Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Bio Protoc ; 10(12): e3662, 2020 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-33659332

RESUMEN

Structural and biochemical studies of proteins require high amounts of stable, purified proteins. Protein stability often depends on the buffer composition, which includes pH and concentration of salts or other solutes such as glycerol, hence an efficient method for identifying optimal buffer conditions for stability would minimize time and resources used for protein purification and further studies. This protocol describes the use of the Thermofluor assay, in combination with a custom 24-condition screen, to identify buffer conditions that increase protein thermostability, using the conserved herpesviral protein UL37 as an example. Detailed instructions on screen conditions, running the Thermofluor MATLAB script, and analyzing the data are provided. In comparison to circular dichroism (CD), the buffer screen in combination with Thermofluor assay provides a faster and more informative method to analyze protein thermostability.

2.
ACS Infect Dis ; 5(3): 460-472, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30608640

RESUMEN

Vaccines and antivirals to combat dengue, Zika, and other flavivirus pathogens present a major, unmet medical need. Vaccine development has been severely challenged by the antigenic diversity of these viruses and the propensity of non-neutralizing, cross-reactive antibodies to facilitate cellular infection and increase disease severity. As an alternative, direct-acting antivirals targeting the flavivirus envelope protein, E, have the potential to act via an analogous mode of action without the risk of antibody-dependent enhancement of infection and disease. We previously discovered that structurally diverse small molecule inhibitors of the dengue virus E protein exhibit varying levels of antiviral activity against other flaviviruses in cell culture. Here, we demonstrate that the broad-spectrum activity of several cyanohydrazones against dengue, Zika, and Japanese encephalitis viruses is due to specific inhibition of E-mediated membrane fusion during viral entry and provide proof of concept for pharmacological inhibition of E as an antiviral strategy in vivo.


Asunto(s)
Antivirales/administración & dosificación , Infecciones por Flavivirus/tratamiento farmacológico , Flavivirus/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Proteínas del Envoltorio Viral/metabolismo , Animales , Antivirales/química , Femenino , Flavivirus/fisiología , Infecciones por Flavivirus/virología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Bibliotecas de Moléculas Pequeñas/química , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/genética , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
3.
Cell Chem Biol ; 25(8): 1006-1016.e8, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-29937406

RESUMEN

Viral envelope proteins are required for productive viral entry and initiation of infection. Although the humoral immune system provides ample evidence for targeting envelope proteins as an antiviral strategy, there are few pharmacological interventions that have this mode of action. In contrast to classical antiviral targets such as viral proteases and polymerases, viral envelope proteins as a class do not have a well-conserved active site that can be rationally targeted with small molecules. We previously identified compounds that inhibit dengue virus by binding to its envelope protein, E. Here, we show that these small molecules inhibit dengue virus fusion and map the binding site of these compounds to a specific pocket on E. We further demonstrate inhibition of Zika, West Nile, and Japanese encephalitis viruses by these compounds, providing pharmacological evidence for the pocket as a target for developing broad-spectrum antivirals against multiple, mosquito-borne flavivirus pathogens.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Infecciones por Flavivirus/tratamiento farmacológico , Flavivirus/efectos de los fármacos , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Virus del Dengue/química , Virus del Dengue/efectos de los fármacos , Virus del Dengue/fisiología , Descubrimiento de Drogas , Flavivirus/química , Flavivirus/fisiología , Infecciones por Flavivirus/metabolismo , Infecciones por Flavivirus/virología , Humanos , Simulación del Acoplamiento Molecular , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas del Envoltorio Viral/química , Replicación Viral/efectos de los fármacos , Virus Zika/química , Virus Zika/efectos de los fármacos , Virus Zika/fisiología
4.
PLoS Pathog ; 13(12): e1006741, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29216315

RESUMEN

A hallmark property of the neurotropic alpha-herpesvirinae is the dissemination of infection to sensory and autonomic ganglia of the peripheral nervous system following an initial exposure at mucosal surfaces. The peripheral ganglia serve as the latent virus reservoir and the source of recurrent infections such as cold sores (herpes simplex virus type I) and shingles (varicella zoster virus). However, the means by which these viruses routinely invade the nervous system is not fully understood. We report that an internal virion component, the pUL37 tegument protein, has a surface region that is an essential neuroinvasion effector. Mutation of this region rendered herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV) incapable of spreading by retrograde axonal transport to peripheral ganglia both in culture and animals. By monitoring the axonal transport of individual viral particles by time-lapse fluorescence microscopy, the mutant viruses were determined to lack the characteristic sustained intracellular capsid motion along microtubules that normally traffics capsids to the neural soma. Consistent with the axonal transport deficit, the mutant viruses did not reach sites of latency in peripheral ganglia, and were avirulent. Despite this, viral propagation in peripheral tissues and in cultured epithelial cell lines remained robust. Selective elimination of retrograde delivery to the nervous system has long been sought after as a means to develop vaccines against these ubiquitous, and sometimes devastating viruses. In support of this potential, we find that HSV-1 and PRV mutated in the effector region of pUL37 evoked effective vaccination against subsequent nervous system challenges and encephalitic disease. These findings demonstrate that retrograde axonal transport of the herpesviruses occurs by a virus-directed mechanism that operates by coordinating opposing microtubule motors to favor sustained retrograde delivery of the virus to the peripheral ganglia. The ability to selectively eliminate the retrograde axonal transport mechanism from these viruses will be useful in trans-synaptic mapping studies of the mammalian nervous system, and affords a new vaccination paradigm for human and veterinary neurotropic herpesviruses.


Asunto(s)
Transporte Axonal/fisiología , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/patogenicidad , Herpesvirus Suido 1/fisiología , Herpesvirus Suido 1/patogenicidad , Proteínas Estructurales Virales/fisiología , Secuencia de Aminoácidos , Animales , Transporte Axonal/genética , Axones/virología , Ganglios/virología , Genes Virales , Herpesvirus Humano 1/genética , Herpesvirus Suido 1/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos DBA , Modelos Moleculares , Mutación , Neuronas/virología , Ratas , Ratas Long-Evans , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/genética , Vacunas Virales/genética , Virulencia/genética , Virulencia/fisiología , Liberación del Virus/genética , Liberación del Virus/fisiología
5.
Antiviral Res ; 147: 124-130, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29051080

RESUMEN

The rapid spread of Zika virus (ZIKV) in recent years has highlighted the severe diseases associated with ZIKV infection, such as Guillain-Barré syndrome in adults and microcephaly in newborns; yet no vaccines or antivirals currently exist to prevent or treat ZIKV infection. We and others have previously identified N-(4-hydroxyphenyl) retinamide (fenretinide or 4-HPR) as an antiviral compound that inhibits dengue virus 2 (DV2) and other flaviviruses by limiting the steady-state accumulation of viral RNA. Here we show that 4-HPR potently inhibits ZIKV in mammalian cell culture and significantly reduces both serum viremia and brain viral burden in a murine model of ZIKV infection. Consistent with previous observations with dengue virus, this antiviral activity is associated with a significant reduction in the steady-state abundance of viral genomic RNA. We show this reduction is due to a major decrease in the rate of viral RNA synthesis, though not via direct inhibition of the activity of the viral replicase. These results establish 4-HPR's mode of action against DV and ZIKV and, taken with previous clinical trials that established 4-HPR's safety and tolerability, illustrate the potential utility of 4-HPR as an agent for treatment of ZIKV infection.


Asunto(s)
Antivirales/farmacología , Fenretinida/farmacología , Replicación Viral/efectos de los fármacos , Infección por el Virus Zika/virología , Virus Zika/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Línea Celular , Virus del Dengue/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Fenretinida/uso terapéutico , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , ARN Viral/metabolismo , Carga Viral/efectos de los fármacos , Ensayo de Placa Viral , Virus Zika/crecimiento & desarrollo , Infección por el Virus Zika/tratamiento farmacológico
6.
Cell Chem Biol ; 23(4): 443-52, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27105280

RESUMEN

Dengue virus infects more than 300 million people annually, yet there is no widely protective vaccine or drugs against the virus. Efforts to develop antivirals against classical targets such as the viral protease and polymerase have not yielded drugs that have advanced to the clinic. Here, we show that the allosteric Abl kinase inhibitor GNF-2 interferes with dengue virus replication via activity mediated by cellular Abl kinases but additionally blocks viral entry via an Abl-independent mechanism. To characterize this newly discovered antiviral activity, we developed disubstituted pyrimidines that block dengue virus entry with structure-activity relationships distinct from those driving kinase inhibition. We demonstrate that biotin- and fluorophore-conjugated derivatives of GNF-2 interact with the dengue glycoprotein, E, in the pre-fusion conformation that exists on the virion surface, and that this interaction inhibits viral entry. This study establishes GNF-2 as an antiviral compound with polypharmacological activity and provides "lead" compounds for further optimization efforts.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Antivirales/química , Virus del Dengue/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Células 3T3 NIH , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-abl/deficiencia , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/química , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/metabolismo
7.
J Virol ; 88(10): 5462-73, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24599989

RESUMEN

UNLABELLED: In cells infected with herpesviruses, two capsid-associated, or inner tegument, proteins, UL37 and UL36, control cytosolic trafficking of capsids by as yet poorly understood mechanisms. Here, we report the crystal structure of the N-terminal half of UL37 from pseudorabies virus, an alphaherpesvirus closely related to herpes simplex viruses and varicella-zoster virus. The structure--the first for any alphaherpesvirus inner tegument protein--reveals an elongated molecule of a complex architecture rich in helical bundles. To explore the function of the UL37 N terminus, we used the three-dimensional framework provided by the structure in combination with evolutionary trace analysis to pinpoint several surface-exposed regions of potential functional importance and test their importance using mutagenesis. This approach identified a novel functional region important for cell-cell spread. These results suggest a novel role for UL37 in intracellular virus trafficking that promotes spread of viral infection, a finding that expands the repertoire of UL37 functions. Supporting this, the N terminus of UL37 shares structural similarity with cellular multisubunit tethering complexes (MTCs), which control vesicular trafficking in eukaryotic cells by tethering transport vesicles to their destination membranes. Our results suggest that UL37 could be the first viral MTC mimic and provide a structural rationale for the importance of UL37 for viral trafficking. We propose that herpesviruses may have co-opted the MTC functionality of UL37 to bring capsids to cytoplasmic budding destinations and further on to cell junctions for spread to nearby cells. IMPORTANCE: To move within an infected cell, viruses encode genes for proteins that interact with host trafficking machinery. In cells infected with herpesviruses, two capsid-associated proteins control the cytosolic movement of capsids by as yet poorly understood mechanisms. Here, we report the crystal structure for the N-terminal half of one of these proteins, UL37. Structure-based mutagenesis revealed a novel function for UL37 in virus trafficking to cell junctions for cell-cell spread. The unexpected structural similarity to components of cellular multisubunit tethering complexes, which control vesicular traffic, suggests that UL37 could be the first viral MTC mimic and provides a structural basis for the importance of UL37 for virus trafficking.


Asunto(s)
Herpesvirus Suido 1/química , Herpesvirus Suido 1/fisiología , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/metabolismo , Liberación del Virus , Secuencia de Aminoácidos , Animales , Línea Celular , Cristalografía por Rayos X , Análisis Mutacional de ADN , Herpesvirus Suido 1/genética , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformación Proteica , Proteínas Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...