Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
Add more filters

Publication year range
1.
Cell ; 184(6): 1604-1620, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740455

ABSTRACT

Historically, emerging viruses appear constantly and have cost millions of human lives. Currently, climate change and intense globalization have created favorable conditions for viral transmission. Therefore, effective antivirals, especially those targeting the conserved protein in multiple unrelated viruses, such as the compounds targeting RNA-dependent RNA polymerase, are urgently needed to combat more emerging and re-emerging viruses in the future. Here we reviewed the development of antivirals with common targets, including those against the same protein across viruses, or the same viral function, to provide clues for development of antivirals for future epidemics.


Subject(s)
Antiviral Agents/therapeutic use , Communicable Diseases, Emerging/drug therapy , Communicable Diseases, Emerging/epidemiology , Molecular Targeted Therapy/methods , Pandemics , Virus Diseases/drug therapy , Virus Diseases/epidemiology , Viruses/enzymology , Animals , Antiviral Agents/pharmacology , Communicable Diseases, Emerging/virology , Humans , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Virus Diseases/virology , Virus Internalization/drug effects
2.
J Virol ; 97(10): e0069623, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37796129

ABSTRACT

IMPORTANCE: Human cytomegalovirus (HCMV) infection is the leading cause of non-heritable birth defects worldwide. HCMV readily infects the early progenitor cell population of the developing brain, and we have found that infection leads to significantly downregulated expression of key neurodevelopmental transcripts. Currently, there are no approved therapies to prevent or mitigate the effects of congenital HCMV infection. Therefore, we used human-induced pluripotent stem cell-derived organoids and neural progenitor cells to elucidate the glycoproteins and receptors used in the viral entry process and whether antibody neutralization was sufficient to block viral entry and prevent disruption of neurodevelopmental gene expression. We found that blocking viral entry alone was insufficient to maintain the expression of key neurodevelopmental genes, but neutralization combined with neurotrophic factor treatment provided robust protection. Together, these studies offer novel insight into mechanisms of HCMV infection in neural tissues, which may aid future therapeutic development.


Subject(s)
Antibodies, Neutralizing , Cytomegalovirus Infections , Cytomegalovirus , Gene Expression , Nerve Growth Factors , Humans , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Cytomegalovirus/drug effects , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Gene Expression/drug effects , Gene Expression/immunology , Induced Pluripotent Stem Cells/cytology , Nerve Growth Factors/pharmacology , Nerve Growth Factors/therapeutic use , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Organoids/cytology , Organoids/metabolism , Organoids/virology , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects
3.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273370

ABSTRACT

Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.


Subject(s)
Antiviral Agents , Viral Envelope Proteins , Virus Internalization , Zika Virus Infection , Zika Virus , Zika Virus/drug effects , Zika Virus/physiology , Virus Internalization/drug effects , Humans , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus Infection/virology , Zika Virus Infection/drug therapy , Antiviral Agents/pharmacology , Animals
4.
J Virol ; 95(16): e0001021, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34037420

ABSTRACT

Respiratory syncytial virus (RSV) has been reported to use CX3CR1 in vitro as a receptor on cultured primary human airway epithelial cultures. To evaluate CX3CR1 as the receptor for RSV in vivo, we used the cotton rat animal model because of its high permissiveness for RSV infection. Sequencing the cotton rat CX3CR1 gene revealed 91% amino acid similarity to human CX3CR1. Previous work found that RSV binds to CX3CR1 via its attachment glycoprotein (G protein) to infect primary human airway cultures. To determine whether CX3CR1-G protein interaction is necessary for RSV infection, recombinant RSVs containing mutations in the CX3CR1 binding site of the G protein were tested in cotton rats. In contrast to wild-type virus, viral mutants did not grow in the lungs of cotton rats. When RSV was incubated with an antibody blocking the CX3CR1 binding site of G protein and subsequently inoculated intranasally into cotton rats, no virus was found in the lungs 4 days postinfection. In contrast, growth of RSV was not affected after preincubation with heparan sulfate (the receptor for RSV on immortalized cell lines). A reduction in CX3CR1 expression in the cotton rat lung through the use of peptide-conjugated morpholino oligomers led to a 10-fold reduction in RSV titers at day 4 postinfection. In summary, these results indicate that CX3CR1 functions as a receptor for RSV in cotton rats and, in combination with data from human airway epithelial cell cultures, strongly suggest that CX3CR1 is a primary receptor for naturally acquired RSV infection. IMPORTANCE The knowledge about a virus receptor is useful to better understand the uptake of a virus into a cell and potentially develop antivirals directed against either the receptor molecule on the cell or the receptor-binding protein of the virus. Among a number of potential receptor proteins, human CX3CR1 has been demonstrated to act as a receptor for respiratory syncytial virus (RSV) on human epithelial cells in tissue culture. Here, we report that the cotton rat CX3CR1, which is similar to the human molecule, acts as a receptor in vivo. This study strengthens the argument that CX3CR1 is a receptor molecule for RSV.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Receptors, Virus/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , Animals , Antibodies, Viral/pharmacology , Binding Sites , CX3C Chemokine Receptor 1/antagonists & inhibitors , CX3C Chemokine Receptor 1/chemistry , Cell Line , Disease Models, Animal , Epithelial Cells/virology , Heparitin Sulfate/metabolism , Humans , Mutation , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/chemistry , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/growth & development , Respiratory Syncytial Virus, Human/metabolism , Respiratory System/metabolism , Respiratory System/virology , Sigmodontinae , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Replication/genetics
5.
Nature ; 535(7610): 169-172, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27362232

ABSTRACT

Ebola viruses (EBOVs) are responsible for repeated outbreaks of fatal infections, including the recent deadly epidemic in West Africa. There are currently no approved therapeutic drugs or vaccines for the disease. EBOV has a membrane envelope decorated by trimers of a glycoprotein (GP, cleaved by furin to form GP1 and GP2 subunits), which is solely responsible for host cell attachment, endosomal entry and membrane fusion. GP is thus a primary target for the development of antiviral drugs. Here we report the first, to our knowledge, unliganded structure of EBOV GP, and high-resolution complexes of GP with the anticancer drug toremifene and the painkiller ibuprofen. The high-resolution apo structure gives a more complete and accurate picture of the molecule, and allows conformational changes introduced by antibody and receptor binding to be deciphered. Unexpectedly, both toremifene and ibuprofen bind in a cavity between the attachment (GP1) and fusion (GP2) subunits at the entrance to a large tunnel that links with equivalent tunnels from the other monomers of the trimer at the three-fold axis. Protein­drug interactions with both GP1 and GP2 are predominately hydrophobic. Residues lining the binding site are highly conserved among filoviruses except Marburg virus (MARV), suggesting that MARV may not bind these drugs. Thermal shift assays show up to a 14 °C decrease in the protein melting temperature after toremifene binding, while ibuprofen has only a marginal effect and is a less potent inhibitor. These results suggest that inhibitor binding destabilizes GP and triggers premature release of GP2, thereby preventing fusion between the viral and endosome membranes. Thus, these complex structures reveal the mechanism of inhibition and may guide the development of more powerful anti-EBOV drugs.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/metabolism , Ebolavirus/chemistry , Toremifene/chemistry , Toremifene/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiviral Agents/pharmacology , Binding Sites , Cell Line , Conserved Sequence , Ebolavirus/drug effects , Endosomes/drug effects , Endosomes/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Ibuprofen/chemistry , Ibuprofen/metabolism , Ibuprofen/pharmacology , Ligands , Marburgvirus/chemistry , Membrane Fusion/drug effects , Models, Molecular , Protein Binding , Protein Stability/drug effects , Protein Structure, Quaternary/drug effects , Protein Subunits/chemistry , Protein Subunits/metabolism , Temperature , Toremifene/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Virus Attachment/drug effects
6.
J Virol ; 94(20)2020 09 29.
Article in English | MEDLINE | ID: mdl-32759318

ABSTRACT

A cascade of protein-protein interactions between four herpes simplex virus (HSV) glycoproteins (gD, gH/gL, and gB) drive fusion between the HSV envelope and host membrane, thereby allowing for virus entry and infection. Specifically, binding of gD to one of its receptors induces a conformational change that allows gD to bind to the regulatory complex gH/gL, which then activates the fusogen gB, resulting in membrane fusion. Using surface plasmon resonance and a panel of anti-gD monoclonal antibodies (MAbs) that sterically blocked the interaction, we previously showed that gH/gL binds directly to gD at sites distinct from the gD receptor binding site. Here, using an analogous strategy, we first evaluated the ability of a panel of uncharacterized anti-gH/gL MAbs to block binding to gD and/or inhibit fusion. We found that the epitopes of four gD-gH/gL-blocking MAbs were located within flexible regions of the gH N terminus and the gL C terminus, while the fifth was placed around gL residue 77. Taken together, our data localized the gD binding region on gH/gL to a group of gH and gL residues at the membrane distal region of the heterodimer. Surprisingly, a second set of MAbs did not block gD-gH/gL binding but instead stabilized the complex by altering the kinetic binding. However, despite this prolonged gD-gH/gL interaction, "stabilizing" MAbs also inhibited cell-cell fusion, suggesting a unique mechanism by which the fusion process is halted. Our findings support targeting the gD-gH/gL interaction to prevent fusion in both therapeutic and vaccine strategies against HSV.IMPORTANCE Key to developing a human HSV vaccine is an understanding of the virion glycoproteins involved in entry. HSV employs multiple glycoproteins for attachment, receptor interaction, and membrane fusion. Determining how these proteins function was resolved, in part, by structural biology coupled with immunological and biologic evidence. After binding, virion gD interacts with a receptor to activate the regulator gH/gL complex, triggering gB to drive fusion. Multiple questions remain, one being the physical location of each glycoprotein interaction site. Using protective antibodies with known epitopes, we documented the long-sought interaction between gD and gH/gL, detailing the region on gD important to create the gD-gH/gL triplex. Now, we have identified the corresponding gD contact sites on gH/gL. Concurrently we discovered a novel mechanism whereby gH/gL antibodies stabilize the complex and inhibit fusion progression. Our model for the gD-gH/gL triplex provides a new framework for studying fusion, which identifies targets for vaccine development.


Subject(s)
Herpesvirus 1, Human/metabolism , Viral Envelope Proteins/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , Membrane Fusion , Sf9 Cells , Spodoptera , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/genetics
7.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32611759

ABSTRACT

Ebola virus (EBOV) entry into cells is mediated by its spike glycoprotein (GP). Following attachment and internalization, virions traffic to late endosomes where GP is cleaved by host cysteine proteases. Cleaved GP then binds its cellular receptor, Niemann-Pick C1. In response to an unknown cellular trigger, GP undergoes conformational rearrangements that drive fusion of viral and endosomal membranes. The temperature-dependent stability (thermostability) of the prefusion conformers of class I viral fusion glycoproteins, including those of filovirus GPs, has provided insights into their propensity to undergo fusion-related rearrangements. However, previously described assays have relied on soluble glycoprotein ectodomains. Here, we developed a simple enzyme-linked immunosorbent assay (ELISA)-based assay that uses the temperature-dependent loss of conformational epitopes to measure thermostability of GP embedded in viral membranes. The base and glycan cap subdomains of all filovirus GPs tested suffered a concerted loss of prefusion conformation at elevated temperatures but did so at different temperature ranges, indicating virus-specific differences in thermostability. Despite these differences, all of these GPs displayed reduced thermostability upon cleavage to GP conformers (GPCL). Surprisingly, acid pH enhanced, rather than decreased, GP thermostability, suggesting it could enhance viral survival in hostile endo/lysosomal compartments. Finally, we confirmed and extended previous findings that some small-molecule inhibitors of filovirus entry destabilize EBOV GP and uncovered evidence that the most potent inhibitors act through multiple mechanisms. We establish the epitope-loss ELISA as a useful tool for studies of filovirus entry, engineering of GP variants with enhanced stability for use in vaccine development, and discovery of new stability-modulating antivirals.IMPORTANCE The development of Ebola virus countermeasures is challenged by our limited understanding of cell entry, especially at the step of membrane fusion. The surface-exposed viral protein, GP, mediates membrane fusion and undergoes major structural rearrangements during this process. The stability of GP at elevated temperatures (thermostability) can provide insights into its capacity to undergo these rearrangements. Here, we describe a new assay that uses GP-specific antibodies to measure GP thermostability under a variety of conditions relevant to viral entry. We show that proteolytic cleavage and acid pH have significant effects on GP thermostability that shed light on their respective roles in viral entry. We also show that the assay can be used to study how small-molecule entry inhibitors affect GP stability. This work provides a simple and readily accessible assay to engineer stabilized GP variants for antiviral vaccines and to discover and improve drugs that act by modulating GP stability.


Subject(s)
Ebolavirus/drug effects , Niemann-Pick C1 Protein/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Viral Fusion Proteins/antagonists & inhibitors , Virion/drug effects , Animals , Binding Sites , Biological Assay , Chlorocebus aethiops , Clomiphene/chemistry , Clomiphene/pharmacology , Ebolavirus/chemistry , Ebolavirus/genetics , Ebolavirus/metabolism , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Hot Temperature , Hydrogen-Ion Concentration , Molecular Docking Simulation , Niemann-Pick C1 Protein/chemistry , Niemann-Pick C1 Protein/genetics , Niemann-Pick C1 Protein/metabolism , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Tertiary , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/chemistry , Tamoxifen/pharmacology , Toremifene/chemistry , Toremifene/pharmacology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virion/chemistry , Virion/genetics , Virion/metabolism
8.
Bioorg Med Chem Lett ; 41: 127983, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33965007

ABSTRACT

We identified and explored the structure-activity relationship (SAR) of a novel heterocyclic chemical series of arenavirus cell entry inhibitors. Optimized lead compounds, including diphenyl-substituted imidazo[1,2-a]pyridines, benzimidazoles, and benzotriazoles exhibited low to sub-nanomolar potency against both pseudotyped and infectious Old and New World arenaviruses, attractive metabolic stability in human and most nonhuman liver microsomes as well as a lack of hERG K + channel or CYP enzyme inhibition. Moreover, the straightforward synthesis of several lead compounds (e.g., the simple high yield 3-step synthesis of imidazo[1,2-a]pyridine 37) could provide a cost-effective broad-spectrum arenavirus therapeutic that may help to minimize the cost-prohibitive burdens associated with treatments for emerging viruses in economically challenged geographical settings.


Subject(s)
Antiviral Agents/pharmacology , Arenavirus/drug effects , Drug Discovery , Heterocyclic Compounds/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Arenavirus/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Humans , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Viral Envelope Proteins/metabolism
9.
J Enzyme Inhib Med Chem ; 36(1): 462-468, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33455472

ABSTRACT

To develop unique small-molecule inhibitors of hepatitis C virus (HCV), thiophen urea (TU) derivatives were synthesised and screened for HCV entry inhibitory activities. Among them, seven TU compounds exhibited portent anti-viral activities against genotypes 1/2 (EC50 < 30 nM) and subsequently, they were further investigated; based on the pharmacological, metabolic, pharmacokinetic, and safety profiles, J2H-1701 was selected as the optimised lead compound as an HCV entry inhibitor. J2H-1701 possesses effective multi-genotypic antiviral activity. The docking results suggested the potential interaction of J2H-1701 with the HCV E2 glycoprotein. These results suggest that J2H-1701 can be a potential candidate drug for the development of HCV entry inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Thiophenes/pharmacology , Urea/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Urea/analogs & derivatives , Urea/chemistry , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects
10.
Molecules ; 26(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652639

ABSTRACT

Hepatitis C is affecting millions of people around the globe annually, which leads to death in very high numbers. After many years of research, hepatitis C virus (HCV) remains a serious threat to the human population and needs proper management. The in silico approach in the drug discovery process is an efficient method in identifying inhibitors for various diseases. In our study, the interaction between Epigallocatechin-3-gallate, a component of green tea, and envelope glycoprotein E2 of HCV is evaluated. Epigallocatechin-3-gallate is the most promising polyphenol approved through cell culture analysis that can inhibit the entry of HCV. Therefore, various in silico techniques have been employed to find out other potential inhibitors that can behave as EGCG. Thus, the homology modelling of E2 protein was performed. The potential lead molecules were predicted using ligand-based as well as structure-based virtual screening methods. The compounds obtained were then screened through PyRx. The drugs obtained were ranked based on their binding affinities. Furthermore, the docking of the topmost drugs was performed by AutoDock Vina, while its 2D interactions were plotted in LigPlot+. The lead compound mms02387687 (2-[[5-[(4-ethylphenoxy) methyl]-4-prop-2-enyl-1,2,4-triazol-3-yl] sulfanyl]-N-[3(trifluoromethyl) phenyl] acetamide) was ranked on top, and we believe it can serve as a drug against HCV in the future, owing to experimental validation.


Subject(s)
Catechin/analogs & derivatives , Hepacivirus/drug effects , Hepatitis C/drug therapy , Viral Envelope Proteins/genetics , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Catechin/chemistry , Catechin/pharmacology , Hepacivirus/genetics , Hepacivirus/pathogenicity , Hepatitis C/virology , Humans , Ligands , Molecular Docking Simulation , Polyphenols/chemistry , Polyphenols/pharmacology , Tea/chemistry , Viral Envelope/chemistry , Viral Envelope Proteins/antagonists & inhibitors , Virus Internalization/drug effects
11.
Biochemistry ; 59(39): 3709-3724, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32876433

ABSTRACT

The World Health Organization has designated Zika virus (ZIKV) as a dangerous, mosquito-borne pathogen that can cause severe developmental defects. The primary goal of this work was identification of small molecules as potential ZIKV inhibitors that target the viral envelope glycoprotein (ZIKV E) involved in membrane fusion and viral entry. A homology model of ZIKV E containing the small molecule ß-octyl glucoside (BOG) was constructed, on the basis of an analogous X-ray structure from dengue virus, and >4 million commercially available compounds were computationally screened using the program DOCK6. A key feature of the screen involved the use of similarity-based scoring to identify inhibitor candidates that make similar interaction energy patterns (molecular footprints) as the BOG reference. Fifty-three prioritized compounds underwent experimental testing using cytotoxicity, cell viability, and tissue culture infectious dose 50% (TCID50) assays. Encouragingly, relative to a known control (NITD008), six compounds were active in both the cell viability assay and the TCID50 infectivity assay, and they showed activity in a third caspase activity assay. In particular, compounds 8 and 15 (tested at 25 µM) and compound 43 (tested at 10 µM) appeared to provide significant protection to infected cells, indicative of anti-ZIKV activity. Overall, the study highlights how similarity-based scoring can be leveraged to computationally identify potential ZIKV E inhibitors that mimic a known reference (in this case BOG), and the experimentally verified hits provide a strong starting point for further refinement and optimization efforts.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus/drug effects , Animals , Chlorocebus aethiops , Drug Discovery , Humans , Molecular Docking Simulation , Vero Cells , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Zika Virus/physiology , Zika Virus Infection/drug therapy , Zika Virus Infection/metabolism , Zika Virus Infection/virology
12.
Biochem Biophys Res Commun ; 530(1): 10-14, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828269

ABSTRACT

COVID-19 is one of the most impactful pandemics in recorded history. As such, the identification of inhibitory drugs against its etiological agent, SARS-CoV-2, is of utmost importance, and in particular, repurposing may provide the fastest route to curb the disease. As the first step in this route, we sought to identify an attractive and viable target in the virus for pharmaceutical inhibition. Using three bacteria-based assays that were tested on known viroporins, we demonstrate that one of its essential components, the E protein, is a potential ion channel and, therefore, is an excellent drug target. Channel activity was demonstrated for E proteins in other coronaviruses, providing further emphasis on the importance of this functionally to the virus' pathogenicity. The results of a screening effort involving a repurposing drug library of ion channel blockers yielded two compounds that inhibit the E protein: Gliclazide and Memantine. In conclusion, as a route to curb viral virulence and abate COVID-19, we point to the E protein of SARS-CoV-2 as an attractive drug target and identify off-label compounds that inhibit it.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Gliclazide/pharmacology , Ion Channels/antagonists & inhibitors , Memantine/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Betacoronavirus/metabolism , COVID-19 , Coronavirus Envelope Proteins , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Drug Discovery , Drug Repositioning , Humans , Ion Channels/metabolism , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , SARS-CoV-2 , Viral Envelope Proteins/metabolism
13.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31092576

ABSTRACT

A key step in the Ebola virus (EBOV) replication cycle involves conformational changes in viral glycoprotein 2 (GP2) which facilitate host-viral membrane fusion and subsequent release of the viral genome. Ebola GP2 plays a critical role in virus entry and has similarities in mechanism and structure to the HIV gp41 protein for which inhibitors have been successfully developed. In this work, a putative binding pocket for the C-terminal heptad repeat in the N-terminal heptad repeat trimer was targeted for identification of small molecules that arrest EBOV-host membrane fusion. Two computational structure-based virtual screens of ∼1.7 M compounds were performed (DOCK program) against a GP2 five-helix bundle, resulting in 165 commercially available compounds purchased for experimental testing. Based on assessment of inhibitory activity, cytotoxicity, and target specificity, four promising candidates emerged with 50% inhibitory concentration values in the 3 to 26 µM range. Molecular dynamics simulations of the two most potent candidates in their DOCK-predicted binding poses indicate that the majority of favorable interactions involve seven highly conserved residues that can be used to guide further inhibitor development and refinement targeting EBOV.IMPORTANCE The most recent Ebola virus disease outbreak, from 2014 to 2016, resulted in approximately 28,000 individuals becoming infected, which led to over 12,000 causalities worldwide. The particularly high pathogenicity of the virus makes paramount the identification and development of promising lead compounds to serve as inhibitors of Ebola infection. To limit viral load, the virus-host membrane fusion event can be targeted through the inhibition of the class I fusion glycoprotein of Ebolavirus In the current work, several promising small-molecule inhibitors that target the glycoprotein GP2 were identified through systematic application of structure-based computational and experimental drug design procedures.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Molecular Mimicry , Viral Envelope Proteins/antagonists & inhibitors , Virus Internalization/drug effects , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cell Line , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding
14.
PLoS Pathog ; 14(8): e1007204, 2018 08.
Article in English | MEDLINE | ID: mdl-30138408

ABSTRACT

Recent studies suggest that some monoclonal antibodies (mAbs) specific for ebolavirus glycoprotein (GP) can protect experimental animals against infections. Most mAbs isolated from ebolavirus survivors appeared to target the glycan cap or the stalk region of the viral GP, which is the envelope protein and the only antigen inducing virus-neutralizing antibody response. Some of the mAbs were demonstrated to be protective in vivo. Here, a panel of mAbs from four individual survivors of ebolavirus infection that target the glycan cap or stem region were selected for investigation of the mechanisms of their antiviral effect. Comparative characterization of the inhibiting effects on multiple steps of viral replication was performed, including attachment, post-attachment, entry, binding at low pH, post-cleavage neutralization of virions, viral trafficking to endosomes, cell-to-cell transmission, viral egress, and inhibition when added early at various time points post-infection. In addition, Fc-domain related properties were characterized, including activation and degranulation of NK cells, antibody-dependent cellular phagocytosis and glycan content. The two groups of mAbs (glycan cap versus stem) demonstrated very different profiles of activities suggesting usage of mAbs with different epitope specificity could coordinate inhibition of multiple steps of filovirus infection through Fab- and Fc-mediated mechanisms, and provide a reliable therapeutic approach.


Subject(s)
Antibodies, Viral/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Viral Envelope Proteins/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Humans
15.
Mol Ther ; 27(5): 974-985, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30962164

ABSTRACT

Zika virus (ZIKV) infection is endemic to several world regions, and many others are at high risk for seasonal outbreaks. Synthetic DNA-encoded monoclonal antibody (DMAb) is an approach that enables in vivo delivery of highly potent mAbs to control infections. We engineered DMAb-ZK190, encoding the mAb ZK190 neutralizing antibody, which targets the ZIKV E protein DIII domain. In vivo-delivered DMAb-ZK190 achieved expression levels persisting >10 weeks in mice and >3 weeks in non-human primate (NHPs), which is protective against ZIKV infectious challenge. This study is the first demonstration of infectious disease control in NHPs following in vivo delivery of a nucleic acid-encoded antibody, supporting the importance of this new platform.


Subject(s)
Antibodies, Neutralizing/pharmacology , DNA/pharmacology , Viral Envelope Proteins/immunology , Zika Virus Infection/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , DNA/immunology , Humans , Mice , Primates , Viral Envelope Proteins/antagonists & inhibitors , Zika Virus/genetics , Zika Virus/immunology , Zika Virus/pathogenicity , Zika Virus Infection/immunology , Zika Virus Infection/therapy , Zika Virus Infection/virology
16.
Mol Ther ; 27(5): 947-959, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30852138

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy is a promising novel therapeutic approach for cancer but also for chronic infection. We have developed a fully human, second-generation CAR directed against the envelope protein of hepatitis B virus on the surface of infected cells (S-CAR). The S-CAR contains a human B cell-derived single-chain antibody fragment and human immunoglobulin G (IgG) spacer, CD28- and CD3-signaling domains that may be immunogenic in mice. Because immunosuppression will worsen the clinical course of chronic hepatitis B, we aimed at developing a preclinical mouse model that is immunocompetent and mimics chronic hepatitis B but nevertheless allows evaluating efficacy and safety of a fully human CAR. The S-CAR grafted on T cells triggered antibody responses in immunocompetent animals, and a co-expressed human-derived safeguard, the truncated epidermal growth factor receptor (EGFRt), even induced B and T cell responses, both limiting the survival of S-CAR-grafted T cells. Total body irradiation and transfer of T cells expressing an analogous, signaling-deficient S-CAR decoy and the safeguard induced immune tolerance toward the human-derived structures. S-CAR T cells transferred after immune recovery persisted and showed long-lasting antiviral effector function. The approach we describe herein will enable preclinical studies of efficacy and safety of fully human CARs in the context of a functional immune system.


Subject(s)
Hepatitis B/therapy , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/immunology , Viral Envelope Proteins/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Disease Models, Animal , Hepatitis B/genetics , Hepatitis B/immunology , Hepatitis B/virology , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Humans , Immunocompetence/drug effects , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Receptors, Chimeric Antigen/administration & dosage , Receptors, Chimeric Antigen/genetics , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Viral Envelope Proteins/antagonists & inhibitors
17.
Molecules ; 25(18)2020 Sep 07.
Article in English | MEDLINE | ID: mdl-32906689

ABSTRACT

Mosquito-borne Zika virus (ZIKV) is a Flavivirus that came under intense study from 2014 to 2016 for its well-known ability to cause congenital microcephaly in fetuses and neurological Guillain-Barré disease in adults. Substantial research on screening antiviral agents against ZIKV and preventing ZIKV infection are globally underway, but Food and Drug Administration (FDA)-approved treatments are not available yet. Compounds from Chinese medicinal herbs may offer an opportunity for potential therapies for anti-ZIKV infection. In this study, we evaluated the antiviral efficacy of harringtonine against ZIKV. Harringtonine possessed anti-ZIKV properties against the binding, entry, replication, and release stage through the virus life cycle. In addition, harringtonine have strong virucidal effects in ZIKV and exhibited prophylaxis antiviral ability prior ZIKV infection. The antiviral activity also observed in the treatment against Japanese encephalitis reporter virus (RP9-GFP strain). Overall, this study demonstrated that harringtonine would be a favorable potential candidate for the development of anti-ZIKV infection therapies.


Subject(s)
Antiviral Agents/pharmacology , Harringtonines/pharmacology , Zika Virus Infection/virology , Zika Virus/drug effects , Animals , Antiviral Agents/chemistry , Cells, Cultured , Chlorocebus aethiops , Harringtonines/chemistry , Humans , Models, Molecular , Molecular Conformation , Structure-Activity Relationship , Vero Cells , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/chemistry , Virus Attachment/drug effects , Virus Internalization/drug effects , Virus Release , Virus Replication/drug effects , Zika Virus/genetics , Zika Virus Infection/drug therapy
18.
Biochem Biophys Res Commun ; 519(2): 330-336, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31514997

ABSTRACT

Pseudorabies virus (PRV), the agent of pseudorabies, has raised considerable attention since 2011 due to the outbreak of emerging PRV variants in China. In the present study, we obtained two monoclonal antibodies (mAbs) known as 2E5 and 5C3 against the glycoprotein E (gE) of a PRV variant (JS-2012 strain). The two mAbs reacted with wild PRV but not the vaccine strain (gE-deleted virus). The 2E5 was located in 161RLRRE165, which was conserved in almost of all PRV strains, while 5C3 in 148EMGIGDY154 was different from almost of all genotype I PRV, in which the 149th amino acid is methionine (M) instead of arginine (R). The two epitopes peptides located in the hydrophilic region and reacted with positive sera against genotype II PRV (JS-2012), which suggests they were likely dominant B-cell epitopes. Furthermore, the mutant peptide 148ERGIGDY154 (genotype I) did not react with the mAb 5C3 or positive sera against genotype II PRV (JS-2012). In conclusion, both mAb 2E5 and 5C3 could be used to identify wild PRV strains from vaccine strains, and mAb 5C3 and the epitope peptide of 5C3 might be used for epidemiological investigation to distinguish genotype II from genotype I PRV.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes, B-Lymphocyte/immunology , Herpesvirus 1, Suid/chemistry , Viral Envelope Proteins/immunology , Animals , Cell Line , Chlorocebus aethiops , Herpesvirus 1, Suid/drug effects , Herpesvirus 1, Suid/immunology , Mice , Peptides/pharmacology , Swine , Vero Cells , Viral Envelope Proteins/antagonists & inhibitors
19.
J Virol ; 92(21)2018 11 01.
Article in English | MEDLINE | ID: mdl-30111563

ABSTRACT

Hepatitis C virus (HCV) infection is a major cause of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. HCV can be sensed by host innate immunity to induce expression of interferons (IFNs) and a number of antiviral effectors. In this study, we found HCV infection induced the expression of neuralized E3 ubiquitin protein ligase 3 (NEURL3), a putative E3 ligase, in a manner that requires the involvement of innate immune sensing but is independent of the IFN action. Furthermore, we showed that NEURL3 inhibited HCV infection while it had little effect on other RNA viruses, including Zika virus (ZIKV), dengue virus (DENV), and vesicular stomatitis virus (VSV). Mechanistic studies demonstrated that NEURL3 inhibited HCV assembly by directly binding HCV envelope glycoprotein E1 to interfere with the E1/E2 heterodimerization, an important prerequisite for virion morphogenesis. Finally, we showed that knockout of NEURL3 significantly enhanced HCV infection. In summary, we identified NEURL3 as a novel inducible antiviral host factor that suppresses HCV assembly. Our results not only shed new insight into how host innate immunity acts against HCV but also revealed a new important biological function for NEURL3.IMPORTANCE The exact biological function of NEURL3, a putative E3 ligase, remains largely unknown. In this study, we found that NEURL3 could be upregulated upon HCV infection in a manner dependent on pattern recognition receptor-mediated innate immune response. NEURL3 inhibits HCV assembly by directly binding viral E1 envelope glycoprotein to disrupt its interaction with E2, an action that requires its Neuralized homology repeat (NHR) domain but not the RING domain. Furthermore, we found that NEURL3 has a pangenotypic anti-HCV activity and interacts with E1 of genotypes 2a, 1b, 3a, and 6a but does not inhibit other closely related RNA viruses, such as ZIKV, DENV, and VSV. To our knowledge, our study is the first report to demonstrate that NEURL3 functions as an antiviral host factor. Our results not only shed new insight into how host innate immunity acts against HCV, but also revealed a new important biological function for NEURL3.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis C/prevention & control , Immunity, Innate/immunology , RNA Virus Infections/virology , Ubiquitin-Protein Ligases/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Dengue Virus/drug effects , HEK293 Cells , Hepacivirus/classification , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/virology , Humans , RNA Virus Infections/drug therapy , RNA Virus Infections/immunology , RNA Viruses/immunology , Vesicular stomatitis Indiana virus/drug effects , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virus Assembly , Zika Virus/drug effects
20.
J Neurovirol ; 25(4): 457-463, 2019 08.
Article in English | MEDLINE | ID: mdl-31140131

ABSTRACT

Varicella-zoster virus (VZV) leads to chicken pox on primary infection and herpes zoster on reactivation. Recent studies suggest that microRNA2911 (MIR2911), honeysuckle (HS)-encoded atypical microRNA, has potential as a therapeutic agent against influenza and EV71 virus infections. Here, we report that MIR2911 directly inhibits VZV replication by targeting the IE62 gene. The luciferase reporter assay and bioinformatics prediction revealed that MIR2911 could target the IE62 gene of VZV. The VZV-encoded IE62 protein expression was inhibited significantly by synthetic MIR2911, while the expression of the mutants, whose MIR2911-binding sites were modified, was not inhibited. The RNA extracted from HS decoction and synthetic MIR2911 considerably suppressed VZV infection. However, it did not influence viral replication of a mutant virus with alterations in the nucleotide sequences of IE62. At the same time, the RNA extracted from HS decoction treated with the anti-MIR2911 antagomir could not inhibit the VZV replication, demonstrating that VZV replication was specifically and sufficiently inhibited by MIR2911. These results indicated that, by targeting the IE62 gene, MIR2911 may effectively inhibit VZV replication. Our results also suggest a potential novel strategy for the treatment and prevention of diseases caused by VZV infection.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 3, Human/drug effects , Immediate-Early Proteins/genetics , Lonicera/chemistry , MicroRNAs/genetics , RNA, Plant/genetics , Trans-Activators/genetics , Viral Envelope Proteins/genetics , Antagomirs/genetics , Antagomirs/metabolism , Antiviral Agents/isolation & purification , Antiviral Agents/metabolism , Cell Line , Drugs, Chinese Herbal/chemistry , Embryo, Mammalian , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression Regulation , Genes, Reporter , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/metabolism , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Mutation , RNA, Plant/antagonists & inhibitors , RNA, Plant/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL