Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
1.
Parasit Vectors ; 17(1): 255, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38863029

ABSTRACT

BACKGROUND: RNA interference (RNAi) is a target-specific gene silencing method that can be used to determine gene functions and investigate host-pathogen interactions, as well as facilitating the development of ecofriendly pesticides. Commercially available transfection reagents (TRs) can improve the efficacy of RNAi. However, we currently lack a product and protocol for the transfection of insect cell lines with long double-stranded RNA (dsRNA). METHODS: We used agarose gel electrophoresis to determine the capacity of eight TRs to form complexes with long dsRNA. A CellTiter-Glo assay was then used to assess the cytotoxicity of the resulting lipoplexes. We also measured the cellular uptake of dsRNA by fluorescence microscopy using the fluorophore Cy3 as a label. Finally, we analyzed the TRs based on their transfection efficacy and compared the RNAi responses of Aedes albopictus C6/36 and U4.4 cells by knocking down an mCherry reporter Semliki Forest virus in both cell lines. RESULTS: The TRs from Biontex (K4, Metafectene Pro, and Metafectene SI+) showed the best complexing capacity and the lowest dsRNA:TR ratio needed for complete complex formation. Only HiPerFect was unable to complex the dsRNA completely, even at a ratio of 1:9. Most of the complexes containing mCherry-dsRNA were nontoxic at 2 ng/µL, but Lipofectamine 2000 was toxic at 1 ng/µL in U4.4 cells and at 2 ng/µL in C6/36 cells. The transfection of U4.4 cells with mCherry-dsRNA/TR complexes achieved significant knockdown of the virus reporter. Comparison of the RNAi response in C6/36 and U4.4 cells suggested that C6/36 cells lack the antiviral RNAi response because there was no significant knockdown of the virus reporter in any of the treatments. CONCLUSIONS: C6/36 cells have an impaired RNAi response as previously reported. This investigation provides valuable information for future RNAi experiments by showing how to mitigate the adverse effects attributed to TRs. This will facilitate the judicious selection of TRs and transfection conditions conducive to RNAi research in mosquitoes.


Subject(s)
Aedes , RNA Interference , RNA, Double-Stranded , Transfection , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Animals , Cell Line , Aedes/genetics , Gene Silencing , Semliki forest virus/genetics , Semliki forest virus/drug effects
2.
Antiviral Res ; 197: 105223, 2022 01.
Article in English | MEDLINE | ID: mdl-34856248

ABSTRACT

Repurposing drugs is a promising strategy to identify therapeutic interventions against novel and re-emerging viruses. Posaconazole is an antifungal drug used to treat invasive aspergillosis and candidiasis. Recently, posaconazole and its structural analog, itraconazole were shown to inhibit replication of multiple viruses by modifying intracellular cholesterol homeostasis. Here, we show that posaconazole inhibits replication of the alphaviruses Semliki Forest virus (SFV), Sindbis virus and chikungunya virus with EC50 values ranging from 1.4 µM to 9.5 µM. Posaconazole treatment led to a significant reduction of virus entry in an assay using a temperature-sensitive SFV mutant, but time-of-addition and RNA transfection assays indicated that posaconazole also inhibits post-entry stages of the viral replication cycle. Virus replication in the presence of posaconazole was partially rescued by the addition of exogenous cholesterol. A transferrin uptake assay revealed that posaconazole considerably slowed down cellular endocytosis. A single point mutation in the SFV E2 glycoprotein, H255R, provided partial resistance to posaconazole as well as to methyl-ß-cyclodextrin, corroborating the effect of posaconazole on cholesterol and viral entry. Our results indicate that posaconazole inhibits multiple steps of the alphavirus replication cycle and broaden the spectrum of viruses that can be targeted in vitro by posaconazole, which could be further explored as a therapeutic agent against emerging viruses.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Drug Repositioning/methods , Triazoles/pharmacology , Virus Replication/drug effects , Alphavirus/classification , Animals , Cell Line , Chikungunya virus/drug effects , Chlorocebus aethiops , Cricetinae , Endocytosis/drug effects , Humans , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Vero Cells , Virus Internalization/drug effects
3.
PLoS Pathog ; 17(5): e1009549, 2021 05.
Article in English | MEDLINE | ID: mdl-33984068

ABSTRACT

The antiviral innate immune response mainly involves type I interferon (IFN) in mammalian cells. The contribution of the RNA silencing machinery remains to be established, but several recent studies indicate that the ribonuclease DICER can generate viral siRNAs in specific conditions. It has also been proposed that type I IFN and RNA silencing could be mutually exclusive antiviral responses. In order to decipher the implication of DICER during infection of human cells with alphaviruses such as the Sindbis virus and Semliki forest virus, we determined its interactome by proteomics analysis. We show that DICER specifically interacts with several double-stranded RNA binding proteins and RNA helicases during viral infection. In particular, proteins such as DHX9, ADAR-1 and the protein kinase RNA-activated (PKR) are enriched with DICER in virus-infected cells. We demonstrate that the helicase domain of DICER is essential for this interaction and that its deletion confers antiviral properties to this protein in an RNAi-independent, PKR-dependent, manner.


Subject(s)
Alphavirus Infections/drug therapy , Antiviral Agents/pharmacology , DEAD-box RNA Helicases/metabolism , Protein Interaction Domains and Motifs/drug effects , Ribonuclease III/metabolism , Semliki forest virus/drug effects , Virus Replication , eIF-2 Kinase/metabolism , Alphavirus Infections/metabolism , Alphavirus Infections/pathology , DEAD-box RNA Helicases/genetics , HEK293 Cells , Humans , Interferon Type I/pharmacology , Ribonuclease III/genetics , eIF-2 Kinase/genetics
4.
Article in English | MEDLINE | ID: mdl-31964798

ABSTRACT

Alphaviruses are arthropod-borne, positive-stranded RNA viruses capable of causing severe disease with high morbidity. Chikungunya virus (CHIKV) is an alphavirus that causes a febrile illness which can progress into chronic arthralgia. The current lack of vaccines and specific treatment for CHIKV infection underscores the need to develop new therapeutic interventions. To discover new antiviral agents, we performed a compound screen in cell culture-based infection models and identified two carbocyclic adenosine analogues, 6'-ß-fluoro-homoaristeromycin (FHA) and 6'-fluoro-homoneplanocin A (FHNA), that displayed potent activity against CHIKV and Semliki Forest virus (SFV) with 50% effective concentrations in the nanomolar range at nontoxic concentrations. The compounds, designed as inhibitors of the host enzyme S-adenosylhomocysteine (SAH) hydrolase, impeded postentry steps in CHIKV and SFV replication. Selection of FHNA-resistant mutants and reverse genetics studies demonstrated that the combination of mutations G230R and K299E in CHIKV nonstructural protein 1 (nsP1) conferred resistance to the compounds. Enzymatic assays with purified wild-type (wt) SFV nsP1 suggested that an oxidized (3'-keto) form, rather than FHNA itself, directly inhibited the MTase activity, while a mutant protein with the K231R and K299E substitutions was insensitive to the compound. Both wt nsP1 and the resistant mutant were equally sensitive to the inhibitory effect of SAH. Our combined data suggest that FHA and FHNA inhibit CHIKV and SFV replication by directly targeting the MTase activity of nsP1, rather than through an indirect effect on host SAH hydrolase. The high potency and selectivity of these novel alphavirus mRNA capping inhibitors warrant further preclinical investigation of these compounds.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Chikungunya virus/physiology , Adenosine/pharmacology , Animals , Chikungunya virus/pathogenicity , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Guanosine Monophosphate/metabolism , Mutation , Phosphorus Radioisotopes , Semliki forest virus/drug effects , Vero Cells , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
5.
Antiviral Res ; 159: 134-142, 2018 11.
Article in English | MEDLINE | ID: mdl-30300716

ABSTRACT

The chikungunya virus (CHIKV) is a mosquito-borne virus that belongs to the genus Alphavirus, family Togaviridae. It is the cause of chikungunya fever in humans, which presents a serious global threat due to its high rate of contagion. The clinical symptoms of CHIKV include fever and persistent, severe arthritis. Micafungin has broad-spectrum fungicidal activity against Candida spp. is a promising echinocandin that was recently approved by the U.S. Food and Drug Administration (FDA) and has demonstrated activity against Candida and Aspergillus. Recent studies have demonstrated the antiviral activity of micafungin; however, the inhibitory effects against CHIKV have yet to be investigated. Our objectives in this study were to explore the antiviral effects of micafungin on CHIKV infection and to elucidate the potential molecular mechanisms of inhibition. We determined that micafungin has the ability to counter CHIKV-induced cytopathic effects. We further discovered that micafungin limits virus replication, release, cell-to-cell transmission, and also slightly affected virus stability during high doses treatment. The efficacy of micafungin was further confirmed against two clinical isolates of CHIKV and two alphaviruses: Sindbis virus (SINV) and Semliki Forest virus (SFV). Our findings suggest that micafungin has considerable potential as a novel inhibitor against the viral replication, and intracellular and extracellular transmission of CHIKV, and has a little effect on virus stability. Our findings also suggest that micafungin could have curative effects on other alphavirus infections.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Micafungin/pharmacology , Alphavirus Infections/drug therapy , Chikungunya Fever/drug therapy , Chikungunya Fever/virology , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Virus Replication/drug effects
6.
Vet J ; 230: 62-64, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29102599

ABSTRACT

The recent outbreak of infection with Zika virus (ZIKV; Flaviviridae) has attracted attention to this previously neglected mosquito-borne pathogen and the need for efficient therapies. Since flavivirus replication is generally known to be dependent on fatty acid biosynthesis, two inhibitors of this pathway, 5-(tetradecyloxyl)-2-furoic acid (TOFA) and cerulenin, were tested for their potentiality to inhibit virus replication. At concentrations previously shown to inhibit the replication of other flaviviruses, neither drug had a significant antiviral affect against ZIKV, but reduced the replication of the non-related mosquito-borne Semliki Forest virus (Togaviridae).


Subject(s)
Antiviral Agents/pharmacology , Cerulenin/pharmacology , Fatty Acid Synthesis Inhibitors/pharmacology , Furans/pharmacology , Hypolipidemic Agents/pharmacology , Semliki forest virus/drug effects , Zika Virus/drug effects , A549 Cells/virology , Dose-Response Relationship, Drug , Humans , Virus Replication/drug effects
7.
ChemMedChem ; 12(8): 613-620, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28334511

ABSTRACT

Novel elongated and shortened derivatives of the peptidomimetic furin inhibitor phenylacetyl-Arg-Val-Arg-4-amidinobenzylamide were synthesized. The most potent compounds, such as Nα (carbamidoyl)Arg-Arg-Val-Arg-4-amidinobenzylamide (Ki =6.2 pm), contain additional basic residues at the N terminus and inhibit furin in the low-picomolar range. Furthermore, to decrease the molecular weight of this inhibitor type, compounds that lack the P5 moiety were prepared. The best inhibitors of this series, 5-(guanidino)valeroyl-Val-Arg-4-amidinobenzylamide and its P3 tert-leucine analogue displayed Ki values of 2.50 and 1.26 nm, respectively. Selected inhibitors, together with our previously described 4-amidinobenzylamide derivatives as references, were tested in cell culture for their activity against furin-dependent infectious pathogens. The propagation of the alphaviruses Semliki Forest virus and chikungunya virus was strongly inhibited in the presence of selected derivatives. Moreover, a significant protective effect of the inhibitors against diphtheria toxin was observed. These results confirm that the inhibition of furin should be a promising approach for the short-term treatment of acute infectious diseases.


Subject(s)
Benzamides/pharmacology , Furin/antagonists & inhibitors , Oligopeptides/pharmacology , Peptidomimetics/pharmacology , Serine Proteinase Inhibitors/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Benzamides/chemical synthesis , Cell Line , Chikungunya virus/drug effects , Chlorocebus aethiops , Cricetinae , Diphtheria Toxin/metabolism , Furin/metabolism , Oligopeptides/chemical synthesis , Peptidomimetics/chemical synthesis , Semliki forest virus/drug effects , Serine Proteinase Inhibitors/chemical synthesis
8.
Article in English | MEDLINE | ID: mdl-27993855

ABSTRACT

As new pathogenic viruses continue to emerge, it is paramount to have intervention strategies that target a common denominator in these pathogens. The fusion of viral and cellular membranes during viral entry is one such process that is used by many pathogenic viruses, including chikungunya virus, West Nile virus, and influenza virus. Obatoclax, a small-molecule antagonist of the Bcl-2 family of proteins, was previously determined to have activity against influenza A virus and also Sindbis virus. Here, we report it to be active against alphaviruses, like chikungunya virus (50% effective concentration [EC50] = 0.03 µM) and Semliki Forest virus (SFV; EC50 = 0.11 µM). Obatoclax inhibited viral entry processes in an SFV temperature-sensitive mutant entry assay. A neutral red retention assay revealed that obatoclax induces the rapid neutralization of the acidic environment of endolysosomal vesicles and thereby most likely inhibits viral fusion. Characterization of escape mutants revealed that the L369I mutation in the SFV E1 fusion protein was sufficient to confer partial resistance against obatoclax. Other inhibitors that target the Bcl-2 family of antiapoptotic proteins inhibited neither viral entry nor endolysosomal acidification, suggesting that the antiviral mechanism of obatoclax does not depend on its anticancer targets. Obatoclax inhibited the growth of flaviviruses, like Zika virus, West Nile virus, and yellow fever virus, which require low pH for fusion, but not that of pH-independent picornaviruses, like coxsackievirus A9, echovirus 6, and echovirus 7. In conclusion, obatoclax is a novel inhibitor of endosomal acidification that prevents viral fusion and that could be pursued as a potential broad-spectrum antiviral candidate.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Endosomes/drug effects , Lysosomes/drug effects , Membrane Fusion/drug effects , Pyrroles/pharmacology , Semliki forest virus/drug effects , Animals , Cell Line , Cell Membrane/drug effects , Cell Membrane/virology , Chikungunya virus/genetics , Chikungunya virus/growth & development , Cricetinae , Drug Resistance, Viral/genetics , Endosomes/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , Gene Expression , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Hydrogen-Ion Concentration/drug effects , Indoles , Lysosomes/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mutation , Neutral Red/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Semliki forest virus/genetics , Semliki forest virus/growth & development , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects , West Nile virus/drug effects , West Nile virus/genetics , West Nile virus/growth & development , Yellow fever virus/drug effects , Yellow fever virus/genetics , Yellow fever virus/growth & development , Zika Virus/drug effects , Zika Virus/genetics , Zika Virus/growth & development
9.
Antiviral Res ; 135: 81-90, 2016 11.
Article in English | MEDLINE | ID: mdl-27742486

ABSTRACT

Chikungunya disease results from an infection with the arbovirus, chikungunya virus (CHIKV). Symptoms of CHIKV include fever and persistent, severe arthritis. In recent years, several antiviral drugs have been evaluated in clinical trials; however, no registered antivirals have been approved for clinical therapy. In this study, we established a high-throughput screening (HTS) system based on CHIKV 26S mediated insect cell fusion inhibition assay. Our screening system was able to search potential anti-CHIKV drugs in vitro. Using this system, four compounds (niclosamide, nitazoxanide, niflumic acid, tolfenamic acid) were identified. These compounds were then further analyzed using a microneutralization assay. We determined that niclosamide and nitazoxanide exhibit ability to against CHIKV-induced CPE. The anti-CHIKV abilities of these compounds were further confirmed by RT-qPCR and IFA. Moreover, niclosamide and nitazoxanide were found to (1) limit virus entry, (2) inhibit both viral release and cell-to-cell transmission, and (3) possess broad anti-alphavius activities, including against two clinical CHIKV isolates and two alphaviruses: Sindbis virus (SINV) and Semliki forest virus (SFV). In conclusion, our findings suggested that niclosamide and nitazoxanide were able to inhibit CHIKV entry and transmission, which might provide a basis for the development of novel human drug therapies against CHIKV and other alphavirus infections.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Drug Discovery , Niclosamide/pharmacology , Thiazoles/pharmacology , Virus Internalization/drug effects , Animals , Cell Line , Chikungunya Fever/transmission , Chikungunya Fever/virology , Chikungunya virus/physiology , High-Throughput Screening Assays , Humans , Nitro Compounds , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Virus Replication/drug effects
10.
Biochem Pharmacol ; 120: 15-21, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27664855

ABSTRACT

Previously, we reported that salicylate-based analogs of bryostatin protect cells from chikungunya virus (CHIKV)-induced cell death. Interestingly, 'capping' the hydroxyl group at C26 of a lead bryostatin analog, a position known to be crucial for binding to and modulation of protein kinase C (PKC), did not abrogate the anti-CHIKV activity of the scaffold, putatively indicating the involvement of a pathway independent of PKC. The work detailed in this study demonstrates that salicylate-derived analog 1 and two capped analogs (2 and 3) are not merely cytoprotective compounds, but act as selective and specific inhibitors of CHIKV replication. Further, a detailed comparative analysis of the effect of the non-capped versus the two capped analogs revealed that compound 1 acts both at early and late stages in the chikungunya virus replication cycle, while the capped analogs only interfere with a later stage process. Co-dosing with the PKC inhibitors sotrastaurin and Gö6976 counteracts the antiviral activity of compound 1 without affecting that of capped analogs 2 and 3, providing further evidence that the latter elicit their anti-CHIKV activity independently of PKC. Remarkably, treatment of CHIKV-infected cells with a combination of compound 1 and a capped analog resulted in a pronounced synergistic antiviral effect. Thus, these salicylate-based bryostatin analogs can inhibit CHIKV replication through a novel, yet still elusive, non-PKC dependent pathway.


Subject(s)
Antiviral Agents/pharmacology , Bryostatins/pharmacology , Chikungunya virus/drug effects , Drug Design , Protein Kinase C/metabolism , Viral Proteins/metabolism , Acetylation , Animals , Antiviral Agents/agonists , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/chemistry , Bryostatins/agonists , Bryostatins/antagonists & inhibitors , Bryostatins/chemistry , Carbazoles/chemistry , Carbazoles/pharmacology , Cell Line , Chikungunya virus/growth & development , Chikungunya virus/metabolism , Chlorocebus aethiops , Drug Synergism , Gene Expression Regulation, Viral/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Methylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Semliki forest virus/drug effects , Semliki forest virus/growth & development , Semliki forest virus/metabolism , Sindbis Virus/drug effects , Sindbis Virus/growth & development , Sindbis Virus/metabolism , Viral Proteins/antagonists & inhibitors , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication/drug effects
11.
Antiviral Res ; 121: 39-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26112648

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes severe and often persistent arthritis. In recent years, millions of people have been infected with this virus for which registered antivirals are still lacking. Using our recently established in vitro assay, we discovered that the approved anti-parasitic drug suramin inhibits CHIKV RNA synthesis (IC50 of ∼5µM). The compound inhibited replication of various CHIKV isolates in cell culture with an EC50 of ∼80µM (CC50>5mM) and was also active against Sindbis virus and Semliki Forest virus. In vitro studies hinted that suramin interferes with (re)initiation of RNA synthesis, whereas time-of-addition studies suggested it to also interfere with a post-attachment early step in infection, possibly entry. CHIKV (nsP4) mutants resistant against favipiravir or ribavirin, which target the viral RNA polymerase, did not exhibit cross-resistance to suramin, suggesting a different mode of action. The assessment of the activity of a variety of suramin-related compounds in cell culture and the in vitro assay for RNA synthesis provided more insight into the moieties required for antiviral activity. The antiviral effect of suramin-containing liposomes was also analyzed. Its approved status makes it worthwhile to explore the use of suramin to prevent and/or treat CHIKV infections.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Chikungunya virus/physiology , Suramin/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Semliki forest virus/drug effects , Sindbis Virus/drug effects
12.
J Nat Prod ; 78(5): 1119-28, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25946116

ABSTRACT

In an effort to identify new potent and selective inhibitors of chikungunya virus and HIV-1 and HIV-2 virus replication, the endemic Mascarene species Stillingia lineata was investigated. LC/MS and bioassay-guided purification of the EtOAc leaf extract using a chikungunya virus-cell-based assay led to the isolation of six new (4-9) and three known (1-3) tonantzitlolones possessing the rare C20-flexibilane skeleton, along with tonantzitloic acid (10), a new linear diterpenoid, and three new (11, 13, and 15) and two known (12 and 14) tigliane-type diterpenoids. The planar structures of the new compounds and their relative configurations were determined by spectroscopic analysis, and their absolute configurations were determined through comparison with literature data and from biogenetic considerations. These compounds were investigated for selective antiviral activity against chikungunya virus (CHIKV), Semliki Forest virus, Sindbis virus, and, for compounds 11-15, the HIV-1 and HIV-2 viruses. Compounds 12-15 were found to be the most potent and are selective inhibitors of CHIKV, HIV-1, and HIV-2 replication. In particular, compound 14 inhibited CHIKV replication with an EC50 value of 1.2 µM on CHIKV and a selectivity index of >240, while compound 15 inhibited HIV-1 and HIV-2 with EC50 values of 0.043 and 0.018 µM, respectively. It was demonstrated further that potency and selectivity are sensitive to the substitution pattern on the tigliane skeleton. The cytotoxic activities of compounds 1-10 were evaluated against the HCT-116, MCF-7, and PC3 cancer cell lines.


Subject(s)
Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Diterpenes/isolation & purification , Diterpenes/pharmacology , Euphorbiaceae/chemistry , Antiviral Agents/chemistry , Chikungunya virus/drug effects , Diterpenes/chemistry , France , HCT116 Cells , HIV-1/drug effects , HIV-2/drug effects , Humans , MCF-7 Cells , Macrocyclic Compounds/pharmacology , Molecular Structure , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Virus Replication/drug effects
13.
J Nat Prod ; 77(6): 1505-12, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24926807

ABSTRACT

Bioassay-guided purification of an EtOAc extract of the whole plant of Euphorbia amygdaloides ssp. semiperfoliata using a chikungunya virus-cell-based assay led to the isolation of six new (1-4, 9, and 10) and six known (5-7, 8, 11, and 12) jatrophane esters. Their planar structures and relative configurations were determined by extensive spectroscopic analysis, and their absolute configurations by X-ray analysis. These compounds were investigated for selective antiviral activity against chikungunya virus (CHIKV), Semliki Forest virus, Sindbis virus, and HIV-1 and HIV-2 viruses. Compound 3 was found to be the most potent and selective inhibitor of the replication of CHIKV and of HIV-1 and HIV-2 (EC50 = 0.76, IC50 = 0.34 and 0.043 µM, respectively). A preliminary structure-activity relationship study demonstrated that potency and selectivity are very sensitive to the substitution pattern on the jatrophane skeleton. Although replication strategies of CHIK and HIV viruses are quite different, the mechanism of action by which these compounds act may involve a similar target for both viruses. The present results provide additional support for a previous hypothesis that the anti-CHIKV activity could involve a PKC-dependent mechanism.


Subject(s)
Alphavirus Infections/drug therapy , Antiviral Agents/isolation & purification , Chikungunya virus/drug effects , Diterpenes/isolation & purification , Diterpenes/pharmacology , Euphorbia/chemistry , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Chikungunya Fever , Diterpenes/chemistry , France , HIV-1/drug effects , HIV-2/drug effects , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Structure-Activity Relationship , Virus Replication/drug effects
14.
Biomacromolecules ; 15(4): 1534-42, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24628489

ABSTRACT

We present two facile approaches for introducing multivalent displays of tyrosine sulfate mimetic ligands on the surface of cellulose nanocrystals (CNCs) for application as viral inhibitors. We tested the efficacy of cellulose nanocrystals, prepared either from cotton fibers or Whatman filter paper, to inhibit alphavirus infectivity in Vero (B) cells. Cellulose nanocrystals were produced by sulfuric acid hydrolysis leading to nanocrystal surfaces decorated with anionic sulfate groups. When the fluorescent marker expressing Semliki Forest virus vector, VA7-EGFP, was incubated with CNCs, strong inhibition of virus infectivity was achieved, up to 100 and 88% for cotton and Whatman CNCs, respectively. When surface sulfate groups of CNCs were exchanged for tyrosine sulfate mimetic groups (i.e. phenyl sulfonates), improved viral inhibition was attained. Our observations suggest that the conjugation of target-specific functionalities to CNC surfaces provides a means to control their antiviral activity. Multivalent CNCs did not cause observable in vitro cytotoxicity to Vero (B) cells or human corneal epithelial (HCE-T) cells, even within the 100% virus-inhibitory concentrations. Based on the similar chemistry of known polyanionic inhibitors, our results suggest the potential application of CNCs as inhibitors of other viruses, such as human immunodeficiency virus (HIV) and herpes simplex viruses.


Subject(s)
Alphavirus Infections/drug therapy , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cellulose/chemical synthesis , Nanoparticles/chemistry , Nanotechnology/methods , Semliki forest virus/drug effects , Tyrosine/analogs & derivatives , Animals , Biomimetics , Cellulose/chemistry , Chlorocebus aethiops , Epithelium, Corneal/cytology , Epithelium, Corneal/drug effects , Genes, erbB-1 , Ligands , Nanoparticles/toxicity , Semliki forest virus/genetics , Spectroscopy, Fourier Transform Infrared , Tyrosine/chemistry , Vero Cells/drug effects , Vero Cells/virology
15.
Phytochemistry ; 84: 160-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22938995

ABSTRACT

The chemical study of the bark and the wood of Trigonostemon cherrieri, a rare endemic plant of New Caledonia, led to the isolation of a series of highly oxygenated daphnane diterpenoid orthoesters (DDO) bearing an uncommon chlorinated moiety: trigocherrins A-F and trigocherriolides A-D. Herein, we describe the isolation and structure elucidation of the DDO (trigocherrins B-F and trigocherriolides A-D). We also report the antiviral activity of trigocherrins A, B and F (1, 2 and 6) and trigocherriolides A, B and C (7-9) against various emerging pathogens: chikungunya virus (CHIKV), Sindbis virus (SINV), Semliki forest virus (SFV) and dengue virus (DENV).


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/drug effects , Dengue Virus/drug effects , Euphorbiaceae/chemistry , Semliki forest virus/drug effects , Sindbis Virus/drug effects , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Diterpenes/chemistry , Diterpenes/isolation & purification , Diterpenes/pharmacology , Esters/chemistry , Esters/isolation & purification , Esters/pharmacology , Microbial Sensitivity Tests , Molecular Conformation , Plant Bark/chemistry , Wood/chemistry
16.
J Virol ; 86(7): 3588-94, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258261

ABSTRACT

The enveloped alphaviruses infect cells via a low-pH-triggered membrane fusion reaction mediated by the viral transmembrane protein E1. During fusion, E1 inserts into the target membrane and refolds to a hairpin-like postfusion conformation in which domain III (DIII) and the juxtamembrane stem pack against a central core trimer. Although zinc has previously been shown to cause a striking block in alphavirus fusion with liposome target membranes, the mechanism of zinc's effect on the E1 fusion protein is not understood. Here we developed a cell culture system to study zinc inhibition of fusion and infection of the alphavirus Semliki Forest virus (SFV). Inclusion of 2 mM ZnCl(2) in the pH 5.75 fusion buffer caused a decrease of ∼5 logs in SFV fusion at the plasma membrane. Fusion was also inhibited by nickel, a chemically related transition metal. Selection for SFV zinc resistance identified a key histidine residue, H333 on E1 DIII, while other conserved E1 histidine residues were not involved. An H333N mutation conferred resistance to both zinc and nickel, with properties in keeping with the known pH-dependent chelation of these metals by histidine. Biochemical studies demonstrated that zinc strongly inhibits formation of the postfusion E1 trimer in wild-type SFV but not in an H333 mutant. Together our results suggest that zinc acts by blocking the fold-back of DIII via its interaction with H333.


Subject(s)
Alphavirus Infections/virology , Down-Regulation/drug effects , Semliki forest virus/drug effects , Semliki forest virus/physiology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects , Zinc/pharmacology , Animals , Cell Line , Cricetinae , Humans , Protein Structure, Tertiary , Semliki forest virus/chemistry , Semliki forest virus/genetics , Viral Fusion Proteins/genetics
17.
PLoS One ; 6(12): e28923, 2011.
Article in English | MEDLINE | ID: mdl-22205980

ABSTRACT

Chikungunya virus (CHIKV), an alphavirus, has recently caused epidemic outbreaks and is therefore considered a re-emerging pathogen for which no effective treatment is available. In this study, a CHIKV replicon containing the virus replicase proteins together with puromycin acetyltransferase, EGFP and Renilla luciferase marker genes was constructed. The replicon was transfected into BHK cells to yield a stable cell line. A non-cytopathic phenotype was achieved by a Pro718 to Gly substitution and a five amino acid insertion within non-structural protein 2 (nsP2), obtained through selection for stable growth. Characterization of the replicon cell line by Northern blotting analysis revealed reduced levels of viral RNA synthesis. The CHIKV replicon cell line was validated for antiviral screening in 96-well format and used for a focused screen of 356 compounds (natural compounds and clinically approved drugs). The 5,7-dihydroxyflavones apigenin, chrysin, naringenin and silybin were found to suppress activities of EGFP and Rluc marker genes expressed by the CHIKV replicon. In a concomitant screen against Semliki Forest virus (SFV), their anti-alphaviral activity was confirmed and several additional inhibitors of SFV with IC50 values between 0.4 and 24 µM were identified. Chlorpromazine and five other compounds with a 10H-phenothiazinyl structure were shown to inhibit SFV entry using a novel entry assay based on a temperature-sensitive SFV mutant. These compounds also reduced SFV and Sindbis virus-induced cytopathic effect and inhibited SFV virion production in virus yield experiments. Finally, antiviral effects of selected compounds were confirmed using infectious CHIKV. In summary, the presented approach for discovering alphaviral inhibitors enabled us to identify potential lead structures for the development of alphavirus entry and replication phase inhibitors as well as demonstrated the usefulness of CHIKV replicon and SFV as biosafe surrogate models for anti-CHIKV screening.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya virus/genetics , Chikungunya virus/physiology , Drug Evaluation, Preclinical/methods , Replicon , Virus Internalization/drug effects , Virus Replication/drug effects , Cell Line , Chikungunya virus/drug effects , Flavonoids/pharmacology , Humans , Phenothiazines/chemistry , Phenothiazines/pharmacology , Semliki forest virus/drug effects , Semliki forest virus/physiology
18.
J Nat Prod ; 72(11): 1917-26, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19839605

ABSTRACT

This paper describes inhibition of Semliki Forest virus (SFV) replication by synthetic derivatives of naturally occurring triterpenoid betulin (1). Chemical modifications were made to OH groups at C-3 and C-28 and to the C-20-C-29 double bond. A set of heterocyclic betulin derivatives was also assayed. A free or acetylated OH group at C-3 was identified as an important structural contributor for anti-SFV activity, 3,28-di-O-acetylbetulin (4) being the most potent derivative (IC50 value 9.1 microM). Betulinic acid (13), 28-O-tetrahydropyranylbetulin (17), and a triazolidine derivative (41) were also shown to inhibit Sindbis virus, with IC50 values of 0.5, 1.9, and 6.1 microM, respectively. The latter three compounds also had significant synergistic effects against SFV when combined with 3'-amino-3'-deoxyadenosine. In contrast to previous work on other viruses, the antiviral activity of 13 was mapped to take place in virus replication phase. The efficacy was also shown to be independent of external guanosine supplementation.


Subject(s)
Semliki forest virus/drug effects , Triterpenes/chemical synthesis , Triterpenes/pharmacology , Virus Replication/drug effects , Guanosine/pharmacology , Molecular Structure , Pentacyclic Triterpenes , Sindbis Virus/drug effects , Triterpenes/chemistry , Betulinic Acid
19.
Immunity ; 30(6): 802-16, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19523849

ABSTRACT

Interferons (IFNs) direct innate and acquired immune responses and, accordingly, are used therapeutically to treat a number of diseases, yet the diverse effects they elicit are not fully understood. Here, we identified the promyelocytic leukemia zinc finger (PLZF) protein as a previously unrecognized component of the IFN response. IFN stimulated an association of PLZF with promyelocytic leukemia protein (PML) and histone deacetylase 1 (HDAC1) to induce a decisive subset of IFN-stimulated genes (ISGs). Consequently, PLZF-deficient mice had a specific ISG expression defect and as a result were more susceptible to viral infection. This susceptibility correlated with a marked decrease in the expression of the key antiviral mediators and an impaired IFN-mediated induction of natural killer cell function. These results provide new insights into the regulatory mechanisms of IFN signaling and the induction of innate antiviral immunity.


Subject(s)
Alphavirus Infections/immunology , Immunity, Innate/genetics , Interferon-alpha/immunology , Killer Cells, Natural/immunology , Kruppel-Like Transcription Factors/metabolism , Alphavirus Infections/genetics , Alphavirus Infections/virology , Animals , Cell Line, Tumor , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression Profiling , Gene Expression Regulation , Histone Deacetylase 1 , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , Interferon-alpha/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Promyelocytic Leukemia Zinc Finger Protein , Semliki forest virus/drug effects , Semliki forest virus/immunology , Signal Transduction/genetics , Signal Transduction/immunology
20.
Biotechnol Lett ; 31(4): 501-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19116690

ABSTRACT

Semliki forest virus (SFV) is a pathogen causing lethal encephalitis in laboratory mice. In this study, we obtained three short hairpin RNAs (shRNAs) which could specifically target SFV sequence in GFP reporting systems and effectively suppress SFV replication in luciferase-containing reporter virus system. At a multiplicity of infection (MOI) of 0.001, the luciferase reporter activity was reduced by 78-92% by shRNA expression plasmids and virus yields reduced 2 to 10-fold at 20 h post-infection. When lentiviral vector-derived shRNAs were employed, the virus titers decreased 8 to 126-fold at 24 h post-infection and 6 to 19-fold at 48 h post-infection and the cell survival was prolonged. These data formed the basis for further in vivo studies of RNA interference in mouse models.


Subject(s)
Antiviral Agents/pharmacology , Lentivirus/genetics , RNA, Small Interfering/pharmacology , Semliki forest virus/drug effects , Semliki forest virus/physiology , Virus Replication , Animals , Cell Line , Cell Survival , Cricetinae , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , Plasmids , RNA, Small Interfering/genetics , Semliki forest virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...