Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Einstein (Sao Paulo) ; 21: eAO0160, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37255058

RESUMEN

OBJECTIVE: This study verified the replication efficiency of the Rocio virus in a primary culture of mouse neural cells. METHODS: Mixed primary cultures (neurons/glia) obtained from the brains of newborn isogenic BALB/c mice were inoculated with Rocio virus on the 7 th day of culture, and the development of cytopathogenic effects was monitored. The infection was confirmed via immunocytochemistry (anti-ROCV), while viral replication was quantified in infected primary cultures. The titration method used depended on the infection period. RESULTS: Rocio virus efficiently infected primary cultured neural cells, with the highest viral titer causing cytopathic changes was observed at 2 days post infection. The virus-infected primary culture survived for up to 7 days post infection, and viral load quantitation showed viral replication kinetics compatible with the cell death kinetics of cultures. CONCLUSION: The findings of this study suggest that mouse neural cell primary cultures support Rocio virus replication and could be used as an alternative system for studying Flavivirus infection in the central nervous system.


Asunto(s)
Infecciones por Flavivirus , Flavivirus , Animales , Ratones , Infecciones por Flavivirus/metabolismo , Infecciones por Flavivirus/patología , Encéfalo , Neuronas/metabolismo , Neuronas/patología , Células Cultivadas
2.
Cell Rep ; 37(11): 110091, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34910910

RESUMEN

Hematophagous arthropods, such as mosquitoes, naturally carry and transmit hundreds of arboviruses to humans. Blood meal is a predominant physical interface that shapes cross-species communications among humans, bloodsuckers, and arboviruses. Here, we identify a human-blood-derived microRNA, hsa-miR-150-5p, that interferes with a mosquito antiviral system to facilitate flavivirus infection and transmission. hsa-miR-150-5p is acquired with a blood meal into the mosquito hemocoel and persists for a prolonged time there. The agomir of hsa-miR-150-5p enhances, whereas the antagomir represses flaviviral infection in mosquitoes and transmission from mice to mosquitoes. Mechanistic studies indicate that hsa-miR-150-5p hijacks the mosquito Argonaute-1-mediated RNA interference system to suppress the expression of some chymotrypsins with potent virucidal activity. Mosquito chymotrypsins are essential for resisting systemic flavivirus infection in hemocoel tissues. Chymotrypsin homologs potentially targeted by miR-150-5p are also found in other hematophagous arthropods, demonstrating a conserved miR-150-5p-mediated cross-species RNAi mechanism that might determine flaviviral transmissibility in nature.


Asunto(s)
Aedes/virología , Quimotripsina/antagonistas & inhibidores , Infecciones por Flavivirus/virología , Flavivirus/aislamiento & purificación , MicroARNs/genética , Mosquitos Vectores/virología , Replicación Viral , Animales , Femenino , Flavivirus/genética , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/patología , Genoma Viral , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/sangre
3.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34873063

RESUMEN

Flaviviruses such as Zika virus and West Nile virus have the potential to cause severe neuropathology if they invade the central nervous system. The type I interferon response is well characterized as contributing to control of flavivirus-induced neuropathogenesis. However, the interferon-stimulated gene (ISG) effectors that confer these neuroprotective effects are less well studied. Here, we used an ISG expression screen to identify Shiftless (SHFL, C19orf66) as a potent inhibitor of diverse positive-stranded RNA viruses, including multiple members of the Flaviviridae (Zika, West Nile, dengue, yellow fever, and hepatitis C viruses). In cultured cells, SHFL functions as a viral RNA-binding protein that inhibits viral replication at a step after primary translation of the incoming genome. The murine ortholog, Shfl, is expressed constitutively in multiple tissues, including the central nervous system. In a mouse model of Zika virus infection, Shfl-/- knockout mice exhibit reduced survival, exacerbated neuropathological outcomes, and increased viral replication in the brain and spinal cord. These studies demonstrate that Shfl is an important antiviral effector that contributes to host protection from Zika virus infection and virus-induced neuropathological disease.


Asunto(s)
Proteínas de Unión al ARN/metabolismo , Infección por el Virus Zika/patología , Virus Zika/metabolismo , Animales , Línea Celular , Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/virología , Flavivirus/genética , Infecciones por Flavivirus/genética , Infecciones por Flavivirus/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/metabolismo , Proteínas de Unión al ARN/genética , Replicación Viral/fisiología , Virus Zika/patogenicidad , Infección por el Virus Zika/genética
4.
Viruses ; 13(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34452347

RESUMEN

The Usutu virus (USUV) is a mosquito-borne zoonotic flavivirus. Despite its continuous circulation in Europe, knowledge on the pathology, cellular and tissue tropism and pathogenetic potential of different circulating viral lineages is still fragmentary. Here, macroscopic and microscopic evaluations are performed in association with the study of cell and tissue tropism and comparison of lesion severity of two circulating virus lineages (Europe 3; Africa 3) in 160 Eurasian blackbirds (Turdus merula) in the Netherlands. Results confirm hepatosplenomegaly, coagulative necrosis and lymphoplasmacytic inflammation as major patterns of lesions and, for the first time, vasculitis as a novel virus-associated lesion. A USUV and Plasmodium spp. co-infection was commonly identified. The virus was associated with lesions by immunohistochemistry and was reported most commonly in endothelial cells and blood circulating and tissue mononucleated cells, suggesting them as a major route of entry and spread. A tropism for mononuclear phagocytes cells was further supported by viral labeling in multinucleated giant cells. The involvement of ganglionic neurons and epithelial cells of the gastrointestinal tract suggests a possible role of oral transmission, while the involvement of feather follicle shafts and bulbs suggests their use as a diagnostic sample for live bird testing. Finally, results suggest similar pathogenicity for the two circulating lineages.


Asunto(s)
Enfermedades de las Aves/virología , Infecciones por Flavivirus/veterinaria , Flavivirus/fisiología , Passeriformes/virología , Animales , Enfermedades de las Aves/patología , Flavivirus/genética , Flavivirus/aislamiento & purificación , Flavivirus/patogenicidad , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/virología , Países Bajos , Fagocitos/virología , Virulencia
5.
Virus Genes ; 57(4): 395-399, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34086153

RESUMEN

According to modern classification, tick-borne flaviviruses have been divided into a mammalian tick-borne virus group and a seabird tick-borne virus group (STBVG). The STBVG includes the Tyuleniy virus, Meaban virus, Saumarez Reef virus, and the recently discovered Kama virus (KAMV). The latter was isolated from Ixodes lividus, an obligate parasitic tick of the sand martin (Riparia riparia), in 1989 in the central part of the Russian Plain. In 2014, based on molecular genetic analysis, it was shown that KAMV is a new virus belonging to STBVG, genus Flavivirus, fam. Flaviviridae. Very little is known about the Kama virus concerning its range, vectors, and reservoir hosts. GenBank contains a single sequence of the complete genome of this virus. In the present study, the complete genome sequences of two strains, isolated in 1983 in the Omsk region (Western Siberia) from gamasid mites in the nests of rooks (Corvus frugilegus), have been determined. Phylogenetic analyses of their genomes showed a close relationship both with each other (approx. 98.9% nucleotide identity) and with KAMV isolated in European Russia (approx. 98.4% nucleotide identity). The ecological features of KAMV that are due to the species of the vector (gamasid mites) and its hosts (colonial birds of the mainland of Eurasia) indicate that KAMV is an atypical representative STBVG.


Asunto(s)
Virus de la Encefalitis Transmitidos por Garrapatas/genética , Infecciones por Flavivirus/genética , Flavivirus/genética , Genoma Viral/genética , Animales , Antígenos Virales/genética , Aves , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Flavivirus/patogenicidad , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/virología , Humanos , Ixodes/genética , Ixodes/virología , Conformación de Ácido Nucleico
6.
Vet Microbiol ; 255: 109033, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33711568

RESUMEN

Tembusu Virus (TMUV), a pathogenic member of Flavivirus family, acts as the causative agent of egg-laying and has severely threatened the duck industry over the past few years. Thus far, the pathogenicity of such virus has been extensively studied, whereas TMUV on immune system has been less comprehensively assessed, especially on ducklings that exhibit more susceptible to TMUV attack. Accordingly, in the present study, 5-day-old ducklings were infected with TMUV-TC2B (104 TCID50) via intravenous injection, and mock ones were inoculated with phosphate-buffered saline (PBS) in identical manner as control. At 1 day-post inoculation (dpi), the innate immunity was strongly activated, and reacted rapidly to TMUV invasion, which was reflected as the significantly up-regulated IFN-stimulated genes (ISGs), especially in immune organs (e.g., thymus, bursa of Fabricius (BF) and spleen). Subsequently, under the continuous monitoring, the levels of IgA, IgM and IgG acting as the representative immunoglobulins (Igs) were constantly higher than those of mock ducklings, demonstrating that humoral immunity also played a major role in anti-virus infection. Despite the immune system activated positively, TMUV still caused systemic infection, and in particular, the immune organs were subject to severe damage in the early infection. With our constant observation, the injury of spleen and BF turned out to be getting more serious, and at 6 dpi, TMUV antigen was widely detected in both of two immune organs by immunohistochemistry (IHC) and main histopathological lesion presented as lymphocytopenia. Moreover, the elevated apoptosis rate of splenic lymphocytes and the alteration of immune organ index also revealed the damage of lymphoid organs and similarly, it is worth noting that severe damages were detected in thymus of TMUV-infected ducklings as well. In brief, the present study systematically described the dynamic damage of immune system after being attacked by TMUV and presented insights into the research of pathogenicity.


Asunto(s)
Inmunidad Adaptativa , Bolsa de Fabricio/patología , Infecciones por Flavivirus/veterinaria , Flavivirus , Bazo/patología , Timo/patología , Animales , Peso Corporal , Encéfalo/patología , Bolsa de Fabricio/virología , Patos , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/virología , Inmunoglobulinas/sangre , Interferones/metabolismo , Riñón/patología , Hígado/patología , Pulmón/patología , Miocardio/patología , Enfermedades de las Aves de Corral/patología , Enfermedades de las Aves de Corral/virología , Bazo/virología , Timo/inmunología
7.
Viruses ; 13(2)2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33498300

RESUMEN

Alfuy (ALFV) is an attenuated flavivirus related to the Murray Valley encephalitis virus (MVEV). We previously identified markers of attenuation in the envelope (E) protein of the prototype strain (ALFV3929), including the hinge region (E273-277) and lack of glycosylation at E154-156. To further determine the mechanisms of attenuation we assessed ALFV3929 binding to glycosaminoglycans (GAG), a known mechanism of flaviviruses attenuation. Indeed, ALFV3929 exhibited reduced binding to GAG-rich cells in the presence of heparin; however, low-passage ALFV isolates were relatively unaffected. Sequence comparisons between ALFV strains and structural modelling incriminated a positively-charged residue (K327) in ALFV3929 as a GAG-binding motif. Substitution of this residue to the corresponding uncharged residue in MVEV (L), using a previously described chimeric virus containing the prM & E genes of ALFV3929 in the backbone of MVEV (MVEV/ALFV-prME), confirmed a role for K327 in enhanced GAG binding. When the wild type residues at E327, E273-277 and E154-156 of ALFV3929 were replaced with the corresponding residues from virulent MVEV, it revealed each motif contributed to attenuation of ALFV3929, with the E327/E273-277 combination most dominant. These data demonstrate that attenuation of ALFV3929 is multifactorial and provide new insights for the rational design of attenuated flavivirus vaccines.


Asunto(s)
Virus de la Encefalitis del Valle Murray/patogenicidad , Virus de la Encefalitis Japonesa (Subgrupo)/patogenicidad , Encefalitis por Arbovirus/virología , Infecciones por Flavivirus/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Secuencias de Aminoácidos , Animales , Encéfalo/patología , Encéfalo/virología , Línea Celular , Virus de la Encefalitis del Valle Murray/química , Virus de la Encefalitis del Valle Murray/metabolismo , Virus de la Encefalitis Japonesa (Subgrupo)/química , Virus de la Encefalitis Japonesa (Subgrupo)/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Subgrupo)/metabolismo , Encefalitis por Arbovirus/patología , Infecciones por Flavivirus/patología , Glicosaminoglicanos/metabolismo , Glicosilación , Heparina/farmacología , Ratones , Mutación , Dominios Proteicos , Pase Seriado , Proteínas del Envoltorio Viral/genética , Ensayo de Placa Viral , Virulencia
8.
PLoS Negl Trop Dis ; 14(10): e0008765, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33044987

RESUMEN

Usutu virus (USUV; Flavivirus), a close phylogenetic and ecological relative of West Nile virus, is a zoonotic virus that can cause neuroinvasive disease in humans. USUV is maintained in an enzootic cycle between Culex mosquitoes and birds. Since the first isolation in 1959 in South Africa, USUV has spread throughout Africa and Europe. Reported human cases have increased over the last few decades, primarily in Europe, with symptoms ranging from mild febrile illness to severe neurological effects. In this study, we investigated whether USUV has become more pathogenic during emergence in Europe. Interferon α/ß receptor knockout (Ifnar1-/-) mice were inoculated with recent USUV isolates from Africa and Europe, as well as the historic 1959 South African strain. The three tested African strains and one European strain from Spain caused 100% mortality in inoculated mice, with similar survival times and histopathology in tissues. Unexpectedly, a European strain from the Netherlands caused only 12% mortality and significantly less histopathology in tissues from mice compared to mice inoculated with the other strains. Viremia was highest in mice inoculated with the recent African strains and lowest in mice inoculated with the Netherlands strain. Based on phylogenetics, the USUV isolates from Spain and the Netherlands were derived from separate introductions into Europe, suggesting that disease outcomes may differ for USUV strains circulating in Europe. These results also suggest that while more human USUV disease cases have been reported in Europe recently, circulating African USUV strains are still a potential major health concern.


Asunto(s)
Infecciones por Flavivirus/virología , Flavivirus/aislamiento & purificación , Flavivirus/patogenicidad , Animales , Culex/virología , Europa (Continente) , Femenino , Flavivirus/clasificación , Flavivirus/genética , Infecciones por Flavivirus/mortalidad , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/transmisión , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Países Bajos , Filogenia , Sudáfrica , España , Virulencia
9.
Viruses ; 12(5)2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32408481

RESUMEN

Usutu virus (USUV) is a mosquito-borne flavivirus, closely related to the West Nile virus (WNV). Similar to WNV, USUV may cause infections in humans, with occasional, but sometimes severe, neurological complications. Further, USUV can be highly pathogenic in wild and captive birds and its circulation in Europe has given rise to substantial avian death. Adequate study models of this virus are still lacking but are critically needed to understand its pathogenesis and virulence spectrum. The chicken embryo is a low-cost, easy-to-manipulate and ethically acceptable model that closely reflects mammalian fetal development and allows immune response investigations, drug screening, and high-throughput virus production for vaccine development. While former studies suggested that this model was refractory to USUV infection, we unexpectedly found that high doses of four phylogenetically distinct USUV strains caused embryonic lethality. By employing immunohistochemistry and quantitative reverse transcriptase-polymerase chain reaction, we demonstrated that USUV was widely distributed in embryonic tissues, including the brain, retina, and feather follicles. We then successfully developed a primary cell line from the chorioallantoic membrane that was permissive to the virus without the need for viral adaptation. We believe the future use of these models would foster a significant understanding of USUV-induced neuropathogenesis and immune response and allow the future development of drugs and vaccines against USUV.


Asunto(s)
Infecciones por Flavivirus/veterinaria , Flavivirus/fisiología , Enfermedades de las Aves de Corral/virología , Animales , Encéfalo/patología , Encéfalo/virología , Línea Celular , Embrión de Pollo , Pollos , Flavivirus/genética , Infecciones por Flavivirus/mortalidad , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/virología , Enfermedades de las Aves de Corral/mortalidad , Enfermedades de las Aves de Corral/patología , Retina/patología , Retina/virología
10.
Vet Microbiol ; 243: 108636, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32273015

RESUMEN

Several duck Tembusu virus (DTMUV) clusters have been identified since its first emergence in 2010. However, the pathogenesis evaluation of DTMUV has been restricted to cluster 2.2 Chinese DTMUVs. In this study, the pathogenesis of a cluster 2.1 Thai DTMUV was investigated in three ages of Cherry Valley ducks (1-, 4- and 27-week-old). In each age, 35 ducks were inoculated with a cluster 2.1 Thai DTMUV and evaluated for clinical signs, virus distribution and shedding, pathology and serological response. Our results demonstrated that all duck ages were susceptible to Thai DTMUV; however, Thai DTMUV induced greater disease severity in younger ducks (1- and 4-week-old) when compared to older ducks (27-week-old) reflected by higher morbidity and mortality rates, and higher degree of pathological severity. Corresponding to these results, longer-term viremia, higher levels of viral loads in tissues and lower neutralizing antibody titers were also observed in younger ducks compared to those in older ducks. However, it should be noted that a significant drop in egg production was found in older ducks, which also indicates the susceptibility to Thai DTMUV in older ducks. Interestingly, prolonged shedding period with high viral loads was observed in older ducks even without showing clinical signs, suggesting the potential role of the older ducks as the carriers of Thai DTMUV. This finding highlights the importance of monitoring DTMUV and preventing the transmission of DTMUV in adult ducks. Overall, this study provides insights into the pathogenesis and infection dynamics of a cluster 2.1 Thai DTMUV in ducks.


Asunto(s)
Anticuerpos Antivirales/sangre , Patos/virología , Infecciones por Flavivirus/veterinaria , Flavivirus/patogenicidad , Enfermedades de las Aves de Corral/virología , Factores de Edad , Animales , Susceptibilidad a Enfermedades , Femenino , Infecciones por Flavivirus/patología , Tailandia , Carga Viral , Viremia
11.
Int J Mol Sci ; 21(7)2020 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-32244328

RESUMEN

The duck Tembusu virus (DTMUV) is a mosquito-borne flavivirus. It causes severe symptoms of egg-drop, as well as neurological symptoms and brain damage in ducks. However, the specific molecular mechanisms of DTMUV-induced neurovirulence and host responses in the brain remain obscure. To better understand the host-pathogen and neuro-immune interactions of DTMUV infection, we conducted high-throughput RNA-sequencing to reveal the transcriptome profiles of DTMUV-infected duck brain. Totals of 117, 212, and 150 differentially expressed genes (DEGs) were identified at 12, 24, and 48 h post infection (hpi). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses uncovered genes and pathways related to the nervous system and immune responses in duck brain. Neuro-related genes, including WNT3A, GATA3, and CHRNA6, were found to be significantly downregulated. RIG-I-like receptors (DHX58, IFIH1) and Toll-like receptors (TLR2 and TLR3) were activated, inducing the expression of 22 interferon stimulated genes (ISGs) and antigen-processing and -presenting genes (TAP1 and TAP2) in the brain. Our research provides comprehensive information for the molecular mechanisms of neuro-immune and host-pathogen interactions of DTMUV.


Asunto(s)
Encéfalo/metabolismo , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/veterinaria , Flavivirus/inmunología , Perfilación de la Expresión Génica/veterinaria , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Neuroinmunomodulación/genética , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2/metabolismo , Miembro 3 de la Subfamilia B de Transportadores de Casetes de Unión a ATP/genética , Animales , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Patos/genética , Patos/inmunología , Flavivirus/patogenicidad , Infecciones por Flavivirus/metabolismo , Infecciones por Flavivirus/patología , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interferones/metabolismo , Neuroinmunomodulación/inmunología , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Transcriptoma , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo
12.
J Virol ; 94(11)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32161171

RESUMEN

Flaviviruses encode one, two, or no N-linked glycosylation sites on their envelope proteins. Glycosylation can impact virus interactions with cell surface attachment factors and also may impact virion stability and virus replication. Envelope protein glycosylation has been identified as a virulence determinant for multiple flaviviruses, but the mechanisms by which glycosylation mediates pathogenesis remain unclear. In this Gem, we summarize current knowledge on flavivirus envelope protein glycosylation and its impact on viral infection and pathogenesis.


Asunto(s)
Infecciones por Flavivirus/metabolismo , Flavivirus , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral , Animales , Flavivirus/patogenicidad , Flavivirus/fisiología , Infecciones por Flavivirus/patología , Glicosilación , Humanos
13.
PLoS Negl Trop Dis ; 14(3): e0008166, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32203536

RESUMEN

Flaviviruses such as yellow fever, dengue or Zika viruses are responsible for significant human and veterinary diseases worldwide. These viruses contain an RNA genome, prone to mutations, which enhances their potential to emerge as pathogens. Bamaga virus (BgV) is a mosquito-borne flavivirus in the yellow fever virus group that we have previously shown to be host-restricted in vertebrates and horizontally transmissible by Culex mosquitoes. Here, we aimed to characterise BgV host-restriction and to investigate the mechanisms involved. We showed that BgV could not replicate in a wide range of vertebrate cell lines and animal species. We determined that the mechanisms involved in BgV host-restriction were independent of the type-1 interferon response and RNAse L activity. Using a BgV infectious clone and two chimeric viruses generated as hybrids between BgV and West Nile virus, we demonstrated that BgV host-restriction occurred post-cell entry. Notably, BgV host-restriction was shown to be temperature-dependent, as BgV replicated in all vertebrate cell lines at 34°C but only in a subset at 37°C. Serial passaging of BgV in Vero cells resulted in adaptive mutants capable of efficient replication at 37°C. The identified mutations resulted in amino acid substitutions in NS4A-S124F, NS4B-N244K and NS5-G2C, all occurring close to a viral protease cleavage site (NS4A/2K and NS4B/NS5). These mutations were reverse engineered into infectious clones of BgV, which revealed that NS4B-N244K and NS5-G2C were sufficient to restore BgV replication in vertebrate cells at 37°C, while NS4A-S124F further increased replication efficiency. When these mutant viruses were injected into immunocompetent mice, alongside BgV and West Nile virus chimeras, infection and neurovirulence were enhanced as determined by clinical scores, seroconversion, micro-neutralisation, viremia, histopathology and immunohistochemistry, confirming the involvement of these residues in the attenuation of BgV. Our studies identify a new mechanism of host-restriction and attenuation of a mosquito-borne flavivirus.


Asunto(s)
Infecciones por Flavivirus/virología , Flavivirus/genética , Flavivirus/patogenicidad , Mutación , Proteínas no Estructurales Virales/genética , Animales , Encéfalo/patología , Encéfalo/virología , Línea Celular , Chlorocebus aethiops , Culicidae/virología , Modelos Animales de Enfermedad , Endorribonucleasas/metabolismo , Femenino , Flavivirus/fisiología , Infecciones por Flavivirus/metabolismo , Infecciones por Flavivirus/patología , Células HEK293 , Humanos , Masculino , Ratones , Mosquitos Vectores/virología , Células Vero , Virulencia/genética , Replicación Viral , Virus del Nilo Occidental/genética
14.
J Virol ; 94(8)2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32024774

RESUMEN

Tembusu virus (TMUV) is a flavivirus responsible for panzootic outbreaks of severe egg-drop and fatal encephalitis of domestic waterfowl in China. Although TMUV can be attenuated by in vitro passaging, experimental evidence supporting the role of specific genetic changes in virulence attenuation is currently lacking. Here, we performed site-directed mutagenesis on five envelope (E) protein amino acid residues in accordance with the attenuated TMUV generated in our recent study. Our results showed that the Thr-to-Lys mutation of residue 367 in E protein (E367) plays a predominant role in viral cell adaptation and virulence attenuation in ducks compared with mutations in other residues. We further demonstrated that the positively charged basic amino acid substitution at E367 enhanced the viral binding affinity for glycosaminoglycans (GAGs) and reduced viremia levels and the efficiency of replication in major target organs in subcutaneously inoculated ducks. Interestingly, the T367K mutation increased viral neutralization sensitivity to the early immune sera. Together, our findings provide the first evidence that a basic amino acid substitution at E367 strongly impacts the in vitro and in vivo infection of TMUV.IMPORTANCE Outbreaks of Tembusu virus (TMUV) infection have caused huge economic losses in the production of domestic waterfowl since the virus was first recognized in China in 2010. To control TMUV infection, a live-attenuated vaccine candidate of TMUV was developed in our previous study, but the mechanisms of virulence attenuation are not fully understood. Here, we found that the Thr-to-Lys substitution at E367 is a crucial determinant of TMUV virulence attenuation in ducks. We demonstrated that the T367K mutation attenuates TMUV through reducing viral replication in the blood, brain, heart (ducklings), and ovaries. These data provide new insights into understanding the pathogenesis of TMUV and the rational development of novel TMUV vaccines.


Asunto(s)
Sustitución de Aminoácidos , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/virología , Flavivirus/genética , Proteínas del Envoltorio Viral/genética , Sustitución de Aminoácidos/inmunología , Animales , Anticuerpos Neutralizantes , Línea Celular , China/epidemiología , Patos/virología , Femenino , Infecciones por Flavivirus/epidemiología , Infecciones por Flavivirus/patología , Mutagénesis Sitio-Dirigida , Mutación , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/mortalidad , Enfermedades de las Aves de Corral/patología , Enfermedades de las Aves de Corral/virología , Carga Viral , Virulencia , Replicación Viral
15.
Viruses ; 12(2)2020 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-32046265

RESUMEN

Usutu virus (USUV) is a mosquito-borne flavivirus that shares many similarities with the closely related West Nile virus (WNV) in terms of ecology and clinical manifestations. Initially distributed in Africa, USUV emerged in Italy in 1996 and managed to co-circulate with WNV in many European countries in a similar mosquito-bird life cycle. The rapid geographic spread of USUV, the seasonal mass mortalities it causes in the European avifauna, and the increasing number of infections with neurological disease both in healthy and immunocompromised humans has stimulated interest in infection studies to delineate USUV pathogenesis. Here, we assessed the pathogenicity of two USUV isolates from a recent Belgian outbreak in immunocompetent mice. The intradermal injection of USUV gave rise to disorientation and paraplegia and was associated with neuronal death in the brain and spinal cord in a single mouse. Intranasal inoculation of USUV could also establish the infection; viral RNA was detected in the brain 15 days post-infection. Overall, this pilot study probes the suitability of this murine model for the study of USUV neuroinvasiveness and the possibility of direct transmission in mammals.


Asunto(s)
Susceptibilidad a Enfermedades/patología , Susceptibilidad a Enfermedades/virología , Infecciones por Flavivirus/patología , Flavivirus/patogenicidad , Animales , Bélgica , Aves/virología , Encéfalo/virología , Modelos Animales de Enfermedad , Femenino , Flavivirus/clasificación , Infecciones por Flavivirus/virología , Inmunocompetencia , Ratones , Proyectos Piloto , ARN Viral/análisis , Médula Espinal/virología
16.
Virus Res ; 274: 197770, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31626874

RESUMEN

Flaviviruses are group of single stranded RNA viruses that cause severe endemic infection and epidemics on a global scale. It presents a significant health impact worldwide and the viruses have the potential to emerge and outbreak in a non-endemic geographical region. Effective vaccines for prophylaxis are only available for several flaviviruses such as Yellow Fever virus, Tick-borne Encephalitis Virus, Dengue Virus and Japanese Encephalitis Virus and there is no antiflaviviral agent being marketed. This review discusses the flavivirus genome, replication cycle, epidemiology, clinical presentation and pathogenesis upon infection. Effective humoral response is critical to confer protective immunity against flaviviruses. Hence, we have also highlighted the immune responses elicited upon infection, various diagnostic facilities available for flaviviral disease and monoclonal antibodies available to date against flavivirus infection.


Asunto(s)
Infecciones por Flavivirus/diagnóstico , Infecciones por Flavivirus/inmunología , Flavivirus/inmunología , Flavivirus/aislamiento & purificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antivirales/inmunología , Antivirales/uso terapéutico , Biomarcadores/sangre , Flavivirus/genética , Flavivirus/fisiología , Infecciones por Flavivirus/tratamiento farmacológico , Infecciones por Flavivirus/patología , Genoma Viral , Humanos , Proteínas no Estructurales Virales/sangre , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/inmunología , Vacunas Virales/uso terapéutico , Replicación Viral
17.
BMC Vet Res ; 15(1): 362, 2019 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-31651323

RESUMEN

BACKGROUND: Tembusu virus (TMUV) usually affects adult ducks, causing a severe drop of egg production. It has also been shown to be pathogenic in commercial Pekin ducklings below 7 weeks of age. Here, we report a TMUV-caused neurological disease in young egg-type ducklings and the pathogenicity of the egg-type duck-origin TMUV isolates in meat-type Pekin ducklings. RESULTS: The disease occurred in 25 to 40-day-old Jinding ducklings in China, and was characterized by paralysis. Gross lesions were lacking and microscopic lesions appeared chiefly in brain and spleen. Inoculation in embryonated duck eggs resulted in isolation of TMUV Y and GL. The clinical signs and microscopic lesions observed in the spontaneously infected egg-type ducks were repeated in Pekin ducklings by experimental infection. Notably, both Y and GL strains caused 100% mortality in the case of 2-day-old inoculation by intracerebral route. High mortalities (80 and 70%) also occurred following infection of the Y virus at 2 days of age by intramuscular route and at 9 days of age by intracerebral route. CONCLUSIONS: These findings demonstrate that the egg-type duck-origin TMUVs exhibit high pathogenicity in Pekin ducklings, and that the severity of the disease in ducklings is dependent on the infection route and the age of birds at the time of infection. The availability of the highly pathogenic TMUV strains provides a useful material with which to begin investigations into the molecular basis of TMUV pathogenicity in ducks.


Asunto(s)
Infecciones por Flavivirus/veterinaria , Flavivirus/patogenicidad , Enfermedades de las Aves de Corral/virología , Factores de Edad , Animales , Línea Celular , Cricetinae , Vías de Administración de Medicamentos/veterinaria , Patos/virología , Flavivirus/genética , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/virología , Parálisis/veterinaria , Parálisis/virología , Enfermedades de las Aves de Corral/patología
18.
Nat Neurosci ; 22(8): 1276-1288, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31235930

RESUMEN

T cells clear virus from the CNS and dynamically regulate brain functions, including spatial learning, through cytokine signaling. Here we determined whether hippocampal T cells that persist after recovery from infection with West Nile virus (WNV) or Zika virus (ZIKV) impact hippocampal-dependent learning and memory. Using newly established models of viral encephalitis recovery in adult animals, we show that in mice that have recovered from WNV or ZIKV infection, T cell-derived interferon-γ (IFN-γ) signaling in microglia underlies spatial-learning defects via virus-target-specific mechanisms. Following recovery from WNV infection, mice showed presynaptic termini elimination with lack of repair, while for ZIKV, mice showed extensive neuronal apoptosis with loss of postsynaptic termini. Accordingly, animals deficient in CD8+ T cells or IFN-γ signaling in microglia demonstrated protection against synapse elimination following WNV infection and decreased neuronal apoptosis with synapse recovery following ZIKV infection. Thus, T cell signaling to microglia drives post-infectious cognitive sequelae that are associated with emerging neurotropic flaviviruses.


Asunto(s)
Trastornos del Conocimiento/psicología , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/psicología , Microglía/inmunología , Sinapsis/inmunología , Sinapsis/patología , Linfocitos T/inmunología , Animales , Apoptosis , Linfocitos T CD8-positivos/inmunología , Trastornos del Conocimiento/etiología , Femenino , Infecciones por Flavivirus/patología , Interferón gamma , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/psicología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interferón/genética , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/psicología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/psicología , Receptor de Interferón gamma
19.
Adv Chronic Kidney Dis ; 26(3): 198-206, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31202392

RESUMEN

The genus Flavivirus comprises approximately 73 viruses, which share several common aspects, such as dimension, structure, nucleic acid properties, and shape in electronic microscopy. Global incidence of flavivirus infection increased dramatically over the last decades, causing large outbreaks in several areas of the world. These viruses are expanding from endemic tropical and subtropical areas to previously nonendemic areas, affecting and causing diseases in millions of individuals worldwide and posing a formidable challenge to public health in several countries. The majority of clinically significant flavivirus-associated infections are mosquito borne (arboviruses-acronym for ARthropod-BOrne VIRUSES), such as dengue, yellow fever, Japanese encephalitis, Zika, and West Nile fever. Most diseases caused by flaviviruses are asymptomatic or manifest as self-limited, mild, undifferentiated febrile diseases. In a limited number of cases, these diseases may evolve to severe inflammatory, multisystem diseases, causing high morbidity and mortality. Some flaviviruses have been consistently identified in kidney tissue and urine and have been clinically associated with kidney diseases. In this review, we will provide an overview of the epidemiology, risk factors, kidney pathology, etiopathogenesis, and outcomes of acute and chronic kidney syndromes associated with dengue, yellow fever, Zika, and West Nile virus disease.


Asunto(s)
Lesión Renal Aguda/epidemiología , Dengue/epidemiología , Insuficiencia Renal Crónica/epidemiología , Fiebre del Nilo Occidental/epidemiología , Fiebre Amarilla/epidemiología , Infección por el Virus Zika/epidemiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/virología , Dengue/metabolismo , Dengue/patología , Flavivirus , Infecciones por Flavivirus/epidemiología , Infecciones por Flavivirus/patología , Humanos , Mosquitos Vectores , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/virología , Factores de Riesgo , Fiebre del Nilo Occidental/metabolismo , Fiebre del Nilo Occidental/patología , Fiebre Amarilla/metabolismo , Fiebre Amarilla/patología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/patología
20.
Int J Mol Sci ; 20(3)2019 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-30736273

RESUMEN

Virus infections of the central nervous system (CNS) can manifest in various forms of inflammation, including that of the brain (encephalitis) and spinal cord (myelitis), all of which may have long-lasting deleterious consequences. Although the knowledge of how different viruses affect neural cells is increasing, understanding of the mechanisms by which cells respond to neurotropic viruses remains fragmented. Several virus types have the ability to infect neural tissue, and astrocytes, an abundant and heterogeneous neuroglial cell type and a key element providing CNS homeostasis, are one of the first CNS cell types to get infected. Astrocytes are morphologically closely aligned with neuronal synapses, blood vessels, and ventricle cavities, and thereby have the capacity to functionally interact with neurons and endothelial cells. In this review, we focus on the responses of astrocytes to infection by neurotropic flaviviruses, including tick-borne encephalitis virus (TBEV), Zika virus (ZIKV), West Nile virus (WNV), and Japanese encephalitis virus (JEV), which have all been confirmed to infect astrocytes and cause multiple CNS defects. Understanding these mechanisms may help design new strategies to better contain and mitigate virus- and astrocyte-dependent neuroinflammation.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/virología , Infecciones por Flavivirus/metabolismo , Infecciones por Flavivirus/virología , Flavivirus/fisiología , Animales , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encefalitis Japonesa/virología , Infecciones por Flavivirus/patología , Infecciones por Flavivirus/transmisión , Humanos , Tropismo Viral , Fiebre del Nilo Occidental/metabolismo , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...