Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Gen Virol ; 102(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33258753

RESUMO

Bunyamwera (BUNV), Batai (BATV) and Ngari (NRIV) are mosquito-borne viruses that are members of the genus Orthobunyavirus in the order Bunyavirales. These three viruses are enveloped with single-stranded, negative-sense RNA genomes consiting of three segments, denoted as Small (S), Medium (M) and Large (L). Ngari is thought to be the natural reassortant progeny of Bunyamwera and Batai viruses. The relationship between these 'parental' viruses and the 'progeny' poses an interesting question, especially given that there is overlap in their respective transmission ecologies, but differences in their infection host ranges and pathogenesis. We compared the in vivo kinetics of these three viruses in a common laboratory system and found no significant difference in growth kinetics. There was, however, a tendency of BATV to have smaller plaques than either BUNV or NRIV. Furthermore, we determined that all three viruses are stable in extracellular conditions and retain infectivity for a week in non-cellular media, which has public health and biosafety implications. The study of this understudied group of viruses addresses a need for basic characterization of viruses that have not yet reached epidemic transmission intensity, but that have the potential due to their infectivity to both human and animal hosts. These results lay the groundwork for future studies of these neglected viruses of potential public and One Health importance.


Assuntos
Infecções por Bunyaviridae/virologia , Culicidae/virologia , Orthobunyavirus/crescimento & desenvolvimento , Orthobunyavirus/genética , Animais , Vírus Bunyamwera/classificação , Vírus Bunyamwera/genética , Genoma Viral , Orthobunyavirus/classificação , Filogenia , RNA Viral/genética
2.
PLoS Negl Trop Dis ; 13(11): e0007346, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31693659

RESUMO

BACKGROUND: Aedes aegypti is a vector mosquito of major public health importance, transmitting arthropod-borne viruses (arboviruses) such as chikungunya, dengue, yellow fever and Zika viruses. Wild mosquito populations are persistently infected at high prevalence with insect-specific viruses that do not replicate in vertebrate hosts. In experimental settings, acute infections with insect-specific viruses have been shown to modulate arbovirus infection and transmission in Ae. aegypti and other vector mosquitoes. However, the impact of persistent insect-specific virus infections, which arboviruses encounter more commonly in nature, has not been investigated extensively. Cell lines are useful models for studying virus-host interactions, however the available Ae. aegypti cell lines are poorly defined and heterogenous cultures. METHODOLOGY/PRINCIPLE FINDINGS: We generated single cell-derived clonal cell lines from the commonly used Ae. aegypti cell line Aag2. Two of the fourteen Aag2-derived clonal cell lines generated harboured markedly and consistently reduced levels of the insect-specific bunyavirus Phasi Charoen-like virus (PCLV) known to persistently infect Aag2 cells. In contrast to studies with acute insect-specific virus infections in cell culture and in vivo, we found that pre-existing persistent PCLV infection had no major impact on the replication of the flaviviruses dengue virus and Zika virus, the alphavirus Sindbis virus, or the rhabdovirus vesicular stomatitis virus. We also performed a detailed characterisation of the morphology, transfection efficiency and immune status of our Aag2-derived clonal cell lines, and have made a clone that we term Aag2-AF5 available to the research community as a well-defined cell culture model for arbovirus-vector interaction studies. CONCLUSIONS/SIGNIFICANCE: Our findings highlight the need for further in vivo studies that more closely recapitulate natural arbovirus transmission settings in which arboviruses encounter mosquitoes harbouring persistent rather than acute insect-specific virus infections. Furthermore, we provide the well-characterised Aag2-derived clonal cell line as a valuable resource to the arbovirus research community.


Assuntos
Aedes/virologia , Arbovírus/crescimento & desenvolvimento , Coinfecção/virologia , Mosquitos Vetores/virologia , Orthobunyavirus/crescimento & desenvolvimento , Replicação Viral , Alphavirus/crescimento & desenvolvimento , Animais , Arbovírus/genética , Sequência de Bases , Técnicas de Cultura de Células/métodos , Linhagem Celular , Vírus da Dengue/crescimento & desenvolvimento , Flavivirus/genética , Flavivirus/crescimento & desenvolvimento , Genoma Viral , Interações Hospedeiro-Patógeno/fisiologia , Orthobunyavirus/genética , Vírus de RNA/genética , Vírus de RNA/crescimento & desenvolvimento , Rhabdoviridae/crescimento & desenvolvimento , Sindbis virus/crescimento & desenvolvimento , Transfecção , Zika virus/crescimento & desenvolvimento
3.
Sci Rep ; 9(1): 12957, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506463

RESUMO

Carcinus maenas is in the top 100 globally invasive species and harbours a wide diversity of pathogens, including viruses. We provide a detailed description for a novel bunyavirus (Carcinus maenas Portunibunyavirus 1) infecting C. maenas from its native range in the Faroe Islands. The virus genome is tripartite, including large (L) (6766 bp), medium (M) (3244 bp) and small (S) (1608 bp) negative sense, single-stranded RNA segments. Individual genomic segments are flanked by 4 bp regions of similarity (CCUG). The segments encode an RNA-dependent RNA-polymerase, glycoprotein, non-structural protein with a Zinc-Finger domain and a nucleoprotein. Most show highest identity to the 'Wenling Crustacean Virus 9' from an unidentified crustacean host. Phylogenomics of crustacean-infecting bunyaviruses place them across multiple bunyavirus families. We discuss the diversity of crustacean bunyaviruses and provide an overview of how these viruses may affect the health and survival of crustacean hosts, including those inhabiting niches outside of their native range.


Assuntos
Braquiúros/virologia , Infecções por Bunyaviridae/virologia , Variação Genética , Genética Populacional , Genoma Viral , Orthobunyavirus/crescimento & desenvolvimento , Filogenia , Sequência de Aminoácidos , Animais , Braquiúros/genética , Infecções por Bunyaviridae/genética , Orthobunyavirus/genética , Orthobunyavirus/isolamento & purificação , Homologia de Sequência
4.
Emerg Microbes Infect ; 8(1): 381-395, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30896304

RESUMO

Orthobunyaviruses are arboviruses (Arthropod Borne Virus) and possess multipartite genomes made up of three negative RNAs corresponding to the small (S), medium (M) and large (L) segments. Reassortment and recombination are evolutionary driving forces of such segmented viruses and lead to the emergence of new strains and species. Retrospective studies based on phylogenetical analysis are able to evaluate these mechanisms at the end of the selection process but fail to address the dynamics of emergence. This issue was addressed using two Orthobunyaviruses infecting ruminants and belonging to the Simbu serogroup: the Sathuperi virus (SATV) and the Shamonda virus (SHAV). Both viruses were associated with abortion, stillbirth and congenital malformations occurring after transplacental transmission and were suspected to spread together in different ruminant and insect populations. This study showed that different viruses related to SHAV and SATV are spreading simultaneously in ruminants and equids of the Sub-Saharan region. Their reassortment and recombination potential was evaluated in mammalian and in insect contexts. A method was set up to determine the genomic background of any clonal progeny viruses isolated after in vitro coinfections assays. All the reassortment combinations were generated in both contexts while no recombinant virus was isolated. Progeny virus populations revealed a high level of reassortment in mammalian cells and a much lower level in insect cells. In vitro selection pressure that mimicked the host switching (insect-mammal) revealed that the best adapted reassortant virus was connected with an advantageous replicative fitness and with the presence of a specific segment.


Assuntos
Infecções por Bunyaviridae/virologia , Orthobunyavirus/genética , Orthobunyavirus/isolamento & purificação , Vírus Reordenados/genética , Vírus Reordenados/isolamento & purificação , Recombinação Genética , Animais , Linhagem Celular , Cricetinae , Insetos , Orthobunyavirus/crescimento & desenvolvimento , Vírus Reordenados/crescimento & desenvolvimento , Estudos Retrospectivos
5.
PLoS Negl Trop Dis ; 12(12): e0006993, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30532189

RESUMO

BACKGROUND: Shuni virus (SHUV) is an orthobunyavirus that belongs to the Simbu serogroup. SHUV was isolated from diverse species of domesticated animals and wildlife, and is associated with neurological disease, abortions, and congenital malformations. Recently, SHUV caused outbreaks among ruminants in Israel, representing the first incursions outside the African continent. The isolation of SHUV from a febrile child in Nigeria and seroprevalence among veterinarians in South Africa suggests that the virus may have zoonotic potential as well. The high pathogenicity, extremely broad tropism, potential transmission via both biting midges and mosquitoes, and zoonotic features warrants prioritization of SHUV for further research. Additional knowledge is essential to accurately determine the risk for animal and human health, and to assess the risk of future epizootics and epidemics. To gain first insights into the potential involvement of arthropod vectors in SHUV transmission, we have investigated the ability of SHUV to infect and disseminate in laboratory-reared biting midges and mosquitoes. METHODOLOGY/PRINCIPAL FINDINGS: Culicoides nubeculosus, C. sonorensis, Culex pipiens pipiens, and Aedes aegypti were orally exposed to SHUV by providing an infectious blood meal. Biting midges showed high infection rates of approximately 40-60%, whereas infection rates of mosquitoes were lower than 2%. SHUV successfully disseminated in both species of biting midges, but no evidence of transmission in orally exposed mosquitoes was found. CONCLUSIONS/SIGNIFICANCE: The results of this study show that different species of Culicoides biting midges are susceptible to infection and dissemination of SHUV, whereas the two mosquito species tested were found not to be susceptible.


Assuntos
Aedes/virologia , Infecções por Bunyaviridae/transmissão , Ceratopogonidae/fisiologia , Culex/virologia , Insetos Vetores/fisiologia , Mosquitos Vetores/fisiologia , Orthobunyavirus/fisiologia , Animais , Infecções por Bunyaviridae/virologia , Ceratopogonidae/virologia , Feminino , Humanos , Insetos Vetores/virologia , Mosquitos Vetores/virologia , Nigéria , Orthobunyavirus/crescimento & desenvolvimento , África do Sul
6.
PLoS Negl Trop Dis ; 12(10): e0006884, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30372452

RESUMO

Orthobunyaviruses such as Cache Valley virus (CVV) and Kairi virus (KRIV) are important animal pathogens. Periodic outbreaks of CVV have resulted in the significant loss of lambs on North American farms, whilst KRIV has mainly been detected in South and Central America with little overlap in geographical range. Vaccines or treatments for these viruses are unavailable. One approach to develop novel vaccine candidates is based on the use of reverse genetics to produce attenuated viruses that elicit immune responses but cannot revert to full virulence. The full genomes of both viruses were sequenced to obtain up to date genome sequence information. Following sequencing, minigenome systems and reverse genetics systems for both CVV and KRIV were developed. Both CVV and KRIV showed a wide in vitro cell host range, with BHK-21 cells a suitable host cell line for virus propagation and titration. To develop attenuated viruses, the open reading frames of the NSs proteins were disrupted. The recombinant viruses with no NSs protein expression induced the production of type I interferon (IFN), indicating that for both viruses NSs functions as an IFN antagonist and that such attenuated viruses could form the basis for attenuated viral vaccines. To assess the potential for reassortment between CVV and KRIV, which could be relevant during vaccination campaigns in areas of overlap, we attempted to produce M segment reassortants by reverse genetics. We were unable to obtain such viruses, suggesting that it is an unlikely event.


Assuntos
Infecções por Bunyaviridae/imunologia , Interações Hospedeiro-Patógeno , Orthobunyavirus/genética , Orthobunyavirus/imunologia , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Genética Reversa/métodos , Animais , Infecções por Bunyaviridae/virologia , Linhagem Celular , Técnicas de Inativação de Genes , Genoma Viral , Especificidade de Hospedeiro , Evasão da Resposta Imune , Imunidade Inata , Orthobunyavirus/crescimento & desenvolvimento , Vírus Reordenados/crescimento & desenvolvimento , Análise de Sequência de DNA , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/genética , Vacinas Virais/isolamento & purificação
7.
PLoS Pathog ; 14(5): e1007047, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29723305

RESUMO

Peribunyaviridae is a large family of RNA viruses with several members that cause mild to severe diseases in humans and livestock. Despite their importance in public heath very little is known about the host cell factors hijacked by these viruses to support assembly and cell egress. Here we show that assembly of Oropouche virus, a member of the genus Orthobunyavirus that causes a frequent arboviral infection in South America countries, involves budding of virus particles toward the lumen of Golgi cisternae. As viral replication progresses, these Golgi subcompartments become enlarged and physically separated from Golgi stacks, forming Oropouche viral factory (Vfs) units. At the ultrastructural level, these virally modified Golgi cisternae acquire an MVB appearance, and while they lack typical early and late endosome markers, they become enriched in endosomal complex required for transport (ESCRT) proteins that are involved in MVB biogenesis. Further microscopy and viral replication analysis showed that functional ESCRT machinery is required for efficient Vf morphogenesis and production of infectious OROV particles. Taken together, our results indicate that OROV attracts ESCRT machinery components to Golgi cisternae to mediate membrane remodeling events required for viral assembly and budding at these compartments. This represents an unprecedented mechanism of how viruses hijack host cell components for coordinated morphogenesis.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Orthobunyavirus/metabolismo , Orthobunyavirus/fisiologia , Técnicas de Cultura de Células , Complexos Endossomais de Distribuição Requeridos para Transporte/fisiologia , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Células HeLa , Humanos , Orthobunyavirus/crescimento & desenvolvimento , Orthobunyavirus/patogenicidade , Vírion/metabolismo , Montagem de Vírus/fisiologia , Liberação de Vírus/fisiologia , Replicação Viral/fisiologia
8.
J Vet Med Sci ; 78(9): 1391-1397, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27181086

RESUMO

The biological function of a nonstructural protein, NSm, of Akabane virus (AKAV) is unknown. In this study, we generated a series of NSm deletion mutant viruses by reverse genetics and compared their phenotypes. The mutant in which the NSm coding region was almost completely deleted could not be rescued, suggesting that NSm plays a role in virus replication. We next generated mutant viruses possessing various partial deletions in NSm and identified several regions critical for virus infectivity. All rescued mutant viruses produced smaller plaques and grew inefficiently in cell culture, compared to the wild-type virus. Interestingly, although the pathogenicity of NSm deletion mutant viruses varied in mice depending on their deletion regions and sizes, more than half the mice died following infection with any mutant virus and the dead mice exhibited encephalitis as in wild-type virus-inoculated mice, indicating their neuroinvasiveness. Abundant viral antigens were detected in the brain tissues of dead mice, whereas appreciable antigen was not observed in those of surviving mice, suggesting a correlation between virus growth rate in the brain and neuropathogenicity in mice. We conclude that NSm affects AKAV replication in vitro as well as in vivo and that it may function as a virulence factor.


Assuntos
Infecções por Bunyaviridae/virologia , Orthobunyavirus/crescimento & desenvolvimento , Proteínas não Estruturais Virais/fisiologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Infecções por Bunyaviridae/patologia , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C , Mutação/genética , Orthobunyavirus/genética , Orthobunyavirus/patogenicidade , Células Vero/virologia
9.
J Virol Methods ; 232: 16-20, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26927704

RESUMO

Akabane disease, caused by the insect-transmitted Akabane virus (AKAV), affects livestock by causing life-threatening deformities or mortality of fetuses. Therefore, Akabane disease has led to notable economic losses in numerous countries, including Japan. In this short communication, a new T7 RNA polymerase-based AKAV reverse genetics system was developed. Using this system, in which three plasmids transcribing antigenomic RNAs were transfected into cells stably expressing T7 polymerase, we successfully reconstituted the live attenuated vaccine TS-C2 strain (named rTTT), and also generated a mutant AKAV (rTTTΔNSs) that lacked the gene encoding the nonstructural NSs protein, which is regarded as a virulence factor. Analysis of growth kinetics revealed that rTTTΔNSs grew at a much slower rate than the rTTT and TS-C2 virus. These results suggest that our established reverse genetics system is a powerful tool that can be used for AKAV vaccine studies with gene-manipulated viruses.


Assuntos
Orthobunyavirus/genética , Orthobunyavirus/fisiologia , Genética Reversa/métodos , Animais , Infecções por Bunyaviridae/veterinária , Infecções por Bunyaviridae/virologia , Linhagem Celular , Japão , Mutação , Orthobunyavirus/crescimento & desenvolvimento , Orthobunyavirus/patogenicidade , Fatores de Virulência/genética
10.
J Biol Chem ; 291(7): 3411-22, 2016 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-26677217

RESUMO

Bunyaviruses are considered to be emerging pathogens facilitated by the segmented nature of their genome that allows reassortment between different species to generate novel viruses with altered pathogenicity. Bunyaviruses are transmitted via a diverse range of arthropod vectors, as well as rodents, and have established a global disease range with massive importance in healthcare, animal welfare, and economics. There are no vaccines or anti-viral therapies available to treat human bunyavirus infections and so development of new anti-viral strategies is urgently required. Bunyamwera virus (BUNV; genus Orthobunyavirus) is the model bunyavirus, sharing aspects of its molecular and cellular biology with all Bunyaviridae family members. Here, we show for the first time that BUNV activates and requires cellular potassium (K(+)) channels to infect cells. Time of addition assays using K(+) channel modulating agents demonstrated that K(+) channel function is critical to events shortly after virus entry but prior to viral RNA synthesis/replication. A similar K(+) channel dependence was identified for other bunyaviruses namely Schmallenberg virus (Orthobunyavirus) as well as the more distantly related Hazara virus (Nairovirus). Using a rational pharmacological screening regimen, two-pore domain K(+) channels (K2P) were identified as the K(+) channel family mediating BUNV K(+) channel dependence. As several K2P channel modulators are currently in clinical use, our work suggests they may represent a new and safe drug class for the treatment of potentially lethal bunyavirus disease.


Assuntos
Antivirais/farmacologia , Vírus Bunyamwera/efeitos dos fármacos , Infecções por Bunyaviridae/tratamento farmacológico , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores , Integração Viral/efeitos dos fármacos , Aedes , Animais , Vírus Bunyamwera/crescimento & desenvolvimento , Vírus Bunyamwera/fisiologia , Infecções por Bunyaviridae/metabolismo , Infecções por Bunyaviridae/virologia , Linhagem Celular , Chlorocebus aethiops , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Humanos , Mesocricetus , Nairovirus/efeitos dos fármacos , Nairovirus/crescimento & desenvolvimento , Nairovirus/fisiologia , Orthobunyavirus/efeitos dos fármacos , Orthobunyavirus/crescimento & desenvolvimento , Orthobunyavirus/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Células Vero
13.
Sci Rep ; 3: 1178, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23378911

RESUMO

During 2011 Schmallenberg virus (SBV) presented as a novel disease of cattle and sheep that had apparently spread through northern Europe over a relatively short period of time, but has yet to infect Scotland. This paper describes the development of a model of SBV spread applied to Scotland in the event of an incursion. This model shows that SBV spread is very sensitive to the temperature, with relatively little spread and few reproductive losses predicted in years with average temperatures but extensive spread (>1 million animals infected) and substantial reproductive losses in the hottest years. These results indicate that it is possible for SBV to spread in Scotland, however spread is limited by climatic conditions and the timing of introduction. Further results show that the transmission kernel shape and extrinsic incubation period parameter have a non-linear effect on disease transmission, so a greater understanding of the SBV transmission parameters is required.


Assuntos
Doenças dos Bovinos/epidemiologia , Orthobunyavirus/crescimento & desenvolvimento , Animais , Bovinos , Doenças dos Bovinos/transmissão , Doenças dos Bovinos/virologia , Insetos Vetores/virologia , Modelos Teóricos , Escócia/epidemiologia , Ovinos/virologia , Temperatura
14.
Zhonghua Yu Fang Yi Xue Za Zhi ; 46(2): 169-72, 2012 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-22490202

RESUMO

OBJECTIVE: To culture, isolate and identify new bunyavirus in Vero cell line. METHODS: Samples of 164 new bunyavirus positive by real time RT-PCR detection and well preserved serum specimens were selected from cases of fever, thrombocytopenia and leukopenia syndrome (FTLS) in Xinyang, Henan province in 2009 - 2011. These sera were cultured in Vero cell line and new bunyavirus were detected by observing cytopathic effect (CPE), Real-time RT-PCR, indirect immunofluorescence assay (IFA) and thin-section electron microscopy observation. A total of 10 positive PCR products were selected randomly for sequencing and the results were compared with sequence in Genbank. RESULTS: Among 164 FTLS serum specimens cultured in Vero cell line, no special CPE were observed and 67 strains (40.85%) were positive detected by Real-time RT-PCR. Nucleic acid similarity of 10 specimens were 97.8% - 100% and there's also a high similarity (> 99%) between specimens and new bunyavirus isolates (Accession No. HQ141600.1). Among 67 positive strains, 58 of them showed specific fluorescence particles by IFA. The viral particles were observed to be spheres with a diameter of 80 - 100 nm by electron microscopy. CONCLUSION: Vero cell line is suitable for culture, isolation and identification of new bunyavirus.


Assuntos
Orthobunyavirus/isolamento & purificação , Células Vero/virologia , Cultura de Vírus/métodos , Animais , Chlorocebus aethiops , Humanos , Orthobunyavirus/crescimento & desenvolvimento , Soro/virologia
15.
Antiviral Res ; 75(2): 121-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17368573

RESUMO

Oropouche, Caraparu, Guama, Guaroa and Tacaiuma viruses (Orthobunyavirus genus) cause human febrile illnesses and/or encephalitis. To achieve a therapeutical agent to prevent and/or treat these diseases we evaluated the antiviral action of Interferon-alpha (IFN-alpha) on these orthobunyaviruses. In vitro results showed that all the studied orthobunyaviruses are susceptible to antiviral action of IFN-alpha, but this susceptibility is limited and dependent on both concentration of drug and treatment period. In vivo results demonstrated that IFN-alpha present antiviral action on Oropouche and Guaroa viruses when used as a prophylactic treatment. Moreover, a treatment initiated 3h after infection prevented the death of Guaroa virus infected-mice. Additionally, mortality of mice was related to the migration and replication of viruses in their brains. Our results suggest that IFN-alpha could be potentially useful in the prevention of diseases caused by Oropouche virus and in the prevention and/or treatment of diseases caused by Guaroa virus.


Assuntos
Infecções por Bunyaviridae/tratamento farmacológico , Interferon-alfa/uso terapêutico , Orthobunyavirus/efeitos dos fármacos , Animais , Animais Lactentes , Antivirais/farmacologia , Antivirais/uso terapêutico , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Infecções por Bunyaviridae/mortalidade , Infecções por Bunyaviridae/prevenção & controle , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Interferon Tipo I/farmacologia , Interferon Tipo I/uso terapêutico , Interferon alfa-2 , Interferon-alfa/farmacologia , Camundongos , Orthobunyavirus/crescimento & desenvolvimento , Proteínas Recombinantes , Taxa de Sobrevida , Fatores de Tempo , Células Vero , Replicação Viral/efeitos dos fármacos
16.
Am J Trop Med Hyg ; 75(5): 1011-6, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17124004

RESUMO

Oropouche, Caraparu, Guama, Guaroa, and Tacaiuma are viruses (genus Orthobunyavirus) that cause human febrile illnesses and encephalitis. The goal of this study was to evaluate the antiviral action of ribavirin on these orthobunyaviruses to achieve a therapeutical agent to treat the diseases caused by these viruses. In vitro results showed that ribavirin (50 microg/mL) had antiviral activity only on the Tacaiuma virus. Addition of guanosine in the culture reversed the antiviral effect of ribavirin on Tacaiuma virus, suggesting that ribavirin inhibited this virus by reducing the intra-cellular guanosine pool. Moreover, ribavirin was not an effective drug in vivo because it was unable to inhibit the death of the mice or virus replication in the brain. The results suggest that ribavirin has no antiviral activity on the Oropouche, Caraparu, Guama, Guaroa, or Tacaiuma viruses; consequently, ribavirin would not be a good therapeutical agent to treat these arboviruses.


Assuntos
Antivirais/farmacologia , Infecções por Bunyaviridae/fisiopatologia , Orthobunyavirus/efeitos dos fármacos , Ribavirina/farmacologia , Animais , Linhagem Celular , Camundongos , Orthobunyavirus/crescimento & desenvolvimento
17.
Virology ; 348(1): 224-32, 2006 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-16445958

RESUMO

Maguari virus (MAGV; genus Orthobunyavirus, family Bunyaviridae) contains a tripartite negative-sense RNA genome. Like all orthobunyaviruses, the medium (M) genome segment encodes a precursor polyprotein (NH(2)-Gn-NSm-Gc-COOH) for the two virion glycoproteins Gn and Gc and a nonstructural protein NSm. The nucleotide sequences of the M segment of wild-type (wt) MAGV, of a temperature-sensitive (ts) mutant, and of two non-ts revertants, R1 and R2, that show electrophoretic mobility differences in their Gc proteins were determined. Twelve amino acid differences (2 in Gn, 10 in Gc) were observed between wt and ts MAGV, of which 9 were maintained in R1 and R2. The M RNA segments of R1 and R2 contained internal deletions, resulting in the removal of the N-terminal 239 residues of Gc (R1) or the C-terminal two thirds of NSm and the N-terminal 431 amino acids of Gc (R2). The sequence data were consistent with analyses of the virion RNAs and virion glycoproteins. These results suggest that neither the N-terminal domain of Gc nor an intact NSm protein is required for the replication of MAGV in tissue culture.


Assuntos
Mutação , Orthobunyavirus/crescimento & desenvolvimento , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , Cricetinae , DNA Complementar/química , DNA Complementar/genética , Eletroforese em Gel de Poliacrilamida , Dados de Sequência Molecular , RNA Viral/genética , Análise de Sequência de DNA , Deleção de Sequência , Proteínas não Estruturais Virais/química , Proteínas Virais/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA