Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 588(7837): 308-314, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33208938

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a neurotropic alphavirus transmitted by mosquitoes that causes encephalitis and death in humans1. VEEV is a biodefence concern because of its potential for aerosol spread and the current lack of sufficient countermeasures. The host factors that are required for VEEV entry and infection remain poorly characterized. Here, using a genome-wide CRISPR-Cas9-based screen, we identify low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3)-a highly conserved yet poorly characterized member of the scavenger receptor superfamily-as a receptor for VEEV. Gene editing of mouse Ldlrad3 or human LDLRAD3 results in markedly reduced viral infection of neuronal cells, which is restored upon complementation with LDLRAD3. LDLRAD3 binds directly to VEEV particles and enhances virus attachment and internalization into host cells. Genetic studies indicate that domain 1 of LDLRAD3 (LDLRAD3(D1)) is necessary and sufficient to support infection by VEEV, and both anti-LDLRAD3 antibodies and an LDLRAD3(D1)-Fc fusion protein block VEEV infection in cell culture. The pathogenesis of VEEV infection is abrogated in mice with deletions in Ldlrad3, and administration of LDLRAD3(D1)-Fc abolishes disease caused by several subtypes of VEEV, including highly virulent strains. The development of a decoy-receptor fusion protein suggests a strategy for the prevention of severe VEEV infection and associated disease in humans.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/metabolismo , Receptores de LDL/metabolismo , Receptores Virales/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/prevención & control , Encefalomielitis Equina Venezolana/virología , Femenino , Prueba de Complementación Genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores Virales/genética , Acoplamiento Viral , Internalización del Virus
2.
Virology ; 539: 121-128, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31733451

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a neurotropic virus that causes significant disease in both humans and equines. Here we characterized the impact of VEEV on signaling pathways regulating cell death in human primary astrocytes. VEEV productively infected primary astrocytes and caused an upregulation of early growth response 1 (EGR1) gene expression at 9 and 18 h post infection. EGR1 induction was dependent on extracellular signal-regulated kinase1/2 (ERK1/2) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), but not on p38 mitogen activated protein kinase (MAPK) or phosphoinositide 3-kinase (PI3K) signaling. Knockdown of EGR1 significantly reduced VEEV-induced apoptosis and impacted viral replication. Knockdown of ERK1/2 or PERK significantly reduced EGR1 gene expression, dramatically reduced viral replication, and increased cell survival as well as rescued cells from VEEV-induced apoptosis. These data indicate that EGR1 activation and subsequent cell death are regulated through ERK and PERK pathways in VEEV infected primary astrocytes.


Asunto(s)
Muerte Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/virología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , eIF-2 Quinasa/metabolismo , Apoptosis , Astrocitos/metabolismo , Astrocitos/patología , Astrocitos/virología , Supervivencia Celular , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/patología , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Transducción de Señal , Replicación Viral , eIF-2 Quinasa/genética
3.
Viruses ; 11(11)2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766138

RESUMEN

Traditional pathogenesis studies of alphaviruses involves monitoring survival, viremia, and pathogen dissemination via serial necropsies; however, molecular imaging shifts this paradigm and provides a dynamic assessment of pathogen infection. Positron emission tomography (PET) with PET tracers targeted to study neuroinflammation (N,N-diethyl-2-[4-phenyl]-5,7-dimethylpyrazolo[1,5-a]pyrimidine-3-acetamide, [18F]DPA-714), apoptosis (caspase-3 substrate, [18F]CP-18), hypoxia (fluormisonidazole, [18F]FMISO), blood-brain barrier (BBB) integrity ([18F]albumin), and metabolism (fluorodeoxyglucose, [18F]FDG) was performed on C3H/HeN mice infected intranasally with 7000 plaque-forming units (PFU) of Venezuelan equine encephalitis virus (VEEV) TC-83. The main findings are as follows: (1) whole-brain [18F]DPA-714 and [18F]CP-18 uptake increased three-fold demonstrating, neuroinflammation and apoptosis, respectively; (2) [18F]albumin uptake increased by 25% across the brain demonstrating an altered BBB; (3) [18F]FMISO uptake increased by 50% across the whole brain indicating hypoxic regions; (4) whole-brain [18F]FDG uptake was unaffected; (5) [18F]DPA-714 uptake in (a) cortex, thalamus, striatum, hypothalamus, and hippocampus increased through day seven and decreased by day 10 post exposure, (b) olfactory bulb increased at day three, peaked day seven, and decreased day 10, and (c) brain stem and cerebellum increased through day 10. In conclusion, intranasal exposure of C3H/HeN mice to VEEV TC-83 results in both time-dependent and regional increases in brain inflammation, apoptosis, and hypoxia, as well as modest decreases in BBB integrity; however, it has no effect on brain glucose metabolism.


Asunto(s)
Apoptosis , Barrera Hematoencefálica/metabolismo , Virus de la Encefalitis Equina Venezolana , Encefalomielitis Equina Venezolana/diagnóstico , Encefalomielitis Equina Venezolana/metabolismo , Hipoxia/metabolismo , Tomografía de Emisión de Positrones , Animales , Biomarcadores , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/virología , Caballos , Procesamiento de Imagen Asistido por Computador , Ratones , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones/métodos , Radiofármacos/química , Radiofármacos/metabolismo
4.
Viruses ; 10(11)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30400156

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is an arbovirus that is associated with robust inflammation that contributes to neurodegenerative phenotypes. In addition to triggering central nervous system (CNS) inflammation, VEEV will also induce mitochondrial dysfunction, resulting in increased cellular apoptosis. In this study, we utilize the TC-83 strain of VEEV to determine the role of mitochondrial oxidative stress in mediating inflammation elicited by murine brain microglial cells. Using an in vitro model, we show that murine microglia are susceptible to TC-83 infection, and that these cells undergo mitochondrial stress as the result of infection. We also indicate that bystander microglia contribute more significantly to the overall inflammatory load than directly infected microglia. Use of a mitochondrial targeted antioxidant, mitoquinone mesylate, greatly reduced the pro-inflammatory cytokines released by both direct infected and bystander microglia. Our data suggest that release of interleukin-1ß, a key instigator of neuroinflammation during VEEV infection, may be the direct result of accumulating mitochondrial stress. This data improves our understanding inflammation elicited by murine microglia and will aid in the development of more accurate in vitro and in vivo murine model of VEEV-induced neuroinflammation.


Asunto(s)
Antioxidantes/metabolismo , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/virología , Microglía/metabolismo , Microglía/virología , Mitocondrias/metabolismo , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalomielitis Equina Venezolana/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Mediadores de Inflamación/metabolismo , Ratones , Estrés Oxidativo , Vacunas Virales/inmunología
5.
Antiviral Res ; 151: 8-19, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29337164

RESUMEN

Although the alphavirus Venezuelan equine encephalitis virus (VEEV) has been the cause of multiple outbreaks resulting in extensive human and equine mortality and morbidity, there are currently no anti-VEEV therapeutics available. VEEV pathogenicity is largely dependent on targeting of the viral capsid protein (CP) to the host cell nucleus through the nuclear transporting importin (Imp) α/ß1 heterodimer. Here we perform a high-throughput screen, combined with nested counterscreens to identify small molecules able to inhibit the Impα/ß1:CP interaction for the first time. Several compounds were able to significantly reduce viral replication in infected cells. Compound G281-1564 in particular could inhibit VEEV replication at low µM concentration, while showing minimal toxicity, with steady state and dynamic quantitative microscopic measurements confirming its ability to inhibit CP nuclear import. This study establishes the principle that inhibitors of CP nucleocytoplasmic trafficking can have potent antiviral activity against VEEV, and represents a platform for future development of safe anti-VEEV compounds with high efficacy and specificity.


Asunto(s)
Antivirales/farmacología , Proteínas de la Cápside/metabolismo , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Encefalomielitis Equina Venezolana/virología , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Replicación Viral/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antivirales/química , Supervivencia Celular , Chlorocebus aethiops , Encefalomielitis Equina Venezolana/metabolismo , Ensayos Analíticos de Alto Rendimiento , Interacciones Huésped-Patógeno/efectos de los fármacos , Concentración 50 Inhibidora , Estructura Molecular , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Células Vero
6.
Mol Cell ; 67(2): 228-238.e5, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28625551

RESUMEN

Circular RNAs (circRNAs) are single-stranded RNAs that are joined head to tail with largely unknown functions. Here we show that transfection of purified in vitro generated circRNA into mammalian cells led to potent induction of innate immunity genes and confers protection against viral infection. The nucleic acid sensor RIG-I is necessary to sense foreign circRNA, and RIG-I and foreign circRNA co-aggregate in cytoplasmic foci. CircRNA activation of innate immunity is independent of a 5' triphosphate, double-stranded RNA structure, or the primary sequence of the foreign circRNA. Instead, self-nonself discrimination depends on the intron that programs the circRNA. Use of a human intron to express a foreign circRNA sequence abrogates immune activation, and mature human circRNA is associated with diverse RNA binding proteins reflecting its endogenous splicing and biogenesis. These results reveal innate immune sensing of circRNA and highlight introns-the predominant output of mammalian transcription-as arbiters of self-nonself identity.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/prevención & control , Tolerancia Inmunológica , Inmunidad Innata , Intrones , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/inmunología , ARN/genética , ARN/inmunología , Animales , Secuencia de Bases , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Proteína 58 DEAD Box/metabolismo , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/metabolismo , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Tolerancia Inmunológica/genética , Inmunidad Innata/genética , Ratones , Conformación de Ácido Nucleico , Unión Proteica , Células RAW 264.7 , ARN/biosíntesis , ARN/química , ARN Circular , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Receptores Inmunológicos , Empalmosomas/inmunología , Empalmosomas/metabolismo , Transfección
7.
Virulence ; 8(8): 1849-1866, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-28075229

RESUMEN

Mitochondria are sentinel organelles that are impacted by various forms of cellular stress, including viral infections. While signaling events associated with mitochondria, including those activated by pathogen associated molecular patterns (PAMPs), are widely studied, alterations in mitochondrial distribution and changes in mitochondrial dynamics are also beginning to be associated with cellular insult. Cells of neuronal origin have been demonstrated to display remarkable alterations in several instances, including neurodegenerative disorders. Venezuelan Equine Encephalitis Virus (VEEV) is a New World alphavirus that infects neuronal cells and contributes to an encephalitic phenotype. We demonstrate that upon infection by the vaccine strain of VEEV (TC-83), astrocytoma cells experience a robust drop in mitochondrial activity, which corresponds with an increased accumulation of reactive oxygen species (ROS) in an infection-dependent manner. Infection status also corresponds with a prominent perinuclear accumulation of mitochondria. Cellular enzymatic machinery, including PINK1 and Parkin, appears to be enriched in mitochondrial fractions as compared with uninfected cells, which is indicative of mitochondrial damage. Dynamin related protein 1 (Drp1), a protein that is associated with mitochondrial fission, demonstrated a modest enrichment in mitochondrial fractions of infected cells. Treatment with an inhibitor of mitochondrial fission, Mdivi-1, led to a decrease in caspase cleavage, suggesting that mitochondrial fission was likely to contribute to apoptosis of infected cells. Finally, our data demonstrate that mitophagy ensues in infected cells. In combination, our data suggest that VEEV infection results in significant changes in the mitochondrial landscape that may influence pathological outcomes in the infected cell.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/virología , Dinámicas Mitocondriales , Animales , Apoptosis , Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/fisiopatología , Caballos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
Virology ; 496: 147-165, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27318152

RESUMEN

Most previous studies of interferon-alpha/beta (IFN-α/ß) response antagonism by alphaviruses have focused upon interruption of IFN-α/ß induction and/or receptor signaling cascades. Infection of mice with Venezuelan equine encephalitis alphavirus (VEEV) or Sindbis virus (SINV) induces serum IFN-α/ß, that elicits a systemic antiviral state in uninfected cells successfully controlling SINV but not VEEV replication. Furthermore, VEEV replication is more resistant than that of SINV to a pre-existing antiviral state in vitro. While host macromolecular shutoff is proposed as a major antagonist of IFN-α/ß induction, the underlying mechanisms of alphavirus resistance to a pre-existing antiviral state are not fully defined, nor is the mechanism for the greater resistance of VEEV. Here, we have separated viral transcription and translation shutoff with multiple alphaviruses, identified the viral proteins that induce each activity, and demonstrated that VEEV nonstructural protein 2-induced translation shutoff is likely a critical factor in enhanced antiviral state resistance of this alphavirus.


Asunto(s)
Resistencia a la Enfermedad , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/virología , Interacciones Huésped-Patógeno , Biosíntesis de Proteínas , Proteínas no Estructurales Virales/metabolismo , Animales , Antivirales/metabolismo , Antivirales/farmacología , Línea Celular , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/mortalidad , Caballos , Humanos , Interferones/biosíntesis , Interferones/farmacología , Ratones , Mutación , Fenotipo , ARN Viral , Proteínas no Estructurales Virales/genética
9.
Arch Virol ; 160(10): 2395-405, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26156105

RESUMEN

Venezuelan equine encephalitis (VEE) is a viral disease transmitted by mosquitoes. The inflammation induced by the VEE virus is associated with a high mortality rate in mice. Angiotensin II (Ang II), a pro-inflammatory molecule, is produced in the normal rat brain. There is no information about the role of this molecule in the inflammatory events occurring during VEE and the effect of inflammation on the mortality rate in VEE-virus-infected rats. This study was designed to determine the role of Ang II in VEE and to analyze the effect of inflammation on mortality in infected rats. Two groups of rats were studied: 1) Virus-infected animals and controls (n = 60) were treated with losartan (a blocker of the Ang II-AT1 receptor) or with pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF-κB) or left untreated and analyzed for morbidity and mortality. 2) Animals treated using the same protocol (n = 30) were sacrificed at day 4 postinfection and analyzed by immunohistochemistry and histopathology and for cytokine production. Increased expression of Ang II, ICAM-1, ED-1 and cytokines (IL-1α, MCP-1, IL-6 and IL-10) in infected animals was observed. The main histopathology findings were dilated capillaries and capillaries with endothelial detachment. Losartan and PDTC reduced the expression of IL-1α, MCP-1, and IL-10, and the number of dilated capillaries and capillaries with endothelial detachment. Survival analysis showed that 100% mortality was reached earlier in infected rats treated with losartan (day 14) or PDTC (day 11) than in untreated animals (day 19). These findings suggest that Ang II plays a role in VEE and that brain inflammation is protective against viral infection.


Asunto(s)
Angiotensina II/metabolismo , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/virología , Angiotensina II/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/mortalidad , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
10.
Brain Res ; 1622: 368-76, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26168898

RESUMEN

Venezuelan equine encephalitis (VEE) virus causes an acute central nervous system infection in human and animals. Melatonin (MLT), minocycline (MIN) and ascorbic acid (AA) have been shown to have antiviral activities in experimental infections; however, the mechanisms involved are poorly studied. Therefore, the aim of this study was to determine the effects of those compounds on the viral titers, NO production and lipid peroxidation in the brain of mice and neuroblastoma cultures infected by VEE virus. Infected mouse (10 LD50) were treated with MLT (500 µg/kg bw), MIN (50mg/kg bw) or AA (50mg/kg bw). Infected neuroblastoma cultures (MOI: 1); MLT: 0.5, 1, 5mM, MIN: 0.1, 0.2, 2 µM or AA: 25, 50, 75 µM. Brains were obtained at days 1, 3 and 5. In addition, survival rate of infected treated mice was also analyzed. Viral replication was determined by the plaque formation technique. NO and lipid peroxidation were measured by Griess׳ reaction and thiobarbituric acid assay respectively. Increased viral replication, NO production and lipid peroxidation were observed in both, infected brain and neuroblastoma cell cultures compared with uninfected controls. Those effects were diminished by the studied treatments. In addition, increased survival rate (50%) in treated infected animals compared with untreated infected mice (0%) was found. MLT, MIN and AA have an antiviral effect involving their anti-oxidant properties, and suggesting a potential use of these compounds for human VEE virus infection.


Asunto(s)
Antivirales/farmacología , Ácido Ascórbico/farmacología , Encefalomielitis Equina Venezolana/tratamiento farmacológico , Melatonina/farmacología , Minociclina/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/mortalidad , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Masculino , Ratones , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Estrés Oxidativo/fisiología , Tasa de Supervivencia , Resultado del Tratamiento , Carga Viral
11.
PLoS One ; 10(4): e0124792, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25927990

RESUMEN

Many viruses have been implicated in utilizing or modulating the Ubiquitin Proteasome System (UPS) to enhance viral multiplication and/or to sustain a persistent infection. The mosquito-borne Venezuelan equine encephalitis virus (VEEV) belongs to the Togaviridae family and is an important biodefense pathogen and select agent. There are currently no approved vaccines or therapies for VEEV infections; therefore, it is imperative to identify novel targets for therapeutic development. We hypothesized that a functional UPS is required for efficient VEEV multiplication. We have shown that at non-toxic concentrations Bortezomib, a FDA-approved inhibitor of the proteasome, proved to be a potent inhibitor of VEEV multiplication in the human astrocytoma cell line U87MG. Bortezomib inhibited the virulent Trinidad donkey (TrD) strain and the attenuated TC-83 strain of VEEV. Additional studies with virulent strains of Eastern equine encephalitis virus (EEEV) and Western equine encephalitis virus (WEEV) demonstrated that Bortezomib is a broad spectrum inhibitor of the New World alphaviruses. Time-of-addition assays showed that Bortezomib was an effective inhibitor of viral multiplication even when the drug was introduced many hours post exposure to the virus. Mass spectrometry analyses indicated that the VEEV capsid protein is ubiquitinated in infected cells, which was validated by confocal microscopy and immunoprecipitation assays. Subsequent studies revealed that capsid is ubiquitinated on K48 during early stages of infection which was affected by Bortezomib treatment. This study will aid future investigations in identifying host proteins as potential broad spectrum therapeutic targets for treating alphavirus infections.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Western Blotting , Bortezomib/farmacología , Supervivencia Celular/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Cobayas , Inmunoprecipitación , Hibridación Fluorescente in Situ , Espectrometría de Masas en Tándem
12.
PLoS One ; 9(2): e86745, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586253

RESUMEN

Venezuelan equine encephalitis virus (VEEV) belongs to the genus Alphavirus, family Togaviridae. VEEV infection is characterized by extensive inflammation and studies from other laboratories implicated an involvement of the NF-κB cascade in the in vivo pathology. Initial studies indicated that at early time points of VEEV infection, the NF-κB complex was activated in cells infected with the TC-83 strain of VEEV. One upstream kinase that contributes to the phosphorylation of p65 is the IKKß component of the IKK complex. Our previous studies with Rift valley fever virus, which exhibited early activation of the NF-κB cascade in infected cells, had indicated that the IKKß component underwent macromolecular reorganization to form a novel low molecular weight form unique to infected cells. This prompted us to investigate if the IKK complex undergoes a comparable macromolecular reorganization in VEEV infection. Size-fractionated VEEV infected cell extracts indicated a macromolecular reorganization of IKKß in VEEV infected cells that resulted in formation of lower molecular weight complexes. Well-documented inhibitors of IKKß function, BAY-11-7082, BAY-11-7085 and IKK2 compound IV, were employed to determine whether IKKß function was required for the production of infectious progeny virus. A decrease in infectious viral particles and viral RNA copies was observed with inhibitor treatment in the attenuated and virulent strains of VEEV infection. In order to further validate the requirement of IKKß for VEEV replication, we over-expressed IKKß in cells and observed an increase in viral titers. In contrast, studies carried out using IKKß(-/-) cells demonstrated a decrease in VEEV replication. In vivo studies demonstrated that inhibitor treatment of TC-83 infected mice increased their survival. Finally, proteomics studies have revealed that IKKß may interact with the viral protein nsP3. In conclusion, our studies have revealed that the host IKKß protein may be critically involved in VEEV replication.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/metabolismo , Quinasa I-kappa B/metabolismo , Animales , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Encefalomielitis Equina Venezolana/patología , Cobayas , Interacciones Huésped-Patógeno , Humanos , Ratones , FN-kappa B/metabolismo , Neuronas/virología , Nitrilos/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonas/farmacología , Carga Viral/efectos de los fármacos , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
13.
J Virol ; 87(22): 12003-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24006447

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a pathogenic alphavirus, which circulates in the Central, South, and North Americas, including the United States, and represents a significant public health threat. In recent years, strong progress has been made in understanding the structure of VEEV virions, but the mechanism of their formation has yet to be investigated. In this study, we analyzed the functions of different capsid-specific domains and its amino-terminal subdomains in viral particle formation. Our data demonstrate that VEEV particles can be efficiently formed directly at the plasma membrane without cytoplasmic nucleocapsid preassembly. The entire amino-terminal domain of VEEV capsid protein was found to be dispensable for particle formation. VEEV variants encoding only the capsid's protease domain efficiently produce genome-free VEEV virus-like particles (VLPs), which are very similar in structure to the wild-type virions. The amino-terminal domain of the VEEV capsid protein contains at least four structurally and functionally distinct subdomains, which mediate RNA packaging and the specificity of packaging in particular. The most positively charged subdomain is a negative regulator of the nucleocapsid assembly. The three other subdomains are not required for genome-free VLP formation but are important regulators of RNA packaging. Our data suggest that the positively charged surface of the VEEV capsid-specific protease domain and the very amino-terminal subdomain are also involved in interaction with viral RNA and play important roles in RNA encapsidation. Finally, we show that VEEV variants with mutated capsid acquire compensatory mutations in either capsid or nsP2 genes.


Asunto(s)
Proteínas de la Cápside/metabolismo , Virus de la Encefalitis Equina Venezolana/fisiología , Encefalomielitis Equina Venezolana/metabolismo , Nucleocápside/metabolismo , ARN Viral/metabolismo , Virión/metabolismo , Ensamble de Virus , Secuencia de Aminoácidos , Animales , Western Blotting , Proteínas de la Cápside/genética , Proliferación Celular , Células Cultivadas , Encefalomielitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/virología , Genoma Viral , Riñón/citología , Riñón/metabolismo , Riñón/virología , Datos de Secuencia Molecular , Mutación/genética , Nucleocápside/genética , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Virión/genética , Replicación Viral
14.
J Pineal Res ; 42(2): 107-12, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17286740

RESUMEN

Melatonin, a potent antioxidant, has shown to be beneficial in murine Venezuelan equine encephalomyelitis (VEE) virus infection. In addition, melatonin can induces the production of interleukin-1 beta (IL-1beta), a cytokine capable of inducing increased expression of inducible nitric oxide synthase; the activity of this enzyme is increased in the brain of mice infected with VEE virus. The aim of this study was to determine the effect of VEE virus on the nitric oxide (NO) production, lipid peroxidation and IL-1beta production in the brain and serum of mice infected with VEE virus, and to investigate the modulatory role of melatonin during this viral infection. Mice were infected with 10 LD(50) of VEE virus and treated with melatonin (500 microg/kg of body weight) starting 3 days before and continuing for 5 days after virus inoculation. Mice were sacrificed on days 1, 3 and 5 postinfection and brains and blood samples were obtained. NO and IL-1beta production and lipid peroxidation levels were measured in perfused brain homogenates and serum. Increased production of brain nitrite was found on days 1, 3 and 5 postinfection and lipid peroxidation products were increased at day 5. Levels of serum nitrite were found elevated on days 3 and 5 postinfection; however, lipid peroxidation products remained similar to basal levels. Melatonin treatment decreased nitrite concentration in brain and serum of infected mice as well as the lipid peroxidation products in the brain. IL-1beta was found to be increased in the brain and serum of infected animals, and melatonin treatment induced higher levels of this cytokine (brain: about 4-fold; serum: about 8-fold). These results may be related to the beneficial effect of melatonin in the VEE experimental disease and address the possible therapeutic potential of the indoleamine in human VEE virus infection.


Asunto(s)
Encéfalo/metabolismo , Regulación hacia Abajo/fisiología , Encefalomielitis Equina Venezolana/metabolismo , Interleucina-1beta/metabolismo , Peroxidación de Lípido/fisiología , Melatonina/fisiología , Óxido Nítrico/antagonistas & inhibidores , Regulación hacia Arriba/fisiología , Animales , Encéfalo/virología , Virus de la Encefalitis Equina Venezolana , Interleucina-1beta/biosíntesis , Masculino , Ratones , Óxido Nítrico/biosíntesis
15.
Neurochem Res ; 26(3): 231-4, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11495546

RESUMEN

In mice infected with the Venezuelan equine encephalomyelitis (VEE) virus and exposed to high intensity light (2500 lux) with a 12 h light: 12 h dark photoperiod, a significant increase in the levels of melatonin in the olfactory bulb was observed. The significance of these findings deserves further studies to understand the mechanisms involved in this effect since the olfactory bulbs have been proposed as first portal for VEE virus entry into the CNS. The increase in melatonin content could represent one of the mechanisms of defense against the viral attack.


Asunto(s)
Encefalomielitis Equina Venezolana/metabolismo , Luz , Melatonina/metabolismo , Bulbo Olfatorio/efectos de la radiación , Animales , Ritmo Circadiano , Relación Dosis-Respuesta en la Radiación , Encefalomielitis Equina Venezolana/fisiopatología , Masculino , Ratones , Bulbo Olfatorio/metabolismo , Bulbo Olfatorio/fisiopatología
16.
Infect Immun ; 69(9): 5709-15, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11500447

RESUMEN

A candidate vaccine against botulinum neurotoxin serotype A (BoNT/A) was developed by using a Venezuelan equine encephalitis (VEE) virus replicon vector. This vaccine vector is composed of a self-replicating RNA containing all of the VEE nonstructural genes and cis-acting elements and also a heterologous immunogen gene placed downstream of the subgenomic 26S promoter in place of the viral structural genes. In this study, the nontoxic 50-kDa carboxy-terminal fragment (H(C)) of the BoNT/A heavy chain was cloned into the replicon vector (H(C)-replicon). Cotransfection of BHK cells in vitro with the H(C)-replicon and two helper RNA molecules, the latter encoding all of the VEE structural proteins, resulted in the assembly and release of propagation-deficient, H(C) VEE replicon particles (H(C)-VRP). Cells infected with H(C)-VRP efficiently expressed this protein when analyzed by either immunofluorescence or by Western blot. To evaluate the immunogenicity of H(C)-VRP, mice were vaccinated with various doses of H(C)-VRP at different intervals. Mice inoculated subcutaneously with H(C)-VRP were protected from an intraperitoneal challenge of up to 100,000 50% lethal dose units of BoNT/A. Protection correlated directly with serum enzyme-linked immunosorbent assay titers to BoNT/A. The duration of the immunity achieved was tested at 6 months and at 1 year postvaccination, and mice challenged at these times remained refractory to challenge with BoNT/A.


Asunto(s)
Vacunas Bacterianas/inmunología , Toxinas Botulínicas Tipo A/inmunología , Botulismo/prevención & control , Encefalomielitis Equina Venezolana/genética , Replicón/genética , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Toxinas Botulínicas Tipo A/genética , Línea Celular , Clostridium botulinum/inmunología , Clostridium botulinum/metabolismo , Encefalomielitis Equina Venezolana/metabolismo , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
17.
Vopr Virusol ; 38(1): 21-4, 1993.
Artículo en Ruso | MEDLINE | ID: mdl-8073740

RESUMEN

A relationship between ribamydil concentration and the intensity of accumulation of peroxidation products was found while storing ribamydil-containing liposomes which indicated a significant pro-oxidant activity of ribamydil. Increasing lethality of guinea pigs and white mice infected with Venezuelan equine encephalomyelitis virus was shown while using liposomal ribamydil containing from 4.78 to 6.82 nmol. ml-1 malonic dialdehyde which appeared to be associated with disordered function of the antioxidant system of the experimental animals.


Asunto(s)
Encefalomielitis Equina Venezolana/tratamiento farmacológico , Peroxidación de Lípido/efectos de los fármacos , Ribavirina/farmacocinética , Fiebre del Valle del Rift/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Portadores de Fármacos , Evaluación Preclínica de Medicamentos , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/mortalidad , Radicales Libres , Cobayas , Liposomas , Malondialdehído/metabolismo , Ratones , Ratas , Ribavirina/administración & dosificación , Fiebre del Valle del Rift/metabolismo , Fiebre del Valle del Rift/mortalidad
18.
J Neural Transm Suppl ; 29: 141-51, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2358799

RESUMEN

In order to evaluate central serotonergic function during viral encephalitis biochemical, behavioural and immunohistofluorescence studies were carried out. Mice were inoculated with the moderate virulent strain of venezuelan equine encephalomyelitis virus, Pixuna. Signs of encephalitis were observed in 50-60% of infected animals. Levels of serotonin and 5-hydroxyindolacetic acid, and the ratio of the indolamine and its metabolite in raphe and cortex did not change with respect to sham-inoculated mice. A differential decrease in turnover rate by pharmacological methods, such as pargyline, p-chlorophenylalanine and probenecid administration, was observed in raphe and cortex. The ratio serotonin turnover rate/steady state concentration of serotonin was only decreased in the raphe of sick animals. The response to 5-methoxy-N,N-dimethyltryptamine was greater in infected animals. The duration of immobility in the swim test was shorter in the infected group. A greater number of viral antigen particles was localized in raphe and periraphe areas than in cortex, brain stem or striatum. The results suggest a serotonin presynaptic deficit, a postsynaptic hyperreactivity of serotonin system, and a region-selective distribution of the virus.


Asunto(s)
Encéfalo/metabolismo , Encefalomielitis Equina/metabolismo , Encefalomielitis Equina Venezolana/metabolismo , Serotonina/metabolismo , Animales , Antígenos Virales/metabolismo , Encefalomielitis Equina Venezolana/inmunología , Masculino , Ratones
19.
J Leukoc Biol ; 45(4): 345-52, 1989 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2467959

RESUMEN

Bone-marrow-culture-derived macrophages killed virally infected cells but not uninfected cells. This activity could be enhanced by preexposure of the macrophages to lipopolysaccharide (LPS), and/or purified interferons. The ability to kill virally infected targets was not restricted to a single cell type or virus. Comparing the ability of activators to induce activity against virally infected targets or tumor (P815) targets, it was found that much lower levels of LPS or alpha/beta-interferon were able to induce cytolytic activity for virally infected cells than were needed for tumor targets. Further, while the antitumor activity did not change significantly with an increase in the time of exposure to activating stimuli from 4 to 24 h, the activity against virally infected cells decreased dramatically with the longer exposure to stimuli.


Asunto(s)
Pruebas Inmunológicas de Citotoxicidad , Encefalomielitis Equina/inmunología , Encefalomielitis Equina Venezolana/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Estomatitis/inmunología , Animales , Anticuerpos Antiidiotipos/fisiología , Médula Ósea , Línea Celular , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad/métodos , Encefalomielitis Equina Venezolana/metabolismo , Interferones/biosíntesis , Interferones/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Estomatitis/metabolismo , Factores de Tiempo
20.
Invest Clin ; 30(1): 31-58, 1989.
Artículo en Español | MEDLINE | ID: mdl-2489274

RESUMEN

The Venezuelan equine encephalomyelitis (VEE) is one the most serious viral infections of the nervous system. It has a wide geographic distribution and may give rise to sequela like mental retardation, amnesia, abortion, epilepsy and hidroanencephaly in infected humans and animals. The pathology of this infection is focused mainly in two tissues: lymphohematopoietic and nervous. The VEE virus has a special cytopathic activity on the nervous cells (glia and neurons) while the lesions produced in the myelin are probably a consequence of the immunological response of the host to the infection. The alterations produced by the VEE virus in different neuronal types can originate changes in the brain concentrations of several neurotransmitters and their receptors. Some biochemical modifications that have also been reported could be due to the cytopathic effect of the virus.


Asunto(s)
Encefalomielitis Equina Venezolana , Encefalomielitis Equina Venezolana/complicaciones , Encefalomielitis Equina Venezolana/epidemiología , Encefalomielitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/patología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...