Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 12(9): e1005880, 2016 09.
Article in English | MEDLINE | ID: mdl-27622505

ABSTRACT

For efficient replication, viruses have developed mechanisms to evade innate immune responses, including the antiviral type-I interferon (IFN-I) system. Nipah virus (NiV), a highly pathogenic member of the Paramyxoviridae family (genus Henipavirus), is known to encode for four P gene-derived viral proteins (P/C/W/V) with IFN-I antagonist functions. Here we report that NiV matrix protein (NiV-M), which is important for virus assembly and budding, can also inhibit IFN-I responses. IFN-I production requires activation of multiple signaling components including the IκB kinase epsilon (IKKε). We previously showed that the E3-ubiquitin ligase TRIM6 catalyzes the synthesis of unanchored K48-linked polyubiquitin chains, which are not covalently attached to any protein, and activate IKKε for induction of IFN-I mediated antiviral responses. Using co-immunoprecipitation assays and confocal microscopy we show here that the NiV-M protein interacts with TRIM6 and promotes TRIM6 degradation. Consequently, NiV-M expression results in reduced levels of unanchored K48-linked polyubiquitin chains associated with IKKε leading to impaired IKKε oligomerization, IKKε autophosphorylation and reduced IFN-mediated responses. This IFN antagonist function of NiV-M requires a conserved lysine residue (K258) in the bipartite nuclear localization signal that is found in divergent henipaviruses. Consistent with this, the matrix proteins of Ghana, Hendra and Cedar viruses were also able to inhibit IFNß induction. Live NiV infection, but not a recombinant NiV lacking the M protein, reduced the levels of endogenous TRIM6 protein expression. To our knowledge, matrix proteins of paramyxoviruses have never been reported to be involved in innate immune antagonism. We report here a novel mechanism of viral innate immune evasion by targeting TRIM6, IKKε and unanchored polyubiquitin chains. These findings expand the universe of viral IFN antagonism strategies and provide a new potential target for development of therapeutic interventions against NiV infections.


Subject(s)
Henipavirus Infections/immunology , I-kappa B Kinase/immunology , Immune Evasion , Interferon Type I/immunology , Nipah Virus/immunology , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Viral Proteins/immunology , A549 Cells , Animals , Chlorocebus aethiops , HeLa Cells , Henipavirus Infections/genetics , Humans , I-kappa B Kinase/genetics , Immunity, Innate , Interferon Type I/genetics , Nipah Virus/genetics , Polyubiquitin/genetics , Polyubiquitin/immunology , Protein Multimerization/genetics , Protein Multimerization/immunology , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology , Vero Cells , Viral Proteins/genetics
2.
PLoS Pathog ; 11(3): e1004739, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25782006

ABSTRACT

The paramyxovirus matrix (M) protein is a molecular scaffold required for viral morphogenesis and budding at the plasma membrane. Transient nuclear residence of some M proteins hints at non-structural roles. However, little is known regarding the mechanisms that regulate the nuclear sojourn. Previously, we found that the nuclear-cytoplasmic trafficking of Nipah virus M (NiV-M) is a prerequisite for budding, and is regulated by a bipartite nuclear localization signal (NLSbp), a leucine-rich nuclear export signal (NES), and monoubiquitination of the K258 residue within the NLSbp itself (NLSbp-lysine). To define whether the sequence determinants of nuclear trafficking identified in NiV-M are common among other Paramyxovirinae M proteins, we generated the homologous NES and NLSbp-lysine mutations in M proteins from the five major Paramyxovirinae genera. Using quantitative 3D confocal microscopy, we determined that the NES and NLSbp-lysine are required for the efficient nuclear export of the M proteins of Nipah virus, Hendra virus, Sendai virus, and Mumps virus. Pharmacological depletion of free ubiquitin or mutation of the conserved NLSbp-lysine to an arginine, which inhibits M ubiquitination, also results in nuclear and nucleolar retention of these M proteins. Recombinant Sendai virus (rSeV-eGFP) bearing the NES or NLSbp-lysine M mutants rescued at similar efficiencies to wild type. However, foci of cells expressing the M mutants displayed marked fusogenicity in contrast to wild type, and infection did not spread. Recombinant Mumps virus (rMuV-eGFP) bearing the homologous mutations showed similar defects in viral morphogenesis. Finally, shotgun proteomics experiments indicated that the interactomes of Paramyxovirinae M proteins are significantly enriched for components of the nuclear pore complex, nuclear transport receptors, and nucleolar proteins. We then synthesize our functional and proteomics data to propose a working model for the ubiquitin-regulated nuclear-cytoplasmic trafficking of cognate paramyxovirus M proteins that show a consistent nuclear trafficking phenotype.


Subject(s)
Cell Nucleus/metabolism , Paramyxovirinae/metabolism , Protein Transport/physiology , Viral Matrix Proteins/metabolism , Amino Acid Sequence , Animals , Chlorocebus aethiops , HeLa Cells , Humans , Imaging, Three-Dimensional , Immunoblotting , Immunoprecipitation , Microscopy, Confocal , Nuclear Localization Signals/metabolism , Transfection , Ubiquitin , Vero Cells
3.
Nat Commun ; 5: 5342, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25405640

ABSTRACT

Zoonotic transmission of lethal henipaviruses (HNVs) from their natural fruit bat reservoirs to humans has only been reported in Australia and South/Southeast Asia. However, a recent study discovered numerous HNV clades in African bat samples. To determine the potential for HNV spillover events among humans in Africa, here we examine well-curated sets of bat (Eidolon helvum, n = 44) and human (n = 497) serum samples from Cameroon for Nipah virus (NiV) cross-neutralizing antibodies (NiV-X-Nabs). Using a vesicular stomatitis virus (VSV)-based pseudoparticle seroneutralization assay, we detect NiV-X-Nabs in 48% and 3-4% of the bat and human samples, respectively. Seropositive human samples are found almost exclusively in individuals who reported butchering bats for bushmeat. Seropositive human sera also neutralize Hendra virus and Gh-M74a (an African HNV) pseudoparticles, as well as live NiV. Butchering bat meat and living in areas undergoing deforestation are the most significant risk factors associated with seropositivity. Evidence for HNV spillover events warrants increased surveillance efforts.


Subject(s)
Chiroptera/virology , Henipavirus Infections/transmission , Henipavirus Infections/virology , Nipah Virus/physiology , Africa , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chiroptera/blood , Chiroptera/immunology , Henipavirus Infections/blood , Henipavirus Infections/immunology , Humans , Neutralization Tests , Nipah Virus/immunology , Zoonoses/blood , Zoonoses/immunology
4.
PLoS Pathog ; 6(11): e1001186, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085610

ABSTRACT

Paramyxoviruses are known to replicate in the cytoplasm and bud from the plasma membrane. Matrix is the major structural protein in paramyxoviruses that mediates viral assembly and budding. Curiously, the matrix proteins of a few paramyxoviruses have been found in the nucleus, although the biological function associated with this nuclear localization remains obscure. We report here that the nuclear-cytoplasmic trafficking of the Nipah virus matrix (NiV-M) protein and associated post-translational modification play a critical role in matrix-mediated virus budding. Nipah virus (NiV) is a highly pathogenic emerging paramyxovirus that causes fatal encephalitis in humans, and is classified as a Biosafety Level 4 (BSL4) pathogen. During live NiV infection, NiV-M was first detected in the nucleus at early stages of infection before subsequent localization to the cytoplasm and the plasma membrane. Mutations in the putative bipartite nuclear localization signal (NLS) and the leucine-rich nuclear export signal (NES) found in NiV-M impaired its nuclear-cytoplasmic trafficking and also abolished NiV-M budding. A highly conserved lysine residue in the NLS served dual functions: its positive charge was important for mediating nuclear import, and it was also a potential site for monoubiquitination which regulates nuclear export of the protein. Concordantly, overexpression of ubiquitin enhanced NiV-M budding whereas depletion of free ubiquitin in the cell (via proteasome inhibitors) resulted in nuclear retention of NiV-M and blocked viral budding. Live Nipah virus budding was exquisitely sensitive to proteasome inhibitors: bortezomib, an FDA-approved proteasome inhibitor for treating multiple myeloma, reduced viral titers with an IC(50) of 2.7 nM, which is 100-fold less than the peak plasma concentration that can be achieved in humans. This opens up the possibility of using an "off-the-shelf" therapeutic against acute NiV infection.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Henipavirus Infections/virology , Nipah Virus/pathogenicity , Ubiquitin/metabolism , Viral Matrix Proteins/metabolism , Virus Assembly/physiology , Amino Acid Sequence , Animals , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Nucleus/drug effects , Chlorocebus aethiops , Cytoplasm/drug effects , Fluorescent Antibody Technique , HeLa Cells , Henipavirus Infections/genetics , Henipavirus Infections/metabolism , Humans , Immunoprecipitation , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Molecular Sequence Data , Mutation/genetics , Nuclear Localization Signals , Protease Inhibitors/pharmacology , Protein Processing, Post-Translational , Protein Transport , Pyrazines/pharmacology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Vero Cells , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/genetics , Virus Assembly/drug effects , Virus Shedding
5.
Vaccine ; 28(32): 5265-71, 2010 Jul 19.
Article in English | MEDLINE | ID: mdl-20538092

ABSTRACT

Two chimeric antibodies (ch) 13D6 and 10C2 against the glycoprotein E of tick-borne encephalitis virus (TBEV) were constructed by fusing variable regions of murine monoclonal antibodies (Mabs) 13D6 and 10C2 to human constant regions. Monovalent analogues of these antibodies in format of single-chain antibodies (scFv or sc) were developed, as well. The ch13D6, ch10C2, sc13D6 and sc10C2 exhibited binding characteristics similar to parental Mabs. Only the ch13D6 and sc13D6 were able to neutralize TBEV infectivity in vitro. The in vitro neutralization provided by ch13D6 suggests that this antibody can be further developed into a potent prophylaxis and therapy for tick-borne encephalitis (TBE) infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Single-Chain Antibodies/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibody Affinity , Encephalitis Viruses, Tick-Borne/immunology , Humans , Mice , Molecular Sequence Data , Neutralization Tests , Plasmids , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Single-Chain Antibodies/genetics , Single-Chain Antibodies/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...