Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Virol ; 97(10): e0059123, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37768084

RESUMEN

IMPORTANCE: Alphaviruses threaten public health continuously, and Getah virus (GETV) is a re-emerging alphavirus that can potentially infect humans. Approved antiviral drugs and vaccines against alphaviruses are few available, but several host antiviral factors have been reported. Here, we used GETV as a model of alphaviruses to screen for additional host factors. Tetrachlorodibenzo-p-dioxin-inducible poly(ADP ribose) polymerase was identified to inhibit GETV replication by inducing ubiquitination of the glycoprotein E2, causing its degradation by recruiting the E3 ubiquitin ligase membrane-associated RING-CH8 (MARCH8). Using GETV as a model virus, focusing on the relationship between viral structural proteins and host factors to screen antiviral host factors provides new insights for antiviral studies on alphaviruses.


Asunto(s)
Alphavirus , Interacciones Microbiota-Huesped , Proteínas de Transporte de Nucleósidos , Poli(ADP-Ribosa) Polimerasas , Transcriptoma , Humanos , Alphavirus/crecimiento & desarrollo , Alphavirus/inmunología , Glicoproteínas/metabolismo , Proteínas de Transporte de Nucleósidos/genética , Proteínas de Transporte de Nucleósidos/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Ubiquitinación , Proteínas Estructurales Virales/metabolismo , Replicación Viral
2.
J Gen Virol ; 102(12)2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34878970

RESUMEN

Although RNA viruses have high mutation rates, host cells and organisms work as selective environments, maintaining the viability of virus populations by eliminating deleterious genotypes. In serial passages of RNA viruses in a single cell line, most of these selective bottlenecks are absent, with no virus circulation and replication in different tissues or host alternation. In this work, Aedes aegypti Aag-2 cells were accidentally infected with Chikungunya virus (CHIKV) and Mayaro virus (MAYV). After numerous passages to achieve infection persistency, the infectivity of these viruses was evaluated in Ae. albopictus C6/36 cells, African green monkey Vero cells and primary-cultured human fibroblasts. While these CHIKV and MAYV isolates were still infectious to mosquito cells, they lost their ability to infect mammalian cells. After genome sequencing, it was observed that CHIKV accumulated many nonsynonymous mutations and a significant deletion in the coding sequence of the hypervariable domain in the nsP3 gene. Since MAYV showed very low titres, it was not sequenced successfully. Persistently infected Aag-2 cells also accumulated high loads of short and recombinant CHIKV RNAs, which seemed to have been originated from virus-derived DNAs. In conclusion, the genome of this CHIKV isolate could guide mutagenesis strategies for the production of attenuated or non-infectious (to mammals) CHIKV vaccine candidates. Our results also reinforce that a paradox is expected during passages of cells persistently infected by RNA viruses: more loosening for the development of more diverse virus genotypes and more pressure for virus specialization to this constant cellular environment.


Asunto(s)
Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/genética , Genoma Viral/genética , Alphavirus/genética , Alphavirus/crecimiento & desarrollo , Animales , Línea Celular , Culicidae , Especificidad del Huésped , Humanos , Mamíferos , Mutación , ARN Viral/genética , Carga Viral/genética , Proteínas no Estructurales Virales/genética , Replicación Viral/genética
3.
PLoS Negl Trop Dis ; 14(4): e0007518, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32287269

RESUMEN

Newly emerging or re-emerging arthropod-borne viruses (arboviruses) are important causes of human morbidity and mortality worldwide. Arboviruses such as Dengue (DENV), Zika (ZIKV), Chikungunya (CHIKV), and West Nile virus (WNV) have undergone extensive geographic expansion in the tropical and sub-tropical regions of the world. In the Americas the main vectors of DENV, ZIKV, and CHIKV are mosquito species adapted to urban environments, namely Aedes aegypti and Aedes albopictus, whereas the main vector of WNV is Culex quinquefasciatus. Given the widespread distribution in the Americas and high permissiveness to arbovirus infection, these mosquito species may play a key role in the epidemiology of other arboviruses normally associated with sylvatic vectors. Here, we test this hypothesis by determining the vector competence of Ae. aegypti, Ae. albopictus, and Cx. quinquefasciatus to Mayaro (MAYV) virus, a sylvatic arbovirus transmitted mainly by Haemagogus janthinomys that has been causing an increasing number of outbreaks in South America, namely in Brazil. Using field mosquitoes from Brazil, female mosquitoes were experimentally infected, and their competence for infection and transmission rates of MAYV was evaluated. We found consistent infection rate for MAYV in Ae. aegypti (57.5%) and Ae. albopictus (61.6%), whereas very low rates were obtained for Cx. quinquefasciatus (2.5%). Concordantly, we observed high potential transmission ability in Ae. aegypti and Ae. albopictus (69.5% and 71.1% respectively), in contrast to Cx. quinquefasciatus, which could not transmit the MAYV. Notably, we found that very low quantities of virus present in the saliva (undetectable by RT-qPCR) were sufficiently virulent to guarantee transmission. Although Ae. aegypti and Ae. albopictus mosquitoes are not the main vectors for MAYV, our studies suggest that these mosquitoes could play a significant role in the transmission of this arbovirus, since both species showed significant vector competence for MAYV (Genotype D), under laboratory conditions.


Asunto(s)
Aedes/virología , Infecciones por Alphavirus/virología , Alphavirus/aislamiento & purificación , Culex/virología , Transmisión de Enfermedad Infecciosa , Alphavirus/genética , Alphavirus/crecimiento & desarrollo , Infecciones por Alphavirus/transmisión , Animales , Brasil , Femenino , Reacción en Cadena en Tiempo Real de la Polimerasa , Saliva/virología , Carga Viral
4.
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
5.
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
6.
J Virol ; 93(24)2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31554691

RESUMEN

Alphaviruses are enveloped, positive-sense RNA viruses that are important causes of viral encephalomyelitis. Sindbis virus (SINV) infects the neurons of rodents and is a model for studying factors that regulate infection of neuronal cells. The outcome of alphavirus infection of the central nervous system is dependent on neuronal maturation status. Differentiated mature neurons survive and control viral replication better than undifferentiated immature neurons. The cellular factors involved in age-dependent susceptibility include higher levels of antiapoptotic and innate immune factors in mature neurons. Because NF-κB pathway activation is required for the initiation of both apoptosis and the host antiviral response, we analyzed the role of NF-κB during SINV infection of differentiated and undifferentiated rat neuronal cells. SINV infection induced canonical NF-κB activation, as evidenced by the degradation of IκBα and the phosphorylation and nuclear translocation of p65. Inhibition or deletion of the upstream IκB kinase substantially reduced SINV replication in differentiated but not in undifferentiated neuronal cells or mouse embryo fibroblasts. NF-κB inhibition did not affect the establishment of infection, replication complex formation, the synthesis of nonstructural proteins, or viral RNA synthesis in differentiated neurons. However, the translation of structural proteins was impaired, phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2α) was decreased, and host protein synthesis was maintained, suggesting that NF-κB activation was involved in the regulation of translation during infection of mature neurons. Inhibition or deletion of double-stranded RNA-activated protein kinase (PKR) also decreased eIF2α phosphorylation, the translation of viral structural proteins, and virus production. Therefore, canonical NF-κB activation synergizes with PKR to promote SINV replication in differentiated neurons by facilitating viral structural protein translation.IMPORTANCE Mosquito-borne alphaviruses are a significant and growing cause of viral encephalomyelitis worldwide. The outcome of alphaviral neuronal infections is host age dependent and greatly affected by neuronal maturation status, with differentiated, mature neurons being more resistant to infection than undifferentiated, immature neurons. The biological factors that change during neuronal maturation and that influence the outcome of viral infection are currently only partially defined. These studies investigated the role of NF-κB in determining the outcome of alphaviral infection in mature and immature neurons. Inhibition of canonical NF-κB activation decreased alphavirus replication in mature neurons by regulating protein synthesis and limiting the production of the viral structural proteins but had little effect on viral replication in immature neurons or fibroblasts. Therefore, NF-κB is a signaling pathway that influences the maturation-dependent outcome of alphaviral infection in neurons and that highlights the importance of cellular context in determining the effects of signal pathway activation.


Asunto(s)
Infecciones por Alphavirus/virología , Alphavirus/efectos de los fármacos , Alphavirus/crecimiento & desarrollo , FN-kappa B/farmacología , Neuronas/virología , Replicación Viral/efectos de los fármacos , Animales , Diferenciación Celular , Línea Celular , Culicidae/virología , Factor 2 Eucariótico de Iniciación/metabolismo , Técnicas de Inactivación de Genes , Ratones , FN-kappa B/genética , Neurogénesis , Fosforilación , ARN Viral/metabolismo , Ratas , Transducción de Señal , Virus Sindbis/efectos de los fármacos , Virus Sindbis/crecimiento & desarrollo , Transcriptoma , eIF-2 Quinasa/metabolismo
7.
Virology ; 535: 227-231, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31325837

RESUMEN

Mayaro virus (MAYV; family Togaviridae, genus Alphavirus) is an emerging global threat that can cause severe clinical manifestations similar to Zika, dengue, and chikungunya viruses. Currently, there is a lack of molecular tools to enable a better understanding of the transmission and pathogenesis of MAYV. Here, we detail the development and characterization of infectious clones of two strains of MAYV that produce infectious virus and replicate in mammalian and mosquito cells similarly to wild-type virus. Additionally, clone-derived viruses produced identical infection rates and phenotypes in CD-1 mice compared to the parental strains. This infectious clone system will provide a resource to the research community to analyze MAYV genetic determinants of virulence, determine vector competence, and develop vaccines.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Alphavirus/genética , ADN Complementario/genética , ADN Viral/genética , Genética Inversa/métodos , Vacunas Virales/aislamiento & purificación , Alphavirus/inmunología , Alphavirus/patogenicidad , Infecciones por Alphavirus/fisiopatología , Infecciones por Alphavirus/prevención & control , Animales , Línea Celular , Clonación Molecular , Enfermedades Transmisibles Emergentes/fisiopatología , Enfermedades Transmisibles Emergentes/prevención & control , Culicidae , ADN Complementario/aislamiento & purificación , ADN Viral/aislamiento & purificación , Humanos , Ratones , Vacunología/métodos , Vacunas Virales/genética , Vacunas Virales/inmunología , Virología/métodos
8.
Antiviral Res ; 168: 82-90, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31150677

RESUMEN

Mayaro virus (MAYV) is a neglected mosquito-borne alphavirus that causes illness similar to Chikungunya (CHIKV), Dengue (DENV) and Zika virus (ZIKV). Currently, there is no specific treatment or vaccine against MAYV infection. To develop an efficient antiviral screening assay for MAYV, we constructed the infectious clones of MAYV strain BeAr 20290 and its eGFP reporter virus. The reporter virus exhibited high replication capacity indistinguishable with the wild type MAYV, and was genetically stable within at least five rounds of passages in BHK-21 cell. The expression of eGFP correlated well with the viral replication. Using the known inhibitor ribavirin, we confirmed that the MAYV-eGFP reporter virus could be used for antiviral screening to identify the specific inhibitors against MAYV. Using the MAYV-eGFP based antiviral assay, we found that the compound 6-Azauridine which had antiviral activity against CHIKV and SFV, showed a significant inhibitory effect on MAYV replication.


Asunto(s)
Alphavirus/efectos de los fármacos , Alphavirus/genética , Antivirales/farmacología , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Alphavirus/crecimiento & desarrollo , Animales , Línea Celular , Cricetinae , Culicidae , Evaluación Preclínica de Medicamentos , Genoma Viral/genética , Inestabilidad Genómica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Replicación Viral/efectos de los fármacos
9.
Nat Commun ; 9(1): 5326, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30552337

RESUMEN

Alphaviruses are enveloped RNA viruses that contain several human pathogens. Due to intrinsic heterogeneity of alphavirus particles, a high resolution structure of the virion is currently lacking. Here we provide a 3.5 Å cryo-EM structure of Sindbis virus, using block based reconstruction method that overcomes the heterogeneity problem. Our structural analysis identifies a number of conserved residues that play pivotal roles in the virus life cycle. We identify a hydrophobic pocket in the subdomain D of E2 protein that is stabilized by an unknown pocket factor near the viral membrane. Residues in the pocket are conserved in different alphaviruses. The pocket strengthens the interactions of the E1/E2 heterodimer and may facilitate virus assembly. Our study provides structural insights into alphaviruses that may inform the design of drugs and vaccines.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Microscopía por Crioelectrón/métodos , Dominios y Motivos de Interacción de Proteínas , Ensamble de Virus , Internalización del Virus , Animales , Chlorocebus aethiops , Cristalografía por Rayos X , Estadios del Ciclo de Vida , Glicoproteínas de Membrana/química , Modelos Moleculares , Conformación Proteica , Virus Sindbis/crecimiento & desarrollo , Virus Sindbis/ultraestructura , Células Vero , Proteínas del Envoltorio Viral/química , Virión/crecimiento & desarrollo , Virión/ultraestructura
10.
Virus Res ; 256: 166-173, 2018 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-30145137

RESUMEN

Two Alphaviruses stand out for their clinical importance in Brazil: chikungunya (CHIKV) and mayaro (MAYV) viruses. Few studies exist on the mechanisms of the immune response after infection by these viruses and neither a treatment nor a vaccine for these pathogens are available. Although their infection does not have a high mortality rate, they can lead to a joint involvement that can persist for months. The aims of this work were the study of the mechanisms of antiviral immune response following in vitro (U937 cells) infection with these viruses; to investigate the characteristics of the infection by these viruses; and to determine possible molecular targets that could serve as antiviral therapies against these pathogens. Several genes were modulated after infection by these viruses, and the three antiviral detection and response pathways were activated (Toll-like, RIG-I and NOD-like). Eotaxin and IL-6 were induced in all experiments. The cellular immune response profile found for each virus was different, with CHIKV activating primarily an inflammatory response (Th1 and Th17) and MAYV inducing a regulatory/suppressive response, an important feature to contain the inflammation resulting from infection. The data acquired by this study could provide an explanation why CHIKV infections, due to activation of the inflammatory response, are more clinically relevant than MAYV infections, which generates mostly an anti-inflammatory response after infection.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Alphavirus/inmunología , Inmunidad Innata , Factores Inmunológicos/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Humanos , Células U937
11.
Antiviral Res ; 157: 57-67, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29981794

RESUMEN

The New World alphaviruses -Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV respectively) - cause a febrile disease that is often lethal in equines and children and leads to long-term neurological sequelae in survivors. Endemic to the Americas, epizootic outbreaks of the three viruses occur sporadically in the continental United States. All three viruses aerosolize readily, replicate to high titers in cell culture, and have low infectious doses. Additionally, there are no FDA-approved vaccines or therapeutics for human use. To address the therapeutic gap, a high throughput assay utilizing a luciferase reporter virus, TC83-luc, was performed to screen a library of commercially available, FDA-approved drugs for antiviral activity. From a group of twenty compounds found to significantly decrease luminescence, the carcinoma therapeutic sorafenib inhibited replication of VEEV-TC83 and TrD in vitro. Additionally, sorafenib inhibited replication of EEEV and two Old World alphaviruses, Sindbis virus and chikungunya virus, at 8 and 16 h post-infection. Sorafenib caused no toxicity in Vero cells, and coupled with a low EC50 value, yielded a selectivity index of >19. Mechanism of actions studies suggest that sorafenib inhibited viral translation through dephosphorylation of several key proteins, including eIF4E and p70S6K, leading to a reduction in viral protein production and overall viral replication.


Asunto(s)
Alphavirus/efectos de los fármacos , Antineoplásicos/farmacología , Antivirales/farmacología , Reposicionamiento de Medicamentos , Sorafenib/farmacología , Replicación Viral/efectos de los fármacos , Alphavirus/crecimiento & desarrollo , Animales , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Luciferasas/análisis , Luciferasas/genética , Mediciones Luminiscentes , Genética Inversa
12.
J Virol ; 92(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29593049

RESUMEN

Alphaviruses are transmitted to humans via bites of infected mosquitoes. Although alphaviruses have caused a wide range of outbreaks and crippling disease, the availability of licensed vaccines or antiviral therapies remains limited. Mosquito vectors such as Aedes and Culex are the main culprits in the transmission of alphaviruses. This review explores how mosquito saliva may promote alphavirus infection. Identifying the roles of mosquito-derived factors in alphavirus pathogenesis will generate novel tools to circumvent and control mosquito-borne alphavirus infections in humans.


Asunto(s)
Aedes/virología , Infecciones por Alphavirus/transmisión , Alphavirus/inmunología , Culex/virología , Saliva/virología , Inmunidad Adaptativa/inmunología , Alphavirus/crecimiento & desarrollo , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/patología , Animales , Humanos , Inmunidad Innata/inmunología , Terapia de Inmunosupresión , Piel/inmunología , Piel/virología
13.
Cell Death Dis ; 9(3): 274, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449555

RESUMEN

Muscle-invasive bladder cancer (MIBC) is associated with low survival and high recurrence rates even in cases in which patients receive systemic treatments, such as surgery and chemotherapy. Here, we found that a naturally existing alphavirus, namely, M1, selectively kills bladder cancer cells but not normal cells, findings supported by our observations of changes in viral replication and MIBC and patient-derived MIBC cell apoptosis. Transcriptome analysis revealed that interferon-stimulated genes (ISGs) are expressed at low levels in sensitive bladder cancer cells and high levels in resistant cells. Knocking down ZC3HAV1 (ZAP), an antiviral factor in ISGs, restores M1 virus reactivity in resistant cells, and overexpressing ZAP partially reverses M1 virus-induced decreases in cell viability in sensitive cells. In orthotopic MIBC mice, tail vein injections of M1 significant inhibit tumor growth and prolong survival period, antitumor effects of M1 are stronger than those of the first-line chemotherapy agent cisplatin (CDDP). Treated tumors display enhanced cleaved-caspase-3 signals, which are representative of cell apoptosis, and decreased Ki-67 signals, which are representative of cell proliferation. Moreover, tissue microarray (TMA) analyses of clinical tumor specimens revealed that up to 45.6% of cases of MIBC presented with low ZAP expression, a finding that is prevalent in advanced MIBC. Our results indicate that the oncolytic virus M1 is a novel agent capable of functioning as a precise and effective therapy for MIBC.


Asunto(s)
Alphavirus/patogenicidad , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Neoplasias de la Vejiga Urinaria/terapia , Anciano , Alphavirus/crecimiento & desarrollo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Interacciones Huésped-Patógeno , Humanos , Antígeno Ki-67/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Virus Oncolíticos/crecimiento & desarrollo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Tiempo , Carga Tumoral , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/virología , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Virol ; 92(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29187545

RESUMEN

Most alphaviruses are mosquito borne and exhibit a broad host range, infecting many different vertebrates, including birds, rodents, equids, humans, and nonhuman primates. Recently, a host-restricted, mosquito-borne alphavirus, Eilat virus (EILV), was described with an inability to infect vertebrate cells based on defective attachment and/or entry, as well as a lack of genomic RNA replication. We investigated the utilization of EILV recombinant technology as a vaccine platform against eastern (EEEV) and Venezuelan equine encephalitis viruses (VEEV), two important pathogens of humans and domesticated animals. EILV chimeras containing structural proteins of EEEV or VEEV were engineered and successfully rescued in Aedes albopictus cells. Cryo-electron microscopy reconstructions at 8 and 11 Å of EILV/VEEV and EILV/EEEV, respectively, showed virion and glycoprotein spike structures similar to those of VEEV-TC83 and other alphaviruses. The chimeras were unable to replicate in vertebrate cell lines or in brains of newborn mice when injected intracranially. Histopathologic examinations of the brain tissues showed no evidence of pathological lesions and were indistinguishable from those of mock-infected animals. A single-dose immunization of either monovalent or multivalent EILV chimera(s) generated neutralizing antibody responses and protected animals against lethal challenge 70 days later. Lastly, a single dose of monovalent EILV chimeras generated protective responses as early as day 1 postvaccination and partial or complete protection by day 6. These data demonstrate the safety, immunogenicity, and efficacy of novel insect-specific EILV-based chimeras as potential EEEV and VEEV vaccines.IMPORTANCE Mostly in the last decade, insect-specific viruses have been discovered in several arbovirus families. However, most of these viruses are not well studied and largely have been ignored. We explored the use of the mosquito-specific alphavirus EILV as an alphavirus vaccine platform in well-established disease models for eastern (EEE) and Venezuelan equine encephalitis (VEE). EILV-based chimeras replicated to high titers in a mosquito cell line yet retained their host range restriction in vertebrates both in vitro and in vivo In addition, the chimeras generated immune responses that were higher than those of other human and/or equine vaccines. These findings indicate the feasibility of producing a safe, efficacious, mono- or multivalent vaccine against the encephalitic alphaviruses VEEV and EEEV. Lastly, these data demonstrate how host-restricted, insect-specific viruses can be engineered to develop vaccines against related pathogenic arboviruses that cause severe disease in humans and domesticated animals.


Asunto(s)
Infecciones por Alphavirus/inmunología , Alphavirus/crecimiento & desarrollo , Virus de la Encefalitis Equina Venezolana/inmunología , Vacunas Virales/inmunología , Alphavirus/inmunología , Alphavirus/aislamiento & purificación , Infecciones por Alphavirus/virología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Microscopía por Crioelectrón , Virus de la Encefalitis Equina Venezolana/genética , Ingeniería Genética , Células HEK293 , Especificidad del Huésped , Humanos , Ratones , Replicación Viral
15.
Virology ; 487: 230-41, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26550947

RESUMEN

Alphaviruses are a family of positive-strand RNA viruses that circulate on all continents between mosquito vectors and vertebrate hosts. Despite a significant public health threat, their biology is not sufficiently investigated, and the mechanisms of alphavirus replication and virus-host interaction are insufficiently understood. In this study, we have applied a variety of experimental systems to further understand the mechanism by which infected cells detect replicating alphaviruses. Our new data strongly suggest that activation of the antiviral response by alphavirus-infected cells is determined by the integrity of viral genes encoding proteins with nuclear functions, and by the presence of two cellular pattern recognition receptors (PRRs), RIG-I and MDA5. No type I IFN response is induced in their absence. The presence of either of these PRRs is sufficient for detecting virus replication. However, type I IFN activation in response to pathogenic alphaviruses depends on the basal levels of RIG-I or MDA5.


Asunto(s)
Alphavirus/inmunología , ARN Helicasas DEAD-box/metabolismo , Interferón beta/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Células 3T3 , Alphavirus/genética , Alphavirus/crecimiento & desarrollo , Animales , Línea Celular , Cricetinae , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Técnicas de Sustitución del Gen , Técnicas de Silenciamiento del Gen , Inmunidad Innata/inmunología , Helicasa Inducida por Interferón IFIH1 , Ratones , Replicación Viral
16.
Emerg Infect Dis ; 21(10): 1742-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26401714

RESUMEN

In 2010, an outbreak of febrile illness with arthralgic manifestations was detected at La Estación village, Portuguesa State, Venezuela. The etiologic agent was determined to be Mayaro virus (MAYV), a reemerging South American alphavirus. A total of 77 cases was reported and 19 were confirmed as seropositive. MAYV was isolated from acute-phase serum samples from 6 symptomatic patients. We sequenced 27 complete genomes representing the full spectrum of MAYV genetic diversity, which facilitated detection of a new genotype, designated N. Phylogenetic analysis of genomic sequences indicated that etiologic strains from Venezuela belong to genotype D. Results indicate that MAYV is highly conserved genetically, showing ≈17% nucleotide divergence across all 3 genotypes and 4% among genotype D strains in the most variable genes. Coalescent analyses suggested genotypes D and L diverged ≈150 years ago and genotype diverged N ≈250 years ago. This virus commonly infects persons residing near enzootic transmission foci because of anthropogenic incursions.


Asunto(s)
Infecciones por Alphavirus/epidemiología , Alphavirus/genética , Evolución Biológica , Biota/inmunología , Brotes de Enfermedades , Alphavirus/crecimiento & desarrollo , Femenino , Humanos , Masculino , Filogenia , Venezuela/epidemiología
17.
J Gen Virol ; 96(10): 3023-3034, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26297344

RESUMEN

Salmonid alphavirus (SAV) is the causative agent of pancreas disease affecting Atlantic salmon and rainbow trout and causes a major burden to the aquaculture industry. This study describes a Norwegian subtype SAV3 virus isolate (SAV3-H10) subjected to serial passages in Chinook salmon embryo cells (CHSE-214) followed by Asian Grouper skin cells (AGK). Two passages from CHSE and one after transfer to AGK cells were chosen for further investigation, based on variation in degree and development of cytopathic effect (CPE). After plaque purification, several in vitro studies were performed. Cell viability after infection, viral replication and ability to cause morphological changes in CHSE and AGK cells was studied for the three isolates. The AGK-transferred isolate was identified with the strongest abilities to reduce cell viability, replicate more and cause more CPE in cell culture when compared with the early and late CHSE-grown isolates. Subsequently, the isolates were tested in an experimental fish challenge, showing higher viral load and higher pathological score for the least cell-cultured isolate. Full-length sequencing of the viral genome of the three isolates revealed divergence in four amino acid positions and the AGK-grown isolate also had a 3 nt deletion in the 3'UTR. In conclusion, we show that cell culture of SAV3-H10 selects for strains inducing earlier CPE in vitro with increased viral replication. In vivo, the effect is reversed, with lower replication levels and lower pathology scores in target organs. This study outlines a path to identify potential virulence motifs of SAV3.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Alphavirus/genética , Salmo salar/virología , Adaptación Biológica , Alphavirus/patogenicidad , Alphavirus/fisiología , Animales , Supervivencia Celular , Efecto Citopatogénico Viral , Células Epiteliales/citología , Células Epiteliales/fisiología , Células Epiteliales/virología , Viabilidad Microbiana , Pase Seriado , Virulencia , Cultivo de Virus , Replicación Viral
18.
Microb Biotechnol ; 7(5): 480-4, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24418177

RESUMEN

Salmonid alphavirus (SAV) causes pancreas disease and sleeping disease in Atlantic salmon (Salmo salar) and rainbow trout (Oncorhynchus mykiss) and confers a major burden to the aquaculture industry. A commercial inactivated whole virus vaccine propagated in a salmon cell line at low temperature provides effective protection against SAV infections. Alphaviruses (family Togaviridae) are generally transmitted between vertebrate hosts via blood-sucking arthropod vectors, typically mosquitoes. SAV is unique in this respect because it can be transmitted directly from fish to fish and has no known invertebrate vector. Here, we show for the first time that SAV is able to complete a full infectious cycle within arthropod cells derived from the Asian tiger mosquito Aedes albopictus. Progeny virus is produced in C6/36 and U4.4. cells in a temperature-dependent manner (at 15 °C but not at 18 °C), can be serially passaged and remains infectious to salmonid Chinook salmon embryo cells. This suggests that SAV is not a vertebrate-restricted alphavirus after all and may have the potential to replicate in invertebrates. The current study also shows the ability of SAV to be propagated in mosquito cells, thereby possibly providing an alternative SAV production system for vaccine applications.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Alphavirus/aislamiento & purificación , Salmonidae/virología , Cultivo de Virus , Aedes , Alphavirus/fisiología , Animales , Línea Celular , Salmón , Temperatura , Vacunas Virales/aislamiento & purificación , Replicación Viral
19.
J Virol ; 86(17): 9484-94, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22761388

RESUMEN

Members of the Alphavirus genus are arboviruses that alternate replication in mosquitoes and vertebrate hosts. In vertebrate cells, the alphavirus resists the activation of antiviral RNA-activated protein kinase (PKR) by the presence of a prominent RNA structure (downstream loop [DLP]) located in viral 26S transcripts, which allows an eIF2-independent translation initiation of these mRNAs. This article shows that DLP structure is essential for replication of Sindbis virus (SINV) in vertebrate cell lines and animals but is dispensable for replication in insect cells, where no ortholog of the vertebrate PKR gene has been found. Sequence comparisons and structural RNA analysis revealed the evolutionary conservation of DLP in SINV and predicted the existence of equivalent DLP structures in many members of the Alphavirus genus. A mutant SINV lacking the DLP structure evolved in murine cells to recover a wild-type phenotype by creating an alternative structure in the RNA that restored the translational independence for eIF2. Genetic, phylogenetic, and biochemical data presented here support an evolutionary scenario for the natural history of alphaviruses, in which the acquisition of DLP structure in their mRNAs probably allowed the colonization of vertebrate host and the consequent geographic expansion of some of these viruses worldwide.


Asunto(s)
Alphavirus/crecimiento & desarrollo , Alphavirus/genética , Biosíntesis de Proteínas , ARN Viral/genética , Vertebrados/virología , Adaptación Fisiológica , Alphavirus/química , Alphavirus/clasificación , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Evolución Molecular , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Secuencias Invertidas Repetidas , Ratones , Datos de Secuencia Molecular , Mariposas Nocturnas , Conformación de Ácido Nucleico , Filogenia , ARN Viral/química , ARN Viral/metabolismo , Replicación Viral
20.
PLoS One ; 7(3): e33007, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427930

RESUMEN

We established a rapid, specific technique for detecting alphaviruses using a replicon-defective reporter gene assay derived from the Sindbis virus XJ-160. The pVaXJ expression vector containing the XJ-160 genome was engineered to form the expression vectors pVaXJ-EGFP expressing enhanced green fluorescence protein (EGFP) or pVaXJ-GLuc expressing Gaussia luciferase (GLuc). The replicon-defective reporter plasmids pVaXJ-EGFPΔnsp4 and pVaXJ-GLucΔnsp4 were constructed by deleting 1139 bp in the non-structural protein 4 (nsP4) gene. The deletion in the nsP4 gene prevented the defective replicons from replicating and expressing reporter genes in transfected BHK-21 cells. However, when these transfected cells were infected with an alphavirus, the non-structural proteins expressed by the alphavirus could act on the defective replicons in trans and induce the expression of the reporter genes. The replicon-defective plasmids were used to visualize the presence of alphavirus qualitatively or detect it quantitatively. Specificity tests showed that this assay could detect a variety of alphaviruses from tissue cultures, while other RNA viruses, such as Japanese encephalitis virus and Tahyna virus, gave negative results with this system. Sensitivity tests showed that the limit of detection (LOD) of this replicon-defective assay is between 1 and 10 PFU for Sindbis viruses. These results indicate that, with the help of the replicon-defective alphavirus detection technique, we can specifically, sensitively, and rapidly detect alphaviruses in tissue cultures. The detection technique constructed here may be well suited for use in clinical examination and epidemiological surveillance, as well as for rapid screening of potential viral biological warfare agents.


Asunto(s)
Infecciones por Alphavirus/diagnóstico , Alphavirus/aislamiento & purificación , Genes Reporteros/genética , Ensayo de Placa Viral/métodos , Alphavirus/crecimiento & desarrollo , Infecciones por Alphavirus/genética , Ingeniería Genética/métodos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Límite de Detección , Sensibilidad y Especificidad , Virus Sindbis/genética , Proteínas no Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...