Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
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
2.
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
3.
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
4.
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.].

5.
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
6.
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
7.
Proc Natl Acad Sci U S A ; 112(32): E4354-63, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26195743

ABSTRACT

Recent advances in biosensing technologies present great potential for medical diagnostics, thus improving clinical decisions. However, creating a label-free general sensing platform capable of detecting multiple biotargets in various clinical specimens over a wide dynamic range, without lengthy sample-processing steps, remains a considerable challenge. In practice, these barriers prevent broad applications in clinics and at patients' homes. Here, we demonstrate the nanoplasmonic electrical field-enhanced resonating device (NE(2)RD), which addresses all these impediments on a single platform. The NE(2)RD employs an immunodetection assay to capture biotargets, and precisely measures spectral color changes by their wavelength and extinction intensity shifts in nanoparticles without prior sample labeling or preprocessing. We present through multiple examples, a label-free, quantitative, portable, multitarget platform by rapidly detecting various protein biomarkers, drugs, protein allergens, bacteria, eukaryotic cells, and distinct viruses. The linear dynamic range of NE(2)RD is five orders of magnitude broader than ELISA, with a sensitivity down to 400 fg/mL This range and sensitivity are achieved by self-assembling gold nanoparticles to generate hot spots on a 3D-oriented substrate for ultrasensitive measurements. We demonstrate that this precise platform handles multiple clinical samples such as whole blood, serum, and saliva without sample preprocessing under diverse conditions of temperature, pH, and ionic strength. The NE(2)RD's broad dynamic range, detection limit, and portability integrated with a disposable fluidic chip have broad applications, potentially enabling the transition toward precision medicine at the point-of-care or primary care settings and at patients' homes.


Subject(s)
Biosensing Techniques/instrumentation , Diagnostic Techniques and Procedures/instrumentation , Electricity , Nanostructures/chemistry , Cell Line, Tumor , Coinfection/diagnosis , Environment , Enzyme-Linked Immunosorbent Assay , Equipment Design , Humans , Hydrogen-Ion Concentration , Limit of Detection , Microfluidics , Osmolar Concentration , Reproducibility of Results , Temperature
8.
J Virol ; 90(9): 4494-4510, 2016 May.
Article in English | MEDLINE | ID: mdl-26912630

ABSTRACT

UNLABELLED: Virus entry into cells is a multistep process that often requires the subversion of subcellular machineries. A more complete understanding of these steps is necessary to develop new antiviral strategies. While studying the potential role of the actin network and one of its master regulators, the small GTPase Cdc42, during Junin virus (JUNV) entry, we serendipitously uncovered the small molecule ZCL278, reported to inhibit Cdc42 function as an entry inhibitor for JUNV and for vesicular stomatitis virus, lymphocytic choriomeningitis virus, and dengue virus but not for the nonenveloped poliovirus. Although ZCL278 did not interfere with JUNV attachment to the cell surface or virus particle internalization into host cells, it prevented the release of JUNV ribonucleoprotein cores into the cytosol and decreased pH-mediated viral fusion with host membranes. We also identified SVG-A astroglial cell-derived cells to be highly permissive for JUNV infection and generated new cell lines expressing fluorescently tagged Rab5c or Rab7a or lacking Cdc42 using clustered regularly interspaced short palindromic repeat (CRISPR)-caspase 9 (Cas9) gene-editing strategies. Aided by these tools, we uncovered that perturbations in the actin cytoskeleton or Cdc42 activity minimally affect JUNV entry, suggesting that the inhibitory effect of ZCL278 is not mediated by ZCL278 interfering with the activity of Cdc42. Instead, ZCL278 appears to redistribute viral particles from endosomal to lysosomal compartments. ZCL278 also inhibited JUNV replication in a mouse model, and no toxicity was detected. Together, our data suggest the unexpected antiviral activity of ZCL278 and highlight its potential for use in the development of valuable new tools to study the intracellular trafficking of pathogens. IMPORTANCE: The Junin virus is responsible for outbreaks of Argentine hemorrhagic fever in South America, where 5 million people are at risk. Limited options are currently available to treat infections by Junin virus or other viruses of the Arenaviridae, making the identification of additional tools, including small-molecule inhibitors, of great importance. How Junin virus enters cells is not yet fully understood. Here we describe new cell culture models in which the cells are susceptible to Junin virus infection and to which we applied CRISPR-Cas9 genome engineering strategies to help characterize early steps during virus entry. We also uncovered ZCL278 to be a new antiviral small molecule that potently inhibits the cellular entry of the Junin virus and other enveloped viruses. Moreover, we show that ZCL278 also functions in vivo, thereby preventing Junin virus replication in a mouse model, opening the possibility for the discovery of ZCL278 derivatives of therapeutic potential.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Drug Discovery , Thiourea/analogs & derivatives , Virus Internalization/drug effects , Actins/metabolism , Animals , Cell Line , Cells, Cultured , Clathrin/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/virology , Gene Knockout Techniques , Hemorrhagic Fever, American/genetics , Hemorrhagic Fever, American/metabolism , Hemorrhagic Fever, American/virology , Humans , Junin virus/drug effects , Junin virus/physiology , Mice , Protein Binding , Protein Transport , Proteolysis , Ribonucleoproteins/metabolism , Thiourea/pharmacology , Viral Load , Viral Proteins/metabolism , Virus Attachment/drug effects , Virus Replication/drug effects , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
9.
Antimicrob Agents Chemother ; 59(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25313218

ABSTRACT

Dengue virus (DENV), a member of the Flaviviridae family, is a mosquito-borne pathogen and the cause of dengue fever. The increasing prevalence of DENV worldwide heightens the need for an effective vaccine and specific antivirals. Due to the dependence of DENV upon the lipid biosynthetic machinery of the host cell, lipid signaling and metabolism present unique opportunities for inhibiting viral replication. We screened a library of bioactive lipids and modulators of lipid metabolism and identified 4-hydroxyphenyl retinamide (4-HPR) (fenretinide) as an inhibitor of DENV in cell culture. 4-HPR inhibits the steady-state accumulation of viral genomic RNA and reduces viremia when orally administered in a murine model of DENV infection. The molecular target responsible for this antiviral activity is distinct from other known inhibitors of DENV but appears to affect other members of the Flaviviridae, including the West Nile, Modoc, and hepatitis C viruses. Although long-chain ceramides have been implicated in DENV replication, we demonstrate that DENV is insensitive to the perturbation of long-chain ceramides in mammalian cell culture and that the effect of 4-HPR on dihydroceramide homeostasis is separable from its antiviral activity. Likewise, the induction of reactive oxygen species by 4-HPR is not required for the inhibition of DENV. The inhibition of DENV in vivo by 4-HPR, combined with its well-established safety and tolerability in humans, suggests that it may be repurposed as a pan-Flaviviridae antiviral agent. This work also illustrates the utility of bioactive lipid screens for identifying critical interactions of DENV and other viral pathogens with host lipid biosynthesis, metabolism, and signal transduction.


Subject(s)
Dengue Virus/growth & development , Dengue/drug therapy , Fenretinide/therapeutic use , Viremia/drug therapy , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Female , HEK293 Cells , Hepacivirus/growth & development , Humans , Mice , Mice, Transgenic , Reactive Oxygen Species/metabolism , Vero Cells , West Nile virus/growth & development
10.
Nature ; 458(7241): 1047-50, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19396146

ABSTRACT

Dengue fever is the most frequent arthropod-borne viral disease of humans, with almost half of the world's population at risk of infection. The high prevalence, lack of an effective vaccine, and absence of specific treatment conspire to make dengue fever a global public health threat. Given their compact genomes, dengue viruses (DENV-1-4) and other flaviviruses probably require an extensive number of host factors; however, only a limited number of human, and an even smaller number of insect host factors, have been identified. Here we identify insect host factors required for DENV-2 propagation, by carrying out a genome-wide RNA interference screen in Drosophila melanogaster cells using a well-established 22,632 double-stranded RNA library. This screen identified 116 candidate dengue virus host factors (DVHFs). Although some were previously associated with flaviviruses (for example, V-ATPases and alpha-glucosidases), most of the DVHFs were newly implicated in dengue virus propagation. The dipteran DVHFs had 82 readily recognizable human homologues and, using a targeted short-interfering-RNA screen, we showed that 42 of these are human DVHFs. This indicates notable conservation of required factors between dipteran and human hosts. This work suggests new approaches to control infection in the insect vector and the mammalian host.


Subject(s)
Conserved Sequence/genetics , Dengue Virus/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/virology , Host-Pathogen Interactions/genetics , Insect Vectors/genetics , Insect Vectors/physiology , Aedes/genetics , Aedes/virology , Animals , Cell Line , Conserved Sequence/physiology , Drosophila melanogaster/physiology , Gene Knockdown Techniques , Genome, Insect/genetics , Humans , RNA Interference , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Virus Replication
11.
Chembiochem ; 15(9): 1317-24, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24828915

ABSTRACT

Many biological experiments are not compatible with the use of immunofluorescence, genetically encoded fluorescent tags, or FRET-based reporters. Conjugation of existing kinase inhibitors to cell-permeable fluorophores can provide a generalized approach to develop fluorescent probes of intracellular kinases. Here, we report the development of a small molecule probe of Src through conjugation of BODIPY to two well-established dual Src-Abl kinase inhibitors, dasatinib and saracatinib. We show that this approach is not successful for saracatinib but that dasatinib-BODIPY largely retains the biological activity of its parent compound and can be used to monitor the presence of Src kinase in individual cells by flow cytometry. It can also be used to track the localization of Src by fixed and live-cell fluorescence microscopy. This strategy could enable generation of additional kinase-specific probes useful in systems not amenable to genetic manipulation or could be used together with fluorescent proteins to enable a multiplexed assay readout.


Subject(s)
Boron Compounds/chemistry , Fluorescence , Molecular Probes/chemistry , src-Family Kinases/chemistry , Animals , Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Boron Compounds/metabolism , Boron Compounds/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Dasatinib , Dose-Response Relationship, Drug , Humans , Mice , Microscopy, Fluorescence , Molecular Probes/metabolism , Molecular Structure , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
12.
J Virol ; 87(13): 7367-81, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23616652

ABSTRACT

In this study, we characterized the antiviral mechanism of action of AZD0530 and dasatinib, two pharmacological inhibitors of host kinases, that also inhibit dengue virus (DV) infection. Using Northern blot and reporter replicon assays, we demonstrated that both small molecules inhibit the DV2 infectious cycle at the step of steady-state RNA replication. In order to identify the cellular target of AZD0530 and dasatinib mediating this anti-DV2 activity, we examined the effects of RNA interference (RNAi)-mediated depletion of the major kinases known to be inhibited by these small molecules. We determined that Fyn kinase, a target of both AZD0530 and dasatinib, is involved in DV2 RNA replication and is probably a major mediator of the anti-DV activity of these compounds. Furthermore, serial passaging of DV2 in the presence of dasatinib led to the identification of a mutation in the transmembrane domain 3 of the NS4B protein that overcomes the inhibition of RNA replication by AZD0530, dasatinib, and Fyn RNAi. Although we observed that dasatinib also inhibits DV2 particle assembly and/or secretion, this activity does not appear to be mediated by Src-family kinases. Together, our results suggest that AZD0530 and dasatinib inhibit DV at the step of viral RNA replication and demonstrate a critical role for Fyn kinase in this viral process. The antiviral activity of these compounds in vitro makes them useful pharmacological tools to validate Fyn or other host kinases as anti-DV targets in vivo.


Subject(s)
Benzodioxoles/pharmacology , Dengue Virus/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/metabolism , Pyrimidines/pharmacology , Quinazolines/pharmacology , RNA, Viral/biosynthesis , Thiazoles/pharmacology , Virus Replication/drug effects , Antibodies, Monoclonal/immunology , Blotting, Northern , Blotting, Western , Cell Survival/drug effects , DNA Primers/genetics , Dasatinib , Dengue Virus/genetics , Dimethyl Sulfoxide , HEK293 Cells , Humans , Mycophenolic Acid , Polymerase Chain Reaction , RNA Interference , Replicon/genetics , Virus Replication/physiology
13.
PLoS Pathog ; 8(4): e1002627, 2012.
Article in English | MEDLINE | ID: mdl-22496653

ABSTRACT

Flavivirus envelope protein (E) mediates membrane fusion and viral entry from endosomes. A low-pH induced, dimer-to-trimer rearrangement and reconfiguration of the membrane-proximal "stem" of the E ectodomain draw together the viral and cellular membranes. We found stem-derived peptides from dengue virus (DV) bind stem-less E trimer and mimic the stem-reconfiguration step in the fusion pathway. We adapted this experiment as a high-throughput screen for small molecules that block peptide binding and thus may inhibit viral entry. A compound identified in this screen, 1662G07, and a number of its analogs reversibly inhibit DV infectivity. They do so by binding the prefusion, dimeric E on the virion surface, before adsorption to a cell. They also block viral fusion with liposomes. Structure-activity relationship studies have led to analogs with submicromolar IC90s against DV2, and certain analogs are active against DV serotypes 1,2, and 4. The compounds do not inhibit the closely related Kunjin virus. We propose that they bind in a previously identified, E-protein pocket, exposed on the virion surface and although this pocket is closed in the postfusion trimer, its mouth is fully accessible. Examination of the E-trimer coordinates (PDB 1OK8) shows that conformational fluctuations around the hinge could open the pocket without dissociating the trimer or otherwise generating molecular collisions. We propose that compounds such as 1662G07 trap the sE trimer in a "pocket-open" state, which has lost affinity for the stem peptide and cannot support the final "zipping up" of the stem.


Subject(s)
Antiviral Agents/pharmacology , Biomimetic Materials/pharmacology , Dengue Virus/metabolism , Peptides/pharmacology , Viral Envelope Proteins/pharmacology , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cell Line , Cricetinae , Dengue Virus/genetics , Humans , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virion/genetics , Virion/metabolism
14.
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
15.
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.

16.
J Virol ; 86(13): 7072-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532681

ABSTRACT

The dengue virus (DV) envelope (E) protein is important in mediating viral entry and assembly of progeny virus during cellular infection. Domains I and III (DI and DIII, respectively) of the DV E protein are connected by a highly conserved but poorly ordered region, the DI/DIII linker. Although the flexibility of the DI/DIII linker is thought to be important for accommodating the structural rearrangements undergone by the E protein during viral entry, the function of the linker in the DV infectious cycle is not well understood. In this study, we performed site-directed mutagenesis on conserved residues in the DI/DIII linker of the DV2 E protein and showed that the resulting mutations had little or no effect on the entry process but greatly affected virus assembly. Biochemical fractionation and immunofluorescence microscopy experiments performed on infectious virus as well as in a virus-like particle (VLP) system indicate that the DI/DIII linker mutants express the DV structural proteins at the sites of particle assembly near the ER but fail to form infectious particles. This defect is not due to disruption of E's interaction with prM and pr in immature and mature virions, respectively. Serial passaging of the DV2 mutant E-Y299F led to the identification of a mutation in the membrane-proximal stem region of E that fully compensates for the assembly defect of this DI/DIII linker mutant. Together, our results suggest a critical and previously unidentified role for the E protein DI/DIII linker region during the DV2 assembly process.


Subject(s)
Dengue Virus/physiology , Mutagenesis, Site-Directed , Viral Envelope Proteins/metabolism , Virus Assembly , Amino Acid Sequence , Cell Line , Dengue Virus/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation , Protein Structure, Tertiary , Viral Envelope Proteins/genetics , Virus Internalization
17.
Proc Natl Acad Sci U S A ; 107(2): 798-802, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20080755

ABSTRACT

To better define the mechanism(s) likely responsible for viral clearance during hepatitis B virus (HBV) infection, viral clearance was studied in a panel of immunodeficient mouse strains that were hydrodynamically transfected with a plasmid containing a replication-competent copy of the HBV genome. Neither B cells nor perforin were required to clear the viral DNA transcriptional template from the liver. In contrast, the template persisted for at least 60 days at high levels in NOD/Scid mice and at lower levels in the absence of CD4(+) and CD8(+) T cells, NK cells, Fas, IFN-gamma (IFN-gamma), IFN-alpha/beta receptor (IFN-alpha/betaR1), and TNF receptor 1 (TNFR1), indicating that each of these effectors was required to eliminate the transcriptional template from the liver. Interestingly, viral replication was ultimately terminated in all lineages except the NOD/Scid mice, suggesting the existence of redundant pathways that inhibit HBV replication. Finally, induction of a CD8(+) T cell response in these animals depended on the presence of CD4(+) T cells. These results are consistent with a model in which CD4(+) T cells serve as master regulators of the adaptive immune response to HBV; CD8(+) T cells are the key cellular effectors mediating HBV clearance from the liver, apparently by a Fas-dependent, perforin-independent process in which NK cells, IFN-gamma, TNFR1, and IFN-alpha/betaR play supporting roles. These results provide insight into the complexity of the systems involved in HBV clearance, and they suggest unique directions for analysis of the mechanism(s) responsible for HBV persistence.


Subject(s)
Hepatitis B virus/genetics , Hepatitis B/immunology , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/epidemiology , DNA, Viral/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Viral , Genome, Viral , Hepatitis B/complications , Hepatitis B/epidemiology , Hepatitis B/genetics , Hepatitis B Core Antigens/genetics , Hepatitis B Surface Antigens/blood , Hepatitis B virus/immunology , Humans , Liver Neoplasms/epidemiology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Transcription, Genetic , Virus Replication
18.
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
19.
J Am Chem Soc ; 134(16): 6896-9, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22480142

ABSTRACT

Hepatitis C virus (HCV) infection has been clinically associated with serum lipid abnormalities, yet our understanding of the effects of HCV on host lipid metabolism and conversely the function of individual lipids in HCV replication remains incomplete. Using liquid chromatography-mass spectrometry metabolite profiling of the HCV JFH1 cell culture infection model, we identified a significant steady-state accumulation of desmosterol, an immediate precursor to cholesterol. Pharmacological inhibition or RNAi-mediated depletion of DHCR7 significantly reduced steady-state HCV protein expression and viral genomic RNA. Moreover, this effect was reversed when cultures were supplemented with exogenous desmosterol. Together, these observations suggest an intimate connection between HCV replication and desmosterol homeostasis and that the enzymes responsible for synthesis of desmosterol may be novel targets for antiviral design.


Subject(s)
Antiviral Agents/pharmacology , Desmosterol/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Lipid Metabolism/drug effects , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Cells, Cultured , Desmosterol/chemistry , Desmosterol/metabolism , Hepacivirus/metabolism , Hepatitis C/metabolism , Humans , Virus Replication/drug effects
20.
PLoS Pathog ; 6(4): e1000851, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20386713

ABSTRACT

The mechanism of membrane fusion by "class II" viral fusion proteins follows a pathway that involves large-scale domain rearrangements of the envelope glycoprotein (E) and a transition from dimers to trimers. The rearrangement is believed to proceed by an outward rotation of the E ectodomain after loss of the dimer interface, followed by a reassociation into extended trimers. The approximately 55-aa-residue, membrane proximal "stem" can then zip up along domain II, bringing together the transmembrane segments of the C-terminus and the fusion loops at the tip of domain II. We find that peptides derived from the stem of dengue-virus E bind stem-less E trimer, which models a conformational intermediate. In vitro assays demonstrate that these peptides specifically block viral fusion. The peptides inhibit infectivity with potency proportional to their affinity for the conformational intermediate, even when free peptide is removed from a preincubated inoculum before infecting cells. We conclude that peptides bind virions before attachment and are carried with virions into endosomes, the compartment in which acidification initiates fusion. Binding depends on particle dynamics, as there is no inhibition of infectivity if preincubation and separation are at 4 degrees C rather than 37 degrees C. We propose a two-step model for the mechanism of fusion inhibition. Targeting a viral entry pathway can be an effective way to block infection. Our data, which support and extend proposed mechanisms for how the E conformational change promotes membrane fusion, suggest strategies for inhibiting flavivirus entry.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/pathogenicity , Peptides/pharmacology , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Animals , Antiviral Agents/metabolism , Dengue Virus/chemistry , Dengue Virus/metabolism , Peptides/metabolism , Viral Envelope Proteins/chemistry , Virion/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL