Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 8(9): e1002929, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028327

ABSTRACT

Vesicular stomatitis virus (VSV) suppresses antiviral responses in infected cells by inhibiting host gene expression at multiple levels, including transcription, nuclear cytoplasmic transport, and translation. The inhibition of host gene expression is due to the activity of the viral matrix (M) protein. Previous studies have shown that M protein interacts with host proteins Rae1 and Nup98 that have been implicated in regulating nuclear-cytoplasmic transport. However, Rae1 function is not essential for host mRNA transport, raising the question of how interaction of a viral protein with a host protein that is not essential for gene expression causes a global inhibition at multiple levels. We tested the hypothesis that there may be multiple M protein-Rae1 complexes involved in inhibiting host gene expression at multiple levels. Using size exclusion chromatography and sedimentation velocity analysis, it was determined that Rae1 exists in high, intermediate, and low molecular weight complexes. The intermediate molecular weight complexes containing Nup98 interacted most efficiently with M protein. The low molecular weight form also interacted with M protein in cells that overexpress Rae1 or cells in which Nup98 expression was silenced. Silencing Rae1 expression had little if any effect on nuclear accumulation of host mRNA in VSV-infected cells, nor did it affect VSV's ability to inhibit host translation. Instead, silencing Rae1 expression reduced the ability of VSV to inhibit host transcription. M protein interacted efficiently with Rae1-Nup98 complexes associated with the chromatin fraction of host nuclei, consistent with an effect on host transcription. These results support the idea that M protein-Rae1 complexes serve as platforms to promote the interaction of M protein with other factors involved in host transcription. They also support the idea that Rae1-Nup98 complexes play a previously under-appreciated role in regulation of transcription.


Subject(s)
Nuclear Matrix-Associated Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Transcription, Genetic , Vesicular stomatitis Indiana virus/metabolism , Viral Matrix Proteins/metabolism , Active Transport, Cell Nucleus , Cell Line , Gene Expression , HEK293 Cells , Humans , Nuclear Matrix-Associated Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Vesicular stomatitis Indiana virus/genetics
2.
J Biol Chem ; 284(7): 4500-9, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19088071

ABSTRACT

Viral matrix (M) proteins bind the nucleoprotein core (nucleocapsid) to host membranes during the process of virus assembly by budding. Previous studies using truncated M proteins had implicated the N-terminal 50 amino acids of the vesicular stomatitis virus M protein in binding both membranes and nucleocapsids and a sequence from amino acids 75-106 as an additional membrane binding region. Structure-based mutations were introduced into these two regions, and their effects on membrane association and incorporation into nucleocapsid-M protein complexes were determined using quantitative assays. The results confirmed that the N terminus of M protein is involved in association with plasma membranes as well as nucleocapsids, although these two activities were differentially affected by individual mutations. Mutations in the 75-106 region affected incorporation into nucleocapsid-M complexes but had only minor effects on association with membranes. The ability of site-specific mutant M proteins to complement growth of temperature-sensitive M mutant virus did not correlate well with the ability to associate with membranes or nucleocapsids, suggesting that complementation involves an additional activity of M protein. Mutants with similar abilities to associate with membranes and nucleocapsids but differing in complementation activity were incorporated into infectious cDNA clones. Infectious virus was repeatedly recovered containing mutant M proteins capable of complementation but was never recovered with mutant M proteins that lacked complementation activity, providing further evidence for a separate activity of M protein that is essential for virus replication.


Subject(s)
Cell Membrane/metabolism , Nucleocapsid/metabolism , Vesiculovirus/physiology , Viral Matrix Proteins/metabolism , Virus Replication/physiology , Animals , Cell Line , Cell Membrane/genetics , Cell Membrane/virology , Cricetinae , Mutation , Nucleocapsid/genetics , Protein Structure, Tertiary/physiology , Viral Matrix Proteins/genetics
3.
Virology ; 362(1): 109-19, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17258257

ABSTRACT

We have analyzed the effectiveness of Hsp90 inhibitors in blocking the replication of negative-strand RNA viruses. In cells infected with the prototype negative strand virus vesicular stomatitis virus (VSV), inhibiting Hsp90 activity reduced viral replication in cells infected at both high and low multiplicities of infection. This inhibition was observed using two Hsp90 inhibitors geldanamycin and radicicol. Silencing of Hsp90 expression using siRNA also reduced viral replication. Hsp90 inhibition changed the half-life of newly synthesized L protein (the large subunit of the VSV polymerase) from >1 h to less than 20 min without affecting the stability of other VSV proteins. Both the inhibition of viral replication and the destabilization of the viral L protein were seen when either geldanamycin or radicicol was added to cells infected with paramyxoviruses SV5, HPIV-2, HPIV-3, or SV41, or to cells infected with the La Crosse bunyavirus. Based on these results, we propose that Hsp90 is a host factor that is important for the replication of many negative strand viruses.


Subject(s)
Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , RNA Viruses/drug effects , Animals , Avulavirus/drug effects , Avulavirus/growth & development , Benzoquinones/pharmacology , Cell Line , Cricetinae , HSP90 Heat-Shock Proteins/genetics , HeLa Cells , Humans , La Crosse virus/drug effects , La Crosse virus/growth & development , Lactams, Macrocyclic/pharmacology , Macrolides/pharmacology , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA Viruses/growth & development , RNA, Small Interfering , RNA-Dependent RNA Polymerase/metabolism , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/growth & development , Viral Proteins/metabolism , Virus Replication/drug effects
4.
J Virol ; 80(8): 3701-11, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16571787

ABSTRACT

The recent solution of the crystal structure of a fragment of the vesicular stomatitis virus matrix (M) protein suggested that amino acids 121 to 124, located on a solvent-exposed loop of the protein, are important for M protein self-association and association with membranes. These residues were mutated from the hydrophobic AVLA sequence to the polar sequence DKQQ. Expression and purification of this mutant from bacteria showed that it was structurally stable and that the mutant M protein had self-association kinetics similar to those of the wild-type M protein. Analysis of the membrane association of M protein in the context of infection with isogenic recombinant viruses showed that both wild-type and mutant M proteins associated with membranes to the same extent. Virus expressing the mutant M protein did show an approximately threefold-lower binding affinity of M protein for nucleocapsid-M complexes. In contrast to the relatively minor effects of the M protein mutation on virus assembly, the mutant virus exhibited growth restriction in MDBK but not BHK cells, a slower induction of apoptosis, and lower viral-protein synthesis. Despite translating less viral protein, the mutant virus produced more viral mRNA, showing that the mutant virus could not effectively promote viral translation. These results demonstrate that the 121-to-124 region of the VSV M protein plays a minor role in virus assembly but is involved in virus-host interactions and VSV replication by augmenting viral-mRNA translation.


Subject(s)
Viral Matrix Proteins/chemistry , Virus Assembly , Animals , Cricetinae , Cytopathogenic Effect, Viral , Hydrophobic and Hydrophilic Interactions , Protein Biosynthesis , RNA, Messenger/genetics , Structure-Activity Relationship , Viral Matrix Proteins/physiology , Virus Replication
5.
J Virol ; 77(8): 4646-57, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12663771

ABSTRACT

The vesicular stomatitis virus (VSV) matrix (M) protein plays a major role in the virus-induced inhibition of host gene expression. It has been proposed that the inhibition of host gene expression by M protein is responsible for suppressing activation of host interferon gene expression. Most wild-type (wt) strains of VSV induce little if any interferon gene expression. Interferon-inducing mutants of VSV have been isolated previously, many of which contain mutations in their M proteins. However, it was not known whether these M protein mutations were responsible for the interferon-inducing phenotype of these viruses. Alternatively, mutations in other genes besides the M gene may enhance the ability of VSV to induce interferons. These hypotheses were tested by transfecting cells with mRNA expressing wt and mutant M proteins in the absence of other viral components and determining their ability to inhibit interferon gene expression. The M protein mutations were the M51R mutation originally found in the tsO82 and T1026R1 mutant viruses, the double substitution V221F and S226R found in the TP3 mutant virus, and the triple substitution E213A, V221F, and S226R found in the TP2 mutant virus. wt M proteins suppressed expression of luciferase from the simian virus 40 promoter and from the beta interferon (IFN-beta) promoter, while M proteins of interferon-inducing viruses were unable to inhibit luciferase expression from either promoter. The M genes of the interferon-inducing mutants of VSV were incorporated into the wt background of a recombinant VSV infectious cDNA clone. The resulting recombinant viruses were tested for their ability to activate interferon gene expression and for their ability to inhibit host RNA and protein synthesis. Each of the recombinant viruses containing M protein mutations induced expression of a luciferase reporter gene driven by the IFN-beta promoter and induced production of interferon bioactivity more effectively than viruses containing wt M proteins. Furthermore, the M protein mutant viruses were defective in their ability to inhibit both host RNA synthesis and host protein synthesis. These data support the idea that wt M protein suppresses interferon gene expression through the general inhibition of host RNA and protein synthesis.


Subject(s)
Gene Expression Regulation , Interferon-beta/metabolism , Proteins/metabolism , RNA/metabolism , Vesicular stomatitis Indiana virus/pathogenicity , Viral Matrix Proteins/metabolism , Animals , Cricetinae , HeLa Cells , Humans , Interferon-beta/genetics , Mutation , Promoter Regions, Genetic , Proteins/genetics , RNA/genetics , Transfection , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/genetics , Viral Matrix Proteins/genetics
6.
Virology ; 295(1): 63-73, 2002 Mar 30.
Article in English | MEDLINE | ID: mdl-12033766

ABSTRACT

In a model system to study factors involved in the establishment of a persistent viral infection that may lead to neurodegenerative diseases, Indiana and New Jersey variants of vesicular stomatitis virus (VSV) with different capacities to infect and persist in human neural cells were studied. Indiana matrix (M) protein mutants and the wild-type New Jersey strain persisted in the human neural cell line H4 for at least 120 days. The Indiana wild-type virus (HR) and a non-M mutant (TP6), both unable to persist, induced apoptosis more strongly than all the other variants tested, as indicated by higher levels of DNA fragmentation and caspase-3-like activity. Transfection of H4 cells with mRNA coding for the VSV M protein confirmed the importance of this protein in the induction of apoptosis. Furthermore, the pan-caspase inhibitor ZVAD-fmk maintained cell survival to about 80%, whereas inhibition of caspase-8, caspase-9, or both only partially protected the cells against death, consistent with the fact that anti-apoptotic molecules from the Bcl-2 family also protect cells from death only partially. These results suggest that VSV activates many pathways of cell death and that an inefficient induction of caspase-3-related apoptosis participates in the establishment of a persistent infection of human neural cells by less virulent VSV variants.


Subject(s)
Apoptosis , Vesicular stomatitis Indiana virus/pathogenicity , Vesiculovirus , Viral Matrix Proteins/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Cell Line , Humans , Mutation , Neuroprotective Agents/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...