Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Viruses ; 10(5)2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29783708

RESUMEN

Alphaviruses are mosquito-transmitted RNA viruses which generally cause acute disease including mild febrile illness, rash, arthralgia, myalgia and more severely, encephalitis. In the mouse, peripheral infection with Semliki Forest virus (SFV) results in encephalitis. With non-virulent strains, infectious virus is detectable in the brain, by standard infectivity assays, for around ten days. As we have shown previously, in severe combined immunodeficient (SCID) mice, infectious virus is detectable for months in the brain. Here we show that in MHC-II-/- mice, with no functional CD4 T-cells, infectious virus is also detectable in the brain for long periods. In contrast, in the brains of CD8-/- mice, virus RNA persists but infectious virus is not detectable. In SCID mice infected with SFV, repeated intraperitoneal administration of anti-SFV immune serum rapidly reduced the titer of infectious virus in the brain to undetectable, however virus RNA persisted. Repeated intraperitoneal passive transfer of immune serum resulted in maintenance of brain virus RNA, with no detectable infectious virus, for several weeks. When passive antibody transfer was stopped, antibody levels declined and infectious virus was again detectable in the brain. In aged immunocompetent mice, previously infected with SFV, immunosuppression of antibody responses many months after initial infection also resulted in renewed ability to detect infectious virus in the brain. In summary, antiviral antibodies control and determine whether infectious virus is detectable in the brain but immune responses cannot clear this infection from the brain. Functional virus RNA capable of generating infectious virus persists and if antibody levels decline, infectious virus is again detectable.


Asunto(s)
Encéfalo/virología , Encefalitis Viral/inmunología , Encefalitis Viral/virología , ARN Viral/inmunología , Virus de los Bosques Semliki/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/uso terapéutico , Encéfalo/inmunología , Línea Celular , Cricetinae , Ciclofosfamida/farmacología , Sueros Inmunes/inmunología , Sueros Inmunes/aislamiento & purificación , Inmunidad Celular/efectos de los fármacos , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos , Ratones SCID , ARN Viral/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/fisiología , Linfocitos T/inmunología , Ensayo de Placa Viral
2.
Artículo en Inglés | MEDLINE | ID: mdl-27993855

RESUMEN

As new pathogenic viruses continue to emerge, it is paramount to have intervention strategies that target a common denominator in these pathogens. The fusion of viral and cellular membranes during viral entry is one such process that is used by many pathogenic viruses, including chikungunya virus, West Nile virus, and influenza virus. Obatoclax, a small-molecule antagonist of the Bcl-2 family of proteins, was previously determined to have activity against influenza A virus and also Sindbis virus. Here, we report it to be active against alphaviruses, like chikungunya virus (50% effective concentration [EC50] = 0.03 µM) and Semliki Forest virus (SFV; EC50 = 0.11 µM). Obatoclax inhibited viral entry processes in an SFV temperature-sensitive mutant entry assay. A neutral red retention assay revealed that obatoclax induces the rapid neutralization of the acidic environment of endolysosomal vesicles and thereby most likely inhibits viral fusion. Characterization of escape mutants revealed that the L369I mutation in the SFV E1 fusion protein was sufficient to confer partial resistance against obatoclax. Other inhibitors that target the Bcl-2 family of antiapoptotic proteins inhibited neither viral entry nor endolysosomal acidification, suggesting that the antiviral mechanism of obatoclax does not depend on its anticancer targets. Obatoclax inhibited the growth of flaviviruses, like Zika virus, West Nile virus, and yellow fever virus, which require low pH for fusion, but not that of pH-independent picornaviruses, like coxsackievirus A9, echovirus 6, and echovirus 7. In conclusion, obatoclax is a novel inhibitor of endosomal acidification that prevents viral fusion and that could be pursued as a potential broad-spectrum antiviral candidate.


Asunto(s)
Antivirales/farmacología , Virus Chikungunya/efectos de los fármacos , Endosomas/efectos de los fármacos , Lisosomas/efectos de los fármacos , Fusión de Membrana/efectos de los fármacos , Pirroles/farmacología , Virus de los Bosques Semliki/efectos de los fármacos , Animales , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/virología , Virus Chikungunya/genética , Virus Chikungunya/crecimiento & desarrollo , Cricetinae , Farmacorresistencia Viral/genética , Endosomas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/virología , Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Indoles , Lisosomas/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutación , Rojo Neutro/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/crecimiento & desarrollo , Virus de la Fiebre Amarilla/efectos de los fármacos , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/crecimiento & desarrollo , Virus Zika/efectos de los fármacos , Virus Zika/genética , Virus Zika/crecimiento & desarrollo
3.
Biochem Pharmacol ; 120: 15-21, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27664855

RESUMEN

Previously, we reported that salicylate-based analogs of bryostatin protect cells from chikungunya virus (CHIKV)-induced cell death. Interestingly, 'capping' the hydroxyl group at C26 of a lead bryostatin analog, a position known to be crucial for binding to and modulation of protein kinase C (PKC), did not abrogate the anti-CHIKV activity of the scaffold, putatively indicating the involvement of a pathway independent of PKC. The work detailed in this study demonstrates that salicylate-derived analog 1 and two capped analogs (2 and 3) are not merely cytoprotective compounds, but act as selective and specific inhibitors of CHIKV replication. Further, a detailed comparative analysis of the effect of the non-capped versus the two capped analogs revealed that compound 1 acts both at early and late stages in the chikungunya virus replication cycle, while the capped analogs only interfere with a later stage process. Co-dosing with the PKC inhibitors sotrastaurin and Gö6976 counteracts the antiviral activity of compound 1 without affecting that of capped analogs 2 and 3, providing further evidence that the latter elicit their anti-CHIKV activity independently of PKC. Remarkably, treatment of CHIKV-infected cells with a combination of compound 1 and a capped analog resulted in a pronounced synergistic antiviral effect. Thus, these salicylate-based bryostatin analogs can inhibit CHIKV replication through a novel, yet still elusive, non-PKC dependent pathway.


Asunto(s)
Antivirales/farmacología , Brioestatinas/farmacología , Virus Chikungunya/efectos de los fármacos , Diseño de Fármacos , Proteína Quinasa C/metabolismo , Proteínas Virales/metabolismo , Acetilación , Animales , Antivirales/agonistas , Antivirales/antagonistas & inhibidores , Antivirales/química , Brioestatinas/agonistas , Brioestatinas/antagonistas & inhibidores , Brioestatinas/química , Carbazoles/química , Carbazoles/farmacología , Línea Celular , Virus Chikungunya/crecimiento & desarrollo , Virus Chikungunya/metabolismo , Chlorocebus aethiops , Sinergismo Farmacológico , Regulación Viral de la Expresión Génica/efectos de los fármacos , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Metilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/química , Proteína Quinasa C/genética , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/química , Pirroles/farmacología , Quinazolinas/química , Quinazolinas/farmacología , Virus de los Bosques Semliki/efectos de los fármacos , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/metabolismo , Virus Sindbis/efectos de los fármacos , Virus Sindbis/crecimiento & desarrollo , Virus Sindbis/metabolismo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/química , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
4.
J Virol ; 87(18): 10295-312, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23864636

RESUMEN

Alphavirus replicase complexes are initially formed at the plasma membrane and are subsequently internalized by endocytosis. During the late stages of infection, viral replication organelles are represented by large cytopathic vacuoles, where replicase complexes bind to membranes of endolysosomal origin. In addition to viral components, these organelles harbor an unknown number of host proteins. In this study, a fraction of modified lysosomes carrying functionally intact replicase complexes was obtained by feeding Semliki Forest virus (SFV)-infected HeLa cells with dextran-covered magnetic nanoparticles and later magnetically isolating the nanoparticle-containing lysosomes. Stable isotope labeling with amino acids in cell culture combined with quantitative proteomics was used to reveal 78 distinct cellular proteins that were at least 2.5-fold more abundant in replicase complex-carrying vesicles than in vesicles obtained from noninfected cells. These host components included the RNA-binding proteins PCBP1, hnRNP M, hnRNP C, and hnRNP K, which were shown to colocalize with the viral replicase. Silencing of hnRNP M and hnRNP C expression enhanced the replication of SFV, Chikungunya virus (CHIKV), and Sindbis virus (SINV). PCBP1 silencing decreased SFV-mediated protein synthesis, whereas hnRNP K silencing increased this synthesis. Notably, the effect of hnRNP K silencing on CHIKV- and SINV-mediated protein synthesis was opposite to that observed for SFV. This study provides a new approach for analyzing the proteome of the virus replication organelle of positive-strand RNA viruses and helps to elucidate how host RNA-binding proteins exert important but diverse functions during positive-strand RNA viral infection.


Asunto(s)
Células Epiteliales/virología , Interacciones Huésped-Patógeno , Lisosomas/virología , Proteoma/análisis , Virus de los Bosques Semliki/fisiología , Replicación Viral , Alphavirus , Virus Chikungunya , Células Epiteliales/química , Células HeLa , Humanos , Marcaje Isotópico , Leporipoxvirus , Lisosomas/química , Magnetismo , Proteómica/métodos , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus Sindbis
5.
J Virol ; 87(4): 2363-6, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23221568

RESUMEN

Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.


Asunto(s)
Glioma/terapia , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/inmunología , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/inmunología , Virus Vaccinia/crecimiento & desarrollo , Virus Vaccinia/inmunología , Animales , Modelos Animales de Enfermedad , Ratones
6.
Cold Spring Harb Protoc ; 2012(8)2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22854567

RESUMEN

The alphaviruses Semliki Forest virus (SFV) and Sindbis virus (SIN) have been used frequently as expression vectors in vitro and in vivo. Usually, these systems consist of replication-deficient vectors that require a helper vector for packaging of recombinant particles. Replication-proficient vectors have also been engineered. Alphaviral vectors can be used as nucleic-acid-based vectors (DNA and RNA) or infectious particles. High-titer viral production is achieved in <2 d. The broad host range of alphaviruses facilitates studies in mammalian and nonmammalian cell lines, primary cells in culture, and in vivo. The strong preference for expression in neuronal cells has made alphaviruses particularly useful in neurobiological studies. Unfortunately, their strong cytotoxic effect on host cells, relatively short-term transient expression patterns, and the reasonably high cost of viral production remain drawbacks. However, novel mutant alphaviruses have shown reduced cytotoxicity and prolonged expression. This protocol describes gene delivery of recombinant alphavirus to hippocampal slice cultures. Organotypic slices are covered by a layer of glial cells that impedes the penetration of viral particles to the neurons. Thus, viral particles should be injected manually into the extracellular space of the tissue.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Hipocampo/virología , Microinyecciones/métodos , Virus de los Bosques Semliki/genética , Virus Sindbis/genética , Animales , Técnicas de Cultivo de Órganos/métodos , Ratas , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus Sindbis/crecimiento & desarrollo , Transducción Genética/métodos
7.
Cold Spring Harb Protoc ; 2012(8)2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22854568

RESUMEN

The alphaviruses Semliki Forest virus (SFV) and Sindbis virus (SIN) have been used frequently as expression vectors in vitro and in vivo. Usually, these systems consist of replication-deficient vectors that require a helper vector for packaging of recombinant particles. Replication-proficient vectors have also been engineered. Alphaviral vectors can be used as nucleic-acid-based vectors (DNA and RNA) or infectious particles. High-titer viral production is achieved in <2 d. The broad host range of alphaviruses facilitates studies in mammalian and nonmammalian cell lines, primary cells in culture, and in vivo. The strong preference for expression in neuronal cells has made alphaviruses particularly useful in neurobiological studies. Unfortunately, their strong cytotoxic effect on host cells, relatively short-term transient expression patterns, and the reasonably high cost of viral production remain drawbacks. However, novel mutant alphaviruses have shown reduced cytotoxicity and prolonged expression. This protocol describes stereotactic microinjection of recombinant alphavirus into rodents. Administration can be performed without any purification or concentration of viral stocks. However, filter-sterilization is recommended to ensure that cell debris or other contaminants are not present.


Asunto(s)
Encéfalo/virología , Técnicas de Transferencia de Gen , Vectores Genéticos , Microinyecciones/métodos , Virus de los Bosques Semliki/genética , Virus Sindbis/genética , Animales , Roedores , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus Sindbis/crecimiento & desarrollo , Transducción Genética/métodos
8.
Cancer Gene Ther ; 19(8): 579-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22722377

RESUMEN

Semliki Forest virus (SFV) vectors are promising tools for cancer gene therapy because they ensure a high level of transgene expression and a rapid and strong cytopathic effect. However, broad tissue tropism and transient expression make it more difficult to develop an optimal cancer treatment strategy. In this study, we have compared the distribution of recombinant SFV particles (recSFV) and naked viral RNA replicon (recRNA) in tumor-free and 4T1 mammary tumor-bearing mice as a consequence of different vector administration strategies. The high potential of SFV recRNA as a biosafe approach for the development of therapeutic treatment was demonstrated. Intravenous (i.v.) inoculation of recRNA provided primary brain targeting in both tumor-free and 4T1 tumor mouse models, but local intratumoral inoculation revealed a high expression level in tumors. Moreover, we observed the predominant tumor targeting of recSFV at a reduced viral dose on i.v. and intraperitoneal (i.p.) virus inoculation, whereas the dose increase led to a broad virus distribution in mice. To prolong transgene expression, we have tested several i.v. and i.p. reinoculation strategies. A detailed evaluation of vector distribution and readministration properties could have an impact on cancer gene therapy clinical trial safety and efficacy.


Asunto(s)
Neoplasias de la Mama , Terapia Genética , Neoplasias Experimentales , Virus de los Bosques Semliki/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos/uso terapéutico , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/genética , Neoplasias Experimentales/terapia , ARN/genética , Replicón/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Transgenes/genética , Virión/genética
9.
J Virol ; 85(6): 2907-17, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21191029

RESUMEN

RNA interference (RNAi) is an important mosquito defense mechanism against arbovirus infection. In this paper we study the processes underlying antiviral RNAi in Aedes albopictus-derived U4.4 mosquito cells infected with Semliki Forest virus (SFV) (Togaviridae; Alphavirus). The production of virus-derived small interfering RNAs (viRNAs) from viral double-stranded RNA (dsRNA) is a key event in this host response. dsRNA could be formed by RNA replication intermediates, by secondary structures in RNA genomes or antigenomes, or by both. Which of these dsRNAs is the substrate for the generation of viRNAs is a fundamental question. Here we used deep sequencing of viRNAs and bioinformatic analysis of RNA secondary structures to gain insights into the characteristics and origins of viRNAs. An asymmetric distribution of SFV-derived viRNAs with notable areas of high-level viRNA production (hot spots) and no or a low frequency of viRNA production (cold spots) along the length of the viral genome with a slight bias toward the production of genome-derived viRNAs over antigenome-derived viRNAs was observed. Bioinformatic analysis suggests that hot spots of viRNA production are rarely but not generally associated with putative secondary structures in the SFV genome, suggesting that most viRNAs are derived from replicative dsRNA. A pattern of viRNAs almost identical to those of A. albopictus cells was observed for Aedes aegypti-derived Aag2 cells, suggesting common mechanisms that lead to viRNA production. Hot-spot viRNAs were found to be significantly less efficient at mediating antiviral RNAi than cold-spot viRNAs, pointing toward a nucleic acid-based viral decoy mechanism to evade the RNAi response.


Asunto(s)
Aedes/fisiología , Aedes/virología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Virus de los Bosques Semliki/crecimiento & desarrollo , Aedes/inmunología , Animales , Línea Celular , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/genética , ARN Viral/genética , ARN Viral/metabolismo , Virus de los Bosques Semliki/genética
10.
Virus Res ; 131(1): 54-64, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17904678

RESUMEN

Semliki Forest virus (SFV, Alphavirus) induce rapid shut down of host cell protein synthesis and apoptotic death of infected vertebrate cells. Data on alphavirus-induced apoptosis are controversial. In this study, the anti-apoptotic bcl-2 gene was placed under the control of duplicated subgenomic promoter or different internal ribosome entry sites (IRES) and expressed using a novel bicistronic SFV vector. The use of IRES containing vectors resulted in high-level Bcl-2 synthesis during the early stages of infection. Nevertheless, in infected BHK-21 cells translational shutdown was almost complete by 6h post-infection, which was similar to infection with appropriate control vectors. These results indicate that very early and high-level bcl-2 expression did not have a protective effect against SFV induced shutdown of host cell translation. No apoptotic cells were detected at those time points for any SFV vectors. Furthermore, Bcl-2 expression did not protect BHK-21 or AT3-neo cells at later time points, and infection of BHK-21 or AT3-neo cells with SFV replicon vectors or with wild-type SFV4 did not lead to release of cytochrome c from mitochondria. Taken together, our data suggest that SFV induced death in BHK-21 or AT3-neo cells is not triggered by the intrinsic pathway of apoptosis.


Asunto(s)
Apoptosis , Genes bcl-2/fisiología , Vectores Genéticos , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/fisiología , Animales , Línea Celular , Células Cultivadas , Regulación Viral de la Expresión Génica , Genes bcl-2/genética , Proteínas Fluorescentes Verdes , Virus de los Bosques Semliki/crecimiento & desarrollo
11.
J Virol Methods ; 148(1-2): 265-70, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18054090

RESUMEN

Semliki Forest virus (SFV, genus Alphavirus) has a broad host range, high efficiency of viral protein expression, and the ability to stimulate an immune response. These properties have made SFV an attractive tool for development of expression vectors, and plasmid clones containing cDNA of the SFV genome often are used. However, instability of these plasmids resulting from cryptic expression of SFV envelope proteins in Escherichia coli represents a problem both for the development of SFV-based vectors and for SFV research. In this study, an infectious plasmid of SFV, pCMV-SFV4, was constructed; its toxic effect was eliminated by intron insertion in the capsid protein encoding region. When transfected into mammalian cells, the plasmid clone was highly infectious and produced virus with properties identical to those of wild-type SFV. The inserted intron was efficiently and properly removed from the RNA genome of SFV. Therefore, this novel and stabilized infectious SFV plasmid represents a superior tool for basic studies of SFV as well as for biotechnological applications.


Asunto(s)
Vectores Genéticos , Intrones , Mutagénesis Insercional , Plásmidos , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/genética , Animales , Proteínas de la Cápside/genética , Línea Celular , Cricetinae , ADN Complementario , ADN Viral/genética , Escherichia coli/genética , Inestabilidad Genómica , Mesocricetus
12.
J Gen Virol ; 88(Pt 11): 3018-3026, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17947525

RESUMEN

Alpha- and flaviviruses contain class II fusion proteins, which form ion-permeable pores in the target membrane during virus entry. The pores generated during entry of the alphavirus Semliki Forest virus have been shown previously to be blocked by lanthanide ions. Here, analyses of the influence of rare earth ions on the entry of the flaviviruses West Nile virus and Uganda S virus revealed an unexpected effect of lanthanide ions. The results showed that a 30 s treatment of cells with an appropriate lanthanide ion changed the cellular chemistry into a state in which the cells no longer supported the multiplication of flaviviruses. This change occurred in cells treated before, during or after infection, did not inhibit multiplication of Semliki Forest virus and did not interfere with host-cell multiplication. The change was generated in vertebrate and insect cells, and was elicited in the presence of actinomycin D. In vertebrate cells, the change was elicited specifically by La(3+), Ce(3+), Pr(3+) and Nd(3+). In insect cells, additional lanthanide ions had this activity. Further analyses showed that lanthanide ion treatment blocked the ability of the host cell to support the replication of flavivirus RNA. These results open two areas of research: the study of molecular alterations induced by lanthanide ion treatment in uninfected cells and the analysis of the resulting modifications of the flavivirus RNA replicase complex. The findings possibly open the way for the development of a general chemotherapy against flavivirus diseases such as Dengue fever, Japanese encephalitis, West Nile fever and yellow fever.


Asunto(s)
Antivirales/farmacología , Flavivirus/crecimiento & desarrollo , Elementos de la Serie de los Lantanoides/farmacología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Chlorocebus aethiops , Cricetinae , Culicidae , ARN Viral/biosíntesis , Virus de los Bosques Semliki/crecimiento & desarrollo , Ensayo de Placa Viral
13.
J Virol ; 81(24): 13631-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17913808

RESUMEN

The budding reactions of a number of enveloped viruses use the cellular machinery involved in the formation of the luminal vesicles of endosomal multivesicular bodies (MVB). Budding of these viruses is dependent on the presence of specific late-domain motifs in membrane-associated viral proteins. Such budding reactions usually involve ubiquitin and are blocked by expression of an ATPase-deficient form of VPS4, a cellular AAA+ ATPase believed to be required late in the MVB pathway for the disassembly/release of the MVB machinery. Here we examined the role of the MVB pathway in the budding of the late-domain-containing rhabdovirus vesicular stomatitis virus (VSV) and the alphavirus Semliki Forest virus (SFV). We tested early and late steps in the MVB pathway by depleting ubiquitin with the proteasome inhibitor MG-132 and by using cell lines inducibly expressing VPS4A or VPS4B protein. As previously shown, VSV budding was strongly dependent on ubiquitin. In contrast to the findings of previous studies with VPS4A, expression of ATPase-deficient mutants of either VPS4A or VPS4B inhibited VSV budding. Inhibition by VPS4 required the presence of the PPPY late domain on the VSV matrix protein and resulted in the accumulation of nonreleased VSV particles at the plasma membrane. In contrast, SFV budding was independent of both ubiquitin and the activity of VPS4, perhaps reflecting the important role of the highly organized envelope protein lattice during alphavirus budding.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Endosomas/ultraestructura , Virus de los Bosques Semliki/crecimiento & desarrollo , Ubiquitina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Vesiculovirus/crecimiento & desarrollo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Alphavirus/genética , Alphavirus/crecimiento & desarrollo , Alphavirus/metabolismo , Alphavirus/patogenicidad , Animales , Línea Celular/ultraestructura , Línea Celular/virología , Cricetinae , Complejos de Clasificación Endosomal Requeridos para el Transporte , Endosomas/metabolismo , Humanos , Leupeptinas/farmacología , Microscopía Electrónica , Rhabdoviridae/genética , Rhabdoviridae/crecimiento & desarrollo , Rhabdoviridae/metabolismo , Rhabdoviridae/patogenicidad , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Virus de los Bosques Semliki/patogenicidad , Ubiquitina/efectos de los fármacos , Ubiquitina/genética , ATPasas de Translocación de Protón Vacuolares , Proteínas de Transporte Vesicular/genética , Vesiculovirus/genética , Vesiculovirus/metabolismo , Vesiculovirus/patogenicidad , Ensamble de Virus
14.
J Virol ; 81(10): 4991-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17344298

RESUMEN

La Crosse virus (LACV) is a mosquito-transmitted member of the Bunyaviridae family that causes severe encephalitis in children. For the LACV nonstructural protein NSs, previous overexpression studies with mammalian cells had suggested two different functions, namely induction of apoptosis and inhibition of RNA interference (RNAi). Here, we demonstrate that mosquito cells persistently infected with LACV do not undergo apoptosis and mount a specific RNAi response. Recombinant viruses that either express (rLACV) or lack (rLACVdelNSs) the NSs gene similarly persisted and were prone to the RNAi-mediated resistance to superinfection. Furthermore, in mosquito cells overexpressed LACV NSs was unable to inhibit RNAi against Semliki Forest virus. In mammalian cells, however, the rLACVdelNSs mutant virus strongly activated the antiviral type I interferon (IFN) system, whereas rLACV as well as overexpressed NSs suppressed IFN induction. Consequently, rLACVdelNSs was attenuated in IFN-competent mouse embryo fibroblasts and animals but not in systems lacking the type I IFN receptor. In situ analyses of mouse brains demonstrated that wild-type and mutant LACV mainly infect neuronal cells and that NSs is able to suppress IFN induction in the central nervous system. Thus, our data suggest little relevance of the NSs-induced apoptosis or RNAi inhibition for growth or pathogenesis of LACV in the mammalian host and indicate that NSs has no function in the insect vector. Since deletion of the viral NSs gene can be fully complemented by inactivation of the host's IFN system, we propose that the major biological function of NSs is suppression of the mammalian innate immune response.


Asunto(s)
Interferón Tipo I/antagonistas & inhibidores , Virus La Crosse/inmunología , Interferencia de ARN , Proteínas no Estructurales Virales/fisiología , Animales , Apoptosis , Encéfalo/patología , Encéfalo/virología , Línea Celular , Cricetinae , Culicidae , Modelos Animales de Enfermedad , Encefalitis de California/virología , Humanos , Inmunohistoquímica , Virus La Crosse/patogenicidad , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Neuronas/virología , Virus de los Bosques Semliki/crecimiento & desarrollo
15.
Antiviral Res ; 75(2): 152-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17395278

RESUMEN

Several viruses have been demonstrated to be the etiologic agent in chronic progressive diseases, associated with persistence; however, major questions concerning the pathogenic mechanisms of viral persistence are still unanswered. With the aim of identifying host cellular proteins that may play a role in viral replication, we established long-term persistently infected human glioblastoma cell lines with mutant measles virus (MV) and analyzed the host proteins by two-dimensional gel electrophoresis (2-DE) with mass spectrometry. We observed significant down-modulation in the expression of mitochondrial short chain enoyl-CoA hydratase (ECHS), which catalyzes the beta-oxidation pathway of fatty acid. Knockdown of this gene by a short interference RNA (siRNA) apparently impaired wild-type MV replication and the cytopathic effects (CPEs) of MV were significantly reduced in siRNA-transfected cells. These findings will shed light upon a new important notion for the interaction between virus replication and lipid metabolism in host cells and might provide a new strategy for virus control.


Asunto(s)
Ácido Graso Sintasas/metabolismo , Virus del Sarampión/crecimiento & desarrollo , Proteínas Mitocondriales/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Replicación Viral , Antivirales/farmacología , Western Blotting , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Electroforesis en Gel Bidimensional , Inhibidores Enzimáticos/farmacología , Compuestos Epoxi/farmacología , Ácido Graso Sintasas/genética , Glioblastoma/enzimología , Glioblastoma/patología , Glioblastoma/virología , Proteínas de Choque Térmico/metabolismo , Humanos , Interferón beta/farmacología , Virus del Sarampión/efectos de los fármacos , Virus del Sarampión/genética , Proteínas Mitocondriales/genética , Chaperonas Moleculares/metabolismo , Mutación , NADH NADPH Oxidorreductasas/genética , ARN Interferente Pequeño/genética , Virus de los Bosques Semliki/efectos de los fármacos , Virus de los Bosques Semliki/crecimiento & desarrollo , Transfección , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo
16.
J Gen Virol ; 88(Pt 4): 1225-1230, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17374766

RESUMEN

Alphavirus-based vector and replicon systems have been extensively used experimentally and are likely to be used in human and animal medicine. Whilst marker genes can be inserted easily under the control of a duplicated subgenomic promoter, these constructs are often genetically unstable. Here, a novel alphavirus construct is described in which an enhanced green fluorescent protein (EGFP) marker gene is inserted into the virus replicase open reading frame between nsP3 and nsP4, flanked by nsP2 protease-recognition sites. This construct has correct processing of the replicase polyprotein, produces viable virus and expresses detectable EGFP fluorescence upon infection of cultured cells and cells of the mouse brain. In comparison to parental virus, the marker virus has an approximately 1 h delay in virus RNA and infectious virus production. Passage of the marker virus in vitro and in vivo demonstrates good genetic stability. Insertion of different markers into this novel construct has potential for various applications.


Asunto(s)
Proteínas Fluorescentes Verdes/biosíntesis , ARN Polimerasa Dependiente del ARN/genética , Virus de los Bosques Semliki/genética , Proteínas no Estructurales Virales/genética , Animales , Encéfalo/virología , Línea Celular , Cricetinae , Genes Reporteros , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Ratones , ARN Polimerasa Dependiente del ARN/fisiología , Virus de los Bosques Semliki/crecimiento & desarrollo , Coloración y Etiquetado , Proteínas no Estructurales Virales/fisiología , Replicación Viral
17.
Virology ; 337(2): 344-52, 2005 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15913697

RESUMEN

Semliki Forest virus (SFV) membrane fusion is mediated by the viral E1 protein at acidic pH and regulated by the dimeric interaction of E1 with the E2 membrane protein. During low pH-triggered fusion, the E2/E1 heterodimer dissociates, freeing E1 to drive membrane fusion. E2 is synthesized as a precursor, p62, which is processed to mature E2 by the cellular protease furin. Both the dissociation of the p62/E1 dimer and the fusion reaction of p62 virus have a more acidic pH threshold than that of the mature E2 virus. We have previously isolated SFV mutations that allow virus growth in furin-deficient cells. Here we have used such pci mutations to compare the interactions of the p62/E1 and E2/E1 dimers. Our data suggest that there is an important p62/E1 dimer interaction site identified by an E2 R250G mutation and that this interaction is maintained after processing to the mature E2 protein.


Asunto(s)
Virus de los Bosques Semliki/fisiología , Proteínas del Envoltorio Viral/metabolismo , Infecciones por Alphavirus/fisiopatología , Animales , Línea Celular , Cricetinae , Dimerización , Electroporación , Concentración de Iones de Hidrógeno , Riñón , Cinética , Fusión de Membrana , Mutación , Péptido Hidrolasas/metabolismo , ARN/genética , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Proteínas del Envoltorio Viral/genética
18.
Virology ; 327(2): 287-96, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15351216

RESUMEN

The enveloped alphavirus Semliki Forest virus (SFV) infects cells via a low pH-triggered membrane fusion reaction mediated by the E1 protein. E1's fusion activity is regulated by its heterodimeric interaction with a companion membrane protein E2. Mature E2 protein is generated by furin processing of the precursor p62. Processing destabilizes the heterodimer, allowing dissociation at acidic pH, E1 conformational changes, and membrane fusion. We used a furin-deficient cell line, FD11, to select for SFV mutants that show increased growth in the absence of p62 processing. We isolated and characterized 7 such pci mutants (p62 cleavage independent), which retained the parental furin cleavage site but showed significant increases in their ability to carry out membrane fusion in the p62 form. Sequence analysis of the pci mutants identified mutations primarily on the E2 protein, and suggested sites important in the interaction of p62 with E1 and the regulation of fusion.


Asunto(s)
Regulación Viral de la Expresión Génica , Fusión de Membrana , Mutación , Precursores de Proteínas/metabolismo , Virus de los Bosques Semliki/patogenicidad , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Cricetinae , Dimerización , Furina/metabolismo , Precursores de Proteínas/química , Precursores de Proteínas/genética , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/crecimiento & desarrollo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética
19.
J Neurovirol ; 9(1): 1-15, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12587064

RESUMEN

Semliki Forest virus (SFV), an enveloped alphavirus of the family Togaviridae, infects a wide range of mammalian host cells. Most strains are neurotropic but differ in virulence. The authors took advantage of the nonpathogenic properties of SFV strain A7(74), cloned recently in their laboratory, and constructed a replication-proficient expression vector to target the central nervous system (CNS) for heterologous gene expression. The vector, termed VA7, was engineered to drive expression of foreign inserts through a second subgenomic promoter inserted in the viral 3' nontranslated region (NTR). Infectious virus was obtained by in vitro transcription and transfection into BHK cells, and was shown to direct synthesis of heterologous proteins in several mammalian cell lines. Although novel expression vehicle is not applicable for targeting specific cell populations within the CNS in its present form, in cultured rat hippocampal slices, VA7 encoding enhanced green fluorescent protein (EGFP) efficiently transduced pyramidal cells, interneurons, and glial cells. With prolonged time post infection, the number of EGFP-expressing neurons in hippocampal slices increased. Mice infected intraperitoneally with the recombinant virus remained completely asymptomatic but showed CNS expression of EGFP as evidenced by immunohistochemistry. SFV A7(74) is a nonintegrating virus, which gives rise to a randomly distributed, patchy infection of the adult CNS that is cleared within 10 days. With the advantage of noninvasive administration, the expression vector described in this work is thus applicable for short-term gene expression in the CNS.


Asunto(s)
Vectores Genéticos , Células Piramidales/virología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/patogenicidad , Animales , Células CHO , Cricetinae , Células Epiteliales/citología , Células Epiteliales/virología , Femenino , Regulación Viral de la Expresión Génica , Glioma , Gliosarcoma , Proteínas Fluorescentes Verdes , Hipocampo/citología , Hipocampo/virología , Indicadores y Reactivos/metabolismo , Riñón/citología , Proteínas Luminiscentes/genética , Melanoma , Ratones , Ratones Endogámicos BALB C , Neuroblastoma , Células Piramidales/citología , Ratas , Virus de los Bosques Semliki/crecimiento & desarrollo , Transducción Genética , Células Tumorales Cultivadas , Virulencia , Replicación Viral
20.
J Neurovirol ; 9(1): 16-28, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12587065

RESUMEN

In central nervous system (CNS) tissue preparations, wild-type Semliki Forest virus (SFV) mainly infects neurons, and in vivo it causes lethal encephalitis in neonatal and adult rodents. The SFV strain A7(74), by contrast, is avirulent in adult rodents, triggering only limited CNS infection. To examine A7(74) infection in hippocampal tissue, the authors constructed a replicon, termed SFV(A774nsP)-GFP, expressing green fluorescent protein. The results were compared to replication-proficient recombinant A7(74) encoding GFP, named VA7-EGFP. As nonstructural gene mutations can confer temperature sensitivity, the authors also tested whether infection was temperature-dependent. Indeed, at 31 degrees C both viral recombinants transduced significantly more baby hamster kidney cells than at 37 degrees C. When rat hippocampal slices and dissociated cells were incubated at 37 degrees C, SFV(A774nsP)-GFP transduced glial cells but virtually no neurons-the opposite of conventional SFV. For VA7-EGFP at 37 degrees C, the preferred GFP-positive cells in hippocampal slices were also non-neuronal cells. At 31 degrees C, however, a more wild-type phenotype was found, with 33% and 94% of the GFP-positive cells being neurons for SFV(A774nsP)-GFP in slices and dissociated cells, respectively, and 94% neurons for VA7-EGFP in slices. Immunochemical and electrophysiological analyses confirmed that at 37 degrees C virtually all cells transduced by SFV(A774nsP)-GFP in slices were astrocytes, while at 31 degrees C they also contained neurons. These results show that in addition to the developmental age, the temperature determines which cell type becomes infected by A7(74). Our data suggest that A7(74) is avirulent in adult animals because it does not readily replicate in mature neurons at body temperature, whereas it still does so at lower temperatures.


Asunto(s)
Astrocitos/virología , Vectores Genéticos , Hipocampo/virología , Células Piramidales/virología , Virus de los Bosques Semliki/genética , Transducción Genética , Animales , Astrocitos/citología , Células Cultivadas , Cricetinae , Genoma Viral , Proteínas Fluorescentes Verdes , Hipocampo/citología , Indicadores y Reactivos/metabolismo , Interneuronas/citología , Interneuronas/virología , Riñón/citología , Proteínas Luminiscentes/genética , Potenciales de la Membrana , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp , Fenotipo , Células Piramidales/citología , Células Piramidales/fisiología , Virus de los Bosques Semliki/crecimiento & desarrollo , Virus de los Bosques Semliki/patogenicidad , Temperatura , Virulencia , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA