Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
PLoS Pathog ; 18(12): e1011022, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36480574

RESUMEN

Rabies virus (RABV) transcription and replication take place within viral factories having liquid properties, called Negri bodies (NBs), that are formed by liquid-liquid phase separation (LLPS). The co-expression of RABV nucleoprotein (N) and phosphoprotein (P) in mammalian cells is sufficient to induce the formation of cytoplasmic biocondensates having properties that are like those of NBs. This cellular minimal system was previously used to identify P domains that are essential for biocondensates formation. Here, we constructed fluorescent versions of N and analyzed by FRAP their dynamics inside the biocondensates formed in this minimal system as well as in NBs of RABV-infected cells using FRAP. The behavior of N appears to be different of P as there was no fluorescence recovery of N proteins after photobleaching. We also identified arginine residues as well as two exposed loops of N involved in condensates formation. Corresponding N mutants exhibited distinct phenotypes in infected cells ranging from co-localization with NBs to exclusion from them associated with a dominant-negative effect on infection. We also demonstrated that in vitro, in crowded environments, purified P as well as purified N0-P complex (in which N is RNA-free) form liquid condensates. We identified P domains required for LLPS in this acellular system. P condensates were shown to associate with liposomes, concentrate RNA, and undergo a liquid-gel transition upon ageing. Conversely, N0-P droplets were disrupted upon incubation with RNA. Taken together, our data emphasize the central role of P in NBs formation and reveal some physicochemical features of P and N0-P droplets relevant for explaining NBs properties such as their envelopment by cellular membranes at late stages of infection and nucleocapsids ejections from the viral factories.


Asunto(s)
Virus de la Rabia , Rabia , Animales , Virus de la Rabia/genética , Virus de la Rabia/metabolismo , Nucleoproteínas/genética , Rabia/metabolismo , Nucleocápside/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Replicación Viral , Mamíferos
2.
J Virol ; 94(6)2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31852780

RESUMEN

The phosphoprotein (P) of the nonsegmented negative-sense RNA viruses is a multimeric modular protein that is essential for RNA transcription and replication. Despite great variability in length and sequence, the architecture of this protein is conserved among the different viral families, with a long N-terminal intrinsically disordered region comprising a nucleoprotein chaperone module, a central multimerization domain (PMD), connected by a disordered linker to a C-terminal nucleocapsid-binding domain. The P protein of vesicular stomatitis virus (VSV) forms dimers, and here we investigate the importance of its dimerization domain, PMD, for viral gene expression and virus growth. A truncated P protein lacking the central dimerization domain (PΔMD) loses its ability to form dimers both in vitro and in a yeast two-hybrid system but conserves its ability to bind N. In a minireplicon system, the truncated monomeric protein performs almost as well as the full-length dimeric protein, while a recombinant virus harboring the same truncation in the P protein has been rescued and follows replication kinetics similar to those seen with the wild-type virus, showing that the dimerization domain of P is dispensable for viral gene expression and virus replication in cell culture. Because RNA viruses have high mutation rates, it is unlikely that a structured domain such as a VSV dimerization domain would persist in the absence of a function(s), but our work indicates that it is not required for the functioning of the RNA polymerase machinery or for the assembly of new viruses.IMPORTANCE The phosphoprotein (P) is an essential and conserved component of all nonsegmented negative-sense RNA viruses, including some major human pathogens (e.g., rabies virus, measles virus, respiratory syncytial virus [RSV], Ebola virus, and Nipah virus). P is a modular protein with intrinsically disordered regions and folded domains that plays specific and similar roles in the replication of the different viruses and, in some cases, hijacks cell components to the advantage of the virus and is involved in immune evasion. All P proteins are multimeric, but the role of this multimerization is still unclear. Here, we demonstrate that the dimerization domain of VSV P is dispensable for the expression of virally encoded proteins and for virus growth in cell culture. This provides new insights into and raises questions about the functioning of the RNA-synthesizing machinery of the nonsegmented negative-sense RNA viruses.


Asunto(s)
Fosfoproteínas/química , Dominios Proteicos , Multimerización de Proteína , Virus de la Estomatitis Vesicular Indiana/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Dimerización , Modelos Moleculares , Nucleocápside/metabolismo , Nucleoproteínas/metabolismo , Fosfoproteínas/genética , Unión Proteica , Conformación Proteica , Multimerización de Proteína/genética , ARN Viral/genética , Alineación de Secuencia , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Replicación Viral
3.
Adv Exp Med Biol ; 1215: 111-127, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31317498

RESUMEN

Replication and assembly of many viruses occur in viral factories which are specialized intracellular compartments formed during viral infection. For rabies virus, those viral factories are called Negri bodies (NBs). NBs are cytoplasmic inclusion bodies in which viral RNAs (mRNAs as well as genomic and antigenomic RNAs) are synthesized. NBs are spherical, they can fuse together, and can reversibly deform when encountering a physical barrier. All these characteristics are similar to those of eukaryotic membrane-less liquid organelles which contribute to the compartmentalization of the cell interior. Indeed, the liquid nature of NBs has been confirmed by FRAP experiments. The co-expression of rabies virus nucleoprotein N and phosphoprotein P is sufficient to induce the formation of cytoplasmic inclusions recapitulating NBs properties. Remarkably, P and N have features similar to those of cellular proteins involved in liquid organelles formation: N is an RNA-binding protein and P contains intrinsically disordered domains. An overview of the literature indicates that formation of liquid viral factories by phase separation is probably common among Mononegavirales. This allows specific recruitment and concentration of viral proteins. Finally, as virus-associated molecular patterns recognized by cellular sensors of RNA virus replication are probably essentially present in the viral factory, there should be a subtle interplay (which remains to be characterized) between those liquid structures and the cellular proteins which trigger the innate immune response.


Asunto(s)
Cuerpos de Inclusión Viral , Virus de la Rabia , Cuerpos de Inclusión Viral/química , Cuerpos de Inclusión Viral/metabolismo , ARN Viral/biosíntesis , Virus de la Rabia/fisiología , Proteínas Virales/metabolismo , Replicación Viral
4.
PLoS Pathog ; 12(10): e1005942, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27749929

RESUMEN

Stress granules (SGs) are membrane-less dynamic structures consisting of mRNA and protein aggregates that form rapidly in response to a wide range of environmental cellular stresses and viral infections. They act as storage sites for translationally silenced mRNAs under stress conditions. During viral infection, SG formation results in the modulation of innate antiviral immune responses, and several viruses have the ability to either promote or prevent SG assembly. Here, we show that rabies virus (RABV) induces SG formation in infected cells, as revealed by the detection of SG-marker proteins Ras GTPase-activating protein-binding protein 1 (G3BP1), T-cell intracellular antigen 1 (TIA-1) and poly(A)-binding protein (PABP) in the RNA granules formed during viral infection. As shown by live cell imaging, RABV-induced SGs are highly dynamic structures that increase in number, grow in size by fusion events, and undergo assembly/disassembly cycles. Some SGs localize in close proximity to cytoplasmic viral factories, known as Negri bodies (NBs). Three dimensional reconstructions reveal that both structures remain distinct even when they are in close contact. In addition, viral mRNAs synthesized in NBs accumulate in the SGs during viral infection, revealing material exchange between both compartments. Although RABV-induced SG formation is not affected in MEFs lacking TIA-1, TIA-1 depletion promotes viral translation which results in an increase of viral replication indicating that TIA-1 has an antiviral effect. Inhibition of PKR expression significantly prevents RABV-SG formation and favors viral replication by increasing viral translation. This is correlated with a drastic inhibition of IFN-B gene expression indicating that SGs likely mediate an antiviral response which is however not sufficient to fully counteract RABV infection.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Cuerpos de Inclusión Viral/virología , Virus de la Rabia , Rabia/virología , Replicación Viral/fisiología , Animales , Western Blotting , Línea Celular , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Inmunidad Innata , Hibridación Fluorescente in Situ , Microscopía Confocal , Rabia/inmunología , Virus de la Rabia/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
J Virol ; 90(14): 6598-6610, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27170750

RESUMEN

UNLABELLED: Multiple cellular pathways are regulated by small ubiquitin-like modifier (SUMO) modification, including ubiquitin-mediated proteolysis, signal transduction, innate immunity, and antiviral defense. In the study described in this report, we investigated the effects of SUMO on the replication of two members of the Rhabdoviridae family, vesicular stomatitis virus (VSV) and rabies virus (RABV). We show that stable expression of SUMO in human cells confers resistance to VSV infection in an interferon-independent manner. We demonstrate that SUMO expression did not alter VSV entry but blocked primary mRNA synthesis, leading to a reduction of viral protein synthesis and viral production, thus protecting cells from VSV-induced cell lysis. MxA is known to inhibit VSV primary transcription. Interestingly, we found that the MxA protein was highly stabilized in SUMO-expressing cells. Furthermore, extracts from cells stably expressing SUMO exhibited an increase in MxA oligomers, suggesting that SUMO plays a role in protecting MxA from degradation, thus providing a stable intracellular pool of MxA available to combat invading viruses. Importantly, MxA depletion in SUMO-expressing cells abrogated the anti-VSV effect of SUMO. Furthermore, SUMO expression resulted in interferon-regulatory factor 3 (IRF3) SUMOylation, subsequently decreasing RABV-induced IRF3 phosphorylation and interferon synthesis. As expected, this rendered SUMO-expressing cells more sensitive to RABV infection, even though MxA was stabilized in SUMO-expressing cells, since its expression did not confer resistance to RABV. Our findings demonstrate opposing effects of SUMO expression on two viruses of the same family, intrinsically inhibiting VSV infection through MxA stabilization while enhancing RABV infection by decreasing IFN induction. IMPORTANCE: We report that SUMO expression reduces interferon synthesis upon RABV or VSV infection. Therefore, SUMO renders cells more sensitive to RABV but unexpectedly renders cells resistant to VSV by blocking primary mRNA synthesis. Unlike the interferon-mediated innate immune response, intrinsic antiviral resistance is mediated by constitutively expressed restriction factors. Among the various anti-VSV restriction factors, only MxA is known to inhibit VSV primary transcription, and we show here that its expression does not alter RABV infection. Interestingly, MxA depletion abolished the inhibition of VSV by SUMO, demonstrating that MxA mediates SUMO-induced intrinsic VSV resistance. Furthermore, MxA oligomerization is known to be critical for its protein stability, and we show that higher levels of oligomers were formed in cells expressing SUMO than in wild-type cells, suggesting that SUMO may play a role in protecting MxA from degradation, providing a stable intracellular pool of MxA able to protect cells from viral infection.


Asunto(s)
Interferón-alfa/farmacología , Proteínas de Resistencia a Mixovirus/farmacología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/farmacología , Estomatitis Vesicular/prevención & control , Virus de la Estomatitis Vesicular Indiana/fisiología , Antivirales/farmacología , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/virología , Células HeLa , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Procesamiento Proteico-Postraduccional , Rabia/metabolismo , Rabia/prevención & control , Rabia/virología , Virus de la Rabia/fisiología , Células Tumorales Cultivadas , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología
6.
J Virol ; 89(3): 1640-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25410852

RESUMEN

UNLABELLED: The rabies virus (RABV) phosphoprotein P is a multifunctional protein: it plays an essential role in viral transcription and replication, and in addition, RABV P has been identified as an interferon antagonist. Here, a yeast two-hybrid screen revealed that RABV P interacts with the focal adhesion kinase (FAK). The binding involved the 106-to-131 domain, corresponding to the dimerization domain of P and the C-terminal domain of FAK containing the proline-rich domains PRR2 and PRR3. The P-FAK interaction was confirmed in infected cells by coimmunoprecipitation and colocalization of FAK with P in Negri bodies. By alanine scanning, we identified a single mutation in the P protein that abolishes this interaction. The mutant virus containing a substitution of Ala for Arg in position 109 in P (P.R109A), which did not interact with FAK, is affected at a posttranscriptional step involving protein synthesis and viral RNA replication. Furthermore, FAK depletion inhibited viral protein expression in infected cells. This provides the first evidence of an interaction of RABV with FAK that positively regulates infection. IMPORTANCE: Rabies virus exhibits a small genome that encodes a limited number of viral proteins. To maintain efficient virus replication, some of them are multifunctional, such as the phosphoprotein P. We and others have shown that P establishes complex networks of interactions with host cell components. These interactions have revealed much about the role of P and about host-pathogen interactions in infected cells. Here, we identified another cellular partner of P, the focal adhesion kinase (FAK). Our data shed light on the implication of FAK in RABV infection and provide evidence that P-FAK interaction has a proviral function.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Interacciones Huésped-Patógeno , Fosfoproteínas/metabolismo , Mapeo de Interacción de Proteínas , Virus de la Rabia/fisiología , Proteínas Estructurales Virales/metabolismo , Replicación Viral , Animales , Línea Celular , Análisis Mutacional de ADN , Humanos , Inmunoprecipitación , Cuerpos de Inclusión Viral/química , Cuerpos de Inclusión Viral/virología , Microscopía Confocal , Chaperonas Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Técnicas del Sistema de Dos Híbridos
7.
PLoS Pathog ; 10(2): e1003975, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586174

RESUMEN

PML/TRIM19, the organizer of nuclear bodies (NBs), has been implicated in the antiviral response to diverse RNA and DNA viruses. Several PML isoforms generated from a single PML gene by alternative splicing, share the same N-terminal region containing the RBCC/tripartite motif but differ in their C-terminal sequences. Recent studies of all the PML isoforms reveal the specific functions of each. The knockout of PML renders mice more sensitive to vesicular stomatitis virus (VSV). Here we report that among PML isoforms (PMLI to PMLVIIb), only PMLIII and PMLIV confer resistance to VSV. Unlike PMLIII, whose anti-VSV activity is IFN-independent, PMLIV can act at two stages: it confers viral resistance directly in an IFN-independent manner and also specifically enhances IFN-ß production via a higher activation of IRF3, thus protecting yet uninfected cells from oncoming infection. PMLIV SUMOylation is required for both activities. This demonstrates for the first time that PMLIV is implicated in innate immune response through enhanced IFN-ß synthesis. Depletion of IRF3 further demonstrates the dual activity of PMLIV, since it abrogated PMLIV-induced IFN synthesis but not PMLIV-induced inhibition of viral proteins. Mechanistically, PMLIV enhances IFN-ß synthesis by regulating the cellular distribution of Pin1 (peptidyl-prolyl cis/trans isomerase), inducing its recruitment to PML NBs where both proteins colocalize. The interaction of SUMOylated PMLIV with endogenous Pin1 and its recruitment within PML NBs prevents the degradation of activated IRF3, and thus potentiates IRF3-dependent production of IFN-ß. Whereas the intrinsic antiviral activity of PMLIV is specific to VSV, its effect on IFN-ß synthesis is much broader, since it affects a key actor of innate immune pathways. Our results show that, in addition to its intrinsic anti-VSV activity, PMLIV positively regulates IFN-ß synthesis in response to different inducers, thus adding PML/TRIM19 to the growing list of TRIM proteins implicated in both intrinsic and innate immunity.


Asunto(s)
Inmunidad Innata/inmunología , Proteínas Nucleares/inmunología , Infecciones por Rhabdoviridae/inmunología , Transducción de Señal/inmunología , Factores de Transcripción/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunoprecipitación , Interferón beta/biosíntesis , Interferón beta/inmunología , Ratones , Ratones Noqueados , Proteína de la Leucemia Promielocítica , Isoformas de Proteínas , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Vesiculovirus
8.
J Infect Dis ; 209(11): 1744-53, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24367042

RESUMEN

BACKGROUND: Rabies virus (RABV) causes rabies disease resulting in >55,000 human deaths/year. The multifunctional RABV P-protein has essential roles in genome replication, and forms interactions with cellular STAT proteins that are thought to underlie viral antagonism of interferon-dependent immunity. However, the molecular details of P-protein-STAT interaction, and its importance to disease are unresolved. METHODS: Studies were performed using sequence/structure analysis, mutagenesis, immunoprecipitation, luciferase and qRT-PCR-based signaling assays, confocal microscopy and reverse genetics/in vivo infection. RESULTS: We identified a hydrophobic pocket of the P-protein C-terminal domain as critical to STAT-binding/antagonism. This interface was found to be functionally and spatially independent of the region responsible for N-protein interaction, which is critical to genome replication. Based on these findings, we generated the first mutant RABV lacking STAT-association. Growth of the virus in vitro was unimpaired, but it lacked STAT-antagonist function and was highly sensitive to interferon. Importantly, growth of the virus was strongly attenuated in brains of infected mice, producing no major neurological symptoms, compared with the invariably lethal wild-type virus. CONCLUSIONS: These data represent direct evidence that P-protein-STAT interaction is critical to rabies, and provide novel insights into the mechanism by which RABV coordinates distinct functions in interferon antagonism and replication.


Asunto(s)
Fosfoproteínas/metabolismo , Virus de la Rabia/metabolismo , Rabia/virología , Factores de Transcripción STAT/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Línea Celular , Femenino , Regulación de la Expresión Génica/inmunología , Genoma Viral , Humanos , Interferones/genética , Interferones/metabolismo , Ratones , Modelos Moleculares , Chaperonas Moleculares , Mutación , Unión Proteica , Conformación Proteica , Factores de Transcripción STAT/genética , Técnicas del Sistema de Dos Híbridos , Replicación Viral
9.
J Virol ; 87(14): 8261-5, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23698294

RESUMEN

Immune evasion by rabies virus depends on targeting of the signal transducers and activator of transcription 1 (STAT1) and STAT2 proteins by the viral interferon antagonist P protein, but targeting of other STAT proteins has not been investigated. Here, we find that P protein associates with activated STAT3 and inhibits STAT3 nuclear accumulation and Gp130-dependent signaling. This is the first report of STAT3 targeting by the interferon antagonist of a virus other than a paramyxovirus, indicating that STAT3 antagonism is important to a range of human-pathogenic viruses.


Asunto(s)
Receptor gp130 de Citocinas/metabolismo , Evasión Inmune/genética , Interferones/antagonistas & inhibidores , Fosfoproteínas/farmacología , Virus de la Rabia/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Estructurales Virales/farmacología , Animales , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/metabolismo , Luciferasas , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Chaperonas Moleculares , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Virus de la Rabia/metabolismo , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/metabolismo , Proteína Fluorescente Roja
10.
J Biol Chem ; 287(33): 28112-21, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22700958

RESUMEN

Regulated nucleocytoplasmic transport of proteins is central to cellular function and dysfunction during processes such as viral infection. Active protein trafficking into and out of the nucleus is dependent on the presence within cargo proteins of intrinsic specific modular signals for nuclear import (nuclear localization signals, NLSs) and export (nuclear export signals, NESs). Rabies virus (RabV) phospho (P) protein, which is largely responsible for antagonising the host anti-viral response, is expressed as five isoforms (P1-P5). The subcellular trafficking of these isoforms is thought to depend on a balance between the activities of a dominant N-terminal NES (N-NES) and a distinct C-terminal NLS (C-NLS). Specifically, the N-NES-containing isoforms P1 and P2 are cytoplasmic, whereas the shorter P3-P5 isoforms, which lack the N-NES, are believed to be nuclear through the activity of the C-NLS. Here, we show for the first time that RabV P contains an additional strong NLS in the N-terminal region (N-NLS), which, intriguingly, overlaps with the N-NES. This arrangement represents a novel nuclear trafficking module where the N-NLS is inactive in P1 but becomes activated in P3, concomitant with truncation of the N-NES, to become the principal targeting signal conferring nuclear accumulation. Understanding this unique switch arrangement of overlapping, co-regulated NES/NLS sequences is vital to delineating the critical role of RabV P protein in viral infection.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Señales de Exportación Nuclear , Señales de Localización Nuclear/metabolismo , Virus de la Rabia/metabolismo , Rabia/metabolismo , Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Citoplasma/genética , Células HeLa , Humanos , Chaperonas Moleculares , Señales de Localización Nuclear/genética , Fosfoproteínas , Rabia/genética , Virus de la Rabia/genética , Proteínas Estructurales Virales
11.
J Virol ; 86(9): 4743-51, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22345440

RESUMEN

The Hsp70 chaperone plays a central role in multiple processes within cells, including protein translation, folding, intracellular trafficking, and degradation. This protein is implicated in the replication of numerous viruses. We have shown that rabies virus infection induced the cellular expression of Hsp70, which accumulated in Negri body-like structures, where viral transcription and replication take place. In addition, Hsp70 is present in both nucleocapsids purified from infected cells and in purified virions. Hsp70 has been shown to interact with the nucleoprotein N. The downregulation of Hsp70, using specific chaperone inhibitors, such as quercetin or RNA interference, resulted in a significant decrease of the amount of viral mRNAs, viral proteins, and virus particles. These results indicate that Hsp70 has a proviral function during rabies virus infection and suggest that Hsp70 is involved in at least one stage(s) of the viral life cycle, such as viral transcription, translation, and/or production. The mechanism by which Hsp70 controls viral infection will be discussed.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Virus de la Rabia/metabolismo , Animales , Línea Celular , Cricetinae , Expresión Génica , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Humanos , Proteínas de la Nucleocápside/metabolismo , Unión Proteica , Interferencia de ARN , ARN Viral/biosíntesis , Virus de la Rabia/genética , Replicación Viral/genética
12.
Cell Rep ; 42(1): 111949, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36640307

RESUMEN

Viruses must overcome the interferon-mediated antiviral response to replicate and propagate into their host. Rabies virus (RABV) phosphoprotein P is known to inhibit interferon induction. Here, using a global mass spectrometry approach, we show that RABV P binds to TBK1, a kinase located at the crossroads of many interferon induction pathways, resulting in innate immunity inhibition. Mutations of TBK1 phosphorylation sites abolish P binding. Importantly, we demonstrate that upon RABV infection or detection of dsRNA by innate immunity sensors, TBK1 and its adaptor proteins NAP1 and SINTBAD form dynamic cytoplasmic condensates that have liquid properties. These condensates can form larger aggregates having ring-like structures in which NAP1 and TBK1 exhibit locally restricted movement. P binding to TBK1 interferes with the formation of these structures. This work demonstrates that proteins of the signaling pathway leading to interferon induction transiently form liquid organelles that can be targeted by viruses.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Virus de la Rabia , Proteínas Serina-Treonina Quinasas/metabolismo , Inmunidad Innata , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Interferones/metabolismo , Factor 3 Regulador del Interferón/metabolismo
13.
J Gen Virol ; 93(Pt 4): 857-865, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22190013

RESUMEN

Vesicular stomatitis virus (VSV) matrix protein (M) has a flexible amino-terminal part that recruits cellular partners. It contains a dynamin-binding site that is required for efficient virus assembly, and two motifs, (24)PPPY(27) and (37)PSAP(40), that constitute potential late domains. Late domains are present in proteins of several enveloped viruses and are involved in the ultimate step of the budding process (i.e. fission between viral and cellular membranes). In baby hamster kidney (BHK)-21 cells, it has been demonstrated that the (24)PPPY(27) motif binds the Nedd4 (neuronal precursor cell-expressed developmentally downregulated 4) E3 ubiquitin ligase for efficient virus budding and that the (37)PSAP(40) motif, although conserved among M proteins of vesiculoviruses, does not possess late-domain activity. In this study, we have re-examined the contribution of the PSAP motif to VSV budding. First, we demonstrate that VSV M indeed binds TSG101 [tumour susceptibility gene 101; a component of the ESCRT1 (endosomal sorting complex required for transport 1)] through its PSAP motif. Second, we analysed the phenotype of several recombinant mutants. We show that a double mutant with point mutations in both the PSAP and the PPPY motifs is impaired compared with a single mutant in the PPPY motif, indicating that the PSAP motif partially compensates for the lack of the PPPY motif. Mutants' phenotypes depend on cell lines: in CERA (chicken embryo-related, Alger clone) cells, a recombinant virus with a single mutation in the PSAP motif was impaired compared with the wild type, and a mutant with a single mutation in the dynamin-binding motif was much less impaired in Vero cells than in BSR (clones of BHK-21) cells. These results have implications for the VSV budding pathway that will be discussed.


Asunto(s)
Vesiculovirus/genética , Proteínas de la Matriz Viral/genética , Animales , Línea Celular , Cricetinae , Dinaminas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Humanos , Microscopía Electrónica , Mutagénesis Sitio-Dirigida , Mutación/genética , Mutación/fisiología , Ubiquitina-Proteína Ligasas Nedd4 , Fenotipo , Ubiquitina-Proteína Ligasas/metabolismo , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/fisiología , Proteínas de la Matriz Viral/fisiología , Ensamble de Virus/genética , Ensamble de Virus/fisiología
14.
J Cell Sci ; 122(Pt 20): 3652-62, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19773364

RESUMEN

Conventional nuclear import is independent of the cytoskeleton, but recent data have shown that the import of specific proteins can be either facilitated or inhibited by microtubules (MTs). Nuclear import of the P-protein from rabies virus involves a MT-facilitated mechanism, but here, we show that P-protein is unique in that it also undergoes MT-inhibited import, with the mode of MT-interaction being regulated by the oligomeric state of the P-protein. This is the first demonstration that a protein can utilise both MT-inhibited and MT-facilitated import mechanisms, and can switch between these different modes of MT interaction to regulate its nuclear trafficking. Importantly, we show that the P-protein exploits MT-dependent mechanisms to manipulate host cell processes by switching the import of the interferon-activated transcription factor STAT1 from a conventional to a MT-inhibited mechanism. This prevents STAT1 nuclear import and signalling in response to interferon, which is vital to the host innate antiviral response. This is the first report of MT involvement in the viral subversion of interferon signalling that is central to virus pathogenicity, and identifies novel targets for the development of antiviral drugs or attenuated viruses for vaccine applications.


Asunto(s)
Antivirales/metabolismo , Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Rabia/virología , Proteínas Estructurales Virales/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Complejo Dinactina , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interferones/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/efectos de los fármacos , Modelos Biológicos , Chaperonas Moleculares , Fosfoproteínas/química , Unión Proteica/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína , Rabia/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Estructurales Virales/química
15.
J Virol ; 84(20): 10719-26, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20702643

RESUMEN

Various reports implicate PML and PML nuclear bodies (NBs) in an intrinsic antiviral response targeting diverse cytoplasmic replicating RNA viruses. PML conjugation to the small ubiquitin-like modifier (SUMO) is required for its localization within NBs. PML displays antiviral effects in vivo, as PML deficiency renders mice more susceptible to infection with the rhabdovirus vesicular stomatitis virus (VSV). Cells derived from these mice are also more sensitive to infection with rabies virus, another member of the rhabdovirus family. Alternative splicing from a single gene results in the synthesis of several PML isoforms, and these are classified into seven groups, designated PMLI to -VII. We report here that expression of PMLIV or PMLIVa, which is missing exon 5, inhibited viral mRNA and protein synthesis, leading to a reduction in viral replication. However, the expression of other nuclear isoforms (PMLI to -VI) and cytoplasmic PMLVIIb failed to impair viral production. This antiviral effect required PMLIV SUMOylation, as it was not observed with PMLIV 3KR, in which the lysines involved in SUMO conjugation were mutated. Thus, PMLIV and PMLIVa may exert this isoform-specific function through interaction with specific NB protein partners via their common C-terminal region.


Asunto(s)
Proteínas Nucleares/inmunología , Virus de la Rabia/patogenicidad , Rabia/inmunología , Rabia/prevención & control , Factores de Transcripción/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN/genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Interferón Tipo I/farmacología , Ratones , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Viral/biosíntesis , ARN Viral/genética , Rabia/genética , Virus de la Rabia/genética , Virus de la Rabia/inmunología , Virus de la Rabia/fisiología , Proteínas Recombinantes , Sumoilación , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Replicación Viral
16.
J Virol ; 84(24): 12609-18, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20943988

RESUMEN

Matrix proteins (M) direct the process of assembly and budding of viruses belonging to the Mononegavirales order. Using the two-hybrid system, the amino-terminal part of vesicular stomatitis virus (VSV) M was shown to interact with dynamin pleckstrin homology domain. This interaction was confirmed by coimmunoprecipitation of both proteins in cells transfected by a plasmid encoding a c-myc-tagged dynamin and infected by VSV. A role for dynamin in the viral cycle (in addition to its role in virion endocytosis) was suggested by the fact that a late stage of the viral cycle was sensitive to dynasore. By alanine scanning, we identified a single mutation of M protein that abolished this interaction and reduced virus yield. The adaptation of mutant virus (M.L4A) occurred rapidly, allowing the isolation of revertants, among which the M protein, despite having an amino acid sequence distinct from that of the wild type, recovered a significant level of interaction with dynamin. This proved that the mutant phenotype was due to the loss of interaction between M and dynamin. The infectious cycle of the mutant virus M.L4A was blocked at a late stage, resulting in a quasi-absence of bullet-shaped viruses in the process of budding at the cell membrane. This was associated with an accumulation of nucleocapsids at the periphery of the cell and a different pattern of VSV glycoprotein localization. Finally, we showed that M-dynamin interaction affects clathrin-dependent endocytosis. Our study suggests that hijacking the endocytic pathway might be an important feature for enveloped virus assembly and budding at the plasma membrane.


Asunto(s)
Dinaminas/metabolismo , Nucleocápside/metabolismo , Vesiculovirus/fisiología , Proteínas de la Matriz Viral/metabolismo , Ensamble de Virus/fisiología , Células Cultivadas , Clatrina/metabolismo , Dinaminas/genética , Endocitosis/fisiología , Endosomas/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Riñón/virología , Mutación/genética , Técnicas del Sistema de Dos Híbridos , Proteínas de la Matriz Viral/genética , Liberación del Virus/fisiología
17.
J Virol ; 84(13): 6699-710, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20427527

RESUMEN

The fixed rabies virus (RV) strain Nishigahara kills adult mice after intracerebral inoculation, whereas the chicken embryo fibroblast cell-adapted strain Ni-CE causes nonlethal infection in adult mice. We previously reported that the chimeric CE(NiP) strain, which has the phosphoprotein (P protein) gene from the Nishigahara strain in the genetic background of the Ni-CE strain, causes lethal infection in adult mice, indicating that the P gene is responsible for the different pathogenicities of the Nishigahara and Ni-CE strains. Previous studies demonstrated that RV P protein binds to the interferon (IFN)-activated transcription factor STAT1 and blocks IFN signaling by preventing its translocation to the nucleus. In this study, we examine the molecular mechanism by which RV P protein determines viral pathogenicity by comparing the IFN antagonist activities of the Nishigahara and Ni-CE P proteins. The results, obtained from both RV-infected cells and cells transfected to express P protein only, show that Ni-CE P protein is significantly impaired for its capacity to block IFN-activated STAT1 nuclear translocation and, consequently, inhibits IFN signaling less efficiently than Nishigahara P protein. Further, it was demonstrated that a defect in the nuclear export of Ni-CE P protein correlates with a defect in its ability to cause the mislocalization of STAT1. These data provide the first evidence that the capacity of the RV P protein to inhibit STAT1 nuclear translocation and IFN signaling correlates with the viral pathogenicity.


Asunto(s)
Interferones/antagonistas & inhibidores , Fosfoproteínas/fisiología , Virus de la Rabia/patogenicidad , Factor de Transcripción STAT1/antagonistas & inhibidores , Proteínas Estructurales Virales/fisiología , Factores de Virulencia/fisiología , Animales , Línea Celular , Femenino , Humanos , Interferones/inmunología , Ratones , Chaperonas Moleculares , Fosfoproteínas/inmunología , Virus de la Rabia/crecimiento & desarrollo , Virus de la Rabia/inmunología , Factor de Transcripción STAT1/inmunología , Carga Viral , Proteínas Estructurales Virales/inmunología , Virulencia , Factores de Virulencia/inmunología
18.
J Virol ; 83(13): 6610-23, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19386703

RESUMEN

After penetrating the host cell, the herpesvirus capsid is transported to the nucleus along the microtubule network and docks to the nuclear pore complex before releasing the viral DNA into the nucleus. The viral and cellular interactions involved in the docking process are poorly characterized. However, the minor capsid protein pUL25 has recently been reported to be involved in viral DNA uncoating. Here we show that herpes simplex virus type 1 (HSV-1) capsids interact with the nucleoporin CAN/Nup214 in infected cells and that RNA silencing of CAN/Nup214 delays the onset of viral DNA replication in the nucleus. We also show that pUL25 interacts with CAN/Nup214 and another nucleoporin, hCG1, and binds to the pUL36 and pUL6 proteins, two other components of the herpesvirus particle that are known to be important for the initiation of infection and viral DNA release. These results identify CAN/Nup214 as being a nuclear receptor for the herpesvirus capsid and pUL25 as being an interface between incoming capsids and the nuclear pore complex and as being a triggering element for viral DNA release into the nucleus.


Asunto(s)
Proteínas de la Cápside/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas de Complejo Poro Nuclear/metabolismo , Poro Nuclear/virología , Proteínas Virales/metabolismo , Animales , Chlorocebus aethiops , Cricetinae , ADN Viral/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Células Vero , Replicación Viral
19.
J Virol ; 83(16): 7948-58, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19494013

RESUMEN

Rabies virus infection induces the formation of cytoplasmic inclusion bodies that resemble Negri bodies found in the cytoplasm of some infected nerve cells. We have studied the morphogenesis and the role of these Negri body-like structures (NBLs) during viral infection. The results indicate that these spherical structures (one or two per cell in the initial stage of infection), composed of the viral N and P proteins, grow during the virus cycle before appearing as smaller structures at late stages of infection. We have shown that the microtubule network is not necessary for the formation of these inclusion bodies but is involved in their dynamics. In contrast, the actin network does not play any detectable role in these processes. These inclusion bodies contain Hsp70 and ubiquitinylated proteins, but they are not misfolded protein aggregates. NBLs, in fact, appear to be functional structures involved in the viral life cycle. Specifically, using in situ fluorescent hybridization techniques, we show that all viral RNAs (genome, antigenome, and every mRNA) are located inside the inclusion bodies. Significantly, short-term RNA labeling in the presence of BrUTP strongly suggests that the NBLs are the sites where viral transcription and replication take place.


Asunto(s)
Cuerpos de Inclusión Viral/virología , Virus de la Rabia/fisiología , Rabia/virología , Transcripción Genética , Replicación Viral , Actinas/metabolismo , Animales , Cricetinae , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Cuerpos de Inclusión Viral/metabolismo , Rabia/metabolismo , Virus de la Rabia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA