Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Molecules ; 27(2)2022 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-35056869

RESUMO

Hendra virus (HeV) belongs to the paramyxoviridae family of viruses which is associated with the respiratory distress, neurological illness, and potential fatality of the affected individuals. So far, no competitive approved therapeutic substance is available for HeV. For that reason, the current research work was conducted to propose some novel compounds, by adopting a Computer Aided Drug Discovery approach, which could be used to combat HeV. The G attachment Glycoprotein (Ggp) of HeV was selected to achieve the primary objective of this study, as this protein makes the entry of HeV possible in the host cells. Briefly, a library of 6000 antiviral compounds was screened for potential drug-like properties, followed by the molecular docking of short-listed compounds with the Protein Data Bank (PDB) structure of Ggp. Docked complexes of top two hits, having maximum binding affinities with the active sites of Ggp, were further considered for molecular dynamic simulations of 200 ns to elucidate the results of molecular docking analysis. MD simulations and Molecular Mechanics Energies combined with the Generalized Born and Surface Area (MMGBSA) or Poisson-Boltzmann and Surface Area (MMPBSA) revealed that both docked complexes are stable in nature. Furthermore, the same methodology was used between lead compounds and HeV Ggp in complex with its functional receptor in human, Ephrin-B2. Surprisingly, no major differences were found in the results, which demonstrates that our identified compounds can also perform their action even when the Ggp is attached to the Ephrin-B2 ligand. Therefore, in light of all of these results, we strongly suggest that compounds (S)-5-(benzylcarbamoyl)-1-(2-(4-methyl-2-phenylpiperazin-1-yl)-2-oxoethyl)-6-oxo-3,6-dihydropyridin-1-ium-3-ide and 5-(cyclohexylcarbamoyl)-1-(2-((2-(3-fluorophenyl)-2-methylpropyl)amino)-2-oxoethyl)-6-oxo-3,6-dihydropyridin-1-ium-3-ide could be considered as potential therapeutic agents against HeV; however, further in vitro and in vivo experiments are required to validate this study.


Assuntos
Antivirais/química , Química Computacional/métodos , Proteínas Virais de Fusão/química , Antivirais/metabolismo , Efrina-B2/química , Efrina-B2/metabolismo , Vírus Hendra/efeitos dos fármacos , Humanos , Ligação de Hidrogênio , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Receptores Virais/química , Receptores Virais/metabolismo , Bibliotecas de Moléculas Pequenas , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/metabolismo , Água/química
2.
Curr Mol Pharmacol ; 13(2): 108-125, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31657692

RESUMO

BACKGROUND: Nipah virus (NiV) and Hendra virus (HeV) of genus Henipavirus are the deadliest zoonotic viruses, which cause severe respiratory ailments and fatal encephalitis in humans and other susceptible animals. The fatality rate for these infections had been alarmingly high with no approved treatment available to date. Viral attachment and fusion with host cell membrane is essential for viral entry and is the most essential event of viral infection. Viral attachment is mediated by interaction of Henipavirus attachment glycoprotein (G) with the host cell receptor: Ephrin B2/B3, while viral fusion and endocytosis are mediated by the combined action of both viral glycoprotein (G) and fusion protein (F). CONCLUSION: This review highlights the mechanism of viral attachment, fusion and also explains the basic mechanism and pathobiology of this infection in humans. The drugs and therapeutics used either experimentally or clinically against NiV and HeV infection have been documented and classified in detail. Some amino acid residues essential for the functionality of G and F proteins were also emphasized. Therapeutic designing to target and block these residues can serve as a promising approach in future drug development against NiV and HeV.


Assuntos
Antivirais/farmacologia , Desenho de Fármacos , Vírus Hendra/efeitos dos fármacos , Vírus Nipah/efeitos dos fármacos , Animais , Vírus Hendra/genética , Infecções por Henipavirus/fisiopatologia , Humanos , Vírus Nipah/genética , Internalização do Vírus/efeitos dos fármacos
3.
Nat Struct Mol Biol ; 26(10): 980-987, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31570878

RESUMO

Nipah virus (NiV) and Hendra virus (HeV) are zoonotic henipaviruses (HNVs) responsible for outbreaks of encephalitis and respiratory illness with fatality rates of 50-100%. No vaccines or licensed therapeutics currently exist to protect humans against NiV or HeV. HNVs enter host cells by fusing the viral and cellular membranes via the concerted action of the attachment (G) and fusion (F) glycoproteins, the main targets of the humoral immune response. Here, we describe the isolation and humanization of a potent monoclonal antibody cross-neutralizing NiV and HeV. Cryo-electron microscopy, triggering and fusion studies show the antibody binds to a prefusion-specific quaternary epitope, conserved in NiV F and HeV F glycoproteins, and prevents membrane fusion and viral entry. This work supports the importance of the HNV prefusion F conformation for eliciting a robust immune response and paves the way for using this antibody for prophylaxis and post-exposure therapy with NiV- and HeV-infected individuals.


Assuntos
Anticorpos Neutralizantes/farmacologia , Antivirais/farmacologia , Vírus Hendra/efeitos dos fármacos , Infecções por Henipavirus/tratamento farmacológico , Vírus Nipah/efeitos dos fármacos , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Células HEK293 , Vírus Hendra/metabolismo , Infecções por Henipavirus/metabolismo , Infecções por Henipavirus/virologia , Humanos , Modelos Moleculares , Vírus Nipah/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus/efeitos dos fármacos
4.
Sci Rep ; 8(1): 7604, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29765101

RESUMO

Nipah and Hendra viruses are recently emerged bat-borne paramyxoviruses (genus Henipavirus) causing severe encephalitis and respiratory disease in humans with fatality rates ranging from 40-75%. Despite the severe pathogenicity of these viruses and their pandemic potential, no therapeutics or vaccines are currently approved for use in humans. Favipiravir (T-705) is a purine analogue antiviral approved for use in Japan against emerging influenza strains; and several phase 2 and 3 clinical trials are ongoing in the United States and Europe. Favipiravir has demonstrated efficacy against a broad spectrum of RNA viruses, including members of the Paramyxoviridae, Filoviridae, Arenaviridae families, and the Bunyavirales order. We now demonstrate that favipiravir has potent antiviral activity against henipaviruses. In vitro, favipiravir inhibited Nipah and Hendra virus replication and transcription at micromolar concentrations. In the Syrian hamster model, either twice daily oral or once daily subcutaneous administration of favipiravir for 14 days fully protected animals challenged with a lethal dose of Nipah virus. This first successful treatment of henipavirus infection in vivo with a small molecule drug suggests that favipiravir should be further evaluated as an antiviral treatment option for henipavirus infections.


Assuntos
Amidas/administração & dosagem , Vírus Hendra/fisiologia , Infecções por Henipavirus/tratamento farmacológico , Vírus Nipah/fisiologia , Pirazinas/administração & dosagem , Administração Oral , Amidas/farmacologia , Animais , Cricetinae , Modelos Animais de Doenças , Feminino , Vírus Hendra/efeitos dos fármacos , Humanos , Injeções Subcutâneas , Vírus Nipah/efeitos dos fármacos , Pirazinas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Resultado do Tratamento , Replicação Viral/efeitos dos fármacos
5.
Sci Rep ; 8(1): 358, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29321677

RESUMO

Hendra virus (HeV) is a paramyxovirus that causes lethal disease in humans, for which no vaccine or antiviral agent is available. HeV V protein is central to pathogenesis through its ability to interact with cytoplasmic host proteins, playing key antiviral roles. Here we use immunoprecipitation, siRNA knockdown and confocal laser scanning microscopy to show that HeV V shuttles to and from the nucleus through specific host nuclear transporters. Spectroscopic and small angle X-ray scattering studies reveal HeV V undergoes a disorder-to-order transition upon binding to either importin α/ß1 or exportin-1/Ran-GTP, dependent on the V N-terminus. Importantly, we show that specific inhibitors of nuclear transport prevent interaction with host transporters, and reduce HeV infection. These findings emphasize the critical role of host-virus interactions in HeV infection, and potential use of compounds targeting nuclear transport, such as the FDA-approved agent ivermectin, as anti-HeV agents.


Assuntos
Vírus Hendra/fisiologia , Infecções por Henipavirus/metabolismo , Infecções por Henipavirus/virologia , Interações Hospedeiro-Patógeno , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Antivirais/química , Antivirais/farmacologia , Núcleo Celular/metabolismo , Descoberta de Drogas , Técnicas de Silenciamento de Genes , Vírus Hendra/efeitos dos fármacos , Infecções por Henipavirus/genética , Humanos , Carioferinas/química , Carioferinas/genética , Carioferinas/metabolismo , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteína Exportina 1
6.
J Virol ; 88(8): 4353-65, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24501399

RESUMO

UNLABELLED: Severe acute respiratory syndrome coronavirus (SARS-CoV) and Ebola, Hendra, and Nipah viruses are members of different viral families and are known causative agents of fatal viral diseases. These viruses depend on cathepsin L for entry into their target cells. The viral glycoproteins need to be primed by protease cleavage, rendering them active for fusion with the host cell membrane. In this study, we developed a novel high-throughput screening assay based on peptides, derived from the glycoproteins of the aforementioned viruses, which contain the cathepsin L cleavage site. We screened a library of 5,000 small molecules and discovered a small molecule that can inhibit the cathepsin L cleavage of all viral peptides with minimal inhibition of cleavage of a host protein-derived peptide (pro-neuropeptide Y). The small molecule inhibited the entry of all pseudotyped viruses in vitro and the cleavage of SARS-CoV spike glycoprotein in an in vitro cleavage assay. In addition, the Hendra and Nipah virus fusion glycoproteins were not cleaved in the presence of the small molecule in a cell-based cleavage assay. Furthermore, we demonstrate that the small molecule is a mixed inhibitor of cathepsin L. Our broad-spectrum antiviral small molecule appears to be an ideal candidate for future optimization and development into a potent antiviral against SARS-CoV and Ebola, Hendra, and Nipah viruses. IMPORTANCE: We developed a novel high-throughput screening assay to identify small molecules that can prevent cathepsin L cleavage of viral glycoproteins derived from SARS-CoV and Ebola, Hendra, and Nipah viruses that are required for their entry into the host cell. We identified a novel broad-spectrum small molecule that could block cathepsin L-mediated cleavage and thus inhibit the entry of pseudotypes bearing the glycoprotein derived from SARS-CoV or Ebola, Hendra, or Nipah virus. The small molecule can be further optimized and developed into a potent broad-spectrum antiviral drug.


Assuntos
Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Ebolavirus/efeitos dos fármacos , Vírus Hendra/efeitos dos fármacos , Ensaios de Triagem em Larga Escala/métodos , Vírus Nipah/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Catepsina L/metabolismo , Ebolavirus/metabolismo , Vírus Hendra/metabolismo , Humanos , Vírus Nipah/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Proteínas do Envelope Viral/metabolismo , Viroses/enzimologia , Viroses/virologia
7.
Curr Drug Targets ; 15(1): 103-13, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24102407

RESUMO

Australia is facing a major national medical challenge with the emergence of the Hendra virus (HeV) as a medically and economically important pathogen of humans and animals. Clinical symptoms of human HeV infection can include fever, hypotension, dizziness, encephalitis, respiratory haemorrhage and edema. The window of opportunity for successful patient treatment remains unknown, but is likely to be very narrow. Currently, very few effective therapeutic options are available for the case management of severe HeV infections or the rapid silencing of local outbreaks. This underscores the need for more activity in the drug discovery arena to develop much needed therapeutics that specifically targets this deadly disease. The structural analysis of HeV is very much in its infancy, which leaves many gaps in our understanding of the biology of HeV and makes structure-guided drug design difficult. Structural studies of the viral RNAdependent- RNA polymerase (RdRp), which is the heart of the viral replication machinery, will set the stage for rational drug design and fill a major gap in our understanding of the HeV replication machinery. This review examines the current knowledge based on the multi-domain architecture of the Hendra RdRp and highlights which essential domain functions represent tangible targets for drug development against this deadly disease.


Assuntos
Antivirais/farmacologia , Vírus Hendra/efeitos dos fármacos , RNA Polimerase Dependente de RNA/efeitos dos fármacos , Animais , Vírus Hendra/enzimologia , Humanos , RNA Polimerase Dependente de RNA/metabolismo
8.
Antiviral Res ; 100(1): 8-13, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23838047

RESUMO

Hendra virus and Nipah virus are bat-borne paramyxoviruses that are the prototypic members of the genus Henipavirus. The henipaviruses emerged in the 1990s, spilling over from their natural bat hosts and causing serious disease outbreaks in humans and livestock. Hendra virus emerged in Australia and since 1994 there have been 7 human infections with 4 case fatalities. Nipah virus first appeared in Malaysia and subsequent outbreaks have occurred in Bangladesh and India. In total, there have been an estimated 582 human cases of Nipah virus and of these, 54% were fatal. Their broad species tropism and ability to cause fatal respiratory and/or neurologic disease in humans and animals make them important transboundary biological threats. Recent experimental findings in animals have demonstrated that a human monoclonal antibody targeting the viral G glycoprotein is an effective post-exposure treatment against Hendra and Nipah virus infection. In addition, a subunit vaccine based on the G glycoprotein of Hendra virus affords protection against Hendra and Nipah virus challenge. The vaccine has been developed for use in horses in Australia and is the first vaccine against a Biosafety Level-4 (BSL-4) agent to be licensed and commercially deployed. Together, these advances offer viable approaches to address Hendra and Nipah virus infection of livestock and people.


Assuntos
Doenças dos Bovinos/tratamento farmacológico , Vírus Hendra/efeitos dos fármacos , Infecções por Henipavirus/tratamento farmacológico , Infecções por Henipavirus/veterinária , Vírus Nipah/efeitos dos fármacos , Vacinas Virais/administração & dosagem , Animais , Anticorpos Monoclonais/uso terapêutico , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/prevenção & controle , Vírus Hendra/genética , Vírus Hendra/imunologia , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/prevenção & controle , Humanos , Vírus Nipah/genética , Vírus Nipah/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
10.
J Virol ; 84(13): 6760-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20357085

RESUMO

The fusion of enveloped viruses with the host cell is driven by specialized fusion proteins to initiate infection. The "class I" fusion proteins harbor two regions, typically two heptad repeat (HR) domains, which are central to the complex conformational changes leading to fusion: the first heptad repeat (HRN) is adjacent to the fusion peptide, while the second (HRC) immediately precedes the transmembrane domain. Peptides derived from the HR regions can inhibit fusion, and one HR peptide, T20 (enfuvirtide), is in clinical use for HIV-1. For paramyxoviruses, the activities of two membrane proteins, the receptor-binding protein (hemagglutinin-neuraminidase [HN] or G) and the fusion protein (F), initiate viral entry. The binding of HN or G to its receptor on a target cell triggers the activation of F, which then inserts into the target cell and mediates the membrane fusion that initiates infection. We have shown that for paramyxoviruses, the inhibitory efficacy of HR peptides is inversely proportional to the rate of F activation. For HIV-1, the antiviral potency of an HRC-derived peptide can be dramatically increased by targeting it to the membrane microdomains where fusion occurs, via the addition of a cholesterol group. We report here that for three paramyxoviruses-human parainfluenza virus type 3 (HPIV3), a major cause of lower respiratory tract diseases in infants, and the emerging zoonotic viruses Hendra virus (HeV) and Nipah virus (NiV), which cause lethal central nervous system diseases-the addition of cholesterol to a paramyxovirus HRC-derived peptide increased antiviral potency by 2 log units. Our data suggest that this enhanced activity is indeed the result of the targeting of the peptide to the plasma membrane, where fusion occurs. The cholesterol-tagged peptides on the cell surface create a protective antiviral shield, target the F protein directly at its site of action, and expand the potential utility of inhibitory peptides for paramyxoviruses.


Assuntos
Antivirais/farmacologia , Vírus Hendra/fisiologia , Vírus Nipah/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Internalização do Vírus/efeitos dos fármacos , Animais , Antivirais/química , Linhagem Celular , Chlorocebus aethiops , Vírus Hendra/efeitos dos fármacos , Humanos , Vírus Nipah/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos
11.
J Gen Virol ; 91(Pt 3): 765-72, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19889926

RESUMO

Hendra virus (HeV) and Nipah virus (NiV) are recently emerged, closely related and highly pathogenic paramyxoviruses that cause severe disease such as encephalitis in animals and humans with fatality rates of up to 75 %. Due to their high case fatality rate following human infection and because of the lack of effective vaccines or therapy, they are classified as Biosafety Level 4 pathogens. A recent study reported that chloroquine, an anti-malarial drug, was effective in preventing NiV and HeV infection in cell culture experiments. In the present study, the antiviral efficacy of chloroquine was analysed, individually and in combination with ribavirin, in the treatment of NiV and HeV infection in in vivo experiments, using a golden hamster model. Although the results confirmed the strong antiviral activity of both drugs in inhibiting viral spread in vitro, they did not prove to be protective in the in vivo model. Ribavirin delayed death from viral disease in NiV-infected hamsters by approximately 5 days, but no significant effect in HeV-infected hamsters was observed. Chloroquine did not protect hamsters when administered either individually or in combination with ribavirin, the latter indicating the lack of a favourable drug-drug interaction.


Assuntos
Antivirais/uso terapêutico , Cloroquina/uso terapêutico , Infecções por Henipavirus/tratamento farmacológico , Infecções por Henipavirus/mortalidade , Ribavirina/uso terapêutico , Animais , Cloroquina/farmacologia , Cricetinae , Modelos Animais de Doenças , Quimioterapia Combinada , Vírus Hendra/efeitos dos fármacos , Humanos , Mesocricetus , Vírus Nipah/efeitos dos fármacos , Ribavirina/farmacologia , Análise de Sobrevida , Resultado do Tratamento
12.
PLoS One ; 4(12): e8266, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-20011515

RESUMO

BACKGROUND: Bats are the suspected natural reservoir hosts for a number of new and emerging zoonotic viruses including Nipah virus, Hendra virus, severe acute respiratory syndrome coronavirus and Ebola virus. Since the discovery of SARS-like coronaviruses in Chinese horseshoe bats, attempts to isolate a SL-CoV from bats have failed and attempts to isolate other bat-borne viruses in various mammalian cell lines have been similarly unsuccessful. New stable bat cell lines are needed to help with these investigations and as tools to assist in the study of bat immunology and virus-host interactions. METHODOLOGY/FINDINGS: Black flying foxes (Pteropus alecto) were captured from the wild and transported live to the laboratory for primary cell culture preparation using a variety of different methods and culture media. Primary cells were successfully cultured from 20 different organs. Cell immortalisation can occur spontaneously, however we used a retroviral system to immortalise cells via the transfer and stable production of the Simian virus 40 Large T antigen and the human telomerase reverse transcriptase protein. Initial infection experiments with both cloned and uncloned cell lines using Hendra and Nipah viruses demonstrated varying degrees of infection efficiency between the different cell lines, although it was possible to infect cells in all tissue types. CONCLUSIONS/SIGNIFICANCE: The approaches developed and optimised in this study should be applicable to bats of other species. We are in the process of generating further cell lines from a number of different bat species using the methodology established in this study.


Assuntos
Técnicas de Cultura de Células/métodos , Linhagem Celular Transformada/citologia , Quirópteros , Animais , Forma Celular/efeitos dos fármacos , Clonagem Molecular , Vírus Hendra/efeitos dos fármacos , Vírus Hendra/fisiologia , Infecções por Henipavirus/virologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Interferons/genética , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/fisiologia , Poli I-C/farmacologia , Vírus 40 dos Símios/genética
13.
Virol J ; 6: 187, 2009 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-19889218

RESUMO

BACKGROUND: Using a recently described monolayer assay amenable to high throughput screening format for the identification of potential Nipah virus and Hendra virus antivirals, we have partially screened a low molecular weight compound library (>8,000 compounds) directly against live virus infection and identified twenty eight promising lead molecules. Initial single blind screens were conducted with 10 microM compound in triplicate with a minimum efficacy of 90% required for lead selection. Lead compounds were then further characterised to determine the median efficacy (IC50), cytotoxicity (CC50) and the in vitro therapeutic index in live virus and pseudotype assay formats. RESULTS: While a number of leads were identified, the current work describes three commercially available compounds: brilliant green, gentian violet and gliotoxin, identified as having potent antiviral activity against Nipah and Hendra virus. Similar efficacy was observed against pseudotyped Nipah and Hendra virus, vesicular stomatitis virus and human parainfluenza virus type 3 while only gliotoxin inhibited an influenza A virus suggesting a non-specific, broad spectrum activity for this compound. CONCLUSION: All three of these compounds have been used previously for various aspects of anti-bacterial and anti-fungal therapy and the current results suggest that while unsuitable for internal administration, they may be amenable to topical antiviral applications, or as disinfectants and provide excellent positive controls for future studies.


Assuntos
Antivirais/farmacologia , Violeta Genciana/farmacologia , Gliotoxina/farmacologia , Vírus Hendra/efeitos dos fármacos , Vírus Nipah/efeitos dos fármacos , Compostos de Amônio Quaternário/farmacologia , Animais , Antivirais/química , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Genoma Viral/efeitos dos fármacos , Violeta Genciana/química , Gliotoxina/química , Estrutura Molecular , Vírus Nipah/genética , Compostos de Amônio Quaternário/química , Células Vero
14.
Virus Res ; 142(1-2): 92-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19428741

RESUMO

We have recently described the development and validation of a high throughput screening assay suitable for henipavirus antiviral identification. While we are confident this assay is robust and effective, we wished to investigate assay performance in a range of alternative cell lines to determine if assay sensitivity and specificity could be improved. We evaluated ten different cell lines for their susceptibility to Hendra and Nipah virus infection and their sensitivity of detection of the effects of the broad spectrum antiviral, ribavirin and nine novel antivirals identified using our initial screening approach. Cell lines were grouped into three categories with respect to viral replication. Virus replicated best in Vero and BSR cells, followed by Hep-2, HeLa, BHK-21 and M17 cells. The lowest levels of RNA replication and viral protein expression were observed in BAEC, MMEC, A549 and ECV304 cells. Eight cell lines appeared to be similarly effective at discriminating the antiviral effects of ribavirin (<2.7-fold difference). The two cells lines most sensitive to the effect of ribavirin (ECV304 and BAEC) also displayed the lowest levels of viral replication while Vero cells were the least sensitive suggesting excess viral replication may limit drug efficacy and cell lines which limit viral replication may result in enhanced antiviral efficacy. However, there was no consistent trend observed with the other nine antivirals tested. While improvements in antiviral sensitivity in other cell lines may indicate an important role in future HTS assays, the slightly lower sensitivity to antiviral detection in Vero cells has inherent advantages in reducing the number of partially effective lead molecules identified during initial screens. Comparison of a panel of 54 novel antiviral compounds identified during routine screening of an in-house compound library in Vero, BHK-21 and BSR cells suggests no clear advantage of screening in either cell type.


Assuntos
Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Vírus Hendra/fisiologia , Vírus Nipah/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Cobaias , Vírus Hendra/efeitos dos fármacos , Humanos , Camundongos , Vírus Nipah/efeitos dos fármacos , Células Vero
15.
J Virol ; 83(10): 5148-55, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19264786

RESUMO

Nipah (NiV) and Hendra (HeV) viruses are emerging zoonotic paramyxoviruses that cause encephalitis in humans, with fatality rates of up to 75%. We designed a new high-throughput screening (HTS) assay for inhibitors of infection based on envelope glycoprotein pseudotypes. The assay simulates multicycle replication and thus identifies inhibitors that target several stages of the viral life cycle, but it still can be carried out under biosafety level 2 (BSL-2) conditions. These features permit a screen for antivirals for emerging viruses and select agents that otherwise would require BSL-4 HTS facilities. The screening of a small compound library identified several effective molecules, including the well-known compound chloroquine, as highly active inhibitors of pseudotyped virus infection. Chloroquine inhibited infection with live HeV and NiV at a concentration of 1 microM in vitro (50% inhibitory concentration, 2 microM), which is less than the plasma concentrations present in humans receiving chloroquine treatment for malaria. The mechanism for chloroquine's antiviral action likely is the inhibition of cathepsin L, a cellular enzyme that is essential for the processing of the viral fusion glycoprotein and the maturation of newly budding virions. Without this processing step, virions are not infectious. The identification of a compound that inhibits a known cellular target that is important for viral maturation but that had not previously been shown to have antiviral activity for henipaviruses highlights the validity of this new screening assay. Given the established safety profile and broad experience with chloroquine in humans, the results described here provide an option for treating individuals infected by these deadly viruses.


Assuntos
Antivirais/farmacologia , Cloroquina/farmacologia , Descoberta de Drogas/métodos , Vírus Hendra/efeitos dos fármacos , Vírus Nipah/efeitos dos fármacos , Animais , Chlorocebus aethiops , Vírus Hendra/fisiologia , Infecções por Henipavirus/tratamento farmacológico , Humanos , Vírus Nipah/fisiologia , Células Vero , Proteínas do Envelope Viral/metabolismo , Replicação Viral
16.
J Virol Methods ; 149(1): 12-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18313148

RESUMO

There are currently no antiviral drugs approved for the highly lethal Biosafety Level 4 pathogens Nipah and Hendra virus. A number of researchers are developing surrogate assays amenable to Biosafety Level 2 biocontainment but ultimately, the development of a high throughput screening method for directly quantifying these viruses in a Biosafety Level 4 environment will be critical for final evaluation of antiviral drugs identified in surrogate assays, in addition to reducing the time required for effective antiviral drug development. By adapting an existing immunoplaque assay and using enzyme linked immunodetection in a microtitre plate format, the current experiments describe a simple two step assay protocol involving an overnight virus inoculation of Vero cell monolayers (with or without antiviral drug treatment) at Biosafety Level 4, followed by cell fixation and virus inactivation enabling removal of plates from the Biosafety Level 4 laboratory and a subsequent immunodetection assay using a chemiluminescent horse radish peroxidase substrate to be performed at Biosafety Level 2. The analytical sensitivity (limit of detection) of this assay is 100 tissue culture infectious dose50/ml of either Nipah or Hendra virus. In addition this assay enables linear quantitation of virus over three orders of magnitude and is unaffected by dimethyl sulfoxide concentrations of 1% or less. Intra-assay coefficients of variation are acceptable (less than 20%) when detecting a minimum of 1000 tissue culture infectious dose50/ml of either virus although inter-assay variation is considerably greater. By an assessment of efficacies of the broad spectrum antiviral Ribavirin and an experimental fusion inhibitory peptide, this assay reveals a good correlation with previously published fluorescent immunodetection assays. The current experiments describe for the first time, a high throughput screening method amenable for direct assessment of live henipavirus antiviral drug activity.


Assuntos
Antivirais/farmacologia , Vírus Hendra/isolamento & purificação , Imunoensaio/métodos , Medições Luminescentes/métodos , Vírus Nipah/isolamento & purificação , Animais , Chlorocebus aethiops , Vírus Hendra/efeitos dos fármacos , Vírus Nipah/efeitos dos fármacos , Sensibilidade e Especificidade , Células Vero
17.
J Virol ; 81(19): 10567-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652384

RESUMO

Hendra virus (HeV) and Nipah virus (NiV) constitute the Henipavirus genus of paramyxoviruses, both fatal in humans and with the potential for subversion as agents of bioterrorism. Binding of the HeV/NiV attachment protein (G) to its receptor triggers a series of conformational changes in the fusion protein (F), ultimately leading to formation of a postfusion six-helix bundle (6HB) structure and fusion of the viral and cellular membranes. The ectodomain of paramyxovirus F proteins contains two conserved heptad repeat regions, the first (the N-terminal heptad repeat [HRN]) adjacent to the fusion peptide and the second (the C-terminal heptad repeat [HRC]) immediately preceding the transmembrane domain. Peptides derived from the HRN and HRC regions of F are proposed to inhibit fusion by preventing activated F molecules from forming the 6HB structure that is required for fusion. We previously reported that a human parainfluenza virus 3 (HPIV3) F peptide effectively inhibits infection mediated by the HeV glycoproteins in pseudotyped-HeV entry assays more effectively than the comparable HeV-derived peptide, and we now show that this peptide inhibits live-HeV and -NiV infection. HPIV3 F peptides were also effective in inhibiting HeV pseudotype virus entry in a new assay that mimics multicycle replication. This anti-HeV/NiV efficacy can be correlated with the greater potential of the HPIV3 C peptide to interact with the HeV F N peptide coiled-coil trimer, as evaluated by thermal unfolding experiments. Furthermore, replacement of a buried glutamic acid (glutamic acid 459) in the C peptide with valine enhances antiviral potency and stabilizes the 6HB conformation. Our results strongly suggest that conserved interhelical packing interactions in the F protein fusion core are important determinants of C peptide inhibitory activity and offer a strategy for the development of more-potent analogs of F peptide inhibitors.


Assuntos
Antivirais/farmacologia , Henipavirus/efeitos dos fármacos , Mimetismo Molecular , Peptídeos/farmacologia , Fosfoproteínas/farmacologia , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas Virais/farmacologia , Internalização do Vírus/efeitos dos fármacos , Sequência de Aminoácidos , Antivirais/química , Linhagem Celular , Sequência Conservada , Vírus Hendra/efeitos dos fármacos , Vírus Hendra/fisiologia , Henipavirus/fisiologia , Humanos , Dados de Sequência Molecular , Mutação , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/fisiologia , Paramyxovirinae/efeitos dos fármacos , Peptídeos/química , Peptídeos/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas Virais/química , Proteínas Virais/genética
18.
Expert Rev Anti Infect Ther ; 4(1): 43-55, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16441208

RESUMO

Hendra and Nipah virus are closely related emerging viruses comprising the Henipavirus genus of the subfamily Paramyxovirinae and are distinguished by their ability to cause fatal disease in both animal and human hosts. In particular, the high mortality and person-to-person transmission associated with the most recent Nipah virus outbreaks, as well as the very recent re-emergence of Hendra virus, has confirmed the importance and necessity of developing effective therapeutic interventions. Much research conducted on the henipaviruses over the past several years has focused on virus entry, including the attachment of virus to the host cell, the identification of the virus receptor and the membrane fusion process between the viral and host cell membranes. These findings have led to the development of possible vaccine candidates, as well as potential antiviral therapeutics. The common link among all of the possible antiviral agents discussed here, which have also been developed and tested, is that they target very early stages of the infection process. The establishment and validation of suitable animal models of Henipavirus infection and pathogenesis are also discussed as they will be crucial in the assessment of the effectiveness of any treatments for Hendra and Nipah virus infection.


Assuntos
Antivirais/uso terapêutico , Infecções por Henipavirus/tratamento farmacológico , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Antivirais/química , Gatos , Cricetinae , Modelos Animais de Doenças , Cães , Desenho de Fármacos , Vírus Hendra/efeitos dos fármacos , Vírus Hendra/imunologia , Vírus Hendra/patogenicidade , Infecções por Henipavirus/fisiopatologia , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/virologia , Humanos , Camundongos , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/imunologia , Vírus Nipah/patogenicidade , Vacinas Virais/química , Vacinas Virais/uso terapêutico
19.
Arch Virol ; 150(3): 521-32, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15526144

RESUMO

Hendra virus is one of two virus species within the newly-formed genus Henipavirus, subfamily Paramxyovirinae. It is a designated select agent with potential biosecurity threat to both human and animal health. Quantitative real-time PCR was used to measure viral RNA synthesis in Vero cells infected by Hendra virus, and to examine the inhibitory effect of ribavirin. It was also used to determine the points of attenuation during transcription of the six viral genes N, P, M, F, G and L by targeting amplicons located towards the 3' end of each gene. Major increases in viral RNA and virus yield occurred between 4 to 8 h and 8 to 10 h post infection, respectively. The effect of ribavirin was examined at a range of concentrations up to 400 microm. At 50 microm, RNA synthesis was reduced 9 fold, and virus yield 58 fold. As expected for a member of the order Mononegavirales, a gradient of transcription was observed in Hendra virus-infected cells. There was significant attenuation at the M-F and G-L junctions, more closely resembling Sendai virus (genus Respirovirus) than measles virus (genus Morbillivirus).


Assuntos
Antivirais/farmacologia , Vírus Hendra/fisiologia , RNA Viral/biossíntese , Ribavirina/farmacologia , Animais , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Amplificação de Genes , Vírus Hendra/efeitos dos fármacos , Reação em Cadeia da Polimerase , RNA Viral/análise , Células Vero , Replicação Viral
20.
Viral Immunol ; 17(2): 210-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15279700

RESUMO

Interferon (IFN) can activate Signal Transducer and Activator of Transcription (STAT) proteins to establish a cellular antiviral response and inhibit virus replication. Many viruses have evolved strategies to inhibit this antiviral mechanism, but paramyxoviruses are unique in their abilities to directly target the IFN-responsive STAT proteins. Hendra virus and Nipah virus (Henipaviruses) are recently emerged paramyxoviruses that are the causative agents of fatal disease outbreaks in Australia and peninsular Malaysia. Similar to other paramyxoviruses, Henipaviruses inhibit IFN signal transduction through a virus-encoded protein called V. Recent studies have shown that Henipavirus V proteins target STAT proteins by inducing the formation of cytoplasmically localized high molecular weight STAT-containing complexes. This sequestration of STAT1 and STAT2 prevents STAT activation and blocks antiviral IFN signaling. As the V proteins are important factors for host evasion, they represent logical targets for therapeutics directed against Henipavirus epidemics.


Assuntos
Antígenos Virais/farmacologia , Vírus Hendra/química , Interferons/farmacologia , Vírus Nipah/química , Transdução de Sinais/efeitos dos fármacos , Proteínas do Capsídeo , Proteínas de Ligação a DNA/metabolismo , Vírus Hendra/efeitos dos fármacos , Humanos , Interferons/antagonistas & inibidores , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA