Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Mol Microbiol ; 115(6): 1229-1243, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33325576

RESUMO

Wolbachia is an obligate intracellular bacterial symbiont prevalent among arthropods and nematodes. To survive and reproduce, Wolbachia interacts with and modifies host subcellular structures, while sensing and responding to changes within the cellular environment. In mutualistic associations, Wolbachia may provision the host with metabolites, or help to maintain the chemical homeostasis of the host cell. Some strains can rapidly invade insect populations by manipulating host reproductive biology, while also preventing viral replication, allowing their use in vector control of arthropod-borne viruses. The Aedes albopictus-derived strain wAlbB is promising in this regard. When transinfected into the Yellow fever mosquito, Aedes aegypti, wAlbB reaches high frequencies within wild populations, and strongly inhibits viral transmission. Despite its obvious potential, much is still unknown about the molecular interactions between Wolbachia and host that enable its use in vector control. Furthermore, most Wolbachia transinfection research to date has focused on host effects. In the current study, we used a cell line model to explore the effect of transinfection of wAlbB from Ae. albopictus to Ae. aegypti. Using RNA sequencing, we show that several genes associated with host-symbiont interactions were downregulated by transinfection, with the greatest downregulation exhibited by prophage-associated genes.


Assuntos
Aedes/microbiologia , Regulação Bacteriana da Expressão Gênica/genética , Simbiose/fisiologia , Wolbachia/genética , Wolbachia/metabolismo , Animais , Antibiose , Proteínas da Membrana Bacteriana Externa/biossíntese , Linhagem Celular , Regulação para Baixo/genética , Expressão Gênica/genética , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Polimorfismo de Nucleotídeo Único/genética , Trocadores de Sódio-Hidrogênio/biossíntese , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/virologia , Replicação Viral/fisiologia , Febre Amarela/transmissão , Vírus da Febre Amarela/crescimento & desenvolvimento
2.
Emerg Microbes Infect ; 9(1): 2256-2265, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32981479

RESUMO

ABSTRACT Mosquito-transmitted flaviviruses cause widespread disease across the world. To provide better molecular tools for drug screens and pathogenesis studies, we report a new approach to produce stable NanoLuc-tagged flaviviruses, including dengue virus serotypes 1-4, Japanese encephalitis virus, yellow fever virus, West Nile virus, and Zika virus. Since the reporter gene is often engineered at the capsid gene region, the capsid sequence must be duplicated to flank the reporter gene; such capsid duplication is essential for viral replication. The conventional approach for stabilizing reporter flaviviruses has been to shorten or modify the duplicated capsid sequence to minimize homologous recombination. No study has examined the effects of capsid duplication length on reporter virus stability. Here we report an optimal length to stabilize reporter flaviviruses. These viruses were stable after ten rounds of cell culture passaging, and in the case of stable NanoLuc-tagged Zika virus (ZIKV C38), the virus replicated to 107 FFU/ml in cell culture and produced robust luciferase signal after inoculation in mosquitoes. Mechanistically, the optimal length of capsid duplication may contain all the cis-acting RNA elements required for viral RNA replication, thus reducing the selection pressure for recombination. Together, these data describe an improved method of constructing optimal reporter flaviviruses.


Assuntos
Biotecnologia/métodos , Proteínas do Capsídeo/genética , Flavivirus/genética , Genes Reporter , Luciferases/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Culicidae , DNA Viral , Vírus da Dengue/genética , Vírus da Dengue/crescimento & desenvolvimento , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/crescimento & desenvolvimento , Flavivirus/crescimento & desenvolvimento , Infecções por Flavivirus/virologia , Humanos , Testes de Neutralização , Células Vero , Replicação Viral , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/crescimento & desenvolvimento , Zika virus/genética , Zika virus/crescimento & desenvolvimento
3.
Methods Mol Biol ; 2095: 141-168, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31858467

RESUMO

The global demand for complex biopharmaceuticals like recombinant proteins, vaccines, or viral vectors is steadily rising. To further improve process productivity and to reduce production costs, process intensification can contribute significantly. The design and optimization of perfusion processes toward very high cell densities require careful selection of strategies for optimal perfusion rate control. In this chapter, various options are discussed to guarantee high cell-specific virus yields and to achieve virus concentrations up to 1010 virions/mL. This includes reactor volume exchange regimes and perfusion rate control based on process variables such as cell concentration and metabolite or by-product concentration. Strategies to achieve high cell densities by perfusion rate control and their experimental implementation are described in detail for pseudo-perfusion or small-scale perfusion bioreactor systems. Suspension cell lines such as MDCK, BHK-21, EB66®, and AGE1.CR.pIX® are used to exemplify production of influenza, yellow fever, Zika, and modified vaccinia Ankara virus.


Assuntos
Técnicas de Cultura Celular por Lotes/instrumentação , Técnicas de Cultura Celular por Lotes/métodos , Reatores Biológicos , Perfusão/métodos , Vacinas/biossíntese , Cultura de Vírus/métodos , Animais , Contagem de Células , Linhagem Celular , Meios de Cultura/química , Meios de Cultura/metabolismo , Vírus da Influenza A/crescimento & desenvolvimento , Vacinas contra Influenza/biossíntese , Vacinas/imunologia , Vacinas/isolamento & purificação , Viroses , Replicação Viral , Vírus da Febre Amarela/crescimento & desenvolvimento , Zika virus/crescimento & desenvolvimento
4.
Mem Inst Oswaldo Cruz ; 114: e190187, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31826129

RESUMO

BACKGROUND: The Yellow Fever (YF) vaccine is produced by the inoculation of embryonated chicken eggs with YF17DD virus on the ninth day of development. Full embryos are collected on the twelfth day of development for vaccine formulation. Skeletal muscle tissue is the main site where biosynthesis of viral particles occurs. OBJECTIVES: This study aimed to analyse the experimental infection of skeletal muscle cells of chicken embryos by the 17DD Yellow Fever virus (YFV) in vivo and in vitro. METHODS: Chicken embryos infected with YF17DD virus were analysed by immunofluorescence using confocal and super-resolution microscopes. Primary cultures of skeletal muscle cells of non-infected chicken embryos were evaluated for susceptibility and permissiveness to YF17DD virus using different protocols. This evaluation was performed based on morphological, viral titration, molecular biology, and colorimetric techniques. FINDINGS: The present work phenotypically characterises embryonic chicken skeletal muscle cells as myogenic precursors expressing the Pax7 transcription factor in some cases. We demonstrated that these cells are susceptible to in vitro infection at different multiplicities of infection (MOIs), reproducing the same infection pattern observed in vivo. Furthermore, myogenic precursors and myoblasts are preferred infection targets, but establishment of infection does not depend on the presence of these cells. The peak of viral production occurred at 48 hpi, with decay occurring 72 hpi, when the cytopathic effect can be observed. MAIN CONCLUSIONS: In conclusion, the primary culture of chicken skeletal muscle cells is a good model for studying muscle cells infected with YF17DD virus. This culture system displays satisfactory emulation of the in vitro phenomenon observed, contributing to our understanding of virus infection dynamics and leading to the development of alternative methods of vaccine production.


Assuntos
Músculo Esquelético/virologia , Vacina contra Febre Amarela/imunologia , Vírus da Febre Amarela/imunologia , Animais , Células Cultivadas , Embrião de Galinha , Imunofluorescência , Cultura de Vírus , Replicação Viral/fisiologia , Vacina contra Febre Amarela/biossíntese , Vírus da Febre Amarela/crescimento & desenvolvimento
5.
PLoS Negl Trop Dis ; 13(8): e0007299, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31412040

RESUMO

BACKGROUND: To be transmitted to vertebrate hosts via the saliva of their vectors, arthropod-borne viruses have to cross several barriers in the mosquito body, including the midgut infection and escape barriers. Yellow fever virus (YFV) belongs to the genus Flavivirus, which includes human viruses transmitted by Aedes mosquitoes, such as dengue and Zika viruses. The live-attenuated YFV-17D vaccine has been used safely and efficiently on a large scale since the end of World War II. Early studies have shown, using viral titration from salivary glands of infected mosquitoes, that YFV-17D can infect Aedes aegypti midgut, but does not disseminate to other tissues. METHODOLOGY/PRINCIPAL FINDINGS: Here, we re-visited this issue using a panel of techniques, such as RT-qPCR, Western blot, immunofluorescence and titration assays. We showed that YFV-17D replication was not efficient in Aedes aegypti midgut, as compared to the clinical isolate YFV-Dakar. Viruses that replicated in the midgut failed to disseminate to secondary organs. When injected into the thorax of mosquitoes, viruses succeeded in replicating into midgut-associated tissues, suggesting that, during natural infection, the block for YFV-17D replication occurs at the basal membrane of the midgut. CONCLUSIONS/SIGNIFICANCE: The two barriers associated with Ae. aegypti midgut prevent YFV-17D replication. Our study contributes to our basic understanding of vector-pathogen interactions and may also aid in the development of non-transmissible live virus vaccines.


Assuntos
Aedes/virologia , Trato Gastrointestinal/virologia , Replicação Viral/efeitos dos fármacos , Vacina contra Febre Amarela/farmacologia , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/crescimento & desenvolvimento , Animais , Linhagem Celular , Trato Gastrointestinal/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Mosquitos Vetores , Glândulas Salivares , Vacinas Atenuadas , Carga Viral , Vírus da Febre Amarela/genética
6.
Vaccine ; 37(24): 3214-3220, 2019 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-31047674

RESUMO

Yellow fever (YF) is a high-lethality viral disease, endemic in tropical regions of South America and Africa, with a population of over 900 million people under risk. A highly effective attenuated vaccine, produced in embryonated eggs, has been used for about 80 years. However, egg-based production limits manufacturing capacity, and vaccine shortage led to the emergency use of a fractional dose (1/5) by the WHO in an outbreak in Africa in 2016 and by Brazilian authorities during an outbreak in 2018. In addition, rare but fatal adverse events of this vaccine have been reported since 2001. These two aspects make clear the need for the development of a new vaccine. In an effort to develop an inactivated YF vaccine, Bio-Manguinhos/FIOCRUZ started developing a new vaccine based on the production of the attenuated 17DD virus in serum-free conditions in Vero cells propagated in bioreactors, followed by chromatography-based purification and ß-propiolactone inactivation. Virus purification was studied in this work. Capture was performed using an anion-exchange membrane adsorber (Sartobind® Q), resulting in a virus recovery of 80.2 ±â€¯4.8% and a residual DNA level of 1.3 ±â€¯1.6 ng/dose, thus in accordance with the recommendations of the WHO (<10 ng/dose). However, the level of host cell proteins (HCP) was still high for a human vaccine, so a second chromatography step was developed based on a multimodal resin (Capto™ Core 700). This step resulted in a virus recovery of 65.7 ±â€¯4.8% and decreased HCP levels to 345 ±â€¯25 ppm. The overall virus recovery in these chromatography steps was 52.7%. SDS-PAGE of the purified sample showed a band with molecular mass of 56 kDa, thus consistent with the virus envelope protein (E) and corresponding to 96.7% of identified proteins. A Western blot stained with an antibody against the E protein showed a single band, confirming the identity of the sample.


Assuntos
Cromatografia/métodos , Cultura de Vírus , Vírus da Febre Amarela/isolamento & purificação , Animais , Chlorocebus aethiops , Vacinas de Produtos Inativados/análise , Células Vero , Vacina contra Febre Amarela , Vírus da Febre Amarela/crescimento & desenvolvimento
7.
Appl Microbiol Biotechnol ; 102(20): 8725-8737, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30091043

RESUMO

A live-attenuated, human vaccine against mosquito-borne yellow fever virus has been available since the 1930s. The vaccine provides long-lasting immunity and consistent mass vaccination campaigns counter viral spread. However, traditional egg-based vaccine manufacturing requires about 12 months and vaccine supplies are chronically close to shortages. In particular, for urban outbreaks, vaccine demand can be covered rarely by global stockpiling. Thus, there is an urgent need for an improved vaccine production platform, ideally transferable to other flaviviruses including Zika virus. Here, we present a proof-of-concept study regarding cell culture-based yellow fever virus 17D (YFV) and wild-type Zika virus (ZIKV) production using duck embryo-derived EB66® cells. Based on comprehensive studies in shake flasks, 1-L bioreactor systems were operated with scalable hollow fiber-based tangential flow filtration (TFF) and alternating tangential flow filtration (ATF) perfusion systems for process intensification. EB66® cells grew in chemically defined medium to cell concentrations of 1.6 × 108 cells/mL. Infection studies with EB66®-adapted virus led to maximum YFV titers of 7.3 × 108 PFU/mL, which corresponds to about 10 million vaccine doses for the bioreactor harvest. For ZIKV, titers of 1.0 × 1010 PFU/mL were achieved. Processes were automated successfully using a capacitance probe to control perfusion rates based on on-line measured cell concentrations. The use of cryo-bags for direct inoculation of production bioreactors facilitates pre-culture preparation contributing to improved process robustness. In conclusion, this platform is a powerful option for next generation cell culture-based flavivirus vaccine manufacturing.


Assuntos
Técnicas de Cultura de Células/métodos , Cultura de Vírus/métodos , Vírus da Febre Amarela/crescimento & desenvolvimento , Zika virus/crescimento & desenvolvimento , Animais , Reatores Biológicos/virologia , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Patos/virologia , Vacinas Virais/imunologia , Cultura de Vírus/instrumentação , Vírus da Febre Amarela/imunologia , Zika virus/imunologia
8.
Nat Commun ; 9(1): 2090, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844387

RESUMO

Evidence of male-to-female sexual transmission of Zika virus (ZIKV) and viral RNA in semen and sperm months after infection supports a potential role for testicular cells in ZIKV propagation. Here, we demonstrate that germ cells (GCs) are most susceptible to ZIKV. We found that only GCs infected by ZIKV, but not those infected by dengue virus and yellow fever virus, produce high levels of infectious virus. This observation coincides with decreased expression of interferon-stimulated gene Ifi44l in ZIKV-infected GCs, and overexpression of Ifi44l results in reduced ZIKV production. Using primary human testicular tissue, we demonstrate that human GCs are also permissive for ZIKV infection and production. Finally, we identified berberine chloride as a potent inhibitor of ZIKV infection in both murine and human testes. Together, these studies identify a potential cellular source for propagation of ZIKV in testes and a candidate drug for preventing sexual transmission of ZIKV.


Assuntos
Antivirais/farmacologia , Berberina/farmacologia , RNA Viral/análise , Doenças Virais Sexualmente Transmissíveis/prevenção & controle , Espermatozoides/virologia , Testículo/virologia , Replicação Viral/efeitos dos fármacos , Infecção por Zika virus/transmissão , Zika virus/crescimento & desenvolvimento , Animais , Antígenos/biossíntese , Proliferação de Células , Células Cultivadas , Chlorocebus aethiops , Proteínas do Citoesqueleto/biossíntese , Vírus da Dengue/crescimento & desenvolvimento , Humanos , Interferon Tipo I/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Viral/isolamento & purificação , Receptor de Interferon alfa e beta/genética , Doenças Virais Sexualmente Transmissíveis/virologia , Testículo/citologia , Células Vero , Replicação Viral/fisiologia , Vírus da Febre Amarela/crescimento & desenvolvimento , Zika virus/isolamento & purificação , Infecção por Zika virus/virologia
9.
Cell Rep ; 21(11): 3032-3039, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29241533

RESUMO

The mosquito-borne flaviviruses include important human pathogens such as dengue, Zika, West Nile, and yellow fever viruses, which pose a serious threat for global health. Recent genetic screens identified endoplasmic reticulum (ER)-membrane multiprotein complexes, including the oligosaccharyltransferase (OST) complex, as critical flavivirus host factors. Here, we show that a chemical modulator of the OST complex termed NGI-1 has promising antiviral activity against flavivirus infections. We demonstrate that NGI-1 blocks viral RNA replication and that antiviral activity does not depend on inhibition of the N-glycosylation function of the OST. Viral mutants adapted to replicate in cells deficient of the OST complex showed resistance to NGI-1 treatment, reinforcing the on-target activity of NGI-1. Lastly, we show that NGI-1 also has strong antiviral activity in primary and disease-relevant cell types. This study provides an example for advancing from the identification of genetic determinants of infection to a host-directed antiviral compound with broad activity against flaviviruses.


Assuntos
Antivirais/farmacologia , Benzamidas/farmacologia , Vírus da Dengue/efeitos dos fármacos , Hexosiltransferases/genética , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Proteínas de Membrana/genética , Sulfonamidas/farmacologia , Replicação Viral/efeitos dos fármacos , Vírus da Dengue/genética , Vírus da Dengue/crescimento & desenvolvimento , Expressão Gênica , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Hexosiltransferases/antagonistas & inibidores , Hexosiltransferases/deficiência , Humanos , Luciferases , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/deficiência , Testes de Sensibilidade Microbiana , Transdução de Sinais , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/crescimento & desenvolvimento , Zika virus/efeitos dos fármacos , Zika virus/genética , Zika virus/crescimento & desenvolvimento
10.
J Virol ; 91(13)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28404855

RESUMO

The molecular mechanisms underlying chikungunya virus (CHIKV) infection are poorly characterized. In this study, we analyzed the host factors involved in CHIKV infection using genome-wide screening. Human haploid HAP1 cells, into which an exon-trapping vector was introduced, were challenged with a vesicular stomatitis virus pseudotype bearing the CHIKV E3 to E1 envelope proteins. Analysis of genes enriched in the cells resistant to the pseudotyped virus infection unveiled a critical role of N-sulfation of heparan sulfate (HS) for the infectivity of the clinically isolated CHIKV Thai#16856 strain to HAP1 cells. Knockout of NDST1 that catalyzes N-sulfation of HS greatly decreased the binding and infectivity of CHIKV Thai#16856 strain but not infectivity of Japanese encephalitis virus (JEV) and yellow fever virus (YFV). While glycosaminoglycans were commonly required for the efficient infectivity of CHIKV, JEV, and YFV, as shown by using B3GAT3 knockout cells, the tropism for N-sulfate was specific to CHIKV. Expression of chondroitin sulfate (CS) in NDST1-knockout HAP1 cells did not restore the binding of CHIKV Thai#16856 strain and the infectivity of its pseudotype but restored the infectivity of authentic CHIKV Thai#16856, suggesting that CS functions at later steps after CHIKV binding. Among the genes enriched in this screening, we found that TM9SF2 is critical for N-sulfation of HS and therefore for CHIKV infection because it is involved in the proper localization and stability of NDST1. Determination of the significance of and the relevant proteins to N-sulfation of HS may contribute to understanding mechanisms of CHIKV propagation, cell tropism, and pathogenesis.IMPORTANCE Recent outbreaks of chikungunya fever have increased its clinical importance. Chikungunya virus (CHIKV) utilizes host glycosaminoglycans to bind efficiently to its target cells. However, the substructure in glycosaminoglycans required for CHIKV infection have not been characterized. Here, we unveil that N-sulfate in heparan sulfate is essential for the efficient infection of a clinical CHIKV strain to HAP1 cells and that chondroitin sulfate does not help the CHIKV binding but does play roles at the later steps in HAP1 cells. We show, by comparing previous reports using Chinese hamster ovary cells, along with another observation that enhanced infectivity of CHIKV bearing Arg82 in envelope E2 does not depend on glycosaminoglycans in HAP1 cells, that the infection manner of CHIKV varies among host cells. We also show that TM9SF2 is required for CHIKV infection to HAP1 cells because it is involved in the N-sulfation of heparan sulfate through ensuring NDST1 activity.


Assuntos
Vírus Chikungunya/fisiologia , Heparitina Sulfato/metabolismo , Proteínas de Membrana/genética , Sulfotransferases/genética , Ligação Viral , Linhagem Celular , Vírus Chikungunya/crescimento & desenvolvimento , Vírus da Encefalite Japonesa (Espécie)/crescimento & desenvolvimento , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Técnicas de Inativação de Genes , Testes Genéticos , Glucuronosiltransferase/genética , Humanos , Proteínas de Membrana/metabolismo , Sulfotransferases/metabolismo , Vírus da Febre Amarela/crescimento & desenvolvimento , Vírus da Febre Amarela/fisiologia
11.
Artigo em Inglês | MEDLINE | ID: mdl-27993855

RESUMO

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


Assuntos
Antivirais/farmacologia , Vírus Chikungunya/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Pirróis/farmacologia , Vírus da Floresta de Semliki/efeitos dos fármacos , Animais , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/virologia , Vírus Chikungunya/genética , Vírus Chikungunya/crescimento & desenvolvimento , Cricetinae , Farmacorresistência Viral/genética , Endossomos/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Expressão Gênica , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Indóis , Lisossomos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutação , Vermelho Neutro/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Vírus da Floresta de Semliki/genética , Vírus da Floresta de Semliki/crescimento & desenvolvimento , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/crescimento & desenvolvimento , Zika virus/efeitos dos fármacos , Zika virus/genética , Zika virus/crescimento & desenvolvimento
12.
Euro Surveill ; 21(39)2016 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-27719755

RESUMO

We assessed the ability of a French population of Aedes albopictus to transmit yellow fever virus (YFV). Batches of 30 to 40 female mosquitoes were analysed at 7, 14 and 21 days post-exposure (dpe). Bodies, heads and saliva were screened for YFV. Infectious viral particles were detected in bodies and heads at 7, 14 and 21 dpe whereas the virus was found in saliva only from 14 dpe. Our results showed that Ae. albopictus can potentially transmit YFV.


Assuntos
Aedes/virologia , Insetos Vetores/virologia , Febre Amarela/transmissão , Vírus da Febre Amarela/crescimento & desenvolvimento , Aedes/classificação , Animais , Feminino , Humanos , Saliva/virologia , Vírus da Febre Amarela/isolamento & purificação
13.
Virology ; 487: 1-10, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26479325

RESUMO

Neurotropic viruses initiate infection in peripheral tissues prior to entry into the central nervous system (CNS). However, mechanisms of dissemination are not completely understood. We used genetically marked viruses to compare dissemination of poliovirus, yellow fever virus 17D (YFV-17D), and reovirus type 3 Dearing in mice from a hind limb intramuscular inoculation site to the sciatic nerve, spinal cord, and brain. While YFV-17D likely entered the CNS via blood, poliovirus and reovirus likely entered the CNS by transport through the sciatic nerve to the spinal cord. We found that dissemination was inefficient in adult immune-competent mice for all three viruses, particularly reovirus. Dissemination of all viruses was more efficient in immune-deficient mice. Although poliovirus and reovirus both accessed the CNS by transit through the sciatic nerve, stimulation of neuronal transport by muscle damage enhanced dissemination only of poliovirus. Our results suggest that these viruses access the CNS using different pathways.


Assuntos
Sistema Nervoso Central/virologia , Orthoreovirus de Mamíferos/patogenicidade , Nervos Periféricos/virologia , Poliovirus/patogenicidade , Vírus da Febre Amarela/patogenicidade , Animais , Linhagem Celular , Cricetinae , Células HeLa , Humanos , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orthoreovirus de Mamíferos/crescimento & desenvolvimento , Poliomielite/patologia , Poliomielite/transmissão , Poliovirus/crescimento & desenvolvimento , Receptor de Interferon alfa e beta/genética , Infecções por Reoviridae/patologia , Infecções por Reoviridae/transmissão , Nervo Isquiático/virologia , Febre Amarela/patologia , Febre Amarela/transmissão , Vírus da Febre Amarela/crescimento & desenvolvimento
14.
PLoS Negl Trop Dis ; 9(9): e0004064, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26371874

RESUMO

The yellow fever (YF) 17D vaccine is one of the most effective human vaccines ever created. The YF vaccine has been produced since 1937 in embryonated chicken eggs inoculated with the YF 17D virus. Yet, little information is available about the infection mechanism of YF 17DD virus in this biological model. To better understand this mechanism, we infected embryos of Gallus gallus domesticus and analyzed their histopathology after 72 hours of YF infection. Some embryos showed few apoptotic bodies in infected tissues, suggesting mild focal infection processes. Confocal and super-resolution microscopic analysis allowed us to identify as targets of viral infection: skeletal muscle cells, cardiomyocytes, nervous system cells, renal tubular epithelium, lung parenchyma, and fibroblasts associated with connective tissue in the perichondrium and dermis. The virus replication was heaviest in muscle tissues. In all of these specimens, RT-PCR methods confirmed the presence of replicative intermediate and genomic YF RNA. This clearer characterization of cell targets in chicken embryos paves the way for future development of a new YF vaccine based on a new cell culture system.


Assuntos
Vacina contra Febre Amarela , Vírus da Febre Amarela/crescimento & desenvolvimento , Estruturas Animais/virologia , Animais , Embrião de Galinha , Histocitoquímica , Vacinas Atenuadas , Replicação Viral
15.
Vaccine ; 33(35): 4288-91, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25930117

RESUMO

In this work, changes in Vero cell cultivation methods have been employed in order to improve cell growth conditions to obtain higher viable cell densities and to increase viral titers. The propagation of the 17DD yellow fever virus (YFV) in Vero cells grown on Cytodex I microcarriers was evaluated in 3-L bioreactor vessels. Prior to the current changes, Vero cells were repeatedly displaying insufficient microcarrier colonization. A modified cultivation process with four changes has resulted in higher cell densities and higher virus titers than previously observed for 17DD YFV.


Assuntos
Reatores Biológicos , Contagem de Células , Técnicas de Cultura de Células , Cultura de Vírus/métodos , Vírus da Febre Amarela/crescimento & desenvolvimento , Animais , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Sobrevivência Celular , Chlorocebus aethiops , Meios de Cultura Livres de Soro , Células Vero , Carga Viral
16.
Vaccine ; 33(35): 4261-8, 2015 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25862300

RESUMO

Yellow fever is an acute infectious disease caused by prototype virus of the genus Flavivirus. It is endemic in Africa and South America where it represents a serious public health problem causing epidemics of hemorrhagic fever with mortality rates ranging from 20% to 50%. There is no available antiviral therapy and vaccination is the primary method of disease control. Although the attenuated vaccines for yellow fever show safety and efficacy it became necessary to develop a new yellow fever vaccine due to the occurrence of rare serious adverse events, which include visceral and neurotropic diseases. The new inactivated vaccine should be safer and effective as the existing attenuated one. In the present study, the immunogenicity of an inactivated 17DD vaccine in C57BL/6 mice was evaluated. The yellow fever virus was produced by cultivation of Vero cells in bioreactors, inactivated with ß-propiolactone, and adsorbed to aluminum hydroxide (alum). Mice were inoculated with inactivated 17DD vaccine containing alum adjuvant and followed by intracerebral challenge with 17DD virus. The results showed that animals receiving 3 doses of the inactivated vaccine (2 µg/dose) with alum adjuvant had neutralizing antibody titers above the cut-off of PRNT50 (Plaque Reduction Neutralization Test). In addition, animals immunized with inactivated vaccine showed survival rate of 100% after the challenge as well as animals immunized with commercial attenuated 17DD vaccine.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacina contra Febre Amarela/imunologia , Febre Amarela/prevenção & controle , Vírus da Febre Amarela/crescimento & desenvolvimento , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Reatores Biológicos/virologia , Chlorocebus aethiops , Desinfetantes/farmacologia , Imunidade Humoral , Esquemas de Imunização , Camundongos Endogâmicos C57BL , Testes de Neutralização , Propiolactona/farmacologia , Análise de Sobrevida , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Células Vero , Cultura de Vírus , Vacina contra Febre Amarela/administração & dosagem , Vírus da Febre Amarela/imunologia , Vírus da Febre Amarela/isolamento & purificação , Vírus da Febre Amarela/patogenicidade
17.
Eksp Klin Farmakol ; 77(10): 38-43, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25518527

RESUMO

Several novel compounds were found to be potent inhibitors of the HCV (JFH-1 isolate) infection in vitro. Human serum did not significantly reduce antiviral activity of the lead compound, AVR560 (< 4-fold). The immunohistochemistry studies with the Huh7 cell line, infectable with the HCV (JFH-1 strain), demonstrated that AVR560 inhibited the early steps of viral infection and blocked the spread of the HCV infection in tissue culture. The cytotoxicity in Huh7 and Vero-76 cell lines was mild. AVR560 proved to be a specific HCV inhibitor and exhibited no activity against other flaviviruses such as yellow fever (strain 17D), West Nile (strain NY99), and dengue (New Guinea type 2) in in vitro infection experiments. AVR560 also did not inhibit any of the tested human CYP450 isozymes (3A4, 1A2, 2C19 and 2D6). In the pharmacokinetic studies in mice, rats and dogs, favorable pharmacokinetic profiles and good oral bioavailability were observed for AV560. Further pre-clinical studies with this novel HCV inhibitor are in progress.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Piperazinas/farmacologia , Piperidinas/farmacologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Sistema Enzimático do Citocromo P-450/metabolismo , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/crescimento & desenvolvimento , Cães , Avaliação Pré-Clínica de Medicamentos , Hepacivirus/fisiologia , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Camundongos , Piperazinas/farmacocinética , Piperidinas/farmacocinética , Ratos , Células Vero , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/crescimento & desenvolvimento , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/crescimento & desenvolvimento
18.
Virus Res ; 176(1-2): 280-4, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23602827

RESUMO

Serial passaging of yellow fever virus 17D in Vero cells was employed to derive seed material for a novel inactivated vaccine, XRX-001. Two independent passaging series identified a novel lysine to arginine mutation at amino acid 160 of the envelope protein, a surface-exposed residue in structural domain I. A third passage series resulted in an isoleucine to methionine mutation at residue 113 of the NS4B protein, a central membrane spanning region of the protein which has previously been associated with Vero cell adaptation of other mosquito-borne flaviviruses. These studies confirm that flavivirus adaptation to growth in Vero cells can be mediated by structural or non-structural protein mutations.


Assuntos
Adaptação Biológica , Mutação de Sentido Incorreto , Inoculações Seriadas , Proteínas não Estruturais Virais/genética , Proteínas Estruturais Virais/genética , Vírus da Febre Amarela/crescimento & desenvolvimento , Vírus da Febre Amarela/genética , Substituição de Aminoácidos , Animais , Chlorocebus aethiops , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , RNA Viral/genética , Análise de Sequência de DNA , Células Vero
19.
Biologicals ; 40(6): 399-404, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23034357

RESUMO

Yellow fever is a disease caused by the prototype virus of the genus Flavivirus and remains endemic in tropical forest regions from Africa and South America, despite the availability of effective vaccines. These are capable of inducing a rapid specific immune response, with the formation of neutralizing antibodies that appear early, are protective and long lasting. The Plaque Reduction Neutralization Test is considered the most sensitive and specific test for quantification of neutralizing antibodies, and the reference method for assessing the protective immune response after vaccination. This study evaluated the reliability (repeatability and reproducibility) and accuracy (sensitivity, specificity and overall accuracy) of micro-PRNT50 and compared its performance with the micro-PRNT90. Although the micro-PRNT50 has showed satisfactory levels of reliability (ICCs ranged from 0.62 to 0.NorNormas e Manuais Técnicosas e Manuais Técnicos6 for repeatability and 0.72 for reproducibility) and accuracy (sensitivity of 91.1%, specificity of 72.9% and overall accuracy of 78%), the micro-PRNT90 showed higher performance, with ICCs for repeatability ranged from 0.78 to 0.79 and 0.81 for reproducibility, sensitivity of 100%, specificity of 94.7% and overall accuracy of 95%. Modifications in the test methodology and changes in the classification criteria in the readings of the results obtained will be important to improve the accuracy of micro-PRNT.


Assuntos
Anticorpos Antivirais/análise , Testes de Neutralização , Ensaio de Placa Viral , Vírus da Febre Amarela/imunologia , Humanos , Limite de Detecção , Reprodutibilidade dos Testes , Vírus da Febre Amarela/crescimento & desenvolvimento
20.
PLoS One ; 7(9): e41707, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028428

RESUMO

Primary clinical isolates of yellow fever virus can be difficult to quantitate by standard in vitro methods because they may not form discernable plaques or induce a measurable cytopathic effect (CPE) on cell monolayers. In our hands, the Dakar strain of yellow fever virus (YFV-Dakar) could not be measured by plaque assay (PA), focus-forming assay (FFA), or by measurement of CPE. For these reasons, we developed a YFV-specific monoclonal antibody (3A8.B6) and used it to optimize a highly sensitive flow cytometry-based tissue culture limiting dilution assay (TC-LDA) to measure levels of infectious virus. The TC-LDA was performed by incubating serial dilutions of virus in replicate wells of C6/36 cells and stained intracellularly for virus with MAb 3A8.B6. Using this approach, we could reproducibly quantitate YFV-Dakar in tissue culture supernatants as well as from the serum of viremic rhesus macaques experimentally infected with YFV-Dakar. Moreover, the TC-LDA approach was >10-fold more sensitive than standard plaque assay for quantitating typical plaque-forming strains of YFV including YFV-17D and YFV-FNV (French neurotropic vaccine). Together, these results indicate that the TC-LDA technique is effective for quantitating both plaque-forming and non-plaque-forming strains of yellow fever virus, and this methodology may be readily adapted for the study and quantitation of other non-plaque-forming viruses.


Assuntos
Citometria de Fluxo , Carga Viral/métodos , Vírus da Febre Amarela/crescimento & desenvolvimento , Aedes , Animais , Anticorpos Antivirais , Linhagem Celular , Feminino , Macaca mulatta , Masculino , Camundongos , Ensaio de Placa Viral , Vírus da Febre Amarela/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...