Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Intervalo de año de publicación
1.
Viruses ; 14(2)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35215938

RESUMEN

Rift Valley fever (RVF) is a zoonotic disease caused by RVF Phlebovirus (RVFV). The RVFV MP-12 vaccine strain is known to exhibit residual virulence in the case of a deficient interferon type 1 response. The hypothesis of this study is that virus replication and severity of lesions induced by the MP-12 strain in immunocompromised mice depend on the specific function of the disturbed pathway. Therefore, 10 strains of mice with deficient innate immunity (B6-IFNARtmAgt, C.129S7(B6)-Ifngtm1Ts/J, B6-TLR3tm1Flv, B6-TLR7tm1Aki, NOD/ShiLtJ), helper T-cell- (CD4tm1Mak), cytotoxic T-cell- (CD8atm1Mak), B-cell- (Igh-Jtm1DhuN?+N2), combined T- and B-cell- (NU/J) and combined T-, B-, natural killer (NK) cell- and macrophage-mediated immunity (NOD.Cg-PrkdcscidIl2rgtm1WjI/SzJ (NSG) mice) were subcutaneously infected with RVFV MP-12. B6-IFNARtmAgt mice were the only strain to develop fatal disease due to RVFV-induced severe hepatocellular necrosis and apoptosis. Notably, no clinical disease and only mild multifocal hepatocellular necrosis and apoptosis were observed in NSG mice, while immunohistochemistry detected the RVFV antigen in the liver and the brain. No or low virus expression and no lesions were observed in the other mouse strains. Conclusively, the interferon type 1 response is essential for early control of RVFV replication and disease, whereas functional NK cells, macrophages and lymphocytes are essential for virus clearance.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Animales , Apoptosis , Femenino , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Hígado/inmunología , Hígado/virología , Macrófagos/inmunología , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos NOD , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/fisiopatología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/virología
2.
J Virol ; 95(23): e0150621, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34495703

RESUMEN

Rift Valley fever virus (RVFV) is an arbovirus found throughout Africa. It causes disease that is typically mild and self-limiting; however, some infected individuals experience severe manifestations, including hepatitis, encephalitis, or even death. Reports of RVFV encephalitis are notable among immunosuppressed individuals, suggesting a role for adaptive immunity in preventing this severe complication. This phenomenon has been modeled in C57BL/6 mice depleted of CD4 T cells prior to infection with DelNSs RVFV (RVFV containing a deletion of nonstructural protein NSs), resulting in late-onset encephalitis accompanied by high levels of viral RNA in the brain in 30% of animals. In this study, we sought to define the specific type(s) of CD4 T cells that mediate protection from RVFV encephalitis. The viral epitopes targeted by CD4 and CD8 T cells were defined in C57BL/6 mice, and tetramers for both CD4 and CD8 T cells were generated. RVFV-specific CD8 T cells were expanded and of a cytotoxic and proliferating phenotype in the liver following infection. RVFV-specific CD4 T cells were identified in the liver and spleen following infection and phenotyped as largely Th1 or Tfh subtypes. Knockout mice lacking various aspects of pathways important in Th1 and Tfh development and function were used to demonstrate that T-bet, CD40, CD40L, and major histocompatibility complex class II (MHC-II) mediated protection from RVFV encephalitis, while gamma interferon (IFN-γ) and interleukin-12 (IL-12) were dispensable. Virus-specific antibody responses correlated with protection from encephalitis in all mouse strains, suggesting that Tfh/B cell interactions modulate clinical outcome in this model. IMPORTANCE The prevention of RVFV encephalitis requires intact adaptive immunity. In this study, we developed reagents to detect RVFV-specific T cells and provide evidence for Tfh cells and CD40/CD40L interactions as critical mediators of this protection.


Asunto(s)
Antígenos CD40 , Ligando de CD40 , Encefalitis Viral/prevención & control , Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Linfocitos T/inmunología , África , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Encéfalo/virología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Epítopos , Femenino , Hígado/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
3.
PLoS Negl Trop Dis ; 15(9): e0009785, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34516560

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus with a wide host range including ruminants and humans. RVFV outbreaks have had devastating effects on public health and the livestock industry in African countries. However, there is no approved RVFV vaccine for human use in non-endemic countries and no FDA-approved antiviral drug for RVFV treatment. The RVFV 78kDa protein (P78), which is a membrane glycoprotein, plays a role in virus dissemination in the mosquito host, but its biological role in mammalian hosts remains unknown. We generated an attenuated RVFV MP-12 strain-derived P78-High virus and a virulent ZH501 strain-derived ZH501-P78-High virus, both of which expressed a higher level of P78 and carried higher levels of P78 in the virion compared to their parental viruses. We also generated another MP-12-derived mutant virus (P78-KO virus) that does not express P78. MP-12 and P78-KO virus replicated to similar levels in fibroblast cell lines and Huh7 cells, while P78-High virus replicated better than MP-12 in Vero E6 cells, fibroblast cell lines, and Huh7 cells. Notably, P78-High virus and P78-KO virus replicated less efficiently and more efficiently, respectively, than MP-12 in macrophage cell lines. ZH501-P78-High virus also replicated poorly in macrophage cell lines. Our data further suggest that inefficient binding of P78-High virus to the cells led to inefficient virus internalization, low virus infectivity and reduced virus replication in a macrophage cell line. P78-High virus and P78-KO virus showed lower and higher virulence than MP-12, respectively, in young mice. ZH501-P78-High virus also exhibited lower virulence than ZH501 in mice. These data suggest that high levels of P78 expression attenuate RVFV virulence by preventing efficient virus replication in macrophages. Genetic alteration leading to increased P78 expression may serve as a novel strategy for the attenuation of RVFV virulence and generation of safe RVFV vaccines.


Asunto(s)
Macrófagos/virología , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/fisiología , Animales , Ratones , Virus de la Fiebre del Valle del Rift/patogenicidad , Proteínas del Envoltorio Viral/genética , Virulencia
4.
Mamm Genome ; 32(1): 30-37, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33420513

RESUMEN

Rift Valley fever (RVF) is an emerging viral zoonosis that primarily affects ruminants and humans. We have previously shown that wild-derived MBT/Pas mice are highly susceptible to RVF virus and that part of this phenotype is controlled by a locus located on distal Chromosome 11. Using congenic strains, we narrowed down the critical interval to a 530 kb region containing five protein-coding genes among which Rnf213 emerged as a potential candidate. We generated Rnf213-deficient mice by CRISPR/CAS9 on the C57BL/6 J background and showed that they were significantly more susceptible to RVF than control mice, with an average survival time post-infection reduced from 7 to 4 days. The human RNF213 gene had been associated with the cerebrovascular Moyamoya disease (MMD or MYMY) but the inactivation of this gene in the mouse resulted only in mild anomalies of the neovascularization. This study provides the first evidence that the Rnf213 gene may also impact the resistance to infectious diseases such as RVF.


Asunto(s)
Adenosina Trifosfatasas/genética , Resistencia a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Fiebre del Valle del Rift/genética , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/fisiología , Ubiquitina-Proteína Ligasas/genética , Animales , Sistemas CRISPR-Cas , Mapeo Cromosómico , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
J Virol ; 93(14)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31043534

RESUMEN

Several host and viral processes contribute to forming infectious virions. Polyamines are small host molecules that play diverse roles in viral replication. We previously demonstrated that polyamines are crucial for RNA viruses; however, the mechanisms by which polyamines function remain unknown. Here, we investigated the role of polyamines in the replication of the bunyaviruses Rift Valley fever virus (vaccine strain MP-12) and La Crosse virus (LACV). We found that polyamine depletion did not impact viral RNA or protein accumulation, despite significant decreases in titer. Viral particles demonstrated no change in morphology, size, or density. Thus, polyamine depletion promotes the formation of noninfectious particles. These particles interfere with virus replication and stimulate innate immune responses. We extended this phenotype to Zika virus; however, coxsackievirus did not similarly produce noninfectious particles. In sum, polyamine depletion results in the accumulation of noninfectious particles that interfere with replication and stimulate immune signaling, with important implications for targeting polyamines therapeutically, as well as for vaccine strategies.IMPORTANCE Bunyaviruses are emerging viral pathogens that cause encephalitis, hemorrhagic fevers, and meningitis. We have uncovered that diverse bunyaviruses require polyamines for productive infection. Polyamines are small, positively charged host-derived molecules that play diverse roles in human cells and in infection. In polyamine-depleted cells, bunyaviruses produce an overabundance of noninfectious particles that are indistinguishable from infectious particles. However, these particles interfere with productive infection and stimulate antiviral signaling pathways. We further find that additional enveloped viruses are similarly sensitive to polyamine depletion but that a nonenveloped enterovirus is not. We posit that polyamines are required to maintain bunyavirus infectivity and that polyamine depletion results in the accumulation of interfering noninfectious particles that limit infectivity. These results highlight a novel means by which bunyaviruses use polyamines for replication and suggest promising means to target host polyamines to reduce virus replication.


Asunto(s)
Poliaminas Biogénicas/inmunología , Infecciones por Bunyaviridae/inmunología , Virus Defectuosos/fisiología , Virus de la Encefalitis de California/fisiología , Virus de la Fiebre del Valle del Rift/fisiología , Virión/fisiología , Replicación Viral/inmunología , Infecciones por Bunyaviridae/genética , Infecciones por Bunyaviridae/patología , Línea Celular Tumoral , Humanos
6.
PLoS One ; 13(7): e0201104, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30028878

RESUMEN

Rift Valley fever (RVF) in humans is usually mild, but, in a subset of cases, can progress to severe hepatic and neurological disease. Rodent models of RVF generally develop acute severe clinical disease. Here, we inoculated humanized NSG-SGM3 mice with Rift Valley fever virus (RVFV) to investigate whether the presence of human immune cells in mice would alter the progression of RVFV infection to more closely model human disease. Despite increased human cytokine expression, including responses mirroring those seen in human disease, and decreased hepatic viral RNA levels at terminal euthanasia, both high- and low-dose RVFV inoculation resulted in lethal disease in all mice with comparable time-to-death as unengrafted mice.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Fiebre del Valle del Rift/inmunología , Fiebre del Valle del Rift/terapia , Enfermedad Aguda , Animales , Antígenos CD34/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Hígado/inmunología , Hígado/virología , Ratones , ARN Viral/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Índice de Severidad de la Enfermedad
7.
Sci Rep ; 8(1): 1925, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386590

RESUMEN

Rift Valley fever virus (RVFV) is a mosquito-borne hemorrhagic fever virus affecting both humans and animals with severe morbidity and mortality and is classified as a potential bioterror agent due to the possible aerosol transmission. At present there is no human vaccine or antiviral therapy available. Thus, there is a great need to develop new antivirals for treatment of RVFV infections. Benzavir-2 was previously identified as potent inhibitor of human adenovirus, herpes simplex virus type 1, and type 2. Here we assess the anti-RVFV activity of benzavir-2 together with four structural analogs and determine pre-clinical pharmacokinetic parameters of benzavir-2. In vitro, benzavir-2 efficiently inhibited RVFV infection, viral RNA production and production of progeny viruses. In vitro, benzavir-2 displayed satisfactory solubility, good permeability and metabolic stability. In mice, benzavir-2 displayed oral bioavailability with adequate maximum serum concentration. Oral administration of benzavir-2 formulated in peanut butter pellets gave high systemic exposure without any observed toxicity in mice. To summarize, our data demonstrated potent anti-RVFV activity of benzavir-2 in vitro together with a promising pre-clinical pharmacokinetic profile. This data support further exploration of the antiviral activity of benzavir-2 in in vivo efficacy models that may lead to further drug development for human use.


Asunto(s)
Antivirales/farmacología , Antivirales/farmacocinética , Benzoatos/farmacología , Benzoatos/farmacocinética , Virus de la Fiebre del Valle del Rift/fisiología , Células A549 , Administración Oral , Animales , Antivirales/administración & dosificación , Antivirales/química , Benzoatos/administración & dosificación , Benzoatos/química , Disponibilidad Biológica , Femenino , Humanos , Ratones Endogámicos BALB C , ARN Viral/genética , Fiebre del Valle del Rift/tratamiento farmacológico , Fiebre del Valle del Rift/prevención & control , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/efectos de los fármacos
8.
Nat Commun ; 9(1): 349, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29367607

RESUMEN

Entry of enveloped viruses relies on insertion of hydrophobic residues of the viral fusion protein into the host cell membrane. However, the intermediate conformations during fusion remain unknown. Here, we address the fusion mechanism of Rift Valley fever virus. We determine the crystal structure of the Gn glycoprotein and fit it with the Gc fusion protein into cryo-electron microscopy reconstructions of the virion. Our analysis reveals how the Gn shields the hydrophobic fusion loops of the Gc, preventing premature fusion. Electron cryotomography of virions interacting with membranes under acidic conditions reveals how the fusogenic Gc is activated upon removal of the Gn shield. Repositioning of the Gn allows extension of Gc and insertion of fusion loops in the outer leaflet of the target membrane. These data show early structural transitions that enveloped viruses undergo during host cell entry and indicate that analogous shielding mechanisms are utilized across diverse virus families.


Asunto(s)
Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Virión/química , Internalización del Virus , Microscopía por Crioelectrón , Cristalografía por Rayos X , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Pliegue de Proteína , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas Virales de Fusión/genética , Virión/metabolismo
9.
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
10.
Viruses ; 8(5)2016 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-27223297

RESUMEN

Rift Valley fever is a mosquito-transmitted, zoonotic disease that infects humans and ruminants. Dendritic cell specific intercellular adhesion molecule 3 (ICAM-3) grabbing non-integrin (DC-SIGN) acts as a receptor for members of the phlebovirus genus. The Rift Valley fever virus (RVFV) glycoproteins (Gn/Gc) encode five putative N-glycan sequons (asparagine (N)-any amino acid (X)-serine (S)/threonine (T)) at positions: N438 (Gn), and N794, N829, N1035, and N1077 (Gc). The N-glycosylation profile and significance in viral infection via DC-SIGN have not been elucidated. Gc N-glycosylation was first evaluated by using Gc asparagine (N) to glutamine (Q) mutants. Subsequently, we generated a series of recombinant RVFV MP-12 strain mutants, which encode N-to-Q mutations, and the infectivity of each mutant in Jurkat cells stably expressing DC-SIGN was evaluated. Results showed that Gc N794, N1035, and N1077 were N-glycosylated but N829 was not. Gc N1077 was heterogeneously N-glycosylated. RVFV Gc made two distinct N-glycoforms: "Gc-large" and "Gc-small", and N1077 was responsible for "Gc-large" band. RVFV showed increased infection of cells expressing DC-SIGN compared to cells lacking DC-SIGN. Infection via DC-SIGN was increased in the presence of either Gn N438 or Gc N1077. Our study showed that N-glycans on the Gc and Gn surface glycoproteins redundantly support RVFV infection via DC-SIGN.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Glicoproteínas/metabolismo , Lectinas Tipo C/metabolismo , Polisacáridos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Proteínas Estructurales Virales/metabolismo , Acoplamiento Viral , Sustitución de Aminoácidos , Glicoproteínas/genética , Humanos , Células Jurkat , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Virus de la Fiebre del Valle del Rift/genética , Proteínas Estructurales Virales/genética
11.
Am J Trop Med Hyg ; 93(6): 1364-1370, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26458775

RESUMEN

In this cross-sectional seroepidemiological study we sought to examine the evidence for circulation of Rift Valley fever virus (RVFV) among herders in Madagascar and Kenya. From July 2010 to June 2012, we enrolled 459 herders and 98 controls (without ruminant exposures) and studied their sera (immunoglobulin G [IgG] and IgM through enzyme-linked immunosorbent assay [ELISA] and plaque reduction neutralization test [PRNT] assays) for evidence of previous RVFV infection. Overall, 59 (12.9%) of 459 herders and 7 (7.1%) of the 98 controls were positive by the IgG ELISA assay. Of the 59 ELISA-positive herders, 23 (38.9%) were confirmed by the PRNT assay (21 from eastern Kenya). Two of the 21 PRNT-positive study subjects also had elevated IgM antibodies against RVFV suggesting recent infection. Multivariate modeling in this study revealed that being seminomadic (odds ratio [OR] = 6.4, 95% confidence interval [CI] = 2.1-15.4) was most strongly associated with antibodies against RVFV. Although we cannot know when these infections occurred, it seems likely that some interepidemic RVFV infections are occurring among herders. As there are disincentives regarding reporting RVFV outbreaks in livestock or wildlife, it may be prudent to conduct periodic, limited, active seroepidemiological surveillance for RVFV infections in herders, especially in eastern Kenya.


Asunto(s)
Fiebre del Valle del Rift/epidemiología , Rumiantes/virología , Adolescente , Adulto , Animales , Anticuerpos Antivirales/inmunología , Ensayo de Inmunoadsorción Enzimática , Epidemias/estadística & datos numéricos , Femenino , Humanos , Inmunoglobulina M/inmunología , Kenia/epidemiología , Madagascar/epidemiología , Masculino , Persona de Mediana Edad , Pruebas de Neutralización , Fiebre del Valle del Rift/diagnóstico , Fiebre del Valle del Rift/etiología , Virus de la Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Factores de Riesgo , Adulto Joven , Zoonosis/epidemiología , Zoonosis/virología
12.
Antiviral Res ; 120: 48-56, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26001632

RESUMEN

Rift Valley Fever Virus (RVFV) belongs to the family Bunyaviridae and is a known cause of epizootics and epidemics in Africa and the Middle East. With no FDA approved therapeutics available to treat RVFV infection, understanding the interactions between the virus and the infected host is crucial to developing novel therapeutic strategies. Here, we investigated the requirement of the ubiquitin-proteasome system (UPS) for the establishment of a productive RVFV infection. It was previously shown that the UPS plays a central role in RVFV multiplication involving degradation of PKR and p62 subunit of TFIIH. Using the FDA-approved proteasome inhibitor Bortezomib, we observed robust inhibition of intracellular and extracellular viral loads. Bortezomib treatment did not affect the nuclear/cytoplasmic distribution of the non-structural S-segment protein (NSs); however, the ability of NSs to form nuclear filaments was abolished as a result of Bortezomib treatment. In silico ubiquitination prediction analysis predicted that known NSs interactors (SAP30, YY1, and mSin3A) have multiple putative ubiquitination sites, while NSs itself was not predicted to be ubiquitinated. Immunoprecipitation studies indicated a decrease in interaction between SAP30 - NSs, and mSin3A - NSs in the context of Bortezomib treatment. This decrease in association between SAP30 - NSs also correlated with a decrease in the ubiquitination status of SAP30 with Bortezomib treatment. Bortezomib treatment, however, resulted in increased ubiquitination of mSin3A, suggesting that Bortezomib dynamically affects the ubiquitination status of host proteins that interact with NSs. Finally, we observed that expression of interferon beta (IFN-ß) was increased in Bortezomib treated cells which indicated that the cellular antiviral mechanism was revived as a result of treatment and may contribute to control of viral multiplication.


Asunto(s)
Antivirales/farmacología , Bortezomib/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 , Células Cultivadas , Células Epiteliales/virología , Humanos , Carga Viral
13.
Cell ; 158(4): 764-777, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25126784

RESUMEN

DEAD-box helicases play essential roles in RNA metabolism across species, but emerging data suggest that they have additional functions in immunity. Through RNAi screening, we identify an evolutionarily conserved and interferon-independent role for the DEAD-box helicase DDX17 in restricting Rift Valley fever virus (RVFV), a mosquito-transmitted virus in the bunyavirus family that causes severe morbidity and mortality in humans and livestock. Loss of Drosophila DDX17 (Rm62) in cells and flies enhanced RVFV infection. Similarly, depletion of DDX17 but not the related helicase DDX5 increased RVFV replication in human cells. Using crosslinking immunoprecipitation high-throughput sequencing (CLIP-seq), we show that DDX17 binds the stem loops of host pri-miRNA to facilitate their processing and also an essential stem loop in bunyaviral RNA to restrict infection. Thus, DDX17 has dual roles in the recognition of stem loops: in the nucleus for endogenous microRNA (miRNA) biogenesis and in the cytoplasm for surveillance against structured non-self-elements.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , MicroARNs/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Animales , Línea Celular Tumoral , ARN Helicasas DEAD-box/inmunología , Proteínas de Drosophila/inmunología , Drosophila melanogaster/metabolismo , Drosophila melanogaster/virología , Humanos , Inmunidad Innata , Secuencias Invertidas Repetidas , ARN Viral/química , Replicación Viral
14.
J Virol ; 88(18): 10883-93, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008937

RESUMEN

UNLABELLED: Bunyavirus genomes comprise a small (S), a medium (M), and a large (L) RNA segment of negative polarity. Although the untranslated regions have been shown to comprise signals required for transcription, replication, and encapsidation, the mechanisms that drive the packaging of at least one S, M, and L segment into a single virion to generate infectious virus are largely unknown. One of the most important members of the Bunyaviridae family that causes devastating disease in ruminants and occasionally humans is the Rift Valley fever virus (RVFV). We studied the flexibility of RVFV genome packaging by splitting the glycoprotein precursor gene, encoding the (NSm)GnGc polyprotein, into two individual genes encoding either (NSm)Gn or Gc. Using reverse genetics, six viruses with a segmented glycoprotein precursor gene were rescued, varying from a virus comprising two S-type segments in the absence of an M-type segment to a virus consisting of four segments (RVFV-4s), of which three are M-type. Despite that all virus variants were able to grow in mammalian cell lines, they were unable to spread efficiently in cells of mosquito origin. Moreover, in vivo studies demonstrated that RVFV-4s is unable to cause disseminated infection and disease in mice, even in the presence of the main virulence factor NSs, but induced a protective immune response against a lethal challenge with wild-type virus. In summary, splitting bunyavirus glycoprotein precursor genes provides new opportunities to study bunyavirus genome packaging and offers new methods to develop next-generation live-attenuated bunyavirus vaccines. IMPORTANCE: Rift Valley fever virus (RVFV) causes devastating disease in ruminants and occasionally humans. Virions capable of productive infection comprise at least one copy of the small (S), medium (M), and large (L) RNA genome segments. The M segment encodes a glycoprotein precursor (GPC) protein that is cotranslationally cleaved into Gn and Gc, which are required for virus entry and fusion. We studied the flexibility of RVFV genome packaging and developed experimental live-attenuated vaccines by applying a unique strategy based on the splitting of the GnGc open reading frame. Several RVFV variants, varying from viruses comprising two S-type segments to viruses consisting of four segments (RVFV-4s), of which three are M-type, could be rescued and were shown to induce a rapid protective immune response. Altogether, the segmentation of bunyavirus GPCs provides a new method for studying bunyavirus genome packaging and facilitates the development of novel live-attenuated bunyavirus vaccines.


Asunto(s)
Genoma Viral , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/genética , Virión/fisiología , Ensamble de Virus , Animales , Culicidae/virología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , ARN Viral/genética , Virus de la Fiebre del Valle del Rift/fisiología , Virión/genética
15.
Virology ; 449: 270-86, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24418562

RESUMEN

Rift Valley fever virus (RVFV) infection is often associated with pronounced liver damage. Previously, our studies revealed altered host phospho-signaling responses (NFκB, MAPK and DNA damage responses) in RVFV infected epithelial cells that correlated with a cellular stress response. Here, we report that RVFV infection of liver cells leads to an increase in reactive oxygen species (ROS). Our data suggests the presence of the viral protein NSs in the mitochondria of infected cells, hence contributing to early increase in ROS. Increased ROS levels correlated with activation of NFκB (p65) and p53 responses, which in conjunction with infection, was also reflected as macromolecular rearrangements observed using size fractionation of protein lysates. Additionally, we documented an increase in cytokine expression and pro-apoptotic gene expression with infection, which was reversed with antioxidant treatment. Collectively, we identified ROS and oxidative stress as critical contributors to apoptosis of liver cells during RVFV infection.


Asunto(s)
Apoptosis , Hígado/citología , Especies Reactivas de Oxígeno/metabolismo , Fiebre del Valle del Rift/metabolismo , Virus de la Fiebre del Valle del Rift/fisiología , Factor de Transcripción ReIA/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Humanos , Hígado/metabolismo , Hígado/virología , Estrés Oxidativo , Fiebre del Valle del Rift/fisiopatología , Fiebre del Valle del Rift/virología , Factor de Transcripción ReIA/genética , Proteína p53 Supresora de Tumor/genética , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
16.
J Virol ; 87(15): 8451-64, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23720721

RESUMEN

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


Asunto(s)
Antígenos de Diferenciación/inmunología , Interacciones Huésped-Patógeno , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Línea Celular , Virus Hantaan/inmunología , Virus Hantaan/fisiología , Orthohantavirus/inmunología , Orthohantavirus/fisiología , Humanos , Interferón-alfa/inmunología , Virus La Crosse/inmunología , Virus La Crosse/fisiología
17.
Anal Biochem ; 438(1): 90-6, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23535274

RESUMEN

To fully understand the interactions of a pathogen with its host, it is necessary to analyze the RNA transcripts of both the host and pathogen throughout the course of an infection. Although this can be accomplished relatively easily on the host side, the analysis of pathogen transcripts is complicated by the overwhelming amount of host RNA isolated from an infected sample. Even with the read depth provided by second-generation sequencing, it is extremely difficult to get enough pathogen reads for an effective gene-level analysis. In this study, we describe a novel capture-based technique and device that considerably enriches for pathogen transcripts from infected samples. This versatile method can, in principle, enrich for any pathogen in any infected sample. To test the technique's efficacy, we performed time course tissue culture infections using Rift Valley fever virus and Francisella tularensis. At each time point, RNA sequencing (RNA-Seq) was performed and the results of the treated samples were compared with untreated controls. The capture of pathogen transcripts, in all cases, led to more than an order of magnitude enrichment of pathogen reads, greatly increasing the number of genes hit, the coverage of those genes, and the depth at which each transcript was sequenced.


Asunto(s)
Francisella tularensis/genética , Francisella tularensis/fisiología , Interacciones Huésped-Patógeno , Virus de la Fiebre del Valle del Rift/genética , Virus de la Fiebre del Valle del Rift/fisiología , Análisis de Secuencia de ARN/métodos , Línea Celular , Perfilación de la Expresión Génica , Humanos , Macrófagos/microbiología , Macrófagos/virología , Hibridación de Ácido Nucleico , ARN Bacteriano/genética , ARN Mensajero/genética , ARN Viral/genética
18.
J Virol ; 87(9): 4846-60, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23408632

RESUMEN

Rift Valley fever virus (RVFV) is an emerging RNA virus with devastating economic and social consequences. Clinically, RVFV induces a gamut of symptoms ranging from febrile illness to retinitis, hepatic necrosis, hemorrhagic fever, and death. It is known that type I interferon (IFN) responses can be protective against severe pathology; however, it is unknown which innate immune receptor pathways are crucial for mounting this response. Using both in vitro assays and in vivo mucosal mouse challenge, we demonstrate here that RNA helicases are critical for IFN production by immune cells and that signaling through the helicase adaptor molecule MAVS (mitochondrial antiviral signaling) is protective against mortality and more subtle pathology during RVFV infection. In addition, we demonstrate that Toll-like-receptor-mediated signaling is not involved in IFN production, further emphasizing the importance of the RNA cellular helicases in type I IFN responses to RVFV.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Interferón beta/inmunología , Membrana Mucosa/virología , Fiebre del Valle del Rift/enzimología , Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/fisiología , Animales , Línea Celular , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Células Dendríticas/inmunología , Células Dendríticas/virología , Femenino , Humanos , Interferón beta/genética , Macrófagos/inmunología , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , Membrana Mucosa/inmunología , Fiebre del Valle del Rift/prevención & control , Fiebre del Valle del Rift/virología , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
19.
J Virol ; 86(23): 12954-70, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22993156

RESUMEN

Rift Valley fever virus (RVFV) is a zoonotic pathogen capable of causing serious morbidity and mortality in both humans and livestock. The lack of efficient countermeasure strategies, the potential for dispersion into new regions, and the pathogenesis in humans and livestock make RVFV a serious public health concern. The receptors, cellular factors, and entry pathways used by RVFV and other members of the family Bunyaviridae remain largely uncharacterized. Here we provide evidence that RVFV strain MP-12 uses dynamin-dependent caveola-mediated endocytosis for cell entry. Caveolae are lipid raft domains composed of caveolin (the main structural component), cholesterol, and sphingolipids. Caveola-mediated endocytosis is responsible for the uptake of a wide variety of host ligands, as well as bacteria, bacterial toxins, and a number of viruses. To determine the cellular entry mechanism of RVFV, we used small-molecule inhibitors, RNA interference (RNAi), and dominant negative (DN) protein expression to inhibit the major mammalian cell endocytic pathways. Inhibitors and RNAi specific for macropinocytosis and clathrin-mediated endocytosis had no effect on RVFV infection. In contrast, inhibitors of caveola-mediated endocytosis, and RNAi targeted to caveolin-1 and dynamin, drastically reduced RVFV infection in multiple cell lines. Expression of DN caveolin-1 also reduced RVFV infection significantly, while expression of DN EPS15, a protein required for the assembly of clathrin-coated pits, and DN PAK-1, an obligate mediator of macropinocytosis, had no significant impact on RVFV infection. These results together suggest that the primary mechanism of RVFV MP-12 uptake is dynamin-dependent, caveolin-1-mediated endocytosis.


Asunto(s)
Caveolas/metabolismo , Endocitosis/fisiología , Virus de la Fiebre del Valle del Rift/fisiología , Internalización del Virus , Animales , Western Blotting , Caveolas/fisiología , Caveolinas/genética , Chlorocebus aethiops , Citometría de Flujo , Proteínas Fluorescentes Verdes , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética
20.
PLoS One ; 7(5): e36327, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22574148

RESUMEN

Rift Valley fever virus (RVFV) is an emerging viral zoonosis that is responsible for devastating outbreaks among livestock and is capable of causing potentially fatal disease in humans. Studies have shown that upon infection, certain viruses have the capability of utilizing particular cellular signaling pathways to propagate viral infection. Activation of p53 is important for the DNA damage signaling cascade, initiation of apoptosis, cell cycle arrest and transcriptional regulation of multiple genes. The current study focuses on the role of p53 signaling in RVFV infection and viral replication. These results show an up-regulation of p53 phosphorylation at several serine sites after RVFV MP-12 infection that is highly dependent on the viral protein NSs. qRT-PCR data showed a transcriptional up-regulation of several p53 targeted genes involved in cell cycle and apoptosis regulation following RVFV infection. Cell viability assays demonstrate that loss of p53 results in less RVFV induced cell death. Furthermore, decreased viral titers in p53 null cells indicate that RVFV utilizes p53 to enhance viral production. Collectively, these experiments indicate that the p53 signaling pathway is utilized during RVFV infection to induce cell death and increase viral production.


Asunto(s)
Apoptosis , Virus de la Fiebre del Valle del Rift/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Replicación Viral , Transporte Activo de Núcleo Celular , Línea Celular , Núcleo Celular/metabolismo , Humanos , Fosforilación , Transducción de Señal , Transcripción Genética , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba , Proteínas no Estructurales Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA