotoxic concentrations for all seven lead compounds. In addition, we found that all seven lead compounds have potent activity against the closely related dengue virus 2 but not vesicular stomatitis virus, an unrelated enveloped virus. Biochemical experiments indicate that these compounds act by preventing E-mediated membrane fusion. This work highlights a new method for the discovery and optimization of direct-acting antivirals targeting the E protein of Zika and other flaviviruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Small Molecule Libraries , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus/drug effects , Dengue Virus/drug effects , Virus Internalization/drug effects
16.
ACS Infect Dis ; 5(3): 460-472, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30608640

ABSTRACT

Vaccines and antivirals to combat dengue, Zika, and other flavivirus pathogens present a major, unmet medical need. Vaccine development has been severely challenged by the antigenic diversity of these viruses and the propensity of non-neutralizing, cross-reactive antibodies to facilitate cellular infection and increase disease severity. As an alternative, direct-acting antivirals targeting the flavivirus envelope protein, E, have the potential to act via an analogous mode of action without the risk of antibody-dependent enhancement of infection and disease. We previously discovered that structurally diverse small molecule inhibitors of the dengue virus E protein exhibit varying levels of antiviral activity against other flaviviruses in cell culture. Here, we demonstrate that the broad-spectrum activity of several cyanohydrazones against dengue, Zika, and Japanese encephalitis viruses is due to specific inhibition of E-mediated membrane fusion during viral entry and provide proof of concept for pharmacological inhibition of E as an antiviral strategy in vivo.


Subject(s)
Antiviral Agents/administration & dosage , Flavivirus Infections/drug therapy , Flavivirus/drug effects , Small Molecule Libraries/administration & dosage , Viral Envelope Proteins/metabolism , Animals , Antiviral Agents/chemistry , Female , Flavivirus/physiology , Flavivirus Infections/virology , Humans , Male , Mice , Mice, Inbred C57BL , Small Molecule Libraries/chemistry , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/genetics , Virus Internalization/drug effects , Virus Replication/drug effects
17.
Sci Signal ; 11(553)2018 10 23.
Article in English | MEDLINE | ID: mdl-30352947

ABSTRACT

This Editorial discusses the state of research on drug resistance in the fields of cancer, infectious disease, and agriculture. Reaching across the aisle for a more cross-collaborative approach may lead to exciting breakthroughs toward tackling the challenges of drug resistance in each field.


Subject(s)
Drug Resistance, Microbial , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Agriculture , Communicable Diseases/drug therapy , Humans , Interdisciplinary Communication , Mutation , Stochastic Processes
18.
Elife ; 72018 07 12.
Article in English | MEDLINE | ID: mdl-29999491

ABSTRACT

Dengue virus (DV) is a compact, icosahedrally symmetric, enveloped particle, covered by 90 dimers of envelope protein (E), which mediates viral attachment and membrane fusion. Fusion requires a dimer-to-trimer transition and membrane engagement of hydrophobic 'fusion loops'. We previously characterized the steps in membrane fusion for the related West Nile virus (WNV), using recombinant, WNV virus-like particles (VLPs) for single-particle experiments (Chao et al., 2014). Trimerization and membrane engagement are rate-limiting; fusion requires at least two adjacent trimers; availability of competent monomers within the contact zone between virus and target membrane creates a trimerization bottleneck. We now report an extension of that work to dengue VLPs, from all four serotypes, finding an essentially similar mechanism. Small-molecule inhibitors of dengue virus infection that target E block its fusion-inducing conformational change. We show that ~12-14 bound molecules per particle (~20-25% occupancy) completely prevent fusion, consistent with the proposed mechanism.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue Virus/physiology , Virus Internalization/drug effects , Antiviral Agents/chemical synthesis , Cell Membrane/metabolism , Models, Biological , Protein Multimerization , Rhodamines/chemical synthesis , Rhodamines/pharmacology , Sulfonic Acids/chemical synthesis , Sulfonic Acids/pharmacology , Viral Envelope Proteins/metabolism , Virosomes/drug effects
19.
ACS Infect Dis ; 4(9): 1395-1406, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30027735

ABSTRACT

Dengue virus is a major human pathogen that infects over 390 million people annually leading to approximately 500 000 hospitalizations due to severe dengue. Since the only marketed vaccine, Dengvaxia, has recently been shown to increase disease severity in those lacking natural immunity, antivirals to prevent or treat dengue  infection represent a large, unmet medical need. Small molecules that target the dengue virus envelope protein, E, on the surface of the virion could act analogously to antibodies by engaging E extracellularly to block infection; however, a shortage of target-based assays suitable for screening and medicinal chemistry studies has limited efforts in this area. Here we demonstrate that the dengue E protein offers a tractable drug target for preventing dengue infection by developing a target-based assay using a recombinantly expressed dengue serotype 2 E protein. We performed a high-throughput screen of ∼20 000 compounds followed by secondary assays to confirm target-binding and antiviral activity and counter-screens to exclude compounds with nonspecific activities. These efforts yielded eight distinct chemical leads that inhibit dengue infection by binding to E and preventing E-mediated membrane fusion with potencies equal to or greater than previously described small molecule inhibitors of E. We show that a subset of these compounds inhibit viruses representative of the other three dengue serotypes and Zika virus. This work provides tools for discovery and optimization of direct-acting antivirals against dengue E and shows that this approach may be useful in developing antivirals with broad-spectrum activity against other flavivirus pathogens.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Dengue Virus/drug effects , Drug Discovery/methods , Small Molecule Libraries/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Dengue/immunology , Dengue/virology , Dengue Virus/genetics , Dengue Virus/physiology , Humans , Small Molecule Libraries/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Zika Virus/drug effects , Zika Virus/physiology
20.
Cell Chem Biol ; 25(8): 1006-1016.e8, 2018 08 16.
Article in English | MEDLINE | ID: mdl-29937406

ABSTRACT

Viral envelope proteins are required for productive viral entry and initiation of infection. Although the humoral immune system provides ample evidence for targeting envelope proteins as an antiviral strategy, there are few pharmacological interventions that have this mode of action. In contrast to classical antiviral targets such as viral proteases and polymerases, viral envelope proteins as a class do not have a well-conserved active site that can be rationally targeted with small molecules. We previously identified compounds that inhibit dengue virus by binding to its envelope protein, E. Here, we show that these small molecules inhibit dengue virus fusion and map the binding site of these compounds to a specific pocket on E. We further demonstrate inhibition of Zika, West Nile, and Japanese encephalitis viruses by these compounds, providing pharmacological evidence for the pocket as a target for developing broad-spectrum antivirals against multiple, mosquito-borne flavivirus pathogens.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Flavivirus Infections/drug therapy , Flavivirus/drug effects , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , Dengue Virus/chemistry , Dengue Virus/drug effects , Dengue Virus/physiology , Drug Discovery , Flavivirus/chemistry , Flavivirus/physiology , Flavivirus Infections/metabolism , Flavivirus Infections/virology , Humans , Molecular Docking Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Envelope Proteins/chemistry , Virus Replication/drug effects , Zika Virus/chemistry , Zika Virus/drug effects , Zika Virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...