Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters








Publication year range
1.
Rev Sci Tech ; 28(1): 219-31, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19618628

ABSTRACT

Mx proteins are interferon-induced large GTPases with antiviral activities. They inhibit a wide range of viruses by blocking early stages of the replication cycles. Importantly, Mx GTPases also suppress the growth of highly pathogenic influenza A viruses, such as currently circulating H5N1 viruses or the pandemic H1N1 virus strain of 1918. In this paper, the authors review the properties of Mx proteins and discuss their role in host defence against highly pathogenic viruses. The authors further suggest that mammalian Mx proteins may normally provide a barrier against zoonotic transmission of avian influenza A viruses and that acquired resistance to the antiviral action of human MxA may be one factor, among many others, that facilitates the spread of pandemic strains in human populations. The presently available evidence suggests that Mx proteins of domestic chickens lack the ability to efficiently combat avian influenza viruses known to cause devastating infections in this species. The deliberate introduction of an antivirally active Mx gene originating from resistant birds or mammals may confer some degree of protection and thus stop commercial birds from serving as amplifying hosts of potentially pandemic influenza virus strains.


Subject(s)
GTP Phosphohydrolases/immunology , GTP Phosphohydrolases/metabolism , Influenza A virus/immunology , Interferons/physiology , Animals , GTP Phosphohydrolases/classification , GTP Phosphohydrolases/genetics , Humans , Influenza A virus/pathogenicity , Phylogeny
2.
Virology ; 363(1): 166-73, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17307214

ABSTRACT

We have previously reported that the addition of interferon (IFN) to the culture medium of Vero cells (which cannot produce IFN) that were infected with the CPI- strain of parainfluenza virus 5 (PIV5, formally known as SV5), that fails to block IFN signaling, rapidly induces alterations in the relative levels of virus mRNA and protein synthesis. In addition, IFN treatment also caused a rapid redistribution of virus proteins and enhanced the formation of cytoplasmic viral inclusion bodies. The most studied IFN-induced genes with known anti-viral activity are MxA, PKR and the Oligo A synthetase/RNase L system. We therefore examined the effects of these proteins on the replication cycle of PIV5. These studies revealed that while these proteins had some anti-viral activity against PIV5 they were not primarily responsible for the very rapid alteration in virus protein synthesis observed following IFN treatment, nor for the IFN-induced formation of virus inclusion bodies, in CPI- infected cells.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Endoribonucleases/metabolism , GTP-Binding Proteins/metabolism , Interferons/immunology , Rubulavirus/immunology , Virus Replication , eIF-2 Kinase/metabolism , Animals , Cell Line, Tumor , Chlorocebus aethiops , Gene Expression Regulation, Viral , Humans , Myxovirus Resistance Proteins , Rubulavirus/physiology , Vero Cells
3.
Exp Cell Res ; 271(2): 286-95, 2001 Dec 10.
Article in English | MEDLINE | ID: mdl-11716541

ABSTRACT

Mx proteins are interferon-induced large GTPases, some of which have antiviral activity against a variety of viruses. The murine Mx1 protein accumulates in the nucleus of interferon-treated cells and is active against members of the Orthomyxoviridae family, such as the influenza viruses and Thogoto virus. The mechanism by which Mx1 exerts its antiviral action is still unclear, but an involvement of undefined nuclear factors has been postulated. Using the yeast two-hybrid system, we identified cellular proteins that interact with Mx1 protein. The Mx1 interactors were mainly nuclear proteins. They included Sp100, Daxx, and Bloom's syndrome protein (BLM), all of which are known to localize to specific subnuclear domains called promyelocytic leukemia protein nuclear bodies (PML NBs). In addition, components of the SUMO-1 protein modification system were identified as Mx1-interacting proteins, namely the small ubiquitin-like modifier SUMO-1 and SAE2, which represents subunit 2 of the SUMO-1 activating enzyme. Analysis of the subcellular localization of Mx1 and some of these interacting proteins by confocal microscopy revealed a close spatial association of Mx1 with PML NBs. This suggests a role of PML NBs and SUMO-1 in the antiviral action of Mx1 and may allow us to discover novel functions of this large GTPase.


Subject(s)
Antigens, Nuclear , Cell Nucleus/enzymology , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins , Interferons/metabolism , Intracellular Signaling Peptides and Proteins , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Proteins/metabolism , SUMO-1 Protein/metabolism , Transcription Factors/metabolism , Viruses/metabolism , Adaptor Proteins, Signal Transducing , Adenosine Triphosphatases/metabolism , Animals , Autoantigens/metabolism , Carrier Proteins/metabolism , Cell Compartmentation/drug effects , Cell Compartmentation/genetics , Cell Nucleus/drug effects , Cell Nucleus/virology , Co-Repressor Proteins , DNA Helicases/metabolism , GTP Phosphohydrolases/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , HeLa Cells/cytology , HeLa Cells/drug effects , HeLa Cells/enzymology , Humans , Immunohistochemistry , Interferons/pharmacology , Mice , Molecular Chaperones , Myxovirus Resistance Proteins , Promyelocytic Leukemia Protein , Proteins/drug effects , RecQ Helicases , Transfection , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Tumor Suppressor Proteins , Two-Hybrid System Techniques
4.
J Interferon Cytokine Res ; 21(9): 663-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11576460

ABSTRACT

Mx proteins belong to the interferon (IFN)-induced antiviral defense. The rat genome contains three Mx genes, ratMx1, ratMx2, and ratMx3. The Mx gene products differ in their subcellular localization and antiviral specificity. The nuclear ratMx1 protein confers resistance to influenza A virus, and the cytoplasmic ratMx2 is active against vesicular stomatitis virus (VSV), whereas the cytoplasmic ratMx3 protein is antivirally inactive. To investigate the antiviral potential of the rat Mx proteins against arboviruses, a phylogenetically diverse group of viruses that frequently infect rodents, we studied the replication of LaCrosse virus (LACV). Rift Valley fever virus (RVFV) (both family Bunyaviridae), and Thogoto virus (THOV) (family Orthomyxoviridae). To that end, we used transfected Vero cells constitutively expressing one of the rat Mx proteins. We observed that the antiviral activity of rat Mx proteins against these arboviruses correlates with their intracellular localization: ratMx1 is active against THOV, which replicates in the nucleus, whereas ratMx2 inhibits bunyaviruses that replicate in the cytoplasm. The results indicate that rats have evolved two Mx proteins to efficiently control viruses with different replication strategies.


Subject(s)
Arboviruses/drug effects , GTP-Binding Proteins , Interferons/metabolism , Proteins/metabolism , Proteins/pharmacology , Rift Valley fever virus/drug effects , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Influenza A virus/drug effects , La Crosse virus/drug effects , Myxovirus Resistance Proteins , Rats , Subcellular Fractions/metabolism , Thogotovirus/drug effects , Transfection , Vero Cells/metabolism , Vesicular stomatitis Indiana virus/drug effects
5.
J Virol ; 75(19): 9282-6, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533190

ABSTRACT

Thogoto virus (THOV) is a tick-transmitted orthomyxovirus with a genome consisting of six negative-stranded RNA segments. To rescue a recombinant THOV, the viral structural proteins were produced from expression plasmids by means of a vaccinia virus expressing the T7 RNA polymerase. Genomic virus RNAs (vRNAs) were generated from plasmids under the control of the RNA polymerase I promoter. Using this system, we could efficiently recover recombinant THOV following transfection of 12 plasmids into 293T cells. To verify the recombinant nature of the rescued virus, specific genetic tags were introduced into two vRNA segments. The availability of this efficient reverse genetics system will allow us to address hitherto-unanswered questions regarding the biology of THOV by manipulating viral genes in the context of infectious virus.


Subject(s)
DNA, Complementary/genetics , Thogotovirus/genetics , Thogotovirus/isolation & purification , Viral Vaccines/genetics , Animals , Cell Line , DNA, Complementary/isolation & purification , Humans , Plasmids , Recombination, Genetic
6.
J Virol ; 74(22): 10785-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11044123

ABSTRACT

Thogoto virus (THOV) is a tick-transmitted orthomyxovirus with a segmented, negative-stranded RNA genome. In this study, we investigated the coding strategy of RNA segment 6 and found that it contains 956 nucleotides and codes for the matrix (M) protein. The full-length cDNA contains a single, long reading frame that lacks a stop codon but has coding capacity for a putative 35-kDa protein. In contrast, the M protein of THOV has an apparent molecular mass of 29 kDa as assessed by polyacrylamide gel electrophoresis. Therefore, we investigated the possibility of posttranscriptional processing of segment 6 transcripts by reverse transcription-PCR and identified a spliced mRNA that contains a stop codon and is translated into the 29-kDa M protein. Interestingly, the nontemplated UGA stop codon is generated by the splicing event itself. Thus, the unusual M coding strategy of THOV resembles that of Influenza C virus.


Subject(s)
RNA Splicing , RNA, Messenger/genetics , Thogotovirus/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Thogotovirus/metabolism
7.
J Virol ; 74(17): 8202-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933733

ABSTRACT

MxA is a large, interferon-induced GTPase with antiviral activity against RNA viruses. It forms large oligomers, but whether oligomerization and GTPase activity are important for antiviral function is not known. The mutant protein MxA(L612K) carries a lysine-for-leucine substitution at position 612 and fails to form oligomers. Here we show that monomeric MxA(L612K) lacks detectable GTPase activity but is capable of inhibiting Thogoto virus in transiently transfected Vero cells or in a Thogoto virus minireplicon system. Likewise, MxA(L612K) inhibited vesicular stomatitis virus multiplication. These findings indicate that MxA monomers are antivirally active and suggest that GTP hydrolysis may not be required for antiviral activity. MxA(L612K) is rapidly degraded in cells, whereas wild-type MxA is stable. We propose that high-molecular-weight MxA oligomers represent a stable intracellular pool from which active MxA monomers are recruited.


Subject(s)
Antiviral Agents/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins , Proteins/metabolism , Amino Acid Substitution , Animals , Antiviral Agents/genetics , COS Cells , Chlorocebus aethiops , Guanosine Triphosphate/metabolism , Humans , Hydrolysis , Mutagenesis, Site-Directed , Mutation , Myxovirus Resistance Proteins , Proteins/genetics , Thogotovirus/physiology , Vero Cells , Vesicular stomatitis Indiana virus/physiology , Virus Replication
8.
J Biol Chem ; 275(10): 7373-7, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10702310

ABSTRACT

A novel protein kinase, Mx-interacting protein kinase (PKM), has been identified in a yeast two-hybrid screen for interaction partners of human MxA, an interferon-induced GTPase with antiviral activity against several RNA viruses. A highly conserved protein kinase domain is present in the N-terminal moiety of PKM, whereas an Mx interaction domain overlaps with C-terminal PEST sequences. PKM has a molecular weight of about 127,000 and exhibits high sequence homology to members of a recently described family of homeodomain-interacting protein kinases. Recombinant PKM has serine/threonine kinase activity that is abolished by a single amino acid substitution in the ATP binding domain (K221W). PKM catalyzes autophosphorylation and phosphorylation of various cellular and viral proteins. PKM is expressed constitutively and colocalizes with the interferon-inducible Sp100 protein and murine Mx1 in discrete nuclear structures known as nuclear bodies.


Subject(s)
Antiviral Agents/analysis , Cell Nucleus/chemistry , GTP-Binding Proteins , Protein Serine-Threonine Kinases/analysis , Proteins/analysis , Amino Acid Sequence , Humans , Molecular Sequence Data , Molecular Weight , Myxovirus Resistance Proteins , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics
9.
J Virol ; 74(1): 560-3, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10590150

ABSTRACT

Human MxA protein accumulates in the cytoplasm of interferon-treated cells and inhibits the multiplication of several RNA viruses, including Thogoto virus (THOV), a tick-borne orthomyxovirus that transcribes and replicates its genome in the cell nucleus. The antiviral mechanism of MxA was investigated by using two alternative minireplicon systems in which recombinant viral ribonucleoprotein complexes (vRNPs) of THOV were reconstituted from cloned cDNAs. A chloramphenicol acetyltransferase reporter minigenome RNA was expressed either by T7 RNA polymerase in the cytoplasm of transfected cells or, alternatively, by RNA polymerase I in the nucleus. The inhibitory effect of MxA was studied in both cellular compartments by coexpressing wild-type MxA or TMxA, an artificial nuclear form of MxA. Our results indicate that both MxA proteins recognize the assembled vRNP rather than the newly synthesized unassembled components. The present findings are consistent with previous data which indicated that cytoplasmic MxA prevents transport of vRNPs into the nucleus, whereas nuclear MxA directly inhibits the viral polymerase activity in the nucleus.


Subject(s)
GTP Phosphohydrolases/physiology , GTP-Binding Proteins , Gene Expression Regulation, Viral/physiology , Genes, Reporter , Proteins/physiology , Ribonucleoproteins/genetics , Thogotovirus/metabolism , Animals , COS Cells , Chloramphenicol O-Acetyltransferase/genetics , Myxovirus Resistance Proteins , Subcellular Fractions/metabolism , Viral Proteins/genetics
10.
FEBS Lett ; 463(1-2): 24-8, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10601631

ABSTRACT

To define domains of the human MxA GTPase involved in GTP hydrolysis and antiviral activity, we used two monoclonal antibodies (mAb) directed against different regions of the molecule. mAb 2C12 recognizes an epitope in the central interactive region of MxA, whereas mAb M143 is directed against the N-terminal G domain. mAb 2C12 greatly stimulated MxA GTPase activity, suggesting that antibody-mediated crosslinking enhances GTP hydrolysis. In contrast, monovalent Fab fragments of 2C12 abolished GTPase activity, most likely by blocking intramolecular interactions required for GTPase activation. Interestingly, intact IgG molecules and Fab fragments of 2C12 both prevented association of MxA with viral nucleocapsids and neutralized MxA antiviral activity in vivo. mAb M143 had no effect on MxA function, indicating that this antibody binds outside functional regions. These data demonstrate that the central region recognized by 2C12 is critical for regulation of GTPase activity and viral target recognition.


Subject(s)
Antiviral Agents/chemistry , GTP Phosphohydrolases/chemistry , GTP-Binding Proteins , Proteins/chemistry , 3T3 Cells , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Binding Sites/immunology , Blotting, Western , Centrifugation, Density Gradient , Dynamins , Escherichia coli/metabolism , Humans , Hydrolysis , Mice , Molecular Sequence Data , Myxovirus Resistance Proteins , Nucleocapsid/metabolism , Precipitin Tests , Protein Binding/drug effects , Proteins/immunology , Sequence Homology, Amino Acid
11.
Arch Virol ; 144(8): 1601-9, 1999.
Article in English | MEDLINE | ID: mdl-10486113

ABSTRACT

Thogoto virus (THOV) represents a new genus in the family Orthomyxoviridae. The three polymerase subunits PB1, PB2, and PA initiate transcription by a unique cap-stealing mechanism that involves the cleavage of only the m(7)GpppAm structure from cellular hnRNAs. Here, we report the cloning of the longest genomic segment of THOV coding for PB2. It comprises 2,375 nucleotides encoding a single large open reading frame flanked by the conserved terminal regions typical for orthomyxoviruses. The deduced amino acid sequence corresponds to a protein of 769 amino acids, with an estimated Mr of 88,042. Expression of the PB2 cDNA in mammalian cells revealed nuclear accumulation of the recombinant protein, consistent with the replication of THOV in the cell nucleus.


Subject(s)
RNA-Dependent RNA Polymerase/chemistry , Thogotovirus/enzymology , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Molecular Sequence Data , Molecular Weight , RNA-Dependent RNA Polymerase/genetics
12.
Proc Natl Acad Sci U S A ; 96(5): 2082-6, 1999 Mar 02.
Article in English | MEDLINE | ID: mdl-10051598

ABSTRACT

Interferon-induced human MxA protein belongs to the dynamin superfamily of large GTPases. It exhibits antiviral activity against a variety of RNA viruses, including Thogoto virus, an influenza virus-like orthomyxovirus transmitted by ticks. Here, we report that MxA blocks the transport of Thogoto virus nucleocapsids into the nucleus, thereby preventing transcription of the viral genome. This interaction can be abolished by a mAb that neutralizes the antiviral activity of MxA. Our results reveal an antiviral mechanism whereby an interferon-induced protein traps the incoming virus and interferes with proper transport of the viral genome to its ultimate target compartment within the infected cell.


Subject(s)
Cell Nucleus/physiology , Cell Nucleus/virology , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins , Interferons/pharmacology , Nucleocapsid Proteins/metabolism , Proteins/metabolism , Thogotovirus/physiology , 3T3 Cells , Animals , Antibodies, Monoclonal/pharmacology , Antiviral Agents/pharmacology , Cell Line , Cricetinae , Cycloheximide/pharmacology , Enzyme Induction , Genome, Viral , Humans , Interferons/physiology , Mice , Models, Biological , Myxovirus Resistance Proteins , Nucleocapsid Proteins/genetics , Protein Biosynthesis , Thogotovirus/genetics , Transcription, Genetic , Virus Replication
13.
J Biol Chem ; 274(7): 4370-6, 1999 Feb 12.
Article in English | MEDLINE | ID: mdl-9933640

ABSTRACT

Human MxA protein is an interferon-induced member of the dynamin superfamily of large GTPases. MxA inhibits the multiplication of several RNA viruses, including Thogoto virus, an influenza virus-like orthomyxovirus transmitted by ticks. Previous studies have indicated that GTP binding is required for antiviral activity, but the mechanism of action is still unknown. Here, we have used an in vitro cosedimentation assay to demonstrate, for the first time, a GTP-dependent interaction between MxA GTPase and a viral target structure. The assay is based on highly active MxA GTPase as effector molecules, Thogoto virus nucleocapsids as viral targets, and guanosine 5'-O-(3-thiotriphosphate) (GTPgammaS) as a stabilizing factor. We show that MxA tightly interacts with viral nucleocapsids by binding to the nucleoprotein component. This interaction requires the presence of GTPgammaS and is mediated by domains in the carboxyl-terminal moiety of MxA. We propose that GTP-bound MxA adopts an antivirally active conformation that allows interaction with viral nucleocapsids, thereby impairing their normal function.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Guanosine Triphosphate/metabolism , Nucleocapsid/metabolism , Proteins/metabolism , Thogotovirus/metabolism , 3T3 Cells , Animals , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Leucine Zippers , Mice , Myxovirus Resistance Proteins , Ribonucleoproteins/metabolism
14.
Virology ; 250(1): 9-18, 1998 Oct 10.
Article in English | MEDLINE | ID: mdl-9770415

ABSTRACT

We have previously shown that the nucleoprotein (NP) of Thogoto virus (THOV), a tick-borne member of the Orthomyxoviridae family, accumulates in the cell nucleus. Here we demonstrate that THOV NP contains a motif (KRxxxxxxxxxKTKK) at amino acid positions 179-193 that represents a classical bipartite nuclear localization signal (NLS). This sequence motif (named cNLS) was able to translocate a cytoplasmic 80-kDa reporter protein into the nucleus. Targeted mutations substituting lysines for alanines in the downstream cluster of the bipartite motif abolished the capacity of cNLS to mediate nuclear import. In contrast, identical mutations had no effect on nuclear localization when introduced into THOV NP, indicating that additional transport signals are present in NP. Amino-acid sequence comparisons revealed that THOV NP lacks the N-terminal nonconvential NLS (named here nNLS), which has been implicated in nuclear import of influenza A virus NP. Accordingly, THOV NP failed to interact in coprecipitation assays with the cellular NPI-1/3 transport factors of the karyopherin alpha family. A highly conserved motif identified in THOV NP was the so-called nuclear accumulation sequence (NAS). Mutating NAS alone, or in combination with cNLS, had no gross effect on the intracellular distribution of the protein, indicating that a functional NAS is not required for nuclear accumulation of THOV NP in mammalian cells. We also studied nuclear transport of influenza A/PR/8/34 virus NP. Interestingly, we found a cNLS motif at amino acid positions 198-216 in addition to the previously described nonconventional nNLS. To further assess the functional role of cNLS, nNLS, and NAS, we analyzed single, double, and triple mutants of influenza A virus NP. When nNLS was destroyed, the protein stayed in the cytoplasm as expected. When NAS was disrupted in addition to nNLS, the double mutant accumulated in the nucleus, suggesting that cNLS was active. Indeed, when cNLS was also inactivated, the triple mutant protein localized again predominantly to the cytoplasm. These findings suggest that NP of orthomyxoviruses have two independent NLSs, namely cNLS and nNLS. They further suggest that NAS and NLSs may assume opposing roles in nucleocytoplasmic transport of NP.


Subject(s)
GTP-Binding Proteins , Influenza A virus/genetics , Nuclear Localization Signals/genetics , Nucleoproteins/genetics , RNA-Binding Proteins , Thogotovirus/genetics , Viral Core Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Chlorocebus aethiops , Conserved Sequence , Molecular Sequence Data , Mutation , Myxovirus Resistance Proteins , Nuclear Proteins/metabolism , Nucleocapsid Proteins , Nucleoproteins/metabolism , Proteins/genetics , Recombinant Fusion Proteins , Vero Cells , Viral Core Proteins/metabolism , alpha Karyopherins
15.
Methods ; 15(3): 255-63, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9735310

ABSTRACT

MxA protein is an interferon-induced GTPase of human cells that inhibits the multiplication of several RNA viruses, including influenza viruses and bunyaviruses. Studies on MxA transgenic mice have shown that MxA is a powerful antiviral agent in vivo. It has been suggested that this cellular protein also protects humans from viral disease, but the mechanism(s) by which MxA exerts its antiviral action is still poorly understood. Using an in vitro cosedimentation assay, we now demonstrate that MxA tightly interacts with components of the ribonucleoprotein complex of Thogoto virus, an influenza-like virus transmitted by ticks. This assay demonstrates for the first time a physical interaction between MxA GTPase and a viral target structure. It is based on three elements, namely, highly active MxA GTPases as effector molecules, viral ribonucleoprotein particles as viral targets, and GTPgammaS as a stabilizing factor. Furthermore, using a simple nuclear translocation assay, we show that human MxA protein forms oligomers in vivo. This assay provides a stringent test for tight association of partner molecules in intact mammalian cells. It not only will be useful for studying physical interactions of MxA with partner molecules, but may also be applicable to other studies on protein-protein interactions in living cells.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins , Guanosine Triphosphate/metabolism , Proteins/metabolism , RNA Viruses/metabolism , Ribonucleoproteins/metabolism , Animals , Biological Transport , Biopolymers , Cell Nucleus/metabolism , Chromatography, Gel , Escherichia coli/genetics , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/isolation & purification , Glutathione Transferase/metabolism , Humans , Mice , Myxovirus Resistance Proteins , Precipitin Tests , Protein Binding , Proteins/genetics , Proteins/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
16.
FEBS Lett ; 432(3): 182-6, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9720921

ABSTRACT

Four cytochrome P450-dependent enzymes, among them dihydroxypterocarpan 6a-hydroxylase (D6aH), are specifically involved in the elicitor-inducible biosynthesis of glyceollins, the phytoalexins of soybean. Here we report that CYP93A1 cDNA, which we isolated previously from elicitor-induced soybean cells, codes for a protein with D6aH activity. Analysis of the catalytic properties of recombinant CYP93A1 expressed in yeast, its NADPH dependency, stereoselectivity and high substrate affinity confirmed that D6aH is the physiological function of CYP93A1. It thus represents the first isoflavonoid-specific CYP to be characterized at the molecular level. In elicitor-treated soybean cells producing phytoalexins, increases in D6aH activity were correlated with elevated transcript levels which indicates that expression of the enzyme is regulated at the level of transcription. Therefore, CYP93A1 cDNA can be used as a specific molecular marker for the inducible defense response against pathogen attack.


Subject(s)
Benzopyrans/metabolism , Cytochrome P-450 Enzyme System/genetics , Glycine max/enzymology , Glycine max/genetics , Plant Proteins/genetics , Amino Acid Sequence , Catalysis , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Plant , Glucans/pharmacology , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/genetics , Molecular Sequence Data , Oligopeptides/chemistry , Plant Extracts/metabolism , Plant Proteins/chemistry , Plant Proteins/drug effects , Protein Structure, Tertiary , Pterocarpans , Recombinant Proteins/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Sesquiterpenes , Soybean Proteins , Glycine max/chemistry , Species Specificity , Substrate Specificity , Terpenes/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Phytoalexins
17.
Rev Sci Tech ; 17(1): 220-30, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9638812

ABSTRACT

Mx proteins are interferon-induced members of the dynamin superfamily of large guanosine triphosphatases. These proteins have attracted attention because some display antiviral activity against pathogenic RNA viruses, for example against members of the orthomyxovirus (influenzavirus) family or the bunyavirus family. Transfected cells and transgenic mice expressing Mx proteins are highly resistant to Mx-sensitive viruses, demonstrating that Mx proteins are powerful antiviral agents. In humans, synthesis of MxA is observed during self-limiting viral infections and may thus promote recovery from disease.


Subject(s)
Antiviral Agents/immunology , GTP-Binding Proteins , Proteins/immunology , RNA Virus Infections/immunology , RNA Viruses/immunology , Animals , Antiviral Agents/chemistry , Antiviral Agents/genetics , Humans , Immunity, Innate , Mice , Myxovirus Resistance Proteins , Proteins/chemistry , Proteins/genetics , RNA Virus Infections/prevention & control
18.
Virus Res ; 58(1-2): 13-20, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9879758

ABSTRACT

Tick-borne Thogoto virus (THOV), the prototype of a new genus in the Orthomyxoviridae family, contains six single-stranded RNA segments of negative polarity. Four of them encode gene products that correspond to the influenza virus PB1, PB2, PA and NP core proteins. Here we describe an in vivo system in which the expression of a THOV model RNA is driven by THOV core proteins synthesized from cloned cDNAs. Our results demonstrated the biological activity of our cloned genes and showed that the three polymerase subunits and the NP are required for gene expression. For comparison, we also used the in vivo reconstituted systems of the influenza A and B viruses. None of the polymerase or NP proteins was active in a heterologous orthomyxovirus core, indicating a high specificity in core assembly and/or function. Interestingly, the THOV polymerase did not recognize the influenza A virus promoter and vice versa.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Orthomyxoviridae/genetics , RNA, Viral , Thogotovirus/enzymology , Viral Proteins/metabolism , Cloning, Molecular , Gene Expression Regulation, Viral , RNA, Viral/genetics , Reassortant Viruses , Templates, Genetic , Thogotovirus/genetics , Viral Core Proteins/genetics , Viral Proteins/genetics
19.
Virus Res ; 50(2): 215-24, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9282786

ABSTRACT

The tick-borne Thogoto virus (THOV) is the type species of a newly recognized fourth genus, Thogotovirus, in the family Orthomyxoviridae. Because of the distant relationship of THOV with the influenza viruses, determination of its genomic information can potentially be used to identify important domains in influenza virus proteins. We have determined the complete nucleotide sequence of the second longest RNA segment of THOV. The molecule comprises 2212 nucleotides with a single large open reading frame encoding a protein of 710 amino acids, estimated Mr 81,284. The protein shares 77% amino acid similarity with the PB1-like protein of Dhori virus, a related tick-borne virus, and 50-53% with the PB1 polymerase proteins of influenza virus A, B and C. All the motifs characteristic of RNA-dependent polymerases were identified including the SSDD motif common to all RNA-dependent RNA polymerases, indicating that the THOV protein is functionally analogous to the influenza virus PB1 proteins and involved in chain elongation. We also report the corrected sequence of the third longest RNA segment of THOV, encoding a protein which shares 44-47% amino acid similarity with the PA-like polymerase proteins of influenza virus A, B and C. The biological significance of conserved domains in these orthomyxovirid proteins is discussed.


Subject(s)
Gammainfluenzavirus/enzymology , Orthomyxoviridae/enzymology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Thogotovirus/enzymology , Thogotovirus/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Nucleus/virology , Cricetinae , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Gammainfluenzavirus/genetics , Gammainfluenzavirus/metabolism , Molecular Sequence Data , Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism , RNA-Dependent RNA Polymerase/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thogotovirus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
20.
J Virol ; 71(4): 2591-9, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9060610

ABSTRACT

Human MxA protein is an interferon-induced 76-kDa GTPase that exhibits antiviral activity against several RNA viruses. Wild-type MxA accumulates in the cytoplasm of cells. TMxA, a modified form of wild-type MxA carrying a foreign nuclear localization signal, accumulates in the cell nucleus. Here we show that MxA protein is translocated into the nucleus together with TMxA when both proteins are expressed simultaneously in the same cell, demonstrating that MxA molecules form tight complexes in living cells. To define domains important for MxA-MxA interaction and antiviral function in vivo, we expressed mutant forms of MxA together with wild-type MxA or TMxA in appropriate cells and analyzed subcellular localization and interfering effects. An MxA deletion mutant, MxA(359-572), formed heterooligomers with TMxA and was translocated to the nucleus, indicating that the region between amino acid positions 359 and 572 contains an interaction domain which is critical for oligomerization of MxA proteins. Mutant T103A with threonine at position 103 replaced by alanine had lost both GTPase and antiviral activities. T103A exhibited a dominant-interfering effect on the antiviral activity of wild-type MxA rendering MxA-expressing cells susceptible to infection with influenza A virus, Thogoto virus, and vesicular stomatitis virus. To determine which sequences are critical for the dominant-negative effect of T103A, we expressed truncated forms of T103A together with wild-type protein. A C-terminal deletion mutant lacking the last 90 amino acids had lost interfering capacity, indicating that an intact C terminus was required. Surprisingly, a truncated version of MxA representing only the C-terminal half of the molecule exerted also a dominant-negative effect on wild-type function, demonstrating that sequences in the C-terminal moiety of MxA are necessary and sufficient for interference. However, all MxA mutants formed hetero-oligomers with TMxA and were translocated to the nucleus, indicating that physical interaction alone is not sufficient for disturbing wild-type function. We propose that dominant-negative mutants directly influence wild-type activity within hetero-oligomers or else compete with wild-type MxA for a cellular or viral target.


Subject(s)
Antiviral Agents/physiology , Proteins/physiology , 3T3 Cells , Animals , Antigens, Viral/analysis , Antiviral Agents/chemistry , Antiviral Agents/genetics , Antiviral Agents/metabolism , Binding Sites , Chlorocebus aethiops , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Genes, Dominant , Humans , Influenza A virus/growth & development , Mice , Mutagenesis , Mutation , Myxovirus Resistance Proteins , Nucleocapsid/genetics , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , RNA, Viral/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Thogotovirus/genetics , Thogotovirus/immunology , Transfection , Vero Cells , Vesicular stomatitis Indiana virus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL