Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nature ; 596(7873): 558-564, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34408324

RESUMEN

Viral pathogens are an ongoing threat to public health worldwide. Analysing their dependence on host biosynthetic pathways could lead to effective antiviral therapies1. Here we integrate proteomic analyses of polysomes with functional genomics and pharmacological interventions to define how enteroviruses and flaviviruses remodel host polysomes to synthesize viral proteins and disable host protein production. We find that infection with polio, dengue or Zika virus markedly modifies polysome composition, without major changes to core ribosome stoichiometry. These viruses use different strategies to evict a common set of translation initiation and RNA surveillance factors from polysomes while recruiting host machineries that are specifically required for viral biogenesis. Targeting these specialized viral polysomes could provide a new approach for antiviral interventions. For example, we find that both Zika and dengue use the collagen proline hydroxylation machinery to mediate cotranslational modification of conserved proline residues in the viral polyprotein. Genetic or pharmacological inhibition of proline hydroxylation impairs nascent viral polyprotein folding and induces its aggregation and degradation. Notably, such interventions prevent viral polysome remodelling and lower virus production. Our findings delineate the modular nature of polysome specialization at the virus-host interface and establish a powerful strategy to identify targets for selective antiviral interventions.


Asunto(s)
Flavivirus/crecimiento & desarrollo , Flavivirus/metabolismo , Interacciones Huésped-Patógeno , Hidroxilación , Procolágeno-Prolina Dioxigenasa/metabolismo , Prolina/metabolismo , Biosíntesis de Proteínas , Línea Celular , Colágeno/química , Colágeno/metabolismo , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Flavivirus/química , Regulación Viral de la Expresión Génica , Genómica , Factores Celulares Derivados del Huésped/antagonistas & inhibidores , Factores Celulares Derivados del Huésped/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Sitios Internos de Entrada al Ribosoma , Chaperonas Moleculares/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Poliovirus/genética , Poliovirus/crecimiento & desarrollo , Polirribosomas/química , Polirribosomas/metabolismo , Agregado de Proteínas , Pliegue de Proteína , Mapas de Interacción de Proteínas , Proteolisis , Proteómica , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
2.
Front Immunol ; 12: 694959, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34421904

RESUMEN

Avian Tembusu virus (TMUV) is a novel flavivirus causing severe egg drop and fatal encephalitis in avian in Asia. In the present study, we screened the structural and functional requirements of TMUV capsid protein (CP) for viral morphogenesis using reverse genetics methods in combination with replicon packaging assays. TMUV-CP showed dramatic functional and structural flexibility, and even though 44 residues were removed from the N-terminus, it was still capable of packaging replicon RNA; in addition, 33 residues were deleted from the C-terminus (containing nearly the entire α4-helix), and infectious particles were still produced, although α4-α4' is supposedly vital for CP dimerization and nucleocapsid formation. We further analyzed two mutants (ΔC20-43 and ΔC64-96 viruses) with relatively large deletions that still replicated well in BHK-21 cells. Our data indicate that internal deletions within CP impaired viral replication or assembly, resulting in attenuated virus proliferation in cells and attenuated virulence in duck embryos, and these deletion mutations are quite stable in cell culture. An in vivo assay indicated that both ΔC20-43 virus and ΔC64-96 virus were highly attenuated in ducklings but still immunogenic. Single-dose immunization with ΔC20-43 virus or ΔC64-96 virus could protect ducklings from a lethal challenge with good antigen clearance. Together, our data shed light on replication/assembly defective TMUV with internal deletions in CP and provide an effective approach to attenuate viral virulence in live vaccines without changing the antigen composition.


Asunto(s)
Proteínas de la Cápside/genética , Infecciones por Flavivirus/prevención & control , Flavivirus/genética , Enfermedades de las Aves de Corral/prevención & control , Eliminación de Secuencia , Vacunas Virales/genética , Ensamble de Virus/genética , Replicación Viral/genética , Animales , Proteínas de la Cápside/inmunología , Línea Celular , Cricetinae , Patos , Flavivirus/crecimiento & desarrollo , Flavivirus/inmunología , Flavivirus/patogenicidad , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Inmunogenicidad Vacunal , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Vacunación , Vacunas Vivas no Atenuadas/administración & dosificación , Vacunas Vivas no Atenuadas/genética , Vacunas Vivas no Atenuadas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Virulencia
3.
Sci Rep ; 11(1): 4674, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33633167

RESUMEN

Classical insect-flaviviruses (cISFVs) and dual host-related insect-specific flavivirus (dISFV) are within the major group of insect-specific flavivirus. Remarkably dISFV are evolutionarily related to some of the pathogenic flavivirus, such as Zika and dengue viruses. The Evolutionary relatedness of dISFV to flavivirus allowed us to investigate the evolutionary principle of host adaptation. Additionally, dISFV can be used for the development of flavivirus vaccines and to explore underlying principles of mammalian pathogenicity. Here we describe the genetic characterization of a novel putative dISFV, termed Guapiaçu virus (GUAPV). Distinct strains of GUAPV were isolated from pools of Aedes terrens and Aedes scapularis mosquitoes. Additionally, we also detected viral GUAPV RNA in a plasma sample of an individual febrile from the Amazon region (North of Brazil). Although GUAPV did not replicate in tested mammalian cells, 3'UTR secondary structures duplication and codon usage index were similar to pathogenic flavivirus.


Asunto(s)
Aedes/virología , Flavivirus/aislamiento & purificación , Mosquitos Vectores/virología , Regiones no Traducidas 3' , Aedes/clasificación , Animales , Secuencia de Bases , Línea Celular , Evolución Molecular , Flavivirus/genética , Flavivirus/crecimiento & desarrollo , Genoma Viral , Humanos , Filogenia , ARN Viral/sangre , Especificidad de la Especie
4.
Avian Pathol ; 50(2): 124-131, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33146547

RESUMEN

Several phylogenetic clusters of duck Tembusu virus (DTMUV) that caused outbreaks in ducks in Asia have been identified since its emergence in 2010, highlighting the need for an efficient host system that can support isolation of all circulating phylogenetic clusters of DTMUV. In this study, various host systems, including different avian embryonated eggs (duck and chicken) and cell cultures (primary duck embryo fibroblast (DEF), primary chicken embryo fibroblast (CEF), baby hamster kidney (BHK-21), African green monkey kidney (Vero) and Aedes albopictus clone C6/36 (C6/36) cells), were evaluated and compared for their ability to support DTMUV isolation and propagation. Our results showed that all host systems were susceptible to DTMUV infection; however, BHK-21 and primary DEF cells supported more efficient replication of DTMUV compared to the other host systems. BHK-21 cells had the highest DTMUV isolation rate when tested with experimental and field clinical samples. All circulating phylogenetic clusters of DTMUV, including clusters 1, 2 and 3, were successfully isolated from duck clinical samples using BHK-21 cells. In conclusion, our findings supported the use of BHK-21 cells as a host system for primary isolation of all circulating phylogenetic clusters of DTMUV from duck clinical samples. This study highlights the importance of selecting the most appropriate host system for efficient isolation and propagation of DTMUV from duck clinical samples.RESEARCH HIGHLIGHTS DTMUV replicated more efficiently in BHK-21 and primary DEF cells than in other host systems tested.BHK-21 cells had the highest DTMUV isolation rate.All DTMUV phylogenetic clusters were successfully isolated from the samples using BHK-21 cells.BHK-21 cells were the most efficient host system for DTMUV isolation.


Asunto(s)
Técnicas de Cultivo de Célula/veterinaria , Pollos/virología , Patos/virología , Infecciones por Flavivirus/veterinaria , Flavivirus/crecimiento & desarrollo , Enfermedades de las Aves de Corral/virología , Animales , Embrión de Pollo , Chlorocebus aethiops , Cricetinae , Fibroblastos/virología , Flavivirus/genética , Infecciones por Flavivirus/virología , Filogenia , Células Vero
5.
Emerg Microbes Infect ; 9(1): 2256-2265, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32981479

RESUMEN

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.


Asunto(s)
Biotecnología/métodos , Proteínas de la Cápside/genética , Flavivirus/genética , Genes Reporteros , Luciferasas/genética , Animales , Línea Celular , Chlorocebus aethiops , Culicidae , ADN Viral , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Flavivirus/crecimiento & desarrollo , Infecciones por Flavivirus/virología , Humanos , Pruebas de Neutralización , Células Vero , Replicación Viral , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/crecimiento & desarrollo , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
6.
Viruses ; 12(6)2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32575394

RESUMEN

Zika virus (ZIKV) is a mosquito-borne pathogen that caused a large outbreak in the Americas in 2015 and 2016. The virus is currently present in tropical areas around the globe and can cause severe disease in humans, including Guillain-Barré syndrome and congenital microcephaly. The tropical yellow fever mosquito, Aedes aegypti, is the main vector in the urban transmission cycles of ZIKV. The discovery of ZIKV in wild-caught Culex mosquitoes and the ability of Culex quinquefasciatus mosquitoes to transmit ZIKV in the laboratory raised the question of whether the common house mosquito Culex pipiens, which is abundantly present in temperate regions in North America, Asia and Europe, could also be involved in ZIKV transmission. In this study, we investigated the vector competence of Cx. pipiens (biotypes molestus and pipiens) from the Netherlands for ZIKV, using Usutu virus as a control. After an infectious blood meal containing ZIKV, none of the tested mosquitoes accumulated ZIKV in the saliva, although 2% of the Cx. pipiens pipiens mosquitoes showed ZIKV-positive bodies. To test the barrier function of the mosquito midgut on virus transmission, ZIKV was forced into Cx. pipiens mosquitoes by intrathoracic injection, resulting in 74% (molestus) and 78% (pipiens) ZIKV-positive bodies. Strikingly, 14% (molestus) and 7% (pipiens) of the tested mosquitoes accumulated ZIKV in the saliva after injection. This is the first demonstration of ZIKV accumulation in the saliva of Cx. pipiens upon forced infection. Nevertheless, a strong midgut barrier restricted virus dissemination in the mosquito after oral exposure and we, therefore, consider Cx. pipiens as a highly inefficient vector for ZIKV.


Asunto(s)
Culex/virología , Saliva/virología , Virus Zika/crecimiento & desarrollo , Virus Zika/aislamiento & purificación , Animales , Línea Celular , Chlorocebus aethiops , Flavivirus/crecimiento & desarrollo , Inyecciones , Mosquitos Vectores/virología , Países Bajos , Especificidad de la Especie , Células Vero , Infección por el Virus Zika/transmisión
7.
Viruses ; 12(5)2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32455871

RESUMEN

We identified and isolated a novel Tembusu virus (TMUV) strain TP1906 (TMUV-TP1906) from a Culexannulus mosquito pool collected from the northern part of Taiwan in 2019. The TMUV-TP1906 genome is a 10,990-nucleotide-long, positive-sense, single-stranded RNA, consisting of a single open reading frame (ORF) encoding a polyprotein of 3425 amino acids, with 5' and 3' untranslated regions (UTRs) of 94 and 618 nucleotides, respectively. The nucleotide sequence of the TMUV-TP1906 of ORF exhibited 93.71% and 91.27% similarity with Sitiawan virus (STWV) and the TMUV prototype strain MM1775, respectively. The 3'-UTR variable region of TMUV-TP1906 showed nucleotide sequence divergence with other TMUV strains. Phylogenetic analysis of the complete ORF and polyprotein sequences revealed that TMUV-TP1906 is most closely related to STWV which causes encephalitis and retarded growth in chickens. We found that the TMUV-TP1906 caused a cytopathic effect (CPE) in the DF-1 chicken fibroblast cell line, while no apparent CPE was observed in Vero and C6/36 cells. In this study, we first identified and isolated a novel TMUV strain in Taiwan. In addition, to our knowledge, it is the first time that the TMUV strain was isolated from the Cx. annulus mosquitoes. Further study is warranted to investigate the host range and virulence of TMUV-TP1906.


Asunto(s)
Flavivirus/clasificación , Flavivirus/genética , Flavivirus/aislamiento & purificación , Genoma Viral , Animales , Secuencia de Bases , Línea Celular , Pollos , Chlorocebus aethiops , Culicidae/virología , Fibroblastos , Flavivirus/crecimiento & desarrollo , Infecciones por Flavivirus/veterinaria , Infecciones por Flavivirus/virología , Especificidad del Huésped , Cinética , Sistemas de Lectura Abierta , Filogenia , Poliproteínas/genética , Enfermedades de las Aves de Corral/virología , Taiwán , Células Vero , Secuenciación Completa del Genoma
8.
PLoS Negl Trop Dis ; 14(4): e0008223, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32324736

RESUMEN

Usutu virus (USUV), an African mosquito-borne flavivirus closely related to West Nile virus, was first isolated in South Africa in 1959. USUV emerged in Europe two decades ago, causing notably massive mortality in Eurasian blackbirds. USUV is attracting increasing attention due to its potential for emergence and its rapid spread in Europe in recent years. Although mainly asymptomatic or responsible for mild clinical signs, USUV was recently described as being associated with neurological disorders in humans such as encephalitis and meningoencephalitis, highlighting the potential health threat posed by the virus. Despite this, USUV pathogenesis remains largely unexplored. The aim of this study was to evaluate USUV neuropathogenicity using in vivo and in vitro approaches. Our results indicate that USUV efficiently replicates in the murine central nervous system. Replication in the spinal cord and brain is associated with recruitment of inflammatory cells and the release of inflammatory molecules as well as induction of antiviral-responses without major modulation of blood-brain barrier integrity. Endothelial cells integrity is also maintained in a human model of the blood-brain barrier despite USUV replication and release of pro-inflammatory cytokines. Furthermore, USUV-inoculated mice developed major ocular defects associated with inflammation. Moreover, USUV efficiently replicates in human retinal pigment epithelium. Our results will help to better characterize the physiopathology related to USUV infection in order to anticipate the potential threat of USUV emergence.


Asunto(s)
Flavivirus/patogenicidad , Modelos Biológicos , Sistema Nervioso/virología , Animales , Encéfalo/virología , Modelos Animales de Enfermedad , Células Endoteliales/virología , Células Epiteliales/virología , Flavivirus/crecimiento & desarrollo , Humanos , Ratones , Epitelio Pigmentado Ocular/virología , Médula Espinal/virología
9.
PLoS One ; 15(4): e0232274, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32330205

RESUMEN

The Flaviviridae is a family of enveloped viruses with a positive-sense single-stranded RNA genome. It contains many viruses that threaten human health, such as Japanese encephalitis virus (JEV) and yellow fever virus (YFV) of the genus Flavivirus as well as hepatitis C virus of the genus Hepacivirus. Cell culture systems highly permissive for the Flaviviridae viruses are very useful for their isolation, propagation, and diagnosis, an understanding of their biology, and the development of vaccines and antiviral agents. Previously, we isolated a human hepatoma HuH-7-derived cell clone, Huh7.5.1-8, which is highly permissive to hepatitis C virus infection. Here, we have characterized flavivirus infection in the Huh7.5.1-8 cell line by comparing with that in the African green monkey kidney-derived Vero cell line, which is permissive for a wide spectrum of viruses. Upon infection with JEV, Huh7.5.1-8 cells produced a higher amount of virus particles early in infection and were more susceptible to virus-induced cell death than Vero cells. Similar outcomes were obtained when the cells were infected with another flavivirus, YFV (17D-204 strain). Quantification of cellular and extracellular viral RNA revealed that high JEV production in Huh7.5.1-8 cells can be attributed to rapid viral replication kinetics and efficient virus release early in infection. In a plaque assay, Huh7.5.1-8 cells developed JEV plaques more rapidly than Vero cells. Although this was not the case with YFV plaques, Huh7.5.1-8 cells developed higher numbers of YFV plaques than Vero cells. Sequence analysis of cDNA encoding an antiviral RNA helicase, RIG-I, showed that Huh7.5.1-8 cells expressed not only a full-length RIG-I mRNA with a known dominant-negative missense mutation but also variants without the mutation. However, the latter mRNAs lacked exon 5/6-12, indicating functional loss of RIG-I in the cells. These characteristics of the Huh7.5.1-8 cell line are helpful for flavivirus detection, titration, and propagation.


Asunto(s)
Carcinoma Hepatocelular/virología , Chlorocebus aethiops/virología , Flavivirus/crecimiento & desarrollo , Animales , Línea Celular , Línea Celular Tumoral , Flavivirus/genética , Infecciones por Flavivirus/virología , Hepacivirus/genética , Humanos , ARN Viral/genética , Células Vero , Replicación Viral/genética
10.
Parasit Vectors ; 13(1): 54, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041638

RESUMEN

BACKGROUND: Wolbachia pipientis are bacterial endosymbionts of arthropods currently being implemented as biocontrol agents to reduce the global burden of arboviral diseases. Some strains of Wolbachia, when introduced into Aedes aegypti mosquitoes, reduce or block the replication of RNA viruses pathogenic to humans. The wAlbB strain of Wolbachia was originally isolated from Aedes albopictus, and when transinfected into Ae. aegypti, persists in mosquitoes under high temperature conditions longer than other strains. The utility of wAlbB to block a broad spectrum of RNA viruses has received limited attention. Here we test the ability of wAlbB to reduce or block the replication of a range of Flavivirus and Alphavirus species in cell culture. METHODS: The C6/36 mosquito cell line was stably infected with the wAlbB strain using the shell-vial technique. The replication of dengue, West Nile and three strains of Zika (genus Flavivirus), and Ross River, Barmah Forest and Sindbis (genus Alphavirus) viruses was compared in wAlbB-infected cells with Wolbachia-free controls. Infectious virus titres were determined using either immunofocus or plaque assays. A general linear model was used to test for significant differences in replication between flaviviruses and alphaviruses. RESULTS: Titres of all viruses were significantly reduced in cell cultures infected with wAlbB versus Wolbachia-free controls. The magnitude of reduction in virus yields varied among virus species and, within species, also among the strains utilized. CONCLUSION: Our results suggest that wAlbB infection of arthropods could be used to reduce transmission of a wide range of pathogenic RNA viruses.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Flavivirus/crecimiento & desarrollo , Interacciones Microbianas , Replicación Viral , Wolbachia , Aedes/microbiología , Aedes/virología , Infecciones por Alphavirus/prevención & control , Animales , Línea Celular/microbiología , Línea Celular/virología , Dengue/prevención & control , Humanos , Insectos Vectores/microbiología , Insectos Vectores/virología , Control Biológico de Vectores , Virosis/prevención & control , Virosis/transmisión , Fiebre del Nilo Occidental/prevención & control , Infección por el Virus Zika/prevención & control
11.
Viruses ; 12(1)2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31947624

RESUMEN

Duck tembusu virus (DTMUV) is a single-stranded, positive-polarity RNA flavivirus that has caused considerable economic losses in China in recent years. Innate immunity represents the first line of defense against invading pathogens and serves as an important role in resisting viral infections. In this study, we found that the infection of ducks by DTMUV triggers Toll-like receptors (TLRs) and (RIG-I)-like receptors (RLRs) signaling pathways and inducing abundant of pro-inflammatory factors and type I interferons (IFNs), in which melanoma differentiation-associated gene 5 (MDA5) and Toll-like receptor 3 (TLR3) play important immunity roles, they can inhibit the replication process of DTMUV via inducing type I IFNs. Moreover, we demonstrated that type I IFNs can inhibit the DTMUV replication process in a time- and dose-dependent manner. Exosomes are small membrane vesicles that have important roles in intercellular communication. MicroRNAs (miRNAs) are small non-coding RNAs that can modulate gene expression and are common substances in exosomes. In our experiment, we successfully isolated DEF cells derived exosome for the first time and explored its function. Firstly, we found the expression of miR-148a-5p is significantly decreased following DTMUV infect. Then we found miR-148a-5p can target TLR3 and down-regulate the expression of TLR3, serving as a negative factor in innate immunity. Unfortunately, we cannot find miRNAs with different expression changes that can target MDA5. Lastly, our experimental results showed that TLR3 was one of the causes of miR-148a-5p reduction, suggesting that the high level of TLR3 after DTMUV infect can both trigger innate immunity and suppress miR-148a-5p to resist DTMUV.


Asunto(s)
Exosomas/metabolismo , Flavivirus/crecimiento & desarrollo , MicroARNs/metabolismo , Receptor Toll-Like 3/metabolismo , Animales , Línea Celular , Patos/virología , Infecciones por Flavivirus/veterinaria , Inmunidad Innata , Replicación Viral
12.
PLoS Negl Trop Dis ; 13(11): e0007346, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31693659

RESUMEN

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.


Asunto(s)
Aedes/virología , Arbovirus/crecimiento & desarrollo , Coinfección/virología , Mosquitos Vectores/virología , Orthobunyavirus/crecimiento & desarrollo , Replicación Viral , Alphavirus/crecimiento & desarrollo , Animales , Arbovirus/genética , Secuencia de Bases , Técnicas de Cultivo de Célula/métodos , Línea Celular , Virus del Dengue/crecimiento & desarrollo , Flavivirus/genética , Flavivirus/crecimiento & desarrollo , Genoma Viral , Interacciones Huésped-Patógeno/fisiología , Orthobunyavirus/genética , Virus ARN/genética , Virus ARN/crecimiento & desarrollo , Rhabdoviridae/crecimiento & desarrollo , Virus Sindbis/crecimiento & desarrollo , Transfección , Virus Zika/crecimiento & desarrollo
13.
Virus Res ; 273: 197739, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31493438

RESUMEN

Duck Tembusu virus (DTMUV) is a new pathogen that produces an acute and potent disease in ducks which has caused serious economic losses in China. In this study, a virulent strain of DTMUV, designated as ZJSBL01, was attenuated by serial passages in BHK-21 cells supplied with 5-Fluorouracil (5-FU) for 50 passages to induce mutation and attenuation. Growth kinetics of different passages of ZJSBL01 strain in BHK-21 cells show that these viruses have similar replication characteristics. The virus was highly attenuated after 40 passages in BHK-21 cells supplied with 5-FU, based on mortality, morbidity, and viral load in inoculated Sheldrake ducklings. In addition, all of the ducklings immunized with ZJSBL01-P40, the virus obtained at passage 40 of ZJSBL01, showed seroconversion on day 14 post inoculation. Moreover, P40 did not cause clinical symptom for layding ducks. Immunization with ZJSBL01-P40 could provide effective protection against the virulent parental ZJSBL01 strain. Seventeen amino acid substitutions were observed in the polyprotein of ZJSBL01-P40 compared with parental ZJSBL01. These results indicate that ZJSBL01-P40 may be a live vaccine candidate for prevention of DTMUV-disease.


Asunto(s)
Patos/virología , Flavivirus/efectos de los fármacos , Flavivirus/genética , Fluorouracilo/farmacología , Pase Seriado , Cultivo de Virus , Animales , Anticuerpos Antivirales/sangre , Flavivirus/crecimiento & desarrollo , Flavivirus/patogenicidad , Inmunización , Cinética , Mutación , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Carga Viral/efectos de los fármacos , Vacunas Virales/administración & dosificación , Vacunas Virales/inmunología , Replicación Viral/efectos de los fármacos
14.
Elife ; 82019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31516121

RESUMEN

Flaviviruses translate their genomes as multi-pass transmembrane proteins at the endoplasmic reticulum (ER) membrane. Here, we show that the ER membrane protein complex (EMC) is indispensable for the expression of viral polyproteins. We demonstrated that EMC was essential for accurate folding and post-translational stability rather than translation efficiency. Specifically, we revealed degradation of NS4A-NS4B, a region rich in transmembrane domains, in absence of EMC. Orthogonally, by serial passaging of virus on EMC-deficient cells, we identified two non-synonymous point mutations in NS4A and NS4B, which rescued viral replication. Finally, we showed a physical interaction between EMC and viral NS4B and that the NS4A-4B region adopts an aberrant topology in the absence of the EMC leading to degradation. Together, our data highlight how flaviviruses hijack the EMC for transmembrane protein biogenesis to achieve optimal expression of their polyproteins, which reinforces a role for the EMC in stabilizing challenging transmembrane proteins during synthesis.


Asunto(s)
Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Flavivirus/crecimiento & desarrollo , Expresión Génica , Interacciones Huésped-Patógeno , Proteínas de la Membrana/metabolismo , Poliproteínas/biosíntesis , Línea Celular , Hepatocitos/virología , Humanos , Procesamiento Proteico-Postraduccional
15.
J Gen Virol ; 100(11): 1580-1586, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31524580

RESUMEN

Here we report the generation of novel chimeric flaviviruses, which express the prM and E proteins of either dengue or Zika viruses on the genomic backbone of Palm Creek virus (PCV), an insect-specific flavivirus. The chimeric virus particles were antigenically indistinguishable from their parental prM-E donors, but were unable to infect vertebrate cells. An additional chimera (PCV structural genes in the backbone of West Nile virus - WNV/PCV-prME) was also unable to infect vertebrate cells, but transfection with RNA from this virus resulted in detectable RNA replication and translation but no infectious virion production. These data suggest multiple blocks at the entry, RNA replication and assembly/release stages of insect-specific flavivirus (ISF) infection in vertebrate cells. Serial passaging of these chimeric viruses in mosquito cells identified amino acid substitutions that may lead to increased replication efficiency. These chimeric viruses provide unique tools to further dissect the mechanisms of the host restriction of ISFs.


Asunto(s)
Flavivirus/crecimiento & desarrollo , Especificidad del Huésped , Tropismo Viral , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Culicidae , Flavivirus/genética , Ratones , Mutación , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Pase Seriado , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo , Internalización del Virus , Liberación del Virus , Replicación Viral
16.
Virus Res ; 273: 197753, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31521764

RESUMEN

Usutu virus (USUV) previously restricted to Africa where it caused mild infections, emerged in 2001 in Europe and caused more severe infections among birds and humans with neurological forms, suggesting an adaptation and increasing virulence. This evolution suggests the need to better understand USUV transmission patterns for assessing risks and to develop control strategies. Phylogenetic analysis conducted in Africa showed low genetic diversity of African USUV strains except for one human and the USUV subtype (USUVsub) strains, which exhibited a deletion in the 3'UTR and nucleotide substitutions throughout the genome. Here we analyzed their viral replication in vitro in mosquito and mammalian cells, and vector competence of Culex quinquefasciatus, compared to a reference strain. Growth kinetics of the different strains showed comparable replication rates however variations in replication and translation efficiency were observed. Vector competence analysis showed that all strains were able to infect Culex quinquefasciatus the main peridomestic Culex species in Africa, with detection of USUV viral genomes and infectious particles. Dissemination and transmission were observed only for USUVsub, but infectious particles were not detected in Culex quinquefasciatus saliva. Our findings suggest that genetic variability can affect USUV in vitro replication in a cell type-dependent manner and in vivo in mosquitoes. In addition, the results show that Culex quinquefasciatus is not competent for the USUV strains analyzed here and also suggest an aborted transmission process for the USUVsub, which requires further investigations.


Asunto(s)
Culex/virología , Infecciones por Flavivirus/transmisión , Flavivirus/genética , Variación Genética , Aedes/virología , África , Animales , Animales Recién Nacidos , Línea Celular , Chlorocebus aethiops , Vectores de Enfermedades , Femenino , Flavivirus/crecimiento & desarrollo , Genoma Viral , Cinética , ARN Viral/genética , Células Vero , Replicación Viral
17.
Viruses ; 11(6)2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31212939

RESUMEN

Chronic viral disease constitutes a major global health problem, with several hundred million people affected and an associated elevated number of deaths. An increasing number of disorders caused by human flaviviruses are related to their capacity to establish a persistent infection. Here we show that Usutu virus (USUV), an emerging zoonotic flavivirus linked to sporadic neurologic disease in humans, can establish a persistent infection in cell culture. Two independent lineages of Vero cells surviving USUV lytic infection were cultured over 82 days (41 cell transfers) without any apparent cytopathology crisis associated. We found elevated titers in the supernatant of these cells, with modest fluctuations during passages but no overall tendency towards increased or decreased infectivity. In addition to full-length genomes, viral RNA isolated from these cells at passage 40 revealed the presence of defective genomes, containing different deletions at the 5' end. These truncated transcripts were all predicted to encode shorter polyprotein products lacking membrane and envelope structural proteins, and most of non-structural protein 1. Treatment with different broad-range antiviral nucleosides revealed that USUV is sensitive to these compounds in the context of a persistent infection, in agreement with previous observations during lytic infections. The exposure of infected cells to prolonged treatment (10 days) with favipiravir and/or ribavirin resulted in the complete clearance of infectivity in the cellular supernatants (decrease of ~5 log10 in virus titers and RNA levels), although modest changes in intracellular viral RNA levels were recorded (<2 log10 decrease). Drug withdrawal after treatment day 10 resulted in a relapse in virus titers. These results encourage the use of persistently-infected cultures as a surrogate system in the identification of improved antivirals against flaviviral chronic disease.


Asunto(s)
Infecciones por Flavivirus/virología , Flavivirus/crecimiento & desarrollo , Modelos Biológicos , Cultivo de Virus/métodos , Replicación Viral , Amidas/administración & dosificación , Amidas/farmacología , Animales , Antivirales/administración & dosificación , Antivirales/farmacología , Chlorocebus aethiops , Infecciones por Flavivirus/tratamiento farmacológico , Pirazinas/administración & dosificación , Pirazinas/farmacología , ARN Viral/genética , Ribavirina/administración & dosificación , Ribavirina/farmacología , Análisis de Secuencia de ADN , Eliminación de Secuencia , Pase Seriado , Resultado del Tratamiento , Células Vero , Carga Viral , Proteínas Virales/genética
18.
Artículo en Inglés | MEDLINE | ID: mdl-30746342

RESUMEN

Several mosquito-borne Flaviviruses such as Japanese encephalitis virus (JEV), West Nile virus (WNV), Dengue Virus (DENV), and Zika virus (ZIKV) can cause severe clinical disease. Being zoonotic, Flaviviruses infect a wide variety of terrestrial vertebrates, which dependent of the virus-host interactions, can enhance ongoing epidemics and maintain the virus in the environment for prolonged periods. Targeted species can vary from amphibians, birds to various mammals, dependent on the virus. For many mosquito-borne flaviviruses the spectrum of targeted species is incompletely understood, in particular with respect to their contribution to the maintenance of virus in certain geographical regions. Furthermore, little is known about virus and host factors contributing to species tropism. The present study utilized human and porcine monocyte-derived dendritic cells (MoDC) as a cell culture model to better understand Flavivirus species tropism and innate immune responses. MoDC were selected based on their presence in the skin and their role as an early target cell for several Flaviviruses and their role as immune sentinels. While differences in viral infectivity and replication were minor when comparing porcine with human MoDC for some of the tested Flaviviruses, a particularly strong replication in human MoDC was found with USUV, while JEV appeared to have a stronger tropism for porcine MoDC. With respect to innate immune responses we found high induction of TNF and IFN-ß in both human and porcine MoDC after infection with JEV, WNV, and USUV, but not with DENV, ZIKV, and Wesselsbron virus. Spondweni virus induced these cytokine responses only in porcine MoDC. Overall, innate immune responses correlated with early infectivity and cytokine production. In conclusion, we demonstrate Flavivirus-dependent differences in the interaction with MoDC. These may play a role in pathogenesis but appear to only partially reflect the expected species tropism.


Asunto(s)
Células Dendríticas/virología , Flavivirus/crecimiento & desarrollo , Flavivirus/aislamiento & purificación , Especificidad del Huésped , Mosquitos Vectores/virología , Animales , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Inmunidad Innata , Porcinos
19.
Artículo en Inglés | MEDLINE | ID: mdl-30234026

RESUMEN

Small-to-medium sized mammals and large animals are lucrative sources of blood meals for ixodid ticks that transmit life-threatening tick-borne flaviviruses (TBFVs). TBFVs have been isolated from various organs obtained from wild-caught Myodes and Apodemus species in Europe and Asia. Thus, these rodents are well-established reservoirs of TBFVs. Wild-caught Peromyscus species have demonstrated seropositivity against Powassan virus, the only TBFV known to circulate in North America, suggesting that they may play an important role in the biology of the virus in this geographic region. However, virus isolation from Peromyscus species is yet to be demonstrated. Wild-caught medium-sized mammals, such as woodchucks (Marmota monax) and skunks (Mephitis mephitis) have also demonstrated seropositivity against POWV, and virus was isolated from apparently healthy animals. Despite the well-established knowledge that small-to-medium sized animals are TBFV reservoirs, specific molecular biology addressing host-pathogen interactions remains poorly understood. Elucidating these interactions will be critical for gaining insight into the mechanism(s) of viral pathogenesis and/or resistance.


Asunto(s)
Reservorios de Enfermedades/virología , Infecciones por Flavivirus/veterinaria , Flavivirus/crecimiento & desarrollo , Mephitidae/virología , Enfermedades de los Roedores/virología , Roedores/virología , Enfermedades por Picaduras de Garrapatas/veterinaria , Animales , Asia , Europa (Continente) , Flavivirus/aislamiento & purificación , Infecciones por Flavivirus/virología , América del Norte , Enfermedades por Picaduras de Garrapatas/virología
20.
Vet Ital ; 53(2): 97-110, 2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28675249

RESUMEN

Usutu (USUV) and West Nile (WNV) are mosquito-borne Flavivirus emerged in Italy in 1996 and 1998, respectively, and reappeared 10 years later. The aim of this work is to review the Italian mosquito species found positive for WNV and USUV between 2008 and 2014. Moreover, the role of mosquitoes in promoting the overwintering of these viruses is discussed, as a result of the mosquito collections performed in Molise region between September 2010 and April 2011. Overall 99,000 mosquitoes were collected: 337 and 457 mosquito pools tested positive by real time reverse transcriptase polymerase chain reaction (real time RT-PCR) for WNV and USUV, respectively. West Nile virus was detected in pools of Culex pipiens s.l. (329), Ochlerotatus caspius (4), Culex modestus (2), and Culex spp. (2). Positive USUV pools were from Cx. pipiens s.l. (435), Aedes albopictus (12), Oc. caspius (5), Culex spp. (2), Anopheles maculipennis s.l. (1), Culiseta annulata (1), and Ochlerotatus detritus (1). In Molise region, 1,694 mosquitoes were collected, and USUV was identi ed in Cx. pipiens s.l., Cs. annulata, and Oc. detritus pools. This paper shows that Cx. pipiens s.l. is the mosquito species most involved in the WNV and USUV circulation in Italy, although other species would also support the spread of both the viruses during Winter.


Asunto(s)
Culex/virología , Infecciones por Flavivirus/veterinaria , Mosquitos Vectores/virología , Fiebre del Nilo Occidental/veterinaria , Animales , Culex/clasificación , Flavivirus/crecimiento & desarrollo , Infecciones por Flavivirus/epidemiología , Infecciones por Flavivirus/transmisión , Italia/epidemiología , Mosquitos Vectores/clasificación , Fiebre del Nilo Occidental/epidemiología , Fiebre del Nilo Occidental/transmisión , Virus del Nilo Occidental/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA