Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(1): 111949, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640307

ABSTRACT

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.


Subject(s)
Protein Serine-Threonine Kinases , Rabies virus , Protein Serine-Threonine Kinases/metabolism , Immunity, Innate , Adaptor Proteins, Signal Transducing/metabolism , Interferons/metabolism , Interferon Regulatory Factor-3/metabolism
2.
PLoS Pathog ; 18(12): e1011022, 2022 12.
Article in English | MEDLINE | ID: mdl-36480574

ABSTRACT

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.


Subject(s)
Rabies virus , Rabies , Animals , Rabies virus/genetics , Rabies virus/metabolism , Nucleoproteins/genetics , Rabies/metabolism , Nucleocapsid/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Virus Replication , Mammals
3.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31852780

ABSTRACT

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.


Subject(s)
Phosphoproteins/chemistry , Protein Domains , Protein Multimerization , Vesicular stomatitis Indiana virus/metabolism , DNA-Directed RNA Polymerases/metabolism , Dimerization , Models, Molecular , Nucleocapsid/metabolism , Nucleoproteins/metabolism , Phosphoproteins/genetics , Protein Binding , Protein Conformation , Protein Multimerization/genetics , RNA, Viral/genetics , Sequence Alignment , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/growth & development , Virus Replication
4.
J Mol Biol ; 431(24): 4959-4977, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31634467

ABSTRACT

The rabies and Ebola viruses recruit the highly conserved host protein LC8 for their own reproductive success. In vivo knockouts of the LC8 recognition motif within the rabies virus phosphoprotein (RavP) result in completely nonlethal viral infections. In this work, we examine the molecular role LC8 plays in viral lethality. We show that RavP and LC8 colocalize in rabies infected cells, and that LC8 interactions are essential for efficient viral polymerase functionality. NMR, SAXS, and molecular modeling demonstrate that LC8 binding to a disordered linker adjacent to an endogenous dimerization domain results in restrictions in RavP domain orientations. The resulting ensemble structure of RavP-LC8 tetrameric complex is similar to that of a related virus phosphoprotein that does not bind LC8, suggesting that with RavP, LC8 binding acts as a switch to induce a more active conformation. The high conservation of the LC8 motif in Lyssavirus phosphoproteins and its presence in other analogous proteins such as the Ebola virus VP35 evinces a broader purpose for LC8 in regulating downstream phosphoprotein functions vital for viral replication.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , Drosophila Proteins/chemistry , Dyneins/chemistry , Lyssavirus/enzymology , Phosphoproteins/chemistry , Viral Proteins/chemistry , Conserved Sequence , DNA-Directed RNA Polymerases/metabolism , Drosophila Proteins/metabolism , Dyneins/metabolism , Enzyme Activation , Host-Pathogen Interactions/immunology , Models, Biological , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Rabies virus/metabolism , STAT1 Transcription Factor/metabolism , Structure-Activity Relationship , Viral Proteins/metabolism
5.
Adv Exp Med Biol ; 1215: 111-127, 2019.
Article in English | MEDLINE | ID: mdl-31317498

ABSTRACT

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.


Subject(s)
Inclusion Bodies, Viral , Rabies virus , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/metabolism , RNA, Viral/biosynthesis , Rabies virus/physiology , Viral Proteins/metabolism , Virus Replication
6.
Antiviral Res ; 168: 51-60, 2019 08.
Article in English | MEDLINE | ID: mdl-31071352

ABSTRACT

Rabies virus (RABV) is a neurotropic virus that causes fatal encephalitis in humans and animals and still kills up to 59,000 people worldwide every year. To date, only preventive or post-exposure vaccination protects against the disease but therapeutics are missing. After screening a library of 80 kinases inhibitors, we identified two compounds as potent inhibitors of RABV infection: tyrphostin 9 and rottlerin. Mechanism of action studies show that both inhibitors interfere with an early step of viral cycle and can prevent viral replication. In presence of tyrphostin 9, the viral entry through endocytosis is disturbed leading to improper delivery of viral particles in cytoplasm, whereas rottlerin is inhibiting the transcription, most likely by decreasing intracellular ATP concentration, and therefore the replication of the viral genome.


Subject(s)
Acetophenones/pharmacology , Benzopyrans/pharmacology , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Rabies virus/drug effects , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Humans , RNA, Viral/biosynthesis , Virus Internalization/drug effects , Virus Replication/drug effects
7.
Viruses ; 10(12)2018 12 03.
Article in English | MEDLINE | ID: mdl-30513968

ABSTRACT

Small Ubiquitin-like MOdifier (SUMO) conjugation to proteins has essential roles in several processes including localization, stability, and function of several players implicated in intrinsic and innate immunity. In human, five paralogs of SUMO are known of which three are ubiquitously expressed (SUMO1, 2, and 3). Infection by rhabdoviruses triggers cellular responses through the activation of pattern recognition receptors, which leads to the production and secretion of interferon. This review will focus on the effects of the stable expression of the different SUMO paralogs or Ubc9 depletion on rhabdoviruses-induced interferon production and interferon signaling pathways as well as on the expression and functions of restriction factors conferring the resistance to rhabdoviruses.


Subject(s)
Rhabdoviridae Infections/immunology , Rhabdoviridae/immunology , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Humans , Immunity, Innate , Interferons/immunology , Mice , Myxovirus Resistance Proteins/genetics , Protein Binding , Rabies virus/immunology , Receptors, Pattern Recognition/immunology , Small Ubiquitin-Related Modifier Proteins/immunology , Sumoylation , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Vesicular stomatitis Indiana virus/immunology , eIF-2 Kinase/genetics
10.
Nat Commun ; 8(1): 58, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28680096

ABSTRACT

Replication of Mononegavirales occurs in viral factories which form inclusions in the host-cell cytoplasm. For rabies virus, those inclusions are called Negri bodies (NBs). We report that NBs have characteristics similar to those of liquid organelles: they are spherical, they fuse to form larger structures, and they disappear upon hypotonic shock. Their liquid phase is confirmed by FRAP experiments. Live-cell imaging indicates that viral nucleocapsids are ejected from NBs and transported along microtubules to form either new virions or secondary viral factories. Coexpression of rabies virus N and P proteins results in cytoplasmic inclusions recapitulating NBs properties. This minimal system reveals that an intrinsically disordered domain and the dimerization domain of P are essential for Negri bodies-like structures formation. We suggest that formation of liquid viral factories by phase separation is common among Mononegavirales and allows specific recruitment and concentration of viral proteins but also the escape to cellular antiviral response.Negative strand RNA viruses, such as rabies virus, induce formation of cytoplasmic inclusions for genome replication. Here, Nikolic et al. show that these so-called Negri bodies (NBs) have characteristics of liquid organelles and they identify the minimal protein domains required for NB formation.

11.
PLoS Pathog ; 12(10): e1005942, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27749929

ABSTRACT

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.


Subject(s)
Host-Parasite Interactions/physiology , Inclusion Bodies, Viral/virology , Rabies virus , Rabies/virology , Virus Replication/physiology , Animals , Blotting, Western , Cell Line , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Immunity, Innate , In Situ Hybridization, Fluorescence , Microscopy, Confocal , Rabies/immunology , Rabies virus/immunology , Real-Time Polymerase Chain Reaction
12.
Sci Rep ; 6: 33493, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27649849

ABSTRACT

Although microtubules (MTs) are known to have important roles in intracellular transport of many viruses, a number of reports suggest that specific viral MT-associated proteins (MAPs) target MTs to subvert distinct MT-dependent cellular processes. The precise functional importance of these interactions and their roles in pathogenesis, however, remain largely unresolved. To assess the association with disease of the rabies virus (RABV) MAP, P3, we quantitatively compared the phenotypes of P3 from a pathogenic RABV strain, Nishigahara (Ni) and a non-pathogenic Ni-derivative strain, Ni-CE. Using confocal/live-cell imaging and dSTORM super-resolution microscopy to quantify protein interactions with the MT network and with individual MT filaments, we found that the interaction by Ni-CE-P3 is significantly impaired compared with Ni-P3. This correlated with an impaired capacity to effect association of the transcription factor STAT1 with MTs and to antagonize interferon (IFN)/STAT1-dependent antiviral signaling. Importantly, we identified a single mutation in Ni-CE-P3 that is sufficient to inhibit MT-association and IFN-antagonist function of Ni-P3, and showed that this mutation alone attenuates the pathogenicity of RABV. These data provide evidence that the viral protein-MT interface has important roles in pathogenesis, suggesting that this interface could provide targets for vaccine/antiviral drug development.


Subject(s)
Immune Evasion , Microtubules/metabolism , Rabies virus/metabolism , Rabies/immunology , Rabies/virology , Viral Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Female , Mice , Mutation/genetics , Protein Binding , Protein Multimerization
13.
J Virol ; 90(14): 6598-6610, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27170750

ABSTRACT

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.


Subject(s)
Interferon-alpha/pharmacology , Myxovirus Resistance Proteins/pharmacology , Small Ubiquitin-Related Modifier Proteins/pharmacology , Vesicular Stomatitis/prevention & control , Vesicular stomatitis Indiana virus/physiology , Antiviral Agents/pharmacology , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/virology , HeLa Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Protein Processing, Post-Translational , Rabies/metabolism , Rabies/prevention & control , Rabies/virology , Rabies virus/physiology , Tumor Cells, Cultured , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology
14.
Viruses ; 7(7): 3675-702, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26198243

ABSTRACT

Interferon (IFN) treatment induces the expression of hundreds of IFN-stimulated genes (ISGs). However, only a selection of their products have been demonstrated to be responsible for the inhibition of rhabdovirus replication in cultured cells; and only a few have been shown to play a role in mediating the antiviral response in vivo using gene knockout mouse models. IFNs inhibit rhabdovirus replication at different stages via the induction of a variety of ISGs. This review will discuss how individual ISG products confer resistance to rhabdoviruses by blocking viral entry, degrading single stranded viral RNA, inhibiting viral translation or preventing release of virions from the cell. Furthermore, this review will highlight how these viruses counteract the host IFN system.


Subject(s)
Rhabdoviridae Infections/immunology , Rhabdoviridae/physiology , Animals , Humans , Interferons/genetics , Interferons/immunology , Rhabdoviridae/genetics , Rhabdoviridae Infections/genetics , Rhabdoviridae Infections/virology
15.
J Virol ; 89(3): 1640-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25410852

ABSTRACT

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.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Host-Pathogen Interactions , Phosphoproteins/metabolism , Protein Interaction Mapping , Rabies virus/physiology , Viral Structural Proteins/metabolism , Virus Replication , Animals , Cell Line , DNA Mutational Analysis , Humans , Immunoprecipitation , Inclusion Bodies, Viral/chemistry , Inclusion Bodies, Viral/virology , Microscopy, Confocal , Molecular Chaperones , Mutagenesis, Site-Directed , Protein Binding , Two-Hybrid System Techniques
16.
PLoS Pathog ; 10(2): e1003975, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586174

ABSTRACT

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.


Subject(s)
Immunity, Innate/immunology , Nuclear Proteins/immunology , Rhabdoviridae Infections/immunology , Signal Transduction/immunology , Transcription Factors/immunology , Tumor Suppressor Proteins/immunology , Animals , Cell Line , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Interferon-beta/biosynthesis , Interferon-beta/immunology , Mice , Mice, Knockout , Promyelocytic Leukemia Protein , Protein Isoforms , Real-Time Polymerase Chain Reaction , Transfection , Vesiculovirus
17.
J Infect Dis ; 209(11): 1744-53, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24367042

ABSTRACT

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.


Subject(s)
Phosphoproteins/metabolism , Rabies virus/metabolism , Rabies/virology , STAT Transcription Factors/metabolism , Viral Structural Proteins/metabolism , Animals , Cell Line , Female , Gene Expression Regulation/immunology , Genome, Viral , Humans , Interferons/genetics , Interferons/metabolism , Mice , Models, Molecular , Molecular Chaperones , Mutation , Protein Binding , Protein Conformation , STAT Transcription Factors/genetics , Two-Hybrid System Techniques , Virus Replication
19.
J Virol ; 87(14): 8261-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23698294

ABSTRACT

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.


Subject(s)
Cytokine Receptor gp130/metabolism , Immune Evasion/genetics , Interferons/antagonists & inhibitors , Phosphoproteins/pharmacology , Rabies virus/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Viral Structural Proteins/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Green Fluorescent Proteins/metabolism , Luciferases , Luminescent Proteins/metabolism , Microscopy, Confocal , Molecular Chaperones , Phosphoproteins/genetics , Phosphoproteins/metabolism , Rabies virus/metabolism , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Red Fluorescent Protein
20.
Nat Commun ; 4: 1429, 2013.
Article in English | MEDLINE | ID: mdl-23385574

ABSTRACT

The typical bullet shape of Rhabdoviruses is thought to rely on the matrix protein for stabilizing the nucleocapsid coil. Here we scrutinize the morphology of purified and recombinant nucleocapsids of vesicular stomatitis virus in vitro. We elucidate pH and ionic strength conditions for their folding into conical tips and further growth into whole bullets, and provide cryo-electron microscopy reconstructions of the bullet tip and the helical trunk. We address conformational variability of the reconstituted nucleocapsids and the issue of constraints imposed by the binding of matrix protein. Our findings bridge the gap between the isolated nucleoprotein-RNA string in its form of an undulating ribbon, and the tight bullet-shaped virion skeleton.


Subject(s)
Nucleocapsid/ultrastructure , Vesicular stomatitis Indiana virus/ultrastructure , Cryoelectron Microscopy , Nucleic Acid Conformation , Nucleoproteins/metabolism , RNA, Viral/ultrastructure , Viral Matrix Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...