Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Virus Res ; 292: 198228, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33188797

RESUMEN

Given the dual life cycle of arboviruses in insect and animal hosts and the importance of serum factors as a first line antiviral defense, we have examined the outcome of interactions between the arbovirus La Crosse Virus (LACV) and human serum. To mimic the life cycle between species, we used LACV derived from insect (I-LACV) and human keratinocyte (HaCaT) cells. Incubation of I-LACV with normal human serum did not result in neutralization, but instead stabilized I-LACV virions and enhanced the amount of infectious virus. Enhanced infectivity was also seen with heat-inactivated serum devoid of complement activity and with serum from a range of animals including mouse, ferret, and non-human primates. Depletion of antibodies from serum resulted in loss of enhancement of infectivity and sucrose gradient sedimentation assays showed IgG co-sedimenting with I-LACV particles. In agreement with our results with I-LACV, HaCaT-derived LACV was not neutralized by complement or antibodies in normal human serum. However, in contrast to I-LACV, HaCaT-derived LACV infectivity was stable when incubated alone and treatment with serum did not enhance infectivity. Our results indicate that LACV derived from insect cells differs substantially from virus derived from human cells, with I-LACV being dependent on serum factors to enhance infectivity. These findings suggest that understanding differential composition of insect versus animal cell-derived LACV may form the foundation for potential new antiviral approaches.


Asunto(s)
Encefalitis de California/virología , Insectos/virología , Queratinocitos/virología , Virus La Crosse/fisiología , Suero/inmunología , Animales , Línea Celular , Modelos Animales de Enfermedad , Encefalitis de California/inmunología , Hurones , Interacciones Huésped-Patógeno , Humanos , Queratinocitos/inmunología , Virus La Crosse/genética , Virus La Crosse/inmunología , Ratones , Pruebas de Neutralización , Primates , Replicación Viral
2.
Viruses ; 11(3)2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917612

RESUMEN

La Crosse virus (LACV) is a mosquito-transmitted arbovirus and the main cause of virus-mediated neurological diseases in children. To date, little is known about the role of C-type lectin receptors (CLRs)-an important class of pattern recognition receptors-in LACV recognition. DC-SIGN remains the only well-described CLR that recognizes LACV. In this study, we investigated the role of additional CLR/LACV interactions. To this end, we applied a flow-through chromatography method for the purification of LACV to perform an unbiased high-throughput screening of LACV with a CLR-hFc fusion protein library. Interestingly, the CARD9-associated CLRs Mincle, Dectin-1, and Dectin-2 were identified to strongly interact with LACV. Since CARD9 is a common adaptor protein for signaling via Mincle, Dectin-1, and Dectin-2, we performed LACV infection of Mincle-/- and CARD9-/- DCs. Mincle-/- and CARD9-/- DCs produced less amounts of proinflammatory cytokines, namely IL-6 and TNF-α, albeit no reduction of the LACV titer was observed. Together, novel CLR/LACV interactions were identified; however, the Mincle/CARD9 axis plays a limited role in early antiviral responses against LACV.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/inmunología , Encefalitis de California/inmunología , Inmunidad Innata , Lectinas Tipo C/inmunología , Proteínas de la Membrana/inmunología , Transducción de Señal/inmunología , Animales , Antivirales , Proteínas Adaptadoras de Señalización CARD/genética , Citocinas/inmunología , Células Dendríticas/inmunología , Virus La Crosse/inmunología , Lectinas Tipo C/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Th17/inmunología
3.
Stem Cells Dev ; 25(8): 648-59, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26906411

RESUMEN

The innate immunity of embryonic stem cells (ESCs) has recently emerged as an important issue in ESC biology and in ESC-based regenerative medicine. We have recently reported that mouse ESCs (mESCs) do not have a functional type I interferon (IFN)-based antiviral innate immunity. They are deficient in expressing IFN in response to viral infection and have limited ability to respond to IFN. Using fibroblasts (FBs) as a cell model, the current study investigated the development of antiviral mechanisms during in vitro differentiation of mESCs. We demonstrate that mESC-differentiated FBs (mESC-FBs) share extensive similarities with naturally differentiated FBs in morphology, marker expression, and growth pattern, but their development of antiviral mechanisms lags behind. Nonetheless, the antiviral mechanisms are inducible during mESC differentiation as demonstrated by the transition of nuclear factor kappa B (NFκB), a key transcription factor for IFN expression, from its inactive state in mESCs to its active state in mESC-FBs and by increased responses of mESC-FBs to viral stimuli and IFN during their continued in vitro propagation. Together with our previously published study, the current data provide important insights into molecular basis for the deficiency of IFN expression in mESCs and the development of antiviral innate immunity during mESC differentiation.


Asunto(s)
Diferenciación Celular , Inmunidad Innata , Células Madre Embrionarias de Ratones/inmunología , Animales , Línea Celular , Virus Chikungunya/inmunología , Chlorocebus aethiops , Técnicas de Cocultivo , Interferón Tipo I/metabolismo , Virus La Crosse/inmunología , Ratones , Células Madre Embrionarias de Ratones/fisiología , Células Madre Embrionarias de Ratones/virología , FN-kappa B/metabolismo , Medicina Regenerativa , Células Vero
4.
J Virol ; 88(19): 11070-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25008929

RESUMEN

UNLABELLED: La Crosse virus (LACV) is the major cause of pediatric viral encephalitis in the United States; however, the mechanisms responsible for age-related susceptibility in the pediatric population are not well understood. Our current studies in a mouse model of LACV infection indicated that differences in myeloid dendritic cell (mDC) responses between weanling and adult mice accounted for susceptibility to LACV-induced neurological disease. We found that type I interferon (IFN) responses were significantly stronger in adult than in weanling mice. Production of these IFNs required both endosomal Toll-like receptors (TLRs) and cytoplasmic RIG-I-like receptors (RLRs). Surprisingly, IFN expression was not dependent on plasmacytoid DCs (pDCs) but rather was dependent on mDCs, which were found in greater number and induced stronger IFN responses in adults than in weanlings. Inhibition of these IFN responses in adults resulted in susceptibility to LACV-induced neurological disease, whereas postinfection treatment with type I IFN provided protection in young mice. These studies provide a definitive mechanism for age-related susceptibility to LACV encephalitis, where mDCs in young mice are insufficiently activated to control peripheral virus replication, thereby allowing virus to persist and eventually cause central nervous system (CNS) disease. IMPORTANCE: La Crosse virus (LACV) is the primary cause of pediatric viral encephalitis in the United States. Although the virus infects both adults and children, over 80% of the reported neurological disease cases are in children. To understand why LACV causes neurological disease primarily in young animals, we used a mouse model where weanling mice, but not adult mice, develop neurological disease following virus infection. We found that an early immune response cell type, myeloid dendritic cells, was critical for protection in adult animals and that these cells were reduced in young animals. Activation of these cells during virus infection or after treatment with type I interferon in young animals provided protection from LACV. Thus, this study demonstrates a reason for susceptibility to LACV infection in young animals and shows that early therapeutic treatment in young animals can prevent neurological disease.


Asunto(s)
Sistema Nervioso Central/inmunología , Células Dendríticas/inmunología , Encefalitis de California/inmunología , Virus La Crosse/inmunología , Células Mieloides/inmunología , Factores de Edad , Animales , Animales Recién Nacidos , Sistema Nervioso Central/virología , Células Dendríticas/virología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalitis de California/mortalidad , Encefalitis de California/virología , Expresión Génica/inmunología , Humanos , Inyecciones Intradérmicas , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Ratones , Células Mieloides/virología , Análisis de Supervivencia , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Replicación Viral
5.
J Biol Chem ; 289(36): 25186-98, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-24966329

RESUMEN

We have recently reported that mouse embryonic stem cells (mESCs) are deficient in expressing type I interferons (IFNs) in response to viral infection and synthetic viral RNA analogs (Wang, R., Wang, J., Paul, A. M., Acharya, D., Bai, F., Huang, F., and Guo, Y. L. (2013) J. Biol. Chem. 288, 15926-15936). Here, we report that mESCs are able to respond to type I IFNs, express IFN-stimulated genes, and mediate the antiviral effect of type I IFNs against La Crosse virus and chikungunya virus. The major signaling components in the IFN pathway are expressed in mESCs. Therefore, the basic molecular mechanisms that mediate the effects of type I IFNs are functional in mESCs; however, these mechanisms may not yet be fully developed as mESCs express lower levels of IFN-stimulated genes and display weaker antiviral activity in response to type I IFNs when compared with fibroblasts. Further analysis demonstrated that type I IFNs do not affect the stem cell state of mESCs. We conclude that mESCs are deficient in type I IFN expression, but they can respond to and mediate the cellular effects of type I IFNs. These findings represent unique and uncharacterized properties of mESCs and are important for understanding innate immunity development and ESC physiology.


Asunto(s)
Antivirales/inmunología , Células Madre Embrionarias/inmunología , Fibroblastos/inmunología , Interferón Tipo I/inmunología , Animales , Antivirales/metabolismo , Antivirales/farmacología , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Células Cultivadas , Virus Chikungunya/inmunología , Virus Chikungunya/fisiología , Chlorocebus aethiops , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/virología , Fibroblastos/metabolismo , Fibroblastos/virología , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/farmacología , Virus La Crosse/inmunología , Virus La Crosse/fisiología , Ratones , Microscopía Confocal , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ubiquitinas/genética , Ubiquitinas/inmunología , Ubiquitinas/metabolismo , Células Vero
6.
J Neurovirol ; 20(2): 150-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23846288

RESUMEN

Viral encephalitis represents a significant, and costly, public health threat particularly for high-risk pediatric populations. An emerging mosquito-borne pathogen endemic to the United States, La Crosse virus (LACV) is one of the most common causes of viral encephalitis in children in the United States. However, no licensed therapeutics or vaccines currently exist for treatment. Hampering development efforts, the host response to LACV and its role in disease pathogenesis has only recently been examined. In this review, we discuss the current understanding of innate immune response in the context of viral pathogenesis and host susceptibility to LACV. In addition, we address the need for a clearer understanding of the early host-virus interactions in LACV infections as it relates to viral pathogenesis in the central nervous system.


Asunto(s)
Sistema Nervioso Central/inmunología , Reservorios de Enfermedades/veterinaria , Encefalitis de California/inmunología , Inmunidad Innata , Virus La Crosse/inmunología , Aedes/virología , Animales , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Niño , Reservorios de Enfermedades/virología , Susceptibilidad a Enfermedades , Vectores de Enfermedades , Encefalitis de California/patología , Encefalitis de California/transmisión , Encefalitis de California/virología , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Interferón Tipo I/biosíntesis , Ratones
7.
J Virol ; 87(15): 8451-64, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720721

RESUMEN

We show that interferon-induced transmembrane protein 1 (IFITM-1), IFITM-2, and IFITM-3 exhibit a broad spectrum of antiviral activity against several members of the Bunyaviridae family, including Rift Valley fever virus (RVFV), La Crosse virus, Andes virus, and Hantaan virus, all of which can cause severe disease in humans and animals. We found that RVFV was restricted by IFITM-2 and -3 but not by IFITM-1, whereas the remaining viruses were equally restricted by all IFITMs. Indeed, at low doses of alpha interferon (IFN-α), IFITM-2 and -3 mediated more than half of the antiviral activity of IFN-α against RVFV. IFITM-2 and -3 restricted RVFV infection mostly by preventing virus membrane fusion with endosomes, while they had no effect on virion attachment to cells, endocytosis, or viral replication kinetics. We found that large fractions of IFITM-2 and IFITM-3 occupy vesicular compartments that are distinct from the vesicles coated by IFITM-1. In addition, although overexpression of all IFITMs expanded vesicular and acidified compartments within cells, there were marked phenotypic differences among the vesicular compartments occupied by IFITMs. Collectively, our data provide new insights into the possible mechanisms by which the IFITM family members restrict distinct viruses.


Asunto(s)
Antígenos de Diferenciación/inmunología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Línea Celular , Virus Hantaan/inmunología , Virus Hantaan/fisiología , Orthohantavirus/inmunología , Orthohantavirus/fisiología , Humanos , Interferón-alfa/inmunología , Virus La Crosse/inmunología , Virus La Crosse/fisiología
8.
Immunity ; 38(4): 705-16, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23499490

RESUMEN

La Crosse virus (LACV), a zoonotic Bunyavirus, is a major cause of pediatric viral encephalitis in the United States. A hallmark of neurological diseases caused by LACV and other encephalitic viruses is the induction of neuronal cell death. Innate immune responses have been implicated in neuronal damage, but no mechanism has been elucidated. By using in vitro studies in primary neurons and in vivo studies in mice, we have shown that LACV infection induced the RNA helicase, RIG-I, and mitochondrial antiviral signaling protein (MAVS) signaling pathway, resulting in upregulation of the sterile alpha and TIR-containing motif 1 (SARM1), an adaptor molecule that we found to be directly involved in neuronal damage. SARM1-mediated cell death was associated with induced oxidative stress response and mitochondrial damage. These studies provide an innate-immune signaling mechanism for virus-induced neuronal death and reveal potential targets for development of therapeutics to treat encephalitic viral infections.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Proteínas del Dominio Armadillo/metabolismo , Proteínas del Citoesqueleto/metabolismo , Encefalitis de California/inmunología , Virus La Crosse/inmunología , Mitocondrias/metabolismo , Neuronas/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteínas del Dominio Armadillo/genética , Células Cultivadas , Proteínas del Citoesqueleto/genética , Encefalitis de California/complicaciones , Encefalitis de California/tratamiento farmacológico , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida , Neuronas/virología , Estrés Oxidativo , Cultivo Primario de Células , Transducción de Señal/inmunología , Regulación hacia Arriba
9.
J Virol ; 86(20): 11223-30, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22875966

RESUMEN

Beta interferon (IFN-ß) is a major component of innate immunity in mammals, but information on the in vivo source of this cytokine after pathogen infection is still scarce. To identify the cell types responsible for IFN-ß production during viral encephalitis, we used reporter mice that express firefly luciferase under the control of the IFN-ß promoter and stained organ sections with luciferase-specific antibodies. Numerous luciferase-positive cells were detected in regions of La Crosse virus (LACV)-infected mouse brains that contained many infected cells. Double-staining experiments with cell-type-specific markers revealed that similar numbers of astrocytes and microglia of infected brains were luciferase positive, whereas virus-infected neurons rarely contained detectable levels of luciferase. Interestingly, if a mutant LACV unable of synthesizing the IFN-antagonistic factor NSs was used for challenge, the vast majority of the IFN-ß-producing cells in infected brains were astrocytes rather than microglia. Similar conclusions were reached in a second series of experiments in which conditional reporter mice expressing the luciferase reporter gene solely in defined cell types were infected with wild-type or mutant LACV. Collectively, our data suggest that glial cells rather than infected neurons represent the major source of IFN-ß in LACV-infected mouse brains. They further indicate that IFN-ß synthesis in astrocytes and microglia is differentially affected by the viral IFN antagonist, presumably due to differences in LACV susceptibility of these two cell types.


Asunto(s)
Astrocitos/inmunología , Encefalitis de California/inmunología , Interferón beta/biosíntesis , Virus La Crosse/inmunología , Microglía/inmunología , Animales , Astrocitos/metabolismo , Astrocitos/virología , Encéfalo/metabolismo , Encéfalo/virología , Interferón beta/genética , Interferón beta/inmunología , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Microglía/virología , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/virología , Regiones Promotoras Genéticas
10.
J Virol ; 86(1): 420-6, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22013033

RESUMEN

La Crosse virus (LACV) and Jamestown Canyon virus (JCV), family Bunyaviridae, are mosquito-borne viruses that are endemic in North America and recognized as etiologic agents of encephalitis in humans. Both viruses belong to the California encephalitis virus serogroup, which causes 70 to 100 cases of encephalitis a year. As a first step in creating live attenuated viral vaccine candidates for this serogroup, we have generated a recombinant LACV expressing the attachment/fusion glycoproteins of JCV. The JCV/LACV chimeric virus contains full-length S and L segments derived from LACV. For the M segment, the open reading frame (ORF) of LACV is replaced with that derived from JCV and is flanked by the untranslated regions of LACV. The resulting chimeric virus retained the same robust growth kinetics in tissue culture as observed for either parent virus, and the virus remains highly infectious and immunogenic in mice. Although both LACV and JCV are highly neurovirulent in 21 day-old mice, with 50% lethal dose (LD50) values of 0.1 and 0.5 log10 PFU, respectively, chimeric JCV/LACV is highly attenuated and does not cause disease even after intracerebral inoculation of 10³ PFU. Parenteral vaccination of mice with 10¹ or 10³ PFU of JCV/LACV protected against lethal challenge with LACV, JCV, and Tahyna virus (TAHV). The chimeric virus was infectious and immunogenic in rhesus monkeys and induced neutralizing antibodies to JCV, LACV, and TAHV. When vaccinated monkeys were challenged with JCV, they were protected against the development of viremia. Generation of highly attenuated yet immunogenic chimeric bunyaviruses could be an efficient general method for development of vaccines effective against these pathogenic viruses.


Asunto(s)
Quimera/inmunología , Virus de la Encefalitis de California/genética , Encefalitis de California/prevención & control , Expresión Génica , Virus La Crosse/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Quimera/genética , Quimera/crecimiento & desarrollo , Encefalitis de California/inmunología , Encefalitis de California/virología , Humanos , Virus La Crosse/genética , Virus La Crosse/crecimiento & desarrollo , Macaca mulatta , Ratones , Vacunación , Proteínas del Envoltorio Viral/genética , Vacunas Virales/genética , Vacunas Virales/inmunología
12.
PLoS Negl Trop Dis ; 4(10): e856, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-21049065

RESUMEN

Mosquitoes rely on RNA interference (RNAi) as their primary defense against viral infections. To this end, the combination of RNAi and invertebrate cell culture systems has become an invaluable tool in studying virus-vector interactions. Nevertheless, a recent study failed to detect an active RNAi response to West Nile virus (WNV) infection in C6/36 (Aedes albopictus) cells, a mosquito cell line frequently used to study arthropod-borne viruses (arboviruses). Therefore, we sought to determine if WNV actively evades the host's RNAi response or if C6/36 cells have a dysfunctional RNAi pathway. C6/36 and Drosophila melanogaster S2 cells were infected with WNV (Flaviviridae), Sindbis virus (SINV, Togaviridae) and La Crosse virus (LACV, Bunyaviridae) and total RNA recovered from cell lysates. Small RNA (sRNA) libraries were constructed and subjected to high-throughput sequencing. In S2 cells, virus-derived small interfering RNAs (viRNAs) from all three viruses were predominantly 21 nt in length, a hallmark of the RNAi pathway. However, in C6/36 cells, viRNAs were primarily 17 nt in length from WNV infected cells and 26-27 nt in length in SINV and LACV infected cells. Furthermore, the origin (positive or negative viral strand) and distribution (position along viral genome) of S2 cell generated viRNA populations was consistent with previously published studies, but the profile of sRNAs isolated from C6/36 cells was altered. In total, these results suggest that C6/36 cells lack a functional antiviral RNAi response. These findings are analogous to the type-I interferon deficiency described in Vero (African green monkey kidney) cells and suggest that C6/36 cells may fail to accurately model mosquito-arbovirus interactions at the molecular level.


Asunto(s)
Aedes/virología , Interacciones Huésped-Patógeno , Virus La Crosse/inmunología , Interferencia de ARN , Virus Sindbis/inmunología , Virus del Nilo Occidental/inmunología , Aedes/inmunología , Animales , Línea Celular , Etiquetas de Secuencia Expresada , Perfilación de la Expresión Génica , Virus La Crosse/crecimiento & desarrollo , Análisis de Secuencia de ADN , Virus Sindbis/crecimiento & desarrollo , Virus del Nilo Occidental/crecimiento & desarrollo
13.
J Immunol ; 183(5): 3229-36, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19667093

RESUMEN

Type I IFN is a major player in innate and adaptive immune responses. Besides, it is involved in organogenesis and tumor development. Generally, IFN responses are amplified by an autocrine loop with IFN-beta as the priming cytokine. However, due to the lack of sensitive detection systems, where and how type I IFN is produced in vivo is still poorly understood. In this study, we describe a luciferase reporter mouse, which allows tracking of IFN-beta gene induction in vivo. Using this reporter mouse, we reveal strong tissue-specific induction of IFN-beta following infection with influenza or La Crosse virus. Importantly, this reporter mouse also allowed us to visualize that IFN-beta is expressed constitutively in several tissues. As suggested before, low amounts of constitutively produced IFN might maintain immune cells in an activated state ready for a timely response to pathogens. Interestingly, thymic epithelial cells were the major source of IFN-beta under noninflammatory conditions. This relatively high constitutive expression was controlled by the NF Aire and might influence induction of tolerance or T cell development.


Asunto(s)
Genes Reporteros , Mediadores de Inflamación/fisiología , Interferón beta/biosíntesis , Interferón beta/genética , Animales , Línea Celular Transformada , Células Cultivadas , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Genes Reporteros/inmunología , Mediadores de Inflamación/metabolismo , Subtipo H7N7 del Virus de la Influenza A/genética , Subtipo H7N7 del Virus de la Influenza A/inmunología , Interferón beta/deficiencia , Virus La Crosse/genética , Virus La Crosse/inmunología , Luciferasas de Luciérnaga/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Enfermedad de Newcastle/genética , Enfermedad de Newcastle/inmunología , Enfermedad de Newcastle/patología , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Timo/inmunología , Timo/patología , Timo/virología
14.
Virol J ; 5: 25, 2008 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-18267012

RESUMEN

BACKGROUND: La Crosse virus (LACV), family Bunyaviridae, was first identified as a human pathogen in 1960 after its isolation from a 4 year-old girl with fatal encephalitis in La Crosse, Wisconsin. LACV is a major cause of pediatric encephalitis in North America and infects up to 300,000 persons each year of which 70-130 result in severe disease of the central nervous system (CNS). As an initial step in the establishment of useful animal models to support vaccine development, we examined LACV infectivity, pathogenesis, and immunogenicity in both weanling mice and rhesus monkeys. RESULTS: Following intraperitoneal inoculation of mice, LACV replicated in various organs before reaching the CNS where it replicates to high titer causing death from neurological disease. The peripheral site where LACV replicates to highest titer is the nasal turbinates, and, presumably, LACV can enter the CNS via the olfactory neurons from nasal olfactory epithelium. The mouse infectious dose50 and lethal dose50 was similar for LACV administered either intranasally or intraperitoneally. LACV was highly infectious for rhesus monkeys and infected 100% of the animals at 10 PFU. However, the infection was asymptomatic, and the monkeys developed a strong neutralizing antibody response. CONCLUSION: In mice, LACV likely gains access to the CNS via the blood stream or via olfactory neurons. The ability to efficiently infect mice intranasally raises the possibility that LACV might use this route to infect its natural hosts. Rhesus monkeys are susceptible to LACV infection and develop strong neutralizing antibody responses after inoculation with as little as 10 PFU. Mice and rhesus monkeys are useful animal models for LACV vaccine immunologic testing although the rhesus monkey model is not optimal.


Asunto(s)
Modelos Animales de Enfermedad , Encefalitis de California/virología , Virus La Crosse/inmunología , Virus La Crosse/patogenicidad , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Línea Celular , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Chlorocebus aethiops , Femenino , Humanos , Virus La Crosse/fisiología , Dosificación Letal Mediana , Macaca mulatta , Ratones , Pruebas de Neutralización , Nariz/virología , Peritoneo/virología , Células Vero , Replicación Viral
15.
J Virol ; 81(10): 4991-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17344298

RESUMEN

La Crosse virus (LACV) is a mosquito-transmitted member of the Bunyaviridae family that causes severe encephalitis in children. For the LACV nonstructural protein NSs, previous overexpression studies with mammalian cells had suggested two different functions, namely induction of apoptosis and inhibition of RNA interference (RNAi). Here, we demonstrate that mosquito cells persistently infected with LACV do not undergo apoptosis and mount a specific RNAi response. Recombinant viruses that either express (rLACV) or lack (rLACVdelNSs) the NSs gene similarly persisted and were prone to the RNAi-mediated resistance to superinfection. Furthermore, in mosquito cells overexpressed LACV NSs was unable to inhibit RNAi against Semliki Forest virus. In mammalian cells, however, the rLACVdelNSs mutant virus strongly activated the antiviral type I interferon (IFN) system, whereas rLACV as well as overexpressed NSs suppressed IFN induction. Consequently, rLACVdelNSs was attenuated in IFN-competent mouse embryo fibroblasts and animals but not in systems lacking the type I IFN receptor. In situ analyses of mouse brains demonstrated that wild-type and mutant LACV mainly infect neuronal cells and that NSs is able to suppress IFN induction in the central nervous system. Thus, our data suggest little relevance of the NSs-induced apoptosis or RNAi inhibition for growth or pathogenesis of LACV in the mammalian host and indicate that NSs has no function in the insect vector. Since deletion of the viral NSs gene can be fully complemented by inactivation of the host's IFN system, we propose that the major biological function of NSs is suppression of the mammalian innate immune response.


Asunto(s)
Interferón Tipo I/antagonistas & inhibidores , Virus La Crosse/inmunología , Interferencia de ARN , Proteínas no Estructurales Virales/fisiología , Animales , Apoptosis , Encéfalo/patología , Encéfalo/virología , Línea Celular , Cricetinae , Culicidae , Modelos Animales de Enfermedad , Encefalitis de California/virología , Humanos , Inmunohistoquímica , Virus La Crosse/patogenicidad , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neuronas/virología , Virus de los Bosques Semliki/crecimiento & desarrollo
16.
Am J Trop Med Hyg ; 74(2): 224-32, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16474075

RESUMEN

La Crosse (LAC) virus (family Bunyaviridae, genus Orthobunyavirus) small (S) segment negative-sense RNA genome (vRNA), positive-sense full-length RNA complement (vcRNA), and subgenomic mRNA were assayed in infected cell cultures and female Aedes (Ochlerotatus) triseriatus mosquito tissues using quantitative PCR (Q-PCR). During persistent infection of C6/36 (Aedes albopictus) and MAT (Aedes triseriatus) cultured cells and cytolytic infection of BHK-21 cultured cells, LAC vRNA was the most abundant RNA species, followed by mRNA and vcRNA. RNA copy numbers per cell were quantified and vRNA correlated to virus titer in cell culture medium. The Q-PCR assay proved more sensitive than reverse transcription (RT)-PCR and immunofluorescence assays (IFA) for detecting LAC virus infection of mosquitoes. After infection of female mosquitoes orally, quantities of LAC RNA increased in ovaries for 6 days, and as ovarian biosynthetic activity quiesced, LAC RNA quantities decreased then remained detectable at a low level. After a second, noninfectious blood meal, quantities of LAC RNA in ovaries increased significantly, quantitatively confirming correlation of LAC virus RNA synthesis with vector metabolic activity. Coregulation of viral replication and mosquito ovary metabolic activity may condition efficient transovarial transmission.


Asunto(s)
Encefalitis de California/virología , Virus La Crosse/genética , ARN Viral/biosíntesis , Aedes/virología , Animales , Células Cultivadas , Chlorocebus aethiops , Cricetinae , Cartilla de ADN , Técnica del Anticuerpo Fluorescente Directa , Regulación Viral de la Expresión Génica , Humanos , Virus La Crosse/inmunología , Virus La Crosse/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Células Vero , Replicación Viral
17.
Intervirology ; 43(4-6): 312-21, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11251387

RESUMEN

For the development of effective conventional vaccines or DNA vaccines against viruses, the availability of suitable animal models is an essential prerequisite. For many recently emerging zoonotic viruses, suitable animal models are still missing. We have established a novel small animal model for DNA vaccines using mice lacking a functional interferon-alpha/beta receptor (IFNAR-1). IFNAR-1-deficient mice are highly susceptible to many different viruses despite their ability to mount a normal humoral and cellular immune response. Taking advantage of this animal model, we show that mice can be completely protected from lethal challenge with a single injection of plasmid DNA encoding the viral envelope proteins G1 and G2. By contrast, vaccination with a plasmid encoding the internal nucleocapsid protein N had little effect. In an effort to enhance the protective immune response to N we assessed the efficacy of vaccination with plasmid DNA encoding N in combination with a plasmid encoding the cytokine IL-12 as adjuvant. IL-12 enhanced the survival of mice following viral challenge, but the effect was independent of N indicating the involvement of components of the innate immune system such as NK cells.


Asunto(s)
Encefalitis de California/prevención & control , Virus La Crosse/inmunología , Plásmidos/inmunología , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Interleucina-12/genética , Interleucina-12/inmunología , Proteínas de la Membrana , Ratones , Receptor de Interferón alfa y beta , Receptores de Interferón/deficiencia , Vacunación , Proteínas del Envoltorio Viral/genética , Vacunas Virales/genética
18.
Hum Gene Ther ; 10(10): 1649-58, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10428210

RESUMEN

La Crosse virus (LACV)-mediated encephalitis is the most frequently reported arboviral disease in the United States, but to date no vaccine against this virus is available. We have established a new animal model, genetically targeted mice lacking a functional interferon type I receptor (IFNAR-1). These mice show an age-independent susceptibility to LACV and develop an acute encephalitis within 6 days of infection, thereby allowing the evaluation of vaccines against LACV. Taking advantage of this knockout mouse model, we have assessed the feasibility of DNA vaccination against this viral disease. Plasmid DNAs, encoding either the virus surface glycoproteins G1 and G2 or the internal nucleocapsid protein N, were used to immunize IFNAR-1-deficient mice. Mice vaccinated with DNA encoding the glycoproteins G1 and G2 produced neutralizing antibodies and exhibited a high degree of protection against challenge with high doses of LACV. Depletion of CD4+ T cells in mice vaccinated with DNA encoding G1/G2 reduced their capacity to control the infection. Virus titration and immunohistological analysis revealed that the protected mice showed no evidence of LACV particles in the brain. This indicates that the vaccine-induced immune response efficiently blocked viral spreading from the primary replication site to the brain. In contrast, immunization with DNA encoding protein N yielded only a partial protective effect that can be attributed to the cellular immune response. Taken together, this study shows that DNA vaccines can be designed to efficiently induce a protective immune response based on neutralizing antibodies and CD4+ T cells.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Encefalitis de California/prevención & control , Virus La Crosse/inmunología , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Animales , Línea Celular , Línea Celular Transformada , Chlorocebus aethiops , Cricetinae , ADN Viral/inmunología , Modelos Animales de Enfermedad , Encefalitis de California/inmunología , Expresión Génica , Glicoproteínas/genética , Glicoproteínas/inmunología , Ratones , Ratones Noqueados , Pruebas de Neutralización , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/inmunología , Vacunación , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
19.
J Virol ; 73(8): 6984-91, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10400797

RESUMEN

The human MxA protein is part of the antiviral state induced by alpha/beta interferon (IFN-alpha/beta). MxA inhibits the multiplication of several RNA viruses in cell culture. However, its antiviral potential in vivo has not yet been fully explored. We have generated MxA-transgenic mice that lack a functional IFN system by crossing MxA-transgenic mice constitutively expressing MxA with genetically targeted (knockout) mice lacking the beta subunit of the IFN-alpha/beta receptor (IFNAR-1(-/-) mice). These mice are an ideal animal model to investigate the unique antiviral activity of human MxA in vivo, because they are unable to express other IFN-induced proteins. Here, we show that MxA confers resistance to Thogoto virus, La Crosse virus, and Semliki Forest virus. No Thogoto virus progeny was detectable in MxA-transgenic mice, indicating an efficient block of virus replication at the primary site of infection. In the case of La Crosse virus, MxA restricted invasion of the central nervous system. In contrast, Semliki Forest virus multiplication in the brain was detectable in both MxA-expressing and nonexpressing IFNAR-1(-/-) mice. However, viral titers were clearly reduced in MxA-transgenic mice. Our results demonstrate that MxA does not need the help of other IFN-induced proteins for activity but is a powerful antiviral agent on its own. Moreover, the results suggest that MxA may protect humans from potential fatal infections by La Crosse virus and other viral pathogens.


Asunto(s)
Antivirales/inmunología , Encefalitis de California/inmunología , Proteínas de Unión al GTP , Interferón-alfa/inmunología , Interferón beta/inmunología , Virus La Crosse/inmunología , Proteínas/inmunología , Receptores de Interferón/inmunología , Infecciones por Alphavirus/inmunología , Animales , Antivirales/biosíntesis , Antivirales/genética , Humanos , Virus La Crosse/crecimiento & desarrollo , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Proteínas de Resistencia a Mixovirus , Infecciones por Orthomyxoviridae/inmunología , Biosíntesis de Proteínas , Proteínas/genética , Receptor de Interferón alfa y beta , Receptores de Interferón/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Thogotovirus/inmunología
20.
J Clin Virol ; 13(1-2): 17-27, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10405888

RESUMEN

BACKGROUND: DNA vaccines have been shown to induce protective immunity against viral infections in different animal models. We have recently demonstrated that DNA vaccine induced protective immunity against influenza A virus and La Crosse virus (LACV) is primarily mediated by humoral immune response. OBJECTIVE: The goal of this study was to investigate whether administration of DNA coding for cytokines such as interleukin 12 (IL-12) and granulocyte-macrophage colony-stimulating factor (GM-CSF) could increase the protective immune response induced by vaccination with DNA coding for viral antigens. STUDY DESIGN: For the influenza A virus or LACV model, C57BL/6 or interferon-alpha/beta receptor (IFNAR-1)-deficient mice, respectively, were vaccinated once or twice with 100 micrograms of DNA encoding viral antigens. At the same time plasmid DNAs (100 micrograms) coding either for mouse GM-CSF or mouse IL-12 were administered. The mice were subsequently challenged with a lethal dose of influenza A virus or LACV and monitored for clinical symptoms (weight loss) and survival. RESULTS: To achieve a high degree of protection (70% survival) two injections of DNA encoding the influenza A virus surface protein hemagglutinin (HA) were required. Intriguingly, administration of DNA coding for IL-12 alone also led to a pronounced protective effect against virus challenge. Co-administration of DNAs encoding IL-12 and HA significantly increased the protective immunity against influenza A virus, while IL-12 expression did not improve protection upon vaccination with DNA coding for the internal nucleocapsid protein N of LACV. Co-injection of DNA coding for mouse GM-CSF and HA also showed an adjuvant effect. CONCLUSIONS: The data clearly indicate that co-administration of DNA encoding cytokines such as IL-12 and GM-CSF with DNA coding for viral antigens has adjuvant effects on the protective immune response against different viral pathogens.


Asunto(s)
Antígenos Virales/inmunología , Encefalitis de California/prevención & control , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Interleucina-12/inmunología , Virus La Crosse/inmunología , Proteínas de la Nucleocápside/inmunología , Vacunas de ADN/inmunología , Animales , Antígenos Virales/genética , Línea Celular , Cricetinae , ADN Viral/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Interleucina-12/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside/genética , Plásmidos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...