Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Virol ; 91(21)2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794043

RESUMEN

There is an urgent need for therapeutic development to combat infections caused by Rift Valley fever virus (RVFV), which causes devastating disease in both humans and animals. In an effort to repurpose drugs for RVFV treatment, our previous studies screened a library of FDA-approved drugs. The most promising candidate identified was the hepatocellular and renal cell carcinoma drug sorafenib. Mechanism-of-action studies indicated that sorafenib targeted a late stage in virus infection and caused a buildup of virions within cells. In addition, small interfering RNA (siRNA) knockdown studies suggested that nonclassical targets of sorafenib are important for the propagation of RVFV. Here we extend our previous findings to identify the mechanism by which sorafenib inhibits the release of RVFV virions from the cell. Confocal microscopy imaging revealed that glycoprotein Gn colocalizes and accumulates within the endoplasmic reticulum (ER) and the transport of Gn from the Golgi complex to the host cell membrane is reduced. Transmission electron microscopy demonstrated that sorafenib caused virions to be present inside large vacuoles inside the cells. p97/valosin-containing protein (VCP), which is involved in membrane remodeling in the secretory pathway and a known target of sorafenib, was found to be important for RVFV egress. Knockdown of VCP resulted in decreased RVFV replication, reduced Gn Golgi complex localization, and increased Gn ER accumulation. The intracellular accumulation of RVFV virions was also observed in cells transfected with siRNA targeting VCP. Collectively, these data indicate that sorafenib causes a disruption in viral egress by targeting VCP and the secretory pathway, resulting in a buildup of virions within dilated ER vesicles.IMPORTANCE In humans, symptoms of RVFV infection mainly include a self-limiting febrile illness. However, in some cases, infected individuals can also experience hemorrhagic fever, neurological disorders, liver failure, and blindness, which could collectively be lethal. The ability of RVFV to expand geographically outside sub-Saharan Africa is of concern, particularly to the Americas, where native mosquito species are capable of virus transmission. Currently, there are no FDA-approved therapeutics to treat RVFV infection, and thus, there is an urgent need to understand the mechanisms by which the virus hijacks the host cell machinery to replicate. The significance of our research is in identifying the cellular target of sorafenib that inhibits RVFV propagation, so that this information can be used as a tool for the further development of therapeutics used to treat RVFV infection.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Fiebre del Valle del Rift/tratamiento farmacológico , Virus de la Fiebre del Valle del Rift/fisiología , Vías Secretoras/efectos de los fármacos , Liberación del Virus/efectos de los fármacos , Adenosina Trifosfatasas/genética , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Proteínas de Ciclo Celular/genética , Chlorocebus aethiops , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Niacinamida/farmacología , Fiebre del Valle del Rift/metabolismo , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Sorafenib , Células Tumorales Cultivadas , Proteína que Contiene Valosina , Células Vero , Virión/efectos de los fármacos , Replicación Viral/efectos de los fármacos
2.
J Virol ; 91(3)2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852852

RESUMEN

The alphaviruses Venezuelan equine encephalitis virus (VEEV), eastern equine encephalitis virus (EEEV), and western equine encephalitis virus (WEEV) are arthropod-borne positive-strand RNA viruses that are capable of causing acute and fatal encephalitis in many mammals, including humans. VEEV was weaponized during the Cold War and is recognized as a select agent. Currently, there are no FDA-approved vaccines or therapeutics for these viruses. The spread of VEEV and other members of this family due to climate change-mediated vector range expansion underscores the need for research aimed at developing medical countermeasures. These viruses utilize programmed -1 ribosomal frameshifting (-1 PRF) to synthesize the viral trans-frame (TF) protein, which has previously been shown to be important for neuropathogenesis in the related Sindbis virus. Here, the alphavirus -1 PRF signals were characterized, revealing novel -1 PRF stimulatory structures. -1 PRF attenuation mildly affected the kinetics of VEEV accumulation in cultured cells but strongly inhibited its pathogenesis in an aerosol infection mouse model. Importantly, the decreased viral titers in the brains of mice infected with the mutant virus suggest that the alphavirus TF protein is important for passage through the blood-brain barrier and/or for neuroinvasiveness. These findings suggest a novel approach to the development of safe and effective live attenuated vaccines directed against VEEV and perhaps other closely related -1 PRF-utilizing viruses. IMPORTANCE: Venezuelan equine encephalitis virus (VEEV) is a select agent that has been weaponized. This arthropod-borne positive-strand RNA virus causes acute and fatal encephalitis in many mammals, including humans. There is no vaccine or other approved therapeutic. VEEV and related alphaviruses utilize programmed -1 ribosomal frameshifting (-1 PRF) to synthesize the viral trans-frame (TF) protein, which is important for neuropathogenesis. -1 PRF attenuation strongly inhibited VEEV pathogenesis in mice, and viral replication analyses suggest that the TF protein is critical for neurological disease. These findings suggest a new approach to the development of safe and effective live attenuated vaccines directed against VEEV and other related viruses.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/genética , Encefalomielitis Equina Venezolana/virología , Sistema de Lectura Ribosómico , Animales , Línea Celular , Femenino , Genoma Viral , Caballos , Humanos , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta , ARN Mensajero/química , ARN Mensajero/genética , ARN Viral , Replicación Viral
3.
Front Cell Infect Microbiol ; 14: 1331755, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38800833

RESUMEN

The mosquito-borne Rift Valley fever virus (RVFV) from the Phenuiviridae family is a single-stranded RNA virus that causes the re-emerging zoonotic disease Rift Valley fever (RVF). Classified as a Category A agent by the NIH, RVFV infection can cause debilitating disease or death in humans and lead to devastating economic impacts by causing abortion storms in pregnant cattle. In a previous study, we showed that the host chaperone protein HSP90 is an RVFV-associated host factor that plays a critical role post viral entry, during the active phase of viral genome replication/transcription. In this study, we have elucidated the molecular mechanisms behind the regulatory effect of HSP90 during infection with RVFV. Our results demonstrate that during the early infection phase, host HSP90 associates with the viral RNA-dependent RNA polymerase (L protein) and prevents its degradation through the proteasome, resulting in increased viral replication.


Asunto(s)
Proteínas HSP90 de Choque Térmico , Complejo de la Endopetidasa Proteasomal , Proteolisis , Virus de la Fiebre del Valle del Rift , Replicación Viral , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Genoma Viral , Humanos , ARN Polimerasa Dependiente del ARN/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Interacciones Huésped-Patógeno , Proteínas Virales/metabolismo , Proteínas Virales/genética , Transcripción Genética , Fiebre del Valle del Rift/virología , Fiebre del Valle del Rift/metabolismo , Línea Celular
4.
Antiviral Res ; 157: 57-67, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29981794

RESUMEN

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


Asunto(s)
Alphavirus/efectos de los fármacos , Antineoplásicos/farmacología , Antivirales/farmacología , Reposicionamiento de Medicamentos , Sorafenib/farmacología , Replicación Viral/efectos de los fármacos , Alphavirus/crecimiento & desarrollo , Animales , Línea Celular , Evaluación Preclínica de Medicamentos/métodos , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Luciferasas/análisis , Luciferasas/genética , Mediciones Luminiscentes , Genética Inversa
5.
Viruses ; 10(4)2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29652799

RESUMEN

Viruses must parasitize host cell translational machinery in order to make proteins for viral progeny. In this study, we sought to use this signal transduction conduit against them by inhibiting multiple kinases that influence translation. Previous work indicated that several kinases involved in translation, including p70 S6K, p90RSK, ERK, and p38 MAPK, are phosphorylated following Rift Valley fever virus (RVFV) infection. Furthermore, inhibiting p70 S6K through treatment with the FDA approved drug rapamycin prevents RVFV pathogenesis in a mouse model of infection. We hypothesized that inhibiting either p70 S6K, p90RSK, or p90RSK’s upstream kinases, ERK and p38 MAPK, would decrease translation and subsequent viral replication. Treatment with the p70 S6K inhibitor PF-4708671 resulted in decreased phosphorylation of translational proteins and reduced RVFV titers. In contrast, treatment with the p90RSK inhibitor BI-D1870, p38MAPK inhibitor SB203580, or the ERK inhibitor PD0325901 alone had minimal influence on RVFV titers. The combination of PF-4708671 and BI-D1870 treatment resulted in robust inhibition of RVFV replication. Likewise, a synergistic inhibition of RVFV replication was observed with p38MAPK inhibitor SB203580 or the ERK inhibitor PD0325901 combined with rapamycin treatment. These findings serve as a proof of concept regarding combination kinase inhibitor treatment for RVFV infection.


Asunto(s)
Antivirales/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/fisiología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Ratones , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Proteínas Ribosómicas/metabolismo
6.
Antiviral Res ; 143: 162-175, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28442428

RESUMEN

Despite over 60 years of research on antiviral drugs, very few are FDA approved to treat acute viral infections. Rift Valley fever virus (RVFV), an arthropod borne virus that causes hemorrhagic fever in severe cases, currently lacks effective treatments. Existing as obligate intracellular parasites, viruses have evolved to manipulate host cell signaling pathways to meet their replication needs. Specifically, translation modulation is often necessary for viruses to establish infection in their host. Here we demonstrated phosphorylation of p70 S6 kinase, S6 ribosomal protein, and eIF4G following RVFV infection in vitro through western blot analysis and in a mouse model of infection through reverse phase protein microarrays (RPPA). Inhibition of p70 S6 kinase through rapamycin treatment reduced viral titers in vitro and increased survival and mitigated clinical disease in RVFV challenged mice. Additionally, the phosphorylation of p70 S6 kinase was decreased following rapamycin treatment in vivo. Collectively these data demonstrate modulating p70 S6 kinase can be an effective antiviral strategy.


Asunto(s)
Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Virus de la Fiebre del Valle del Rift/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirolimus/antagonistas & inhibidores , Animales , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Replicación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Factor 4G Eucariótico de Iniciación/metabolismo , Femenino , Inmunohistoquímica , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/patología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/crecimiento & desarrollo , Virus de la Fiebre del Valle del Rift/patogenicidad , Sirolimus/metabolismo , Sirolimus/uso terapéutico , Análisis de Supervivencia , Células Vero , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
7.
PLoS Negl Trop Dis ; 10(11): e0005122, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27902702

RESUMEN

The capsid structural protein of the New World alphavirus, Venezuelan equine encephalitis virus (VEEV), interacts with the host nuclear transport proteins importin α/ß1 and CRM1. Novel selective inhibitor of nuclear export (SINE) compounds, KPT-185, KPT-335 (verdinexor), and KPT-350, target the host's primary nuclear export protein, CRM1, in a manner similar to the archetypical inhibitor Leptomycin B. One major limitation of Leptomycin B is its irreversible binding to CRM1; which SINE compounds alleviate because they are slowly reversible. Chemically inhibiting CRM1 with these compounds enhanced capsid localization to the nucleus compared to the inactive compound KPT-301, as indicated by immunofluorescent confocal microscopy. Differences in extracellular versus intracellular viral RNA, as well as decreased capsid in cell free supernatants, indicated the inhibitors affected viral assembly, which led to a decrease in viral titers. The decrease in viral replication was confirmed using a luciferase-tagged virus and through plaque assays. SINE compounds had no effect on VEEV TC83_Cm, which encodes a mutated form of capsid that is unable to enter the nucleus. Serially passaging VEEV in the presence of KPT-185 resulted in mutations within the nuclear localization and nuclear export signals of capsid. Finally, SINE compound treatment also reduced the viral titers of the related eastern and western equine encephalitis viruses, suggesting that CRM1 maintains a common interaction with capsid proteins across the New World alphavirus genus.


Asunto(s)
Infecciones por Alphavirus/virología , Alphavirus/efectos de los fármacos , Antivirales/farmacología , Proteínas de la Cápside/metabolismo , Replicación Viral/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de los fármacos , Alphavirus/genética , Alphavirus/fisiología , Animales , Proteínas de la Cápside/genética , Núcleo Celular/virología , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/genética , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Ensamble de Virus/efectos de los fármacos , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA