Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Viruses ; 13(9)2021 09 08.
Article in English | MEDLINE | ID: mdl-34578374

ABSTRACT

Ebola virus (EBOV), one of the most infectious human viruses and a leading cause of viral hemorrhagic fever, imposes a potential public health threat with several recent outbreaks. Despite the difficulties associated with working with this pathogen in biosafety level-4 containment, a protective vaccine and antiviral therapeutic were recently approved. However, the high mortality rate of EBOV infection underscores the necessity to continuously identify novel antiviral strategies to help expand the scope of prophylaxis/therapeutic management against future outbreaks. This includes identifying antiviral agents that target EBOV entry, which could improve the management of EBOV infection. Herein, using EBOV glycoprotein (GP)-pseudotyped particles, we screened a panel of natural medicinal extracts, and identified the methanolic extract of Perilla frutescens (PFME) as a robust inhibitor of EBOV entry. We show that PFME dose-dependently impeded EBOV GP-mediated infection at non-cytotoxic concentrations, and exerted the most significant antiviral activity when both the extract and the pseudoparticles are concurrently present on the host cells. Specifically, we demonstrate that PFME could block viral attachment and neutralize the cell-free viral particles. Our results, therefore, identified PFME as a potent inhibitor of EBOV entry, which merits further evaluation for development as a therapeutic strategy against EBOV infection.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Ebolavirus/physiology , Perilla frutescens/chemistry , Plant Extracts/pharmacology , Viral Envelope Proteins , Virus Internalization/drug effects , Ebolavirus/chemistry , Ebolavirus/genetics , HEK293 Cells , Humans , Methanol/chemistry , Methanol/pharmacology , Plant Extracts/chemistry , Viral Envelope Proteins/genetics
2.
Viruses ; 12(9)2020 08 22.
Article in English | MEDLINE | ID: mdl-32842671

ABSTRACT

Filoviruses, including Ebola virus (EBOV) and Marburg virus (MARV), cause severe hemorrhagic fever in humans and nonhuman primates with high mortality rates. There is no approved therapy against these deadly viruses. Antiviral drug development has been hampered by the requirement of a biosafety level (BSL)-4 facility to handle infectious EBOV and MARV because of their high pathogenicity to humans. In this study, we aimed to establish a surrogate animal model that can be used for anti-EBOV and -MARV drug screening under BSL-2 conditions by focusing on the replication-competent recombinant vesicular stomatitis virus (rVSV) pseudotyped with the envelope glycoprotein (GP) of EBOV (rVSV/EBOV) and MARV (rVSV/MARV), which has been investigated as vaccine candidates and thus widely used in BSL-2 laboratories. We first inoculated mice, rats, and hamsters intraperitoneally with rVSV/EBOV and found that only hamsters showed disease signs and succumbed within 4 days post-infection. Infection with rVSV/MARV also caused lethal infection in hamsters. Both rVSV/EBOV and rVSV/MARV were detected at high titers in multiple organs including the liver, spleen, kidney, and lungs of infected hamsters, indicating acute and systemic infection resulting in fatal outcomes. Therapeutic effects of passive immunization with an anti-EBOV neutralizing antibody were specifically observed in rVSV/EBOV-infected hamsters. Thus, this animal model is expected to be a useful tool to facilitate in vivo screening of anti-filovirus drugs targeting the GP molecule.


Subject(s)
Disease Models, Animal , Ebolavirus/genetics , Marburgvirus/genetics , Vesicular Stomatitis/virology , Vesiculovirus/genetics , Viral Envelope Proteins/genetics , Animals , Antibodies, Viral/administration & dosage , Cricetinae , Disease Susceptibility , Drug Evaluation, Preclinical , Ebolavirus/immunology , Mesocricetus , Mice , Rats , Vaccines, Synthetic , Vesicular Stomatitis/pathology , Vesicular Stomatitis/prevention & control , Vesicular Stomatitis/therapy , Vesiculovirus/pathogenicity , Viral Envelope Proteins/immunology , Viral Load
3.
Viruses ; 13(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396288

ABSTRACT

Filoviruses, such as Ebola virus and Marburg virus, are of significant human health concern. From 2013 to 2016, Ebola virus caused 11,323 fatalities in Western Africa. Since 2018, two Ebola virus disease outbreaks in the Democratic Republic of the Congo resulted in 2354 fatalities. Although there is progress in medical countermeasure (MCM) development (in particular, vaccines and antibody-based therapeutics), the need for efficacious small-molecule therapeutics remains unmet. Here we describe a novel high-throughput screening assay to identify inhibitors of Ebola virus VP40 matrix protein association with viral particle assembly sites on the interior of the host cell plasma membrane. Using this assay, we screened nearly 3000 small molecules and identified several molecules with the desired inhibitory properties. In secondary assays, one identified compound, sangivamycin, inhibited not only Ebola viral infectivity but also that of other viruses. This finding indicates that it is possible for this new VP40-based screening method to identify highly potent MCMs against Ebola virus and its relatives.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Nucleoproteins/antagonists & inhibitors , Viral Core Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Ebolavirus/genetics , Gene Expression Regulation, Viral/drug effects , HEK293 Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Medical Countermeasures , Molecular Structure , Nucleoproteins/chemistry , Pyrimidine Nucleosides/pharmacology , Vero Cells , Viral Core Proteins/chemistry , Virus Release/drug effects
4.
ACS Infect Dis ; 5(1): 35-48, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30516045

ABSTRACT

The Ebola virus has a grave potential to destabilize civil society as we know it. The past few deadly Ebola outbreaks were unprecedented in size: The 2014-15 Ebola West Africa outbreak saw the virus spread from the epicenter through to Guinea, Sierra Leone, Nigeria, Congo, and Liberia. The 2014-15 Ebola West Africa outbreak was associated with almost 30,000 suspected or confirmed cases and over 11,000 documented deaths. The more recent 2018 outbreak in the Democratic Republic of Congo has so far resulted in 216 suspected or confirmed cases and 139 deaths. There is a general acceptance within the World Health Organization (WHO) and the Ebola outbreak response community that future outbreaks will become increasingly more frequent and more likely to involve intercontinental transmission. The magnitude of the recent outbreaks demonstrated in dramatic fashion the shortcomings of our mass casualty disease response capabilities and lack of therapeutic modalities for supporting Ebola outbreak prevention and control. Currently, there are no approved drugs although vaccines for human Ebola virus infection are in the trial phases and some potential treatments have been field tested most recently in the Congo Ebola outbreak. Treatment is limited to pain management and supportive care to counter dehydration and lack of oxygen. This underscores the critical need for effective antiviral drugs that specifically target this deadly disease. This review examines the current approaches for the discovery of anti-Ebola small molecule or biological therapeutics, their viral targets, mode of action, and contemporary platforms, which collectively form the backbone of the anti-Ebola drug discovery pipeline.


Subject(s)
Antiviral Agents/pharmacology , Biological Therapy , Drug Discovery , Hemorrhagic Fever, Ebola/drug therapy , Small Molecule Libraries/pharmacology , Africa, Western , Antiviral Agents/isolation & purification , Clinical Trials as Topic , Disease Outbreaks/prevention & control , Ebolavirus/drug effects , Ebolavirus/genetics , Humans , Research
5.
Antiviral Res ; 156: 46-54, 2018 08.
Article in English | MEDLINE | ID: mdl-29870771

ABSTRACT

Ebola virus (EBOV) causes a severe haemorrhagic fever in humans and has a mortality rate over 50%. With no licensed drug treatments available, EBOV poses a significant threat. Investigations into possible therapeutics have been severely hampered by the classification of EBOV as a BSL4 pathogen. Here, we describe a drug discovery pathway combining in silico screening of compounds predicted to bind to a hydrophobic pocket on the nucleoprotein (NP); with a robust and rapid EBOV minigenome assay for inhibitor validation at BSL2. One compound (MCCB4) was efficacious (EC50 4.8 µM), exhibited low cytotoxicity (CC50 > 100 µM) and was specific, with no effect on either a T7 RNA polymerase driven firefly luciferase or a Bunyamwera virus minigenome. Further investigations revealed that this small molecule inhibitor was able to outcompete established replication complexes, an essential aspect for a potential EBOV treatment.


Subject(s)
Antiviral Agents/isolation & purification , Antiviral Agents/pharmacology , Ebolavirus/genetics , Transcription, Genetic/drug effects , Virus Replication/drug effects , Animals , Antiviral Agents/toxicity , Cell Line , Drug Evaluation, Preclinical/methods , Ebolavirus/physiology , Molecular Dynamics Simulation , Nucleoproteins/metabolism , Protein Binding , Viral Proteins/metabolism
6.
Antiviral Res ; 150: 193-201, 2018 02.
Article in English | MEDLINE | ID: mdl-29294299

ABSTRACT

Ebola virus (EBOV) is an enveloped negative-sense, single-stranded RNA virus of the filovirus family that causes severe disease in humans. Approved therapies for EBOV disease are lacking. EBOV RNA synthesis is carried out by a virus-encoded complex with RNA-dependent RNA polymerase activity that is required for viral propagation. This complex and its activities are therefore potential antiviral targets. To identify potential lead inhibitors of EBOV RNA synthesis, a library of small molecule compounds was screened against a previously established assay of EBOV RNA synthesis, the EBOV minigenome assay (MGA), in 384 well microplate format. The screen identified 56 hits that inhibited EBOV MGA activity by more than 70% while exhibiting less than 20% cell cytotoxicity. Inhibitory chemical scaffolds included angelicin derivatives, derivatives of the antiviral compound GSK983 and benzoquinolines. Structure-activity relationship (SAR) studies of the benzoquinoline scaffold produced ∼50 analogs and led to identification of an optimized compound, SW456, with a submicromolar IC50 in the EBOV MGA and antiviral activity against infectious EBOV in cell culture. The compound was also active against a MGA for another deadly filovirus, Marburg virus. It also exhibited antiviral activity towards a negative-sense RNA virus from the rhabdovirus family, vesicular stomatitis virus, and a positive-sense RNA virus, Zika virus. Overall, these data demonstrate the potential of the EBOV MGA to identify anti-EBOV compounds and identifies the benzoquinoline series as a broad-spectrum antiviral lead.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , High-Throughput Screening Assays , Quinolines/pharmacology , Virus Replication/drug effects , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Drug Evaluation, Preclinical , Ebolavirus/genetics , Humans , Quinolines/chemistry , Small Molecule Libraries
7.
Expert Opin Biol Ther ; 18(2): 159-173, 2018 02.
Article in English | MEDLINE | ID: mdl-29148858

ABSTRACT

INTRODUCTION: The Ebola virus (EBOV) disease epidemic during 2014-16 in West Africa has accelerated the clinical development of several vaccine candidates that have demonstrated efficacy in the gold standard nonhuman primate (NHP) model, namely cynomolgus macaques. AREAS COVERED: This review discusses the pre-clinical research and if available, clinical evaluation of the currently available EBOV vaccine candidates, while emphasizing the translatability of pre-clinical data generated in the NHP model to clinical data in humans. EXPERT OPINION: Despite the existence of many successful EBOV vaccine candidates in the pre-clinical stages, only two platforms became the focus of Phase 2/3 efficacy trials in Liberia, Sierra Leone, and Guinea near the peak of the epidemic: the Vesicular stomatitis virus (VSV)-vectored vaccine and the chimpanzee adenovirus type 3 (ChAd3)-vectored vaccine. The results of three distinct clinical trials involving these candidates may soon pave the way for a licensed, safe and efficacious EBOV vaccine to help combat future epidemics.


Subject(s)
Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/prevention & control , Animals , Drug Evaluation, Preclinical , Ebolavirus/genetics , Ebolavirus/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Humans , Macaca fascicularis , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, Virus-Like Particle/immunology
8.
J Virol ; 90(19): 8720-8, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27440895

ABSTRACT

UNLABELLED: Ebola virus (EBOV) is a highly contagious lethal pathogen. As a biosafety level 4 (BSL-4) agent, however, EBOV is restricted to costly BSL-4 laboratories for experimentation, thus significantly impeding the evaluation of EBOV vaccines and drugs. Here, we report an EBOV-like particle (EBOVLP)-based luciferase reporter system that enables the evaluation of anti-EBOV agents in vitro and in vivo outside BSL-4 facilities. Cotransfection of HEK293T cells with four plasmids encoding the proteins VP40, NP, and GP of EBOV and firefly luciferase (Fluc) resulted in the production of Fluc-containing filamentous particles that morphologically resemble authentic EBOV. The reporter EBOVLP was capable of delivering Fluc into various cultured cells in a GP-dependent manner and was recognized by a conformation-dependent anti-EBOV monoclonal antibody (MAb). Significantly, inoculation of mice with the reporter EBOVLP led to the delivery of Fluc protein into target cells and rapid generation of intense bioluminescence signals that could be blocked by the administration of EBOV neutralizing MAbs. This BSL-4-free reporter system should facilitate high-throughput screening for anti-EBOV drugs targeting viral entry and efficacy testing of candidate vaccines. IMPORTANCE: Ebola virus (EBOV) researches have been limited to costly biosafety level 4 (BSL-4) facilities due to the lack of animal models independent of BSL-4 laboratories. In this study, we reveal that a firefly luciferase-bearing EBOV-like particle (EBOVLP) with typical filamentous EBOV morphology is capable of delivering the reporter protein into murine target cells both in vitro and in vivo Moreover, we demonstrate that the reporter delivery can be inhibited both in vitro and in vivo by a known anti-EBOV protective monoclonal antibody, 13C6. Our work provides a BSL-4-free system that can facilitate the in vivo evaluation of anti-EBOV antibodies, drugs, and vaccines. The system may also be useful for mechanistic study of the viral entry process.


Subject(s)
Antiviral Agents/isolation & purification , Ebola Vaccines/immunology , Ebolavirus/drug effects , Endocytosis , Genes, Reporter , Luciferases/analysis , Virosomes/metabolism , Animals , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Ebolavirus/genetics , Luciferases/genetics , Mice , Virosomes/drug effects , Virosomes/genetics , Virosomes/immunology
9.
Infect Dis Poverty ; 5: 12, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26888469

ABSTRACT

BACKGROUND: The Ebola virus is highly pathogenic and destructive to humans and other primates. The Ebola virus encodes viral protein 40 (VP40), which is highly expressed and regulates the assembly and release of viral particles in the host cell. Because VP40 plays a prominent role in the life cycle of the Ebola virus, it is considered as a key target for antiviral treatment. However, there is currently no FDA-approved drug for treating Ebola virus infection, resulting in an urgent need to develop effective antiviral inhibitors that display good safety profiles in a short duration. METHODS: This study aimed to screen the effective lead candidate against Ebola infection. First, the lead molecules were filtered based on the docking score. Second, Lipinski rule of five and the other drug likeliness properties are predicted to assess the safety profile of the lead candidates. Finally, molecular dynamics simulations was performed to validate the lead compound. RESULTS: Our results revealed that emodin-8-beta-D-glucoside from the Traditional Chinese Medicine Database (TCMD) represents an active lead candidate that targets the Ebola virus by inhibiting the activity of VP40, and displays good pharmacokinetic properties. CONCLUSION: This report will considerably assist in the development of the competitive and robust antiviral agents against Ebola infection.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Drug Evaluation, Preclinical , Ebolavirus/chemistry , Ebolavirus/genetics , Ebolavirus/metabolism , HEK293 Cells , Hemorrhagic Fever, Ebola , Humans , Molecular Docking Simulation , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Curr Gene Ther ; 16(1): 47-55, 2016.
Article in English | MEDLINE | ID: mdl-26785737

ABSTRACT

Previously unidentified viruses, such as Middle East respiratory syndrome coronavirus, continue to emerge and threaten populations, while powerful new techniques have identified many new human and animal viruses. Similarly, existing viruses, from Ebola virus to chikungunya virus, are reemerging and spreading to new geographical regions. These viruses often pose a challenge for researchers to study due to their highly pathogenic nature. Lentiviral and rhabdoviral pseudotypes are excellent tools for studying enveloped viruses and have contributed to many recent advances in areas such as receptor usage, viral entry and serology. In particular, pseudotypes allow the safe study of unknown or highly pathogenic viruses. They also allow the initial characterization of aspects of infection such as cellular tropism for difficult to culture viruses. In this review we will introduce various pseudotyping systems for emerging viruses, including chikungunya virus, Ebola virus, SARS and MERS coronaviruses and Nipah virus, as well as their use in diverse studies including drug screening and antibody neutralization. We will also discuss the limitations and potential caveats using pseudotypes.


Subject(s)
Drug Evaluation, Preclinical/methods , Genetic Therapy/methods , Genetic Vectors , Viral Envelope Proteins/genetics , Viral Tropism/physiology , Antibodies, Neutralizing/genetics , Coronavirus/genetics , Ebolavirus/genetics , Nipah Virus/genetics , Viral Vaccines
11.
Curr Top Med Chem ; 16(13): 1530-5, 2016.
Article in English | MEDLINE | ID: mdl-26369818

ABSTRACT

Regulation of protein expression by non-coding RNAs typically involves effects on mRNA degradation and/or ribosomal translation. The possibility of virus-host mRNA-mRNA antisense tethering interactions (ATI) as a gain-of-function strategy, via the capture of functional RNA motifs, has not been hitherto considered. We present evidence that ATIs may be exploited by certain RNA viruses in order to tether the mRNAs of host selenoproteins, potentially exploiting the proximity of a captured host selenocysteine insertion sequence (SECIS) element to enable the expression of virally-encoded selenoprotein modules, via translation of in-frame UGA stop codons as selenocysteine. Computational analysis predicts thermodynamically stable ATIs between several widely expressed mammalian selenoprotein mRNAs (e.g., isoforms of thioredoxin reductase) and specific Ebola virus mRNAs, and HIV-1 mRNA, which we demonstrate via DNA gel shift assays. The probable functional significance of these ATIs is further supported by the observation that, in both viruses, they are located in close proximity to highly conserved in-frame UGA stop codons at the 3' end of open reading frames that encode essential viral proteins (the HIV-1 nef protein and the Ebola nucleoprotein). Significantly, in HIV/AIDS patients, an inverse correlation between serum selenium and mortality has been repeatedly documented, and clinical benefits of selenium in the context of multi-micronutrient supplementation have been demonstrated in several well-controlled clinical trials. Hence, in the light of our findings, the possibility of a similar role for selenium in Ebola pathogenesis and treatment merits serious investigation.


Subject(s)
Ebolavirus/genetics , HIV-1/genetics , RNA, Antisense/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Viral/genetics , Selenium/metabolism , Selenoproteins/genetics , Humans , Thermodynamics
12.
J Virol ; 90(5): 2345-55, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26676781

ABSTRACT

UNLABELLED: Ebola virus (EBOV) is an RNA virus that can cause hemorrhagic fever with high fatality rates, and there are no approved vaccines or therapies. Typically, RNA viruses have high spontaneous mutation rates, which permit rapid adaptation to selection pressures and have other important biological consequences. However, it is unknown if filoviruses exhibit high mutation frequencies. Ultradeep sequencing and a recombinant EBOV that carries the gene encoding green fluorescent protein were used to determine the spontaneous mutation frequency of EBOV. The effects of the guanosine analogue ribavirin during EBOV infections were also assessed. Ultradeep sequencing revealed that the mutation frequency for EBOV was high and similar to those of other RNA viruses. Interestingly, significant genetic diversity was not observed in viable viruses, implying that changes were not well tolerated. We hypothesized that this could be exploited therapeutically. In vitro, the presence of ribavirin increased the error rate, and the 50% inhibitory concentration (IC50) was 27 µM. In a mouse model of ribavirin therapy given pre-EBOV exposure, ribavirin treatment corresponded with a significant delay in time to death and up to 75% survival. In mouse and monkey models of therapy given post-EBOV exposure, ribavirin treatment also delayed the time to death and increased survival. These results demonstrate that EBOV has a spontaneous mutation frequency similar to those of other RNA viruses. These data also suggest a potential for therapeutic use of ribavirin for human EBOV infections. IMPORTANCE: Ebola virus (EBOV) causes a severe hemorrhagic disease with high case fatality rates; there are no approved vaccines or therapies. We determined the spontaneous mutation frequency of EBOV, which is relevant to understanding the potential for the virus to adapt. The frequency was similar to those of other RNA viruses. Significant genetic diversity was not observed in viable viruses, implying that changes were not well tolerated. We hypothesized that this could be exploited therapeutically. Ribavirin is a viral mutagen approved for treatment of several virus infections; it is also cheap and readily available. In cell culture, we showed that ribavirin was effective at reducing production of infectious EBOV. In mouse and monkey models of therapy given post-EBOV exposure, ribavirin treatment delayed the time to death and increased survival. These data provide a better understanding of EBOV spontaneous mutation and suggest that ribavirin may have great value in the context of human disease.


Subject(s)
Ebolavirus/genetics , Genetic Variation , Mutation Rate , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Disease Models, Animal , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Hemorrhagic Fever, Ebola/drug therapy , High-Throughput Nucleotide Sequencing , Humans , Inhibitory Concentration 50 , Macaca fascicularis , Male , Mice, Inbred BALB C , Microbial Sensitivity Tests , RNA, Viral/genetics , Ribavirin/administration & dosage , Ribavirin/pharmacology , Sequence Analysis, DNA , Survival Analysis , Treatment Outcome
13.
Science ; 349(6249): 739-42, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26249231

ABSTRACT

The latest Ebola virus (EBOV) epidemic spread rapidly through Guinea, Sierra Leone, and Liberia, creating a global public health crisis and accelerating the assessment of experimental therapeutics and vaccines in clinical trials. One of those vaccines is based on recombinant vesicular stomatitis virus expressing the EBOV glycoprotein (VSV-EBOV), a live-attenuated vector with marked preclinical efficacy. Here, we provide the preclinical proof that VSV-EBOV completely protects macaques against lethal challenge with the West African EBOV-Makona strain. Complete and partial protection was achieved with a single dose given as late as 7 and 3 days before challenge, respectively. This indicates that VSV-EBOV may protect humans against EBOV infections in West Africa with relatively short time to immunity, promoting its use for immediate public health responses.


Subject(s)
Ebola Vaccines/administration & dosage , Ebolavirus/immunology , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/prevention & control , Viral Proteins/immunology , Animals , Drug Evaluation, Preclinical , Ebola Vaccines/immunology , Ebolavirus/genetics , Genetic Vectors , Glycoproteins/genetics , Macaca , Vesiculovirus , Viral Proteins/genetics
14.
Virology ; 484: 259-264, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26122472

ABSTRACT

Previous studies have demonstrated the potential application of reverse genetics technology in studying a broad range of aspects of viral biology, including gene regulation, protein function, cell entry, and pathogenesis. Here, we describe a highly efficient reverse genetics system used to generate recombinant Ebola virus (EBOV) based on a recent isolate from a human patient infected during the 2014-2015 outbreak in Western Africa. We also rescued a recombinant EBOV expressing a fluorescent reporter protein from a cleaved VP40 protein fusion. Using this virus and an inexpensive method to quantitate the expression of the foreign gene, we demonstrate its potential usefulness as a tool for screening antiviral compounds and measuring neutralizing antibodies.


Subject(s)
Ebolavirus/genetics , Green Fluorescent Proteins/biosynthesis , Reverse Genetics/methods , Africa, Western , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Antiviral Agents/isolation & purification , Drug Evaluation, Preclinical/methods , Ebolavirus/isolation & purification , Genes, Reporter , Green Fluorescent Proteins/genetics , Hemorrhagic Fever, Ebola/virology , Humans , Molecular Sequence Data , RNA, Viral/genetics , Sequence Analysis, DNA , Staining and Labeling
15.
Vaccine ; 33(15): 1830-8, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25728317

ABSTRACT

Dengue infection is on the rise in many endemic areas of the tropics. Vaccination remains the most realistic strategy for prevention of this potentially fatal viral disease but there is currently no effective vaccine that could protect against all four known serotypes of the dengue virus. This study describes the generation and testing of a novel vaccination approach against dengue based on recombinant immune complexes (RIC). We modelled the dengue RIC on the existing Ebola RIC (Phoolcharoen, et al. Proc Natl Acad Sci USA 2011;108(Dec (51)):20695) but with a key modification that allowed formation of a universal RIC platform that can be easily adapted for use for other pathogens. This was achieved by retaining only the binding epitope of the 6D8 ant-Ebola mAb, which was then fused to the consensus dengue E3 domain (cEDIII), resulting in a hybrid dengue-Ebola RIC (DERIC). We expressed human and mouse versions of these molecules in tobacco plants using a geminivirus-based expression system. Following purification from the plant extracts by protein G affinity chromatography, DERIC bound to C1q component of complement, thus confirming functionality. Importantly, following immunization of mice, DERIC induced a potent, virus-neutralizing anti-cEDIII humoral immune response without exogenous adjuvants. We conclude that these self-adjuvanting immunogens have the potential to be developed as a novel vaccine candidate for dengue infection, and provide the basis for a universal RIC platform for use with other antigens.


Subject(s)
Adjuvants, Immunologic , Antibodies, Viral/immunology , Antigen-Antibody Complex/immunology , Dengue Vaccines/immunology , Dengue Virus/immunology , Dengue/prevention & control , Vaccination/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antigen-Antibody Complex/administration & dosage , Antigen-Antibody Complex/genetics , Cell Line , Complement C1q/immunology , Dengue Vaccines/administration & dosage , Dengue Vaccines/genetics , Dengue Vaccines/isolation & purification , Ebolavirus/genetics , Ebolavirus/immunology , Epitopes/immunology , Geminiviridae/genetics , Humans , Immunity, Humoral , Mice , Plant Leaves , Nicotiana , Viral Envelope Proteins/immunology
16.
Expert Rev Anti Infect Ther ; 12(10): 1253-63, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25169588

ABSTRACT

Filoviruses cause severe hemorrhagic fevers with case fatality rates of up to 90%, for which no antivirals are currently available. Their categorization as biosafety level 4 agents restricts work with infectious viruses to a few maximum containment laboratories worldwide, which constitutes a significant obstacle for the development of countermeasures. Reverse genetics facilitates the generation of recombinant filoviruses, including reporter-expressing viruses, which have been increasingly used for drug screening and development in recent years. Further, reverse-genetics based lifecycle modeling systems allow modeling of the filovirus lifecycle without the need for a maximum containment laboratory and have recently been optimized for use in high-throughput assays. The availability of these reverse genetics-based tools will significantly improve our ability to find novel antivirals against filoviruses.


Subject(s)
Antiviral Agents/pharmacology , Filoviridae Infections/drug therapy , Filoviridae/genetics , Models, Theoretical , Reverse Genetics/methods , Antiviral Agents/therapeutic use , Drug Discovery , Drug Evaluation, Preclinical , Ebolavirus/drug effects , Ebolavirus/genetics , Ebolavirus/physiology , Filoviridae/drug effects , Filoviridae/physiology , Genome, Viral , Humans , Microbial Sensitivity Tests , Virus Replication
17.
Antiviral Res ; 106: 86-94, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24713118

ABSTRACT

Marburg virus (MARV) and Ebola virus (EBOV), members of the family Filoviridae, represent a significant challenge to global public health. Currently, no licensed therapies exist to treat filovirus infections, which cause up to 90% mortality in human cases. To facilitate development of antivirals against these viruses, we established two distinct screening platforms based on MARV and EBOV reverse genetics systems that express secreted Gaussia luciferase (gLuc). The first platform is a mini-genome replicon to screen viral replication inhibitors using gLuc quantification in a BSL-2 setting. The second platform is complementary to the first and expresses gLuc as a reporter gene product encoded in recombinant infectious MARV and EBOV, thereby allowing for rapid quantification of viral growth during treatment with antiviral compounds. We characterized these viruses by comparing luciferase activity to virus production, and validated luciferase activity as an authentic real-time measure of viral growth. As proof of concept, we adapt both mini-genome and infectious virus platforms to high-throughput formats, and demonstrate efficacy of several antiviral compounds. We anticipate that both approaches will prove highly useful in the development of anti-filovirus therapies, as well as in basic research on the filovirus life cycle.


Subject(s)
Antiviral Agents/isolation & purification , Drug Evaluation, Preclinical/methods , Ebolavirus/drug effects , Marburgvirus/drug effects , Reverse Genetics/methods , Animals , Antiviral Agents/pharmacology , Cell Line , Ebolavirus/genetics , Ebolavirus/physiology , Genes, Reporter , Luciferases/analysis , Luciferases/genetics , Marburgvirus/genetics , Marburgvirus/physiology , Virus Replication/drug effects
18.
Antiviral Res ; 99(3): 207-13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23751367

ABSTRACT

Ebola virus (EBOV) causes a severe hemorrhagic fever with case fatality rates of up to 90%, for which no antiviral therapies are available. Antiviral screening is hampered by the fact that development of cytopathic effect, the easiest means to detect infection with wild-type EBOV, is relatively slow. To overcome this problem we generated a recombinant EBOV carrying a luciferase reporter. Using this virus we show that EBOV entry is rapid, with viral protein expression detectable within 2 h after infection. Further, luminescence-based assays were developed to allow highly sensitive titer determination within 48 h. As a proof-of-concept for its utility in antiviral screening we used this virus to assess neutralizing antibodies and siRNAs, with significantly faster screening times than currently available wild-type or recombinant viruses. The availability of this recombinant virus will allow for more rapid and quantitative evaluation of antivirals against EBOV, as well as the study of details of the EBOV life cycle.


Subject(s)
Antiviral Agents/pharmacology , Biological Assay/methods , Drug Evaluation, Preclinical/methods , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/virology , Luciferases/genetics , Ebolavirus/genetics , Ebolavirus/metabolism , Gene Expression/drug effects , Genes, Reporter/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Humans , Luciferases/antagonists & inhibitors , Luciferases/metabolism
19.
Chem Biol ; 20(3): 424-33, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23521799

ABSTRACT

There are no approved therapeutics for the most deadly nonsegmented negative-strand (NNS) RNA viruses, including Ebola (EBOV). To identify chemical scaffolds for the development of broad-spectrum antivirals, we undertook a prototype-based lead identification screen. Using the prototype NNS virus, vesicular stomatitis virus (VSV), multiple inhibitory compounds were identified. Three compounds were investigated for broad-spectrum activity and inhibited EBOV infection. The most potent, CMLDBU3402, was selected for further study. CMLDBU3402 did not show significant activity against segmented negative-strand RNA viruses, suggesting proscribed broad-spectrum activity. Mechanistic analysis indicated that CMLDBU3402 blocked VSV viral RNA synthesis and inhibited EBOV RNA transcription, demonstrating a consistent mechanism of action against genetically distinct viruses. The identification of this chemical backbone as a broad-spectrum inhibitor of viral RNA synthesis offers significant potential for the development of new therapies for highly pathogenic viruses.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Ebolavirus/genetics , RNA, Viral/biosynthesis , Vesiculovirus/drug effects , Vesiculovirus/genetics , Animals , Cell Line , Drug Evaluation, Preclinical , Ebolavirus/growth & development , Ebolavirus/physiology , Gene Expression Regulation, Viral/drug effects , Humans , Transcription, Genetic/drug effects , Vesiculovirus/growth & development , Vesiculovirus/physiology , Virus Replication/drug effects
20.
Proc Natl Acad Sci U S A ; 105(4): 1129-33, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18212124

ABSTRACT

Ebola virus (EBOV), a public health concern in Africa and a potential biological weapon, is classified as a biosafety level-4 agent because of its high mortality rate and the lack of approved vaccines and antivirals. Basic research into the mechanisms of EBOV pathogenicity and the development of effective countermeasures are restricted by the current biosafety classification of EBOVs. We therefore developed biologically contained EBOV that express a reporter gene instead of the VP30 gene, which encodes an essential transcription factor. A Vero cell line that stably expresses VP30 provides this essential protein in trans and biologically confines the virus to its complete replication cycle in this cell line. This complementation approach is highly efficient because biologically contained EBOVs lacking the VP30 gene grow to titers similar to those obtained with wild-type virus. Moreover, EBOVs lacking the VP30 gene are indistinguishable in their morphology from wild-type virus and are genetically stable, as determined by sequence analysis after seven serial passages in VP30-expressing Vero cells. We propose that this system provides a safe means to handle EBOV outside a biosafety level-4 facility and will stimulate critical studies on the EBOV life cycle as well as large-scale screening efforts for compounds with activity against this lethal virus.


Subject(s)
Ebolavirus/physiology , Virus Replication/physiology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Drug Evaluation, Preclinical , Ebola Vaccines/biosynthesis , Ebola Vaccines/genetics , Ebola Vaccines/immunology , Ebolavirus/drug effects , Ebolavirus/genetics , Ebolavirus/immunology , Humans , Pilot Projects , Serial Passage , Transcription Factors/deficiency , Transcription Factors/genetics , Vero Cells , Viral Proteins/genetics , Virus Replication/drug effects , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL