Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Phys Chem B ; 127(14): 3175-3186, 2023 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-37001021

RESUMEN

Although Venezuelan equine encephalitis virus (VEEV) is a life-threatening pathogen with a capacity for epidemic outbreaks, there are no FDA-approved VEEV antivirals for humans. VEEV cytotoxicity is partially attributed to the formation of a tetrameric complex between the VEEV capsid protein, the nuclear import proteins importin-α and importin-ß, and the nuclear export protein CRM1, which together block trafficking through the nuclear pore complex. Experimental studies have identified small molecules from the CL6662 scaffold as potential inhibitors of the viral nuclear localization signal (NLS) sequence binding to importin-α. However, little is known about the molecular mechanism of CL6662 inhibition. To address this issue, we employed all-atom replica exchange molecular dynamics simulations to probe, in atomistic detail, the binding mechanism of CL6662 ligands to importin-α. Three ligands, including G281-1485 and two congeners with varying hydrophobicities, were considered. We investigated the distribution of ligand binding poses, their locations, and ligand specificities measured by the strength of binding interactions. We found that G281-1485 binds nonspecifically without forming well-defined binding poses throughout the NLS binding site. Binding of the less hydrophobic congener becomes strongly on-target with respect to the NLS binding site but remains nonspecific. However, a more hydrophobic congener is a strongly specific binder and the only ligand out of three to form a well-defined binding pose, while partially overlapping with the NLS binding site. On the basis of free energy estimates, we argue that all three ligands weakly compete with the viral NLS sequence for binding to importin-α in an apparent compromise to preserve host NLS binding. We further show that all-atom replica exchange binding simulations are a viable tool for studying ligands binding nonspecifically without forming well-defined binding poses.


Asunto(s)
Virus de la Encefalitis Equina Venezolana , alfa Carioferinas , Animales , Caballos , Humanos , alfa Carioferinas/química , alfa Carioferinas/metabolismo , Virus de la Encefalitis Equina Venezolana/metabolismo , Simulación de Dinámica Molecular , Ligandos , Señales de Localización Nuclear/química , Señales de Localización Nuclear/metabolismo , Núcleo Celular/metabolismo , Sitios de Unión , Unión Proteica
2.
Virology ; 565: 13-21, 2022 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-34626907

RESUMEN

Eastern equine encephalitis virus (EEEV), western equine encephalitis virus (WEEV) and Venezuelan equine encephalitis virus (VEEV) can cause fatal encephalitis in humans and equids. Some MAbs to the E1 glycoprotein are known to be cross-reactive, weakly neutralizing in vitro but can protect from disease in animal models. We investigated the mechanism of neutralization of VEEV infection by the broadly cross-reactive E1-specific MAb 1A4B-6. 1A4B-6 protected 3-week-old Swiss Webster mice prophylactically from lethal VEEV challenge. Likewise, 1A4B-6 inhibited virus growth in vitro at a pre-attachment step after virions were incubated at 37 °C and inhibited virus-mediated cell fusion. Amino acid residue N100 in the fusion loop of E1 protein was identified as critical for binding. The potential to elicit broadly cross-reactive MAbs with limited virus neutralizing activity in vitro but that can inhibit virus entry and protect animals from infection merits further exploration for vaccine and therapeutic developmental research.


Asunto(s)
Anticuerpos Antivirales/inmunología , Virus de la Encefalitis Equina Venezolana/inmunología , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/virología , Proteínas del Envoltorio Viral/inmunología , Replicación Viral/efectos de los fármacos , Alphavirus/inmunología , Infecciones por Alphavirus/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Línea Celular , Chlorocebus aethiops , Reacciones Cruzadas , Encefalomielitis Equina Venezolana/terapia , Glicoproteínas/inmunología , Inmunoterapia , Ratones , Unión Proteica , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Virión/inmunología , Virión/metabolismo
3.
Sci Rep ; 10(1): 21491, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293592

RESUMEN

Venezuelan equine encephalitis virus (VEEV), a New World alphavirus of the Togaviridae family of viruses causes periodic outbreaks of disease in humans and equines. Disease following VEEV infection manifests as a febrile illness with flu-like symptoms, which can progress to encephalitis and cause permanent neurological sequelae in a small number of cases. VEEV is classified as a category B select agent due to ease of aerosolization and high retention of infectivity in the aerosol form. Currently, there are no FDA-approved vaccines or therapeutics available to combat VEEV infection. VEEV infection in vivo is characterized by extensive systemic inflammation that can exacerbate infection by potentially increasing the susceptibility of off-site cells to infection and dissemination of the virus. Hence, a therapeutic targeting both the infection and associated inflammation represents an unmet need. We have previously demonstrated that host defense peptides (HDPs), short peptides that are key components of the innate immune response, exhibit antiviral activity against a multitude of viruses including VEEV. In this study, we designed synthetic peptides derived from indolicidin, a naturally occurring HDP, and tested their efficacy against VEEV. Two candidate synthetic peptides inhibited VEEV replication by approximately 1000-fold and decreased the expression of inflammatory mediators such as IL1α, IL1ß, IFNγ, and TNFα at both the gene and protein expression levels. Furthermore, an increase in expression levels of genes involved in chemotaxis of leukocytes and anti-inflammatory genes such as IL1RN was also observed. Overall, we conclude that our synthetic peptides inhibit VEEV replication and the inflammatory burden associated with VEEV infection.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Virus de la Encefalitis Equina Venezolana/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Péptidos Catiónicos Antimicrobianos/síntesis química , Péptidos Catiónicos Antimicrobianos/química , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/genética , Caballos , Humanos , Inflamación , Ratones , Células Vero
4.
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
5.
Viruses ; 12(9)2020 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-32933112

RESUMEN

Venezuelan equine encephalitis virus (VEEV), a mosquito transmitted alphavirus of the Togaviridae family, can cause a highly inflammatory and encephalitic disease upon infection. Although a category B select agent, no FDA-approved vaccines or therapeutics against VEEV currently exist. We previously demonstrated NF-κB activation and macromolecular reorganization of the IKK complex upon VEEV infection in vitro, with IKKß inhibition reducing viral replication. Mass spectrometry and confocal microscopy revealed an interaction between IKKß and VEEV non-structural protein 3 (nsP3). Here, using western blotting, a cell-free kinase activity assay, and mass spectrometry, we demonstrate that IKKß kinase activity can directly phosphorylate VEEV nsP3 at sites 204/5, 142, and 134/5. Alanine substitution mutations at sites 204/5, 142, or 134/5 reduced VEEV replication by >30-100,000-fold corresponding to a severe decrease in negative-strand synthesis. Serial passaging rescued viral replication and negative-strand synthesis, and sequencing of revertant viruses revealed reversion to the wild-type TC-83 phosphorylation capable amino acid sequences at nsP3 sites 204/5, 142, and 135. Generation of phosphomimetic mutants using aspartic acid substitutions at site 204/5 resulted in rescue of both viral replication and negative-strand RNA production, whereas phosphomimetic mutant 134/5 rescued viral replication but failed to restore negative-strand RNA levels, and phosphomimetic mutant 142 did not rescue VEEV replication. Together, these data demonstrate that IKKß can phosphorylate VEEV nsP3 at sites 204/5, 142, and 134/5, and suggest that phosphorylation is essential for negative-strand RNA synthesis at site 204/5, but may be important for infectious particle production at site 134/5.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Equina Venezolana/efectos de los fármacos , Virus de la Encefalitis Equina Venezolana/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas no Estructurales Virales/metabolismo , Aedes , Animales , Línea Celular , Chlorocebus aethiops , Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana , Humanos , Mutación , FN-kappa B/metabolismo , Fosforilación , Células Vero , Proteínas no Estructurales Virales/genética , Replicación Viral/efectos de los fármacos
6.
PLoS Pathog ; 16(3): e1008282, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32150585

RESUMEN

Protein phosphorylation plays an important role during the life cycle of many viruses. Venezuelan equine encephalitis virus (VEEV) capsid protein has recently been shown to be phosphorylated at four residues. Here those studies are extended to determine the kinase responsible for phosphorylation and the importance of capsid phosphorylation during the viral life cycle. Phosphorylation site prediction software suggests that Protein Kinase C (PKC) is responsible for phosphorylation of VEEV capsid. VEEV capsid co-immunoprecipitated with PKCδ, but not other PKC isoforms and siRNA knockdown of PKCδ caused a decrease in viral replication. Furthermore, knockdown of PKCδ by siRNA decreased capsid phosphorylation. A virus with capsid phosphorylation sites mutated to alanine (VEEV CPD) displayed a lower genomic copy to pfu ratio than the parental virus; suggesting more efficient viral assembly and more infectious particles being released. RNA:capsid binding was significantly increased in the mutant virus, confirming these results. Finally, VEEV CPD is attenuated in a mouse model of infection, with mice showing increased survival and decreased clinical signs as compared to mice infected with the parental virus. Collectively our data support a model in which PKCδ mediated capsid phosphorylation regulates viral RNA binding and assembly, significantly impacting viral pathogenesis.


Asunto(s)
Proteínas de la Cápside/metabolismo , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/enzimología , Proteína Quinasa C-delta/metabolismo , ARN Viral/metabolismo , Animales , Cápside/metabolismo , Proteínas de la Cápside/genética , Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/virología , Femenino , Caballos , Interacciones Huésped-Patógeno , Ratones , Ratones Endogámicos C3H , Fosforilación , Unión Proteica , Proteína Quinasa C-delta/genética , ARN Viral/genética
7.
J Struct Biol ; 206(1): 119-127, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30825649

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a new world alphavirus which can be involved in several central nervous system disorders such as encephalitis and meningitis. The VEEV genome codes for 4 non-structural proteins (nsP), of which nsP3 contains a Macro domain. Macro domains (MD) can be found as stand-alone proteins or embedded within larger proteins in viruses, bacteria and eukaryotes. Their most common feature is the binding of ADP-ribose (ADPr), while several macro domains act as ribosylation writers, erasers or readers. Alphavirus MD erase ribosylation but their precise contribution in viral replication is still under investigation. NMR-driven titration experiments of ADPr in solution with the VEEV macro domain (in apo- and complex state) show that it adopts a suitable conformation for ADPr binding. Specific experiments indicate that the flexibility of the loops ß5-α3 and α3-ß6 is critical for formation of the complex and assists a wrapping mechanism for ADPr binding. Furthermore, along with this sequence of events, the VEEV MD undergoes a conformational exchange process between the apo state and a low-populated "dark" conformational state.


Asunto(s)
Adenosina Difosfato Ribosa/química , Virus de la Encefalitis Equina Venezolana/metabolismo , Simulación de Dinámica Molecular , Dominios Proteicos , Proteínas no Estructurales Virales/química , Adenosina Difosfato Ribosa/metabolismo , Animales , Virus de la Encefalitis Equina Venezolana/genética , Caballos , Humanos , Espectroscopía de Resonancia Magnética , Conformación Molecular , Unión Proteica , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
8.
Stem Cell Res Ther ; 8(1): 217, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28969679

RESUMEN

BACKGROUND: Dysfunction of the retinal pigment epithelium (RPE) is implicated in numerous forms of retinal degeneration. The readily accessible environment of the eye makes it particularly suitable for the transplantation of RPE cells, which can now be derived from autologous induced pluripotent stem cells (iPSCs), to treat retinal degeneration. For RPE transplantation to become feasible in the clinic, patient-specific somatic cells should be reprogrammed to iPSCs without the introduction of reprogramming genes into the genome of the host cell, and then subsequently differentiated into RPE cells that are well characterized for safety and functionality prior to transplantation. METHODS: We have reprogrammed human dermal fibroblasts to iPSCs using nonintegrating RNA, and differentiated the iPSCs toward an RPE fate (iPSC-RPE), under Good Manufacturing Practice (GMP)-compatible conditions. RESULTS: Using highly sensitive assays for cell polarity, structure, organelle trafficking, and function, we found that iPSC-RPE cells in culture exhibited key characteristics of native RPE. Importantly, we demonstrate for the first time with any stem cell-derived RPE cell that live cells are able to support dynamic organelle transport. This highly sensitive test is critical for RPE cells intended for transplantation, since defects in intracellular motility have been shown to promote RPE pathogenesis akin to that found in macular degeneration. To test their capabilities for in-vivo transplantation, we injected the iPSC-RPE cells into the subretinal space of a mouse model of retinal degeneration, and demonstrated that the transplanted cells are capable of rescuing lost RPE function. CONCLUSIONS: This report documents the successful generation, under GMP-compatible conditions, of human iPSC-RPE cells that possess specific characteristics of healthy RPE. The report adds to a growing literature on the utility of human iPSC-RPE cells for cell culture investigations on pathogenicity and for therapeutic transplantation, by corroborating findings of others, and providing important new information on essential RPE cell biological properties.


Asunto(s)
Reprogramación Celular/genética , Virus de la Encefalitis Equina Venezolana/genética , Células Epiteliales/efectos de los fármacos , Fibroblastos/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Degeneración Retiniana/terapia , Animales , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/metabolismo , Células Epiteliales/citología , Células Epiteliales/fisiología , Células Epiteliales/trasplante , Fibroblastos/citología , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Inyecciones Intraoculares , Péptidos y Proteínas de Señalización Intercelular/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Cultivo Primario de Células , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/fisiología , Piel/citología
9.
Viruses ; 9(10)2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28961161

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a New World alphavirus that is vectored by mosquitos and cycled in rodents. It can cause disease in equines and humans characterized by a febrile illness that may progress into encephalitis. Like the capsid protein of other viruses, VEEV capsid is an abundant structural protein that binds to the viral RNA and interacts with the membrane-bound glycoproteins. It also has protease activity, allowing cleavage of itself from the growing structural polypeptide during translation. However, VEEV capsid protein has additional nonstructural roles within the host cell functioning as the primary virulence factor for VEEV. VEEV capsid inhibits host transcription and blocks nuclear import in mammalian cells, at least partially due to its complexing with the host CRM1 and importin α/ß1 nuclear transport proteins. VEEV capsid also shuttles between the nucleus and cytoplasm and is susceptible to inhibitors of nuclear trafficking, making it a promising antiviral target. Herein, the role of VEEV capsid in viral replication and pathogenesis will be discussed including a comparison to proteins of other alphaviruses.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Virus de la Encefalitis Equina Venezolana/patogenicidad , ARN Viral/metabolismo , Replicación Viral , Transporte Activo de Núcleo Celular , Animales , Cápside/química , Proteínas de la Cápside/genética , Línea Celular , Virus de la Encefalitis Equina del Este , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/metabolismo , Virus de la Encefalitis Equina del Oeste , Caballos , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Virulencia , Replicación Viral/genética , Proteína Exportina 1
10.
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
11.
Antiviral Res ; 131: 49-60, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27105836

RESUMEN

The mosquito-borne New World alphavirus, Venezuelan equine encephalitis virus (VEEV) is a Category B select agent with no approved vaccines or therapies to treat infected humans. Therefore it is imperative to identify novel targets that can be targeted for effective therapeutic intervention. We aimed to identify and validate interactions of VEEV nonstructural protein 3 (nsP3) with host proteins and determine the consequences of these interactions to viral multiplication. We used a HA tagged nsP3 infectious clone (rTC-83-nsP3-HA) to identify and validate two RNA helicases: DDX1 and DDX3 that interacted with VEEV-nsP3. In addition, DDX1 and DDX3 knockdown resulted in a decrease in infectious viral titers. Furthermore, we propose a functional model where the nsP3:DDX3 complex interacts with the host translational machinery and is essential in the viral life cycle. This study will lead to future investigations in understanding the importance of VEEV-nsP3 to viral multiplication and apply the information for the discovery of novel host targets as therapeutic options.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Virus de la Encefalitis Equina Venezolana/metabolismo , Interacciones Huésped-Patógeno , Proteínas no Estructurales Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Virus de la Encefalitis Equina Venezolana/química , Técnicas de Silenciamiento del Gen , Humanos , ARN Helicasas/metabolismo , Células Vero , Carga Viral , Replicación Viral
12.
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
13.
Biomol NMR Assign ; 9(2): 247-51, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25291978

RESUMEN

Macro domains consist of 130-190 amino acid residues and appear to be highly conserved in all kingdoms of life. Intense research on this field has shown that macro domains bind ADP-ribose and other similar molecules, but their exact function still remains intangible. Macro domains are highly conserved in the Alphavirus genus and the Venezuelan equine encephalitis virus (VEEV) is a member of this genus that causes fatal encephalitis to equines and humans. In this study we report the high yield recombinant expression and preliminary solution NMR study of the macro domain of VEEV. An almost complete sequence-specific assignment of its (1)H, (15)N and (13)C resonances was obtained and its secondary structure predicted by TALOS+. The protein shows a unique mixed α/ß-fold.


Asunto(s)
Espectroscopía de Resonancia Magnética con Carbono-13 , Virus de la Encefalitis Equina Venezolana/metabolismo , Resonancia Magnética Nuclear Biomolecular , Espectroscopía de Protones por Resonancia Magnética , Proteínas no Estructurales Virales/química , Secuencia de Aminoácidos , Datos de Secuencia Molecular , Isótopos de Nitrógeno , Estructura Terciaria de Proteína , Alineación de Secuencia
14.
J Mol Graph Model ; 29(3): 347-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21036084

RESUMEN

The outbreaks of chikungunya (CHIKV) and venezuelan equine encephalitis (VEEV) viral infections in humans have emerged or re-emerged in various countries of "Africa and southeast Asia", and "central and south America", respectively. At present, no drug or vaccine is available for the treatment and therapy of both viral infections, but the non-structural protein, nsP3, is a potential target for the design of potent inhibitors that fit at the adenosine-binding site of its macro domain. Here, so as to understand the fundamental basis of the particular interactions between the ADP-ribose bound to the nsP3 amino acid residues at the binding site, molecular dynamics simulations were applied. The results show that these two nsP3 domains share a similar binding pattern for accommodating the ADP-ribose. The ADP-ribose phosphate unit showed the highest degree of stabilization through hydrogen bond interactions with the nsP3 V33 residue and the consequent amino acid residues 110-114. The adenine base of ADP-ribose was specifically recognized by the conserved nsP3 residue D10. Additionally, the ribose and the diphosphate units were found to play more important roles in the CHIKV nsP3-ADP-ribose complex, while the ter-ribose was more important in the VEEV complex. The slightly higher binding affinity of ADP-ribose toward the nsP3 macro domain of VEEV, as predicted by the simulation results, is in good agreement with previous experimental data. These simulation results provide useful information to further assist in drug design and development for these two important viruses.


Asunto(s)
Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Virus Chikungunya/química , Virus de la Encefalitis Equina Venezolana/química , Virus de la Encefalitis Equina Venezolana/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Animales , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Estructura Molecular , Termodinámica , Proteínas no Estructurales Virales/genética
15.
Biochemistry (Mosc) ; 74(12): 1328-36, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19961413

RESUMEN

Polyclonal and monoclonal antibodies (MABs) to human laminin-binding protein (LBP) can efficiently block the penetration of some alpha- and flaviviruses into the cell. A panel of 13 types of MABs to human recombinant LBP was used for more detailed study of the mechanism of this process. Competitive analysis has shown that MABs to LBP can be divided into six different competition groups. MABs 4F6 and 8E4 classified under competition groups 3 and 4 can inhibit the replication of Venezuelan equine encephalitis virus (VEEV), which is indicative of their interaction with the receptor domain of LBP providing for binding with virions. According to enzyme immunoassay and immunoblotting data, polyclonal anti-idiotypic antibodies to MABs 4F6 and 8E4 modeling paratopes of the LBP receptor domain can specifically interact with VEEV E2 protein and tick-borne encephalitis virus (TBEV) E protein. Mapping of binding sites of MABs 4F6 and 8E4 with LBP by constructing short deletion fragments of the human LBP molecule has shown that MAB 8E4 interacts with the fragment of amino acid residues 187-210, and MAB 4F6 interacts with the fragment of residues 263-278 of LBP protein, which is represented by two TEDWS peptides separated by four amino acid residues. This suggested that the LBP receptor domain interacting with VEEV E2 and TBEV E viral proteins is located at the C-terminal fragment of the LBP molecule. A model of the spatial structure of the LBP receptor domain distally limited by four linear loops (two of which are represented by experimentally mapped regions of amino acid residues 187-210 and 263-278) as well as the central beta-folded region turning into the alpha-helical site including residues 200-216 of the LBP molecule and providing for the interaction with the laminin-1 molecule has been proposed.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/metabolismo , Virus de la Encefalitis Transmitidos por Garrapatas/metabolismo , Receptores de Laminina/metabolismo , Receptores Virales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Anticuerpos Antiidiotipos/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Sitios de Unión , Chlorocebus aethiops , Cristalografía por Rayos X , Humanos , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Receptores de Laminina/química , Receptores de Laminina/genética , Receptores Virales/química , Receptores Virales/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos
16.
Virology ; 387(1): 211-21, 2009 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-19278709

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is one of the most pathogenic members of the Alphavirus genus in the Togaviridae family. Viruses in the VEEV serocomplex continuously circulate in the Central and South America. The only currently available attenuated strain VEEV TC-83 is being used only for vaccination of at-risk laboratory workers and military personnel. Its attenuated phenotype was shown to rely only on two point mutations, one of which, G3A, was found in the 5' untranslated region (5'UTR) of the viral genome. Our data demonstrate that the G3A mutation strongly affects the secondary structure of VEEV 5'UTR, but has only a minor effect on translation. The indicated mutation increases replication of the viral genome, downregulates transcription of the subgenomic RNA, and, thus, affects the ratio of genomic and subgenomic RNA synthesis. These findings and the previously reported G3A-induced, higher sensitivity of VEEV TC-83 to IFN-alpha/beta suggest a plausible explanation for its attenuated phenotype.


Asunto(s)
Regiones no Traducidas 5'/genética , Virus de la Encefalitis Equina Venezolana/genética , Genoma Viral/genética , Biosíntesis de Proteínas/genética , ARN Viral/biosíntesis , Proteínas Virales/metabolismo , Animales , Línea Celular , Cricetinae , Virus de la Encefalitis Equina Venezolana/metabolismo , Virus de la Encefalitis Equina Venezolana/patogenicidad , Regulación Viral de la Expresión Génica , Ratones , Células 3T3 NIH , ARN Viral/genética , Replicación Viral
17.
J Virol ; 83(9): 4275-86, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19225006

RESUMEN

Venezuelan equine encephalitis virus (VEE) replicon particles (VRP) were used to model the initial phase of VEE-induced encephalitis in the mouse brain. VRP can target and infect cells as VEE, but VRP do not propagate beyond the first infected cell due to the absence of the structural genes. Direct intracranial inoculation of VRP into mice induced acute encephalitis with signs similar to the neuronal phase of wild-type VEE infection and other models of virus-induced encephalitis. Using the previously established VRP-mRNP tagging system, a new method to distinguish the host responses in infected cells from those in uninfected bystander cell populations, we detected a robust and rapid innate immune response in the central nervous system (CNS) by infected neurons and uninfected bystander cells. Moreover, this innate immune response in the CNS compromised blood-brain barrier integrity, created an inflammatory response, and directed an adaptive immune response characterized by proliferation and activation of microglia cells and infiltration of inflammatory monocytes, in addition to CD4(+) and CD8(+) T lymphocytes. Taken together, these data suggest that a naïve CNS has an intrinsic potential to induce an innate immune response that could be crucial to the outcome of the infection by determining the composition and dynamics of the adaptive immune response. Furthermore, these results establish a model for neurotropic virus infection to identify host and viral factors that contribute to invasion of the brain, the mechanism(s) whereby the adaptive immune response can clear the infection, and the role of the host innate response in these processes.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/crecimiento & desarrollo , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/virología , Virión/crecimiento & desarrollo , Animales , Proliferación Celular , Citocinas/metabolismo , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/patología , Femenino , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Microglía/metabolismo , Microglía/patología , ARN Viral/genética , Virión/genética
18.
PLoS One ; 3(7): e2709, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18628938

RESUMEN

BACKGROUND: The Venezuelan equine encephalitis (VEE) virus replicon system was used to produce virus-like replicon particles (VRP) packaged with a number of different VEE-derived glycoprotein (GP) coats. The GP coat is believed to be responsible for the cellular tropism noted for VRP and it is possible that different VEE GP coats may have different affinities for cells. We examined VRP packaged in four different VEE GP coats for their ability to infect cells in vitro and to induce both humoral and cellular immune responses in vivo. METHODOLOGY/PRINCIPAL FINDINGS: The VRP preparations were characterized to determine both infectious units (IU) and genome equivalents (GE) prior to in vivo analysis. VRP packaged with different VEE GP coats demonstrated widely varying GE/IU ratios based on Vero cell infectivity. BALB/c mice were immunized with the different VRP based on equal GE titers and the humoral and cellular responses to the expressed HIV gag gene measured. The magnitude of the immune responses measured in mice revealed small but significant differences between different GP coats when immunization was based on GE titers. CONCLUSIONS/SIGNIFICANCE: We suggest that care should be taken when alternative coat proteins are used to package vector-based systems as the titers determined by cell culture infection may not represent accurate particle numbers and in turn may not accurately represent actual in vivo dose.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/metabolismo , Replicón , Animales , Encefalomielitis Equina Venezolana/virología , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos , Genoma , Glicoproteínas/química , Sistema Inmunológico , Ratones , Ratones Endogámicos BALB C , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Virol ; 81(24): 13552-65, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17913819

RESUMEN

The encephalitogenic New World alphaviruses, including Venezuelan (VEEV), eastern (EEEV), and western equine encephalitis viruses, constitute a continuing public health threat in the United States. They circulate in Central, South, and North America and have the ability to cause fatal disease in humans and in horses and other domestic animals. We recently demonstrated that these viruses have developed the ability to interfere with cellular transcription and use it as a means of downregulating a cellular antiviral response. The results of the present study suggest that the N-terminal, approximately 35-amino-acid-long peptide of VEEV and EEEV capsid proteins plays the most critical role in the downregulation of cellular transcription and development of a cytopathic effect. The identified VEEV-specific peptide C(VEE)33-68 includes two domains with distinct functions: the alpha-helix domain, helix I, which is critically involved in supporting the balance between the presence of the protein in the cytoplasm and nucleus, and the downstream peptide, which might contain a functional nuclear localization signal(s). The integrity of both domains not only determines the intracellular distribution of the VEEV capsid but is also essential for direct capsid protein functioning in the inhibition of transcription. Our results suggest that the VEEV capsid protein interacts with the nuclear pore complex, and this interaction correlates with the protein's ability to cause transcriptional shutoff and, ultimately, cell death. The replacement of the N-terminal fragment of the VEEV capsid by its Sindbis virus-specific counterpart in the VEEV TC-83 genome does not affect virus replication in vitro but reduces cytopathogenicity and results in attenuation in vivo. These findings can be used in designing a new generation of live, attenuated, recombinant vaccines against the New World alphaviruses.


Asunto(s)
Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/farmacología , Virus de la Encefalitis Equina Venezolana/patogenicidad , Proteínas/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Proteínas de la Cápside/genética , Supervivencia Celular , Cricetinae , Efecto Citopatogénico Viral , Virus de la Encefalitis Equina Venezolana/genética , Virus de la Encefalitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/mortalidad , Encefalomielitis Equina Venezolana/patología , Encefalomielitis Equina Venezolana/virología , Femenino , Inmunización , Ratones , Mutación , Proteínas/genética
20.
J Virol ; 81(19): 10268-79, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17652399

RESUMEN

Many RNA viruses, which replicate predominantly in the cytoplasm, have nuclear components that contribute to their life cycle or pathogenesis. We investigated the intracellular localization of the multifunctional nonstructural protein 2 (nsP2) in mammalian cells infected with Venezuelan equine encephalitis virus (VEE), an important, naturally emerging zoonotic alphavirus. VEE nsP2 localizes to both the cytoplasm and the nucleus of mammalian cells in the context of infection and also when expressed alone. Through the analysis of a series of enhanced green fluorescent protein fusions, a segment of nsP2 that completely localizes to the nucleus of mammalian cells was identified. Within this region, mutation of the putative nuclear localization signal (NLS) PGKMV diminished, but did not obliterate, the ability of the protein to localize to the nucleus, suggesting that this sequence contributes to the nuclear localization of VEE nsP2. Furthermore, VEE nsP2 specifically interacted with the nuclear import protein karyopherin-alpha1 but not with karyopherin-alpha2, -3, or -4, suggesting that karyopherin-alpha1 transports nsP2 to the nucleus during infection. Additionally, a novel nuclear export signal (NES) was identified, which included residues L526 and L528 of VEE nsP2. Leptomycin B treatment resulted in nuclear accumulation of nsP2, demonstrating that nuclear export of nsP2 is mediated via the CRM1 nuclear export pathway. Disruption of either the NLS or the NES in nsP2 compromised essential viral functions. Taken together, these results establish the bidirectional transport of nsP2 across the nuclear membrane, suggesting that a critical function of nsP2 during infection involves its shuttling between the cytoplasm and the nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Virus de la Encefalitis Equina Venezolana/metabolismo , Señales de Localización Nuclear/genética , Proteínas no Estructurales Virales/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Núcleo Celular/química , Citoplasma/química , Citoplasma/metabolismo , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Señales de Localización Nuclear/metabolismo , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas no Estructurales Virales/análisis , Proteínas no Estructurales Virales/genética , alfa Carioferinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...