Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Biotechnol ; 274: 33-39, 2018 May 20.
Article in English | MEDLINE | ID: mdl-29577966

ABSTRACT

Viral hepatitis caused by the hepatitis C virus (HCV) affects millions of people worldwide. The non-structural protein 3 (NS3), one of the most conserved proteins in HCV, is the target of many therapeutic studies. The NS3 protease domain (NS3p) has a range of cytotoxic T lymphocyte (CTL) epitopes, and synthesizing the protein inside the cells is the most appropriate way to present it to the immune system. We developed a tool to study this kind of presentation, using two vectored particle (VP) systems, one based on the Semliki Forest virus (SFV) and the other on HCV pseudoparticles (HCVpp), both carrying the protease domain of the NS3 gene. In addition to producing the particles, we developed a method to quantify these VPs using qRT-PCR. We produced batches of approximately 2.4 × 104 SFV-NS3p/µL and 4.0 × 102 HCVpp-NS3p/µL. BHK-21 and HuH-7 cells treated with the VPs expressed the NS3 protein, thus showing the functionality of this system.


Subject(s)
Cloning, Molecular/methods , Hepacivirus/enzymology , Transfection/methods , Viral Nonstructural Proteins/genetics , Animals , Cell Line , Cricetinae , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , HEK293 Cells , Hepacivirus/genetics , Humans , Plasmids/genetics , Protein Domains , Semliki forest virus/enzymology , Semliki forest virus/genetics , Viral Load , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
2.
Viruses ; 9(10)2017 10 07.
Article in English | MEDLINE | ID: mdl-28991178

ABSTRACT

Semliki Forest virus (SFV) is an arthropod-borne alphavirus that induces membrane invaginations (spherules) in host cells. These harbor the viral replication complexes (RC) that synthesize viral RNA. Alphaviruses have four replicase or nonstructural proteins (nsPs), nsP1-4, expressed as polyprotein P1234. An early RC, which synthesizes minus-strand RNA, is formed by the polyprotein P123 and the polymerase nsP4. Further proteolytic cleavage results in a late RC consisting of nsP1-4 and synthesizing plus strands. Here, we show that only the late RCs are highly active in RNA synthesis in vitro. Furthermore, we demonstrate that active RCs can be isolated from both virus-infected cells and cells transfected with the wild-type replicase in combination with a plasmid expressing a template RNA. When an uncleavable polyprotein P123 and polymerase nsP4 were expressed together with a template, high levels of minus-strand RNA were produced in cells, but RCs isolated from these cells were hardly active in vitro. Furthermore, we observed that the uncleavable polyprotein P123 and polymerase nsP4, which have previously been shown to form spherules even in the absence of the template, did not replicate an exogenous template. Consequently, we hypothesize that the replicase proteins were sequestered in spherules and were no longer able to recruit a template.


Subject(s)
Polyproteins/metabolism , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Semliki forest virus/enzymology , Semliki forest virus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Arthropods/virology , Gene Expression Regulation, Viral , Protein Processing, Post-Translational , RNA, Viral/metabolism , Semliki forest virus/genetics , Virus Replication
3.
J Biochem ; 159(5): 497-508, 2016 May.
Article in English | MEDLINE | ID: mdl-26711239

ABSTRACT

Various proteins synthesized by ribosomes are imported into specific organelles. To elucidate the behavior of protein domains during import, we developed a folding probe, in which the capsid protease (CP) domain of the Semliki Forest virus was connected to enhanced green fluorescent protein (EGFP). The probe was fused to appropriate N-terminal organelle-targeting signal sequences and expressed in cultured cells. When the entire CP-domain was present in the cytosol, it became folded and cleaved off the following EGFP-domain. Once cleaved, EGFP stability was not affected by upstream sequences. Based on EGFP localization, we estimated the extent of CP-domain folding in the cytosolic space. When fused to mitochondrial hydrophobic multispanning membrane protein ABCB10, more than half of the EGFP remained in the cytoplasm, whereas most of the CP-portion was in the mitochondrial fraction. When fused to the endoplasmic reticulum (ER) signal, the cleaved EGFP was observed only in the ER fraction, confirming that the CP-domain cannot fold on the cytoplasmic side during cotranslational ER translocation. Thus, import of the ABCB10 molecule was not as tightly coupled with chain elongation as ER translocation. Use of this probe to quantitatively examine stop-translocation at the ER translocon in living cells revealed that positively charged residues on the translocating nascent chain stall at the ER translocon.


Subject(s)
Capsid Proteins/metabolism , Endoplasmic Reticulum/metabolism , Peptide Hydrolases/metabolism , Protein Folding , Semliki forest virus/enzymology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , COS Cells , Capsid Proteins/genetics , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Peptide Hydrolases/genetics , Protein Domains , Protein Transport/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Semliki forest virus/genetics
4.
J Virol ; 87(18): 10207-20, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23864614

ABSTRACT

Semliki Forest virus (genus Alphavirus) is an important model for studying regulated nonstructural (ns) polyprotein processing. In this study, we evaluated the strictness of the previously outlined cleavage rules, accounting for the timing and outcome of each of three cleavages within the ns polyprotein P1234, and assessed the significance of residues P6 to P4 within the cleavage sites using an alanine scanning approach. The processing of the 1/2 and 3/4 sites was most strongly affected following changes in residues P5 and P4, respectively. However, none of the mutations had a detectable effect on the processing of the 2/3 site. An analysis of recombinant viruses bearing combinations of mutations in cleavage sites revealed tolerance toward the cooccurrence of native and mutated cleavage sites within the same polyprotein, suggesting a remarkable plasticity of the protease recognition pocket. Even in a virus in which all of the cleavage sequences were replaced with alanines in the P6, P5, and P4 positions, the processing pattern was largely preserved, without leading to reversion of cleavage site mutations. Instead, the emergence of second-site mutations was identified, among which Q706R/L in nsP2 was confirmed to be associated with the recognition of the P4 position within the modified cleavage sites. Our results imply that the spatial arrangement of the viral replication complex inherently contributes to scissile-site presentation for the protease, alleviating stringent sequence recognition requirements yet ensuring the precision and the correct order of processing events. Obtaining a proper understanding of the consequences of cleavage site manipulations may provide new tools for taming alphaviruses.


Subject(s)
Peptide Hydrolases/metabolism , Polyproteins/metabolism , Semliki forest virus/enzymology , Viral Proteins/metabolism , Amino Acid Substitution , DNA Mutational Analysis , Mutagenesis, Site-Directed , Peptide Hydrolases/genetics , Proteolysis , Semliki forest virus/genetics , Substrate Specificity
5.
J Virol ; 86(13): 7180-91, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22514352

ABSTRACT

The Old World alphaviruses are emerging human pathogens with an ability to cause widespread epidemics. The latest epidemic of Chikungunya virus, from 2005 to 2007, affected over 40 countries in Africa, Asia, and Europe. The Old World alphaviruses are highly cytopathic and known to evade the cellular antiviral response by inducing global inhibition of transcription in vertebrate cells. This function was shown to be mediated by their nonstructural nsP2 protein; however, the detailed mechanism of this phenomenon has remained unknown. Here, we report that nsP2 proteins of Sindbis, Semliki Forest, and Chikungunya viruses inhibit cellular transcription by inducing rapid degradation of Rpb1, a catalytic subunit of the RNAPII complex. This degradation of Rpb1 is independent of the nsP2-associated protease activity, but, instead, it proceeds through nsP2-mediated Rpb1 ubiquitination. This function of nsP2 depends on the integrity of the helicase and S-adenosylmethionine (SAM)-dependent methyltransferase-like domains, and point mutations in either of these domains abolish Rpb1 degradation. We go on to show that complete degradation of Rpb1 in alphavirus-infected cells occurs within 6 h postinfection, before other previously described virus-induced changes in cell physiology, such as apoptosis, autophagy, and inhibition of STAT1 phosphorylation, are detected. Since Rpb1 is a subunit that catalyzes the polymerase reaction during RNA transcription, degradation of Rpb1 plays an indispensable role in blocking the activation of cellular genes and downregulating cellular antiviral response. This indicates that the nsP2-induced degradation of Rpb1 is a critical mechanism utilized by the Old World alphaviruses to subvert the cellular antiviral response.


Subject(s)
Chikungunya virus/enzymology , Cysteine Endopeptidases/metabolism , Immune Evasion , Proteolysis , RNA Polymerase II/antagonists & inhibitors , Semliki forest virus/enzymology , Sindbis Virus/enzymology , Animals , Catalytic Domain , Cell Line , Chikungunya virus/pathogenicity , Cricetinae , Immunity, Innate , Mice , RNA Polymerase II/metabolism , Semliki forest virus/pathogenicity , Sindbis Virus/pathogenicity , Ubiquitination
6.
J Virol ; 86(1): 553-65, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031949

ABSTRACT

Semliki Forest virus (SFV) is a member of the Alphavirus genus, which produces its replicase proteins in the form of a nonstructural (ns) polyprotein precursor P1234. The maturation of the replicase occurs in a temporally controlled manner by protease activity of nsP2. The template preference and enzymatic capabilities of the alphaviral replication complex have a very important connection with its composition, which is irreversibly altered by proteolysis. The final cleavage of the 2/3 site in the ns polyprotein apparently leads to significant rearrangements within the replication complex and thus denotes the "point of no return" for viral replication progression. Numerous studies have devised rules for when and how ns protease acts, but how the alphaviral 2/3 site is recognized remained largely unexplained. In contrast to the other two cleavage sites within the ns polyprotein, the 2/3 site evidently lacks primary sequence elements in the vicinity of the scissile bond sufficient for specific protease recognition. In this study, we sought to investigate the molecular details of the regulation of the 2/3 site processing in the SFV ns polyprotein. We present evidence that correct macromolecular assembly, presumably strengthened by exosite interactions rather than the functionality of the individual nsP2 protease, is the driving force for specific substrate targeting. We conclude that structural elements within the macrodomain of nsP3 are used for precise positioning of a substrate recognition sequence at the catalytic center of the protease and that this process is coordinated by the exact N-terminal end of nsP2, thus representing a unique regulation mechanism used by alphaviruses.


Subject(s)
Polyproteins/metabolism , RNA-Dependent RNA Polymerase/metabolism , Semliki forest virus/enzymology , Viral Proteins/metabolism , Alphavirus Infections/virology , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Molecular Sequence Data , Polyproteins/chemistry , Polyproteins/genetics , Protein Processing, Post-Translational , Proteolysis , RNA-Dependent RNA Polymerase/genetics , Semliki forest virus/chemistry , Semliki forest virus/genetics , Semliki forest virus/physiology , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication
7.
J Mol Biol ; 376(3): 721-35, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18177892

ABSTRACT

The N-terminal segment of the Semliki Forest virus polyprotein is an intramolecular serine protease that cleaves itself off after the invariant Trp267 from a viral polyprotein and generates the mature capsid protein. After this autoproteolytic cleavage, the free carboxylic group of Trp267 interacts with the catalytic triad (His145, Asp167 and Ser219) and inactivates the enzyme. We have deleted the last 1-7 C-terminal residues of the mature capsid protease to investigate whether removal of Trp267 regenerates enzymatic activity. Although the C-terminally truncated polypeptides do not adopt a defined three-dimensional structure and show biophysical properties observed in natively unfolded proteins, they efficiently catalyse the hydrolysis of aromatic amino acid esters, with higher catalytic efficiency for tryptophan compared to tyrosine esters and k(cat)/K(M) values up to 5 x 10(5) s(-1) M(-1). The enzymatic mechanism of these deletion variants is typical of serine proteases. The pH enzyme activity profile shows a pK(a1)=6.9, and the Ser219Ala substitution destroys the enzymatic activity. In addition, the fast release of the first product of the enzymatic reaction is followed by a steady-state second phase, indicative of formation and breakdown of a covalent acyl-enzyme intermediate. The rates of acylation and deacylation are k(2)=4.4+/-0.6 s(-1) and k(3)=1.6+/-0.5 s(-1), respectively, for a tyrosine derivative ester substrate, and the amplitude of the burst phase indicates that 95% of the enzyme molecules are active. In summary, our data provide further evidence for the potential catalytic activity of natively unfolded proteins, and provide the basis for engineering of alphavirus capsid proteins towards hydrolytic enzymes with novel specificities.


Subject(s)
Capsid Proteins/chemistry , Semliki forest virus/enzymology , Serine Endopeptidases/chemistry , Catalysis , Gene Deletion , Kinetics , Mutagenesis , Protein Conformation , Protein Folding , Serine Endopeptidases/genetics
8.
J Virol ; 81(16): 8677-84, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17553895

ABSTRACT

The type I interferons (IFNs) are potent mediators of antiviral immunity, and many viruses have developed means to block their expression or their effects. Semliki Forest virus (SFV) infection induces rapid and profound silencing of host cell gene expression, a process believed to be important for the inhibition of the IFN response. In SFV-infected cells, a large proportion of the nonstructural protein nsp2 is found in the nucleus, but a role for this localization has not been described. In this work we demonstrate that a viral mutant, SFV4-RDR, in which the nuclear localization sequence of nsp2 has been rendered inactive, induces a significantly more robust IFN response in infected cells. This mutant virus replicates at a rate similar to that of the parental SFV4 strain and also shuts off host cell gene expression to similar levels, indicating that the general cellular shutoff is not responsible for the inhibition of IFN expression. Further, the rate of virus-induced nuclear translocation of early IFN transcription factors was not found to differ between the wild-type and mutant viruses, indicating that the effect of nsp2 is at a later stage. These results provide novel information about the mode of action of this viral IFN antagonist.


Subject(s)
Cysteine Endopeptidases/metabolism , Interferon Type I/metabolism , Semliki forest virus/physiology , Animals , Cell Nucleus/enzymology , Cells, Cultured , Cysteine Endopeptidases/analysis , Cysteine Endopeptidases/genetics , Gene Expression Regulation , Interferon Type I/genetics , Mice , Nuclear Localization Signals , Semliki forest virus/enzymology , Semliki forest virus/genetics , Transcription Factors/metabolism , Virus Replication
9.
Biopolymers ; 85(5-6): 422-37, 2007.
Article in English | MEDLINE | ID: mdl-17211889

ABSTRACT

Abeta fibrils, which are central to the pathology of Alzheimer's disease, form a cross-beta-structure that contains likely parallel beta-sheets with a salt bridge between residues Asp23 and Lys28. Recent studies suggest that soluble oligomers of amyloid peptides have neurotoxic effects in cell cultures, raising the interest in studying the structures of these intermediate forms. Here, we present three models of possible soluble Abeta forms based on the sequences similarities, assumed to support local structural similarities, of the Abeta peptide with fragments of three proteins (adhesin, Semliki Forest virus capsid protein, and transthyretin). These three models share a similar structure in the C-terminal region composed of two beta-strands connected by a loop, which contain the Asp23-Lys28 salt bridge. This segment is also structurally well conserved in Abeta fibril forms. Differences between the three monomeric models occur in the N-terminal region and in the C-terminal tail. These three models might sample some of the most stable conformers of the soluble Abeta peptide within oligomeric assemblies, which were modeled here in the form of dimers, trimers, tetramers, and hexamers. The consistency of these models is discussed with respect to available experimental and theoretical data.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Adhesins, Escherichia coli/chemistry , Amino Acid Sequence , Capsid Proteins/chemistry , Dimerization , Humans , Models, Molecular , Molecular Sequence Data , Prealbumin/chemistry , Protein Conformation , Protein Folding , Semliki forest virus/chemistry , Semliki forest virus/enzymology , Sequence Alignment
10.
J Virol ; 80(11): 5413-22, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16699022

ABSTRACT

The C-terminal cysteine protease domain of Semliki Forest virus nonstructural protein 2 (nsP2) regulates the virus life cycle by sequentially cleaving at three specific sites within the virus-encoded replicase polyprotein P1234. The site between nsP3 and nsP4 (the 3/4 site) is cleaved most efficiently. Analysis of Semliki Forest virus-specific cleavage sites with shuffled N-terminal and C-terminal half-sites showed that the main determinants of cleavage efficiency are located in the region preceding the cleavage site. Random mutagenesis analysis revealed that amino acid residues in positions P4, P3, P2, and P1 of the 3/4 cleavage site cannot tolerate much variation, whereas in the P5 position most residues were permitted. When mutations affecting cleavage efficiency were introduced into the 2/3 and 3/4 cleavage sites, the resulting viruses remained viable but had similar defects in P1234 processing as observed in the in vitro assay. Complete blockage of the 3/4 cleavage was found to be lethal. The amino acid in position P1' had a significant effect on cleavage efficiency, and in this regard the protease markedly preferred a glycine residue over the tyrosine natively present in the 3/4 site. Therefore, the cleavage sites represent a compromise between protease recognition and other requirements of the virus life cycle. The protease recognizes at least residues P4 to P1', and the P4 arginine residue plays an important role in the fast cleavage of the 3/4 site.


Subject(s)
Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Semliki forest virus/enzymology , Viral Nonstructural Proteins/metabolism , Semliki forest virus/genetics , Substrate Specificity , Viral Nonstructural Proteins/chemistry
11.
FEBS Lett ; 580(5): 1502-8, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16466719

ABSTRACT

The function of Semliki Forest Virus nsP2 protease was investigated by site-directed mutagenesis. Mutations were introduced in its protease domain, Pro39, and the mutated proteins were expressed in Escherichia coli, purified and their activity in vitro was compared to that of the wild type Pro39. Mutations M781T, A662T and G577R, found in temperature-sensitive virus strains, rendered the enzyme temperature-sensitive in vitro as well. Five conserved residues were required for the proteolytic activity of Pro39. Changes affecting Cys478, His548, and Trp549 resulted in complete inactivation of the enzyme, whereas the replacements N600D and N605D significantly impaired its activity. The importance of Trp549 for the proteolytic cleavage specificity is discussed and a new structural motif involved in substrate recognition by cysteine proteases is proposed.


Subject(s)
Cysteine Endopeptidases/genetics , Semliki forest virus/enzymology , Amino Acid Substitution , Conserved Sequence , Cysteine Endopeptidases/metabolism , Mutagenesis, Site-Directed , Semliki forest virus/genetics , Substrate Specificity , Temperature , Viral Proteins/genetics , Viral Proteins/metabolism
12.
J Biol Chem ; 278(43): 41636-45, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-12917405

ABSTRACT

The replication of most positive-strand RNA viruses and retroviruses is regulated by proteolytic processing. Alphavirus replicase proteins are synthesized as a polyprotein, called P1234, which is cleaved into nsP1, nsP2, nsP3, and nsP4 by the carboxyl-terminal protease domain of nsP2. The cleavage intermediate P123+nsP4 synthesizes minus-strand copies of the viral RNA genome, whereas the completely processed complex is required for plus-strand synthesis. To understand the mechanisms responsible for this sequential proteolysis, we analyzed in vitro translated Semliki Forest virus polyproteins containing noncleavable processing sites or various deletions. Processing of each of the three sites in vitro required a different type of activity. Site 3/4 was cleaved in trans by nsP2, its carboxyl-terminal fragment Pro39, and by all polyprotein proteases. Site 1/2 was cleaved in cis with a half-life of about 20-30 min. Site 2/3 was cleaved rapidly in trans but only after release of nsP1 from the polyprotein exposing an "activator" sequence present in the amino terminus of nsP2. Deletion of amino-terminal amino acids of nsP2 or addition of extra amino acid residues to its amino terminus specifically inhibited the protease activity that processes the 2/3 site. This sequence of delayed processing of P1234 would explain the accumulation of P123 plus nsP4, the early short-lived minus-strand replicase. The polyprotein stage would allow correct assembly and membrane association of the RNA-polymerase complex. Late in infection free nsP2 would cleave at site 2/3 yielding P12 and P34, the products of which, nsP1-4, are distributed to the plasma membrane, nucleus, cytoplasmic aggregates, and proteasomes, respectively.


Subject(s)
Gene Expression Regulation, Viral , Polyproteins/biosynthesis , RNA-Dependent RNA Polymerase/biosynthesis , Semliki forest virus/enzymology , Cysteine Endopeptidases/metabolism , Half-Life , Kinetics , Mutation , Peptide Mapping , Polyproteins/genetics , Polyproteins/metabolism , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Tissue Distribution
13.
J Gen Virol ; 84(Pt 6): 1525-1533, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12771422

ABSTRACT

It has been shown previously that an avirulent Semliki Forest virus (SFV) clone, rA774, engineered to carry the nsP3 gene of the virulent clone SFV4 becomes highly neurovirulent and is lethal for adult BALB/c mice. rA774, like several other alphaviruses, has an opal termination codon close to the 5' end of nsP3 (aa 469), while SFV4 has an arginine residue at this position. Mutation of the opal codon to an arginine residue increases the virulence of rA774 but does not reconstruct the severe neurovirulence of SFV4. Additionally, nsP3 amino acid sequences differ between these two strains by eight amino acids and by a deletion of seven amino acids in the C-terminal third of rA774 nsP3. This study shows that neurovirulence can be reconstituted gradually by exchanging individual amino acids and is fully retained when combinations of two nsP3 mutations, V(11)-->I and L(201)-->F, V(11)-->I and D(249)-->N, A(48)-->E and G(70)-->A or T(435)-->A and F(442)-->L, are introduced into an rA774 derivative carrying R(469). The critical role of the arginine codon for neurovirulence was confirmed further by the acquisition of a fully lethal phenotype following the introduction of R(469) into a moderately virulent rA774 recombinant carrying the SFV4 nsP1 and nsP2 genes. In conclusion, virulence determinants in SFV are distributed over a wide region of the nonstructural genes.


Subject(s)
RNA-Binding Proteins/genetics , Semliki forest virus/genetics , Viral Nonstructural Proteins/genetics , Alphavirus Infections/virology , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Cricetinae , Female , Genes, Viral , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Phenotype , RNA-Dependent RNA Polymerase/genetics , Semliki forest virus/enzymology , Semliki forest virus/pathogenicity , Sequence Deletion , Sequence Homology, Amino Acid , Virulence/genetics
14.
Virology ; 297(1): 19-30, 2002 May 25.
Article in English | MEDLINE | ID: mdl-12083832

ABSTRACT

To determine molecular viral components which can induce innate immune responses in human peripheral blood mononuclear cells (PBMC), we investigated the anti-neoplastic agent Newcastle disease virus (NDV) and its two spike proteins hemagglutinin-neuraminidase (HN) and fusion protein (F). NDV was an excellent inducer in PBMC of IFN-alpha production and capable of inducing upregulation of plasma membrane expression of tumor necrosis factor related apoptosis inducing ligand (TRAIL). Viral replication was not required for these responses because NDV inactivated for 5 min by UV was as good as live NDV. NDV-modified and paraformaldehyde-fixed BHK cells could also trigger IFN-alpha and TRAIL induction, indicating that contacts of responder cells with NDV-modified cell surfaces are sufficient to induce these activities in PBMC. Antibodies against HN but not F were able to block these responses. Finally we could show that HN but not F induced IFN-alpha and TRAIL in PBMC. This was possible through the use of respective gene transfectants generated with the help of Semliki Forest virus (SFV) replicase-based DNA recombinant expression systems. Upon contact with BHK cells expressing HN but not F at their cell surface, human PBMC produced IFN-alpha and some cells, including monocytes and T lymphocytes, upregulated cell surface TRAIL expression.


Subject(s)
Interferon-alpha/biosynthesis , Leukocytes, Mononuclear/immunology , Membrane Glycoproteins/biosynthesis , Newcastle disease virus/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Apoptosis Regulatory Proteins , Cell Line , Cells, Cultured , DNA-Directed DNA Polymerase , Fluorescent Antibody Technique , Genetic Vectors , HN Protein , Humans , Interferon-alpha/analysis , Leukocytes, Mononuclear/virology , Membrane Glycoproteins/analysis , Recombination, Genetic , Semliki forest virus/enzymology , Semliki forest virus/genetics , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Necrosis Factor-alpha/analysis , Viral Fusion Proteins
15.
Scand J Immunol ; 54(1-2): 117-24, 2001.
Article in English | MEDLINE | ID: mdl-11439157

ABSTRACT

Development of nucleic acid-based vaccines against parasitic diseases shows great promise, although certain concerns about safety aspects of conventional DNA vaccines have been raised. This study presents a comparison of antibody responses induced in mice by DNA and RNA-based immunization with vectors encoding a part of the P. falciparum antigen Pf332. Two types of plasmids were used, one conventional DNA plasmid containing a cytomegalovirus promoter and one suicidal DNA plasmid encoding the Semliki Forest virus (SFV) replicase. RNA, encoding the SFV replicase and the relevant antigen, was delivered either as naked RNA or packaged in SFV suicide particles. In general, the antibody responses induced by the DNA plasmids were low and peaking after three injections, the conventional plasmid giving the highest responses. Also the RNA delivered in SFV particles consistently induced antibody responses, although comparatively low. Analyses of the ratio of immunoglobulin (Ig)G1/IgG2a subclasses in the responses indicated that all plasmids resulted in a bias for a Th2-type of response, while the SFV-particles elicited a Th1 type of response. Importantly, all these immunogens induced an immunological memory, which could be efficiently activated by a booster injection with the corresponding protein, with unchanged patterns of IgG subclasses.


Subject(s)
DNA, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/immunology , Female , Genetic Vectors , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Plasmids , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , RNA, Viral , Semliki forest virus/enzymology , Semliki forest virus/genetics , Vaccination
16.
Life Sci ; 67(20): 2473-84, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-11065170

ABSTRACT

Human UDP-glucuronosyltransferases (UGTs) 1A6 and 1A9 were expressed using Semliki Forest virus (SFV) vectors. Infection of chinese hamster lung fibroblast V79 cells with recombinant SFV-UGT viruses resulted in efficient protein expression as detected by metabolic labeling, Western blot analyses and immunofluorescence microscopy. The expression of UGT 1A6 and UGT1A9 in the SFV-infected cells was approximately two fold higher than in a stable V79 cell line. No UGT signal was detected in noninfected cells. In addition, SFV-UGT viruses also efficiently infected other mammalian cells, such as baby hamster kidney (BHK), chinese hamster ovary (CHO) and human lung (WI-26 VA4) cells leading to high production of recombinant enzyme. The measurement of enzyme activities and kinetic parameters using p-nitrophenol and nitrocatechol (entacapone) as substrates for UGT1A6 and UGT1A9, respectively, showed that the overall kinetic properties of the enzymes produced by the two systems were similar. We conclude that the SFV expression system represents an efficient, fast and versatile method for production of metabolic enzymes for in vitro assays.


Subject(s)
Gene Expression , Glucuronosyltransferase/biosynthesis , Glucuronosyltransferase/genetics , Semliki forest virus/genetics , Animals , CHO Cells/enzymology , CHO Cells/virology , Catechols/metabolism , Cells, Cultured , Cricetinae , DNA Primers/chemistry , Fibroblasts/enzymology , Fibroblasts/virology , Genetic Vectors , Humans , Kidney/enzymology , Kidney/virology , Lung/enzymology , Lung/virology , Nitriles , Nitrophenols/metabolism , RNA, Messenger/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Semliki forest virus/enzymology , Substrate Specificity , Transfection , UDP-Glucuronosyltransferase 1A9
17.
J Biol Chem ; 275(48): 37853-9, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10984480

ABSTRACT

The RNA replication complex of Semliki Forest virus is bound to cytoplasmic membranes via the mRNA-capping enzyme Nsp1. Here we have studied the structure and liposome interactions of a synthetic peptide (245)GSTLYTESRKLLRSWHLPSV(264) corresponding to the membrane binding domain of Nsp1. The peptide interacted with liposomes only if negatively charged lipids were present that induced a structural change in the peptide from a random coil to a partially alpha-helical conformation. NMR structure shows that the alpha-helix is amphipathic, the hydrophobic surface consisting of several leucines, a valine, and a tryptophan moiety (Trp-259). Fluorescence studies revealed that this tryptophan intercalates in the bilayer to the depth of the ninth and tenth carbons of lipid acyl chains. Mutation W259A altered the mode of bilayer association of the peptide and abolished its ability to compete for membrane association of intact Nsp1, demonstrating its crucial role in the membrane association and function of Nsp1.


Subject(s)
Membrane Fusion , RNA Caps , Amino Acid Sequence , Cell Membrane/enzymology , Models, Molecular , Molecular Sequence Data , Protein Conformation , Semliki forest virus/enzymology , Sequence Homology, Amino Acid , Tryptophan/chemistry
18.
J Biol Chem ; 275(23): 17281-7, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10748213

ABSTRACT

Both genomic and subgenomic RNAs of the Alphavirus have m(7)G(5')ppp(5')N (cap0 structure) at their 5' end. Previously it has been shown that Alphavirus-specific nonstructural protein Nsp1 has guanine-7N-methyltransferase and guanylyltransferase activities needed in the synthesis of the cap structure. During normal cap synthesis the 5' gamma-phosphate of the nascent viral RNA chain is removed by a specific RNA 5'-triphosphatase before condensation with GMP, delivered by the guanylyltransferase. Using a novel RNA triphosphatase assay, we show here that nonstructural protein Nsp2 (799 amino acids) of Semliki Forest virus specifically cleaves the gamma,beta-triphosphate bond at the 5' end of RNA. The same activity was demonstrated for Nsp2 of Sindbis virus, as well as for the amino-terminal fragment of Semliki Forest virus Nsp2-N (residues 1-470). The carboxyl-terminal part of Semliki Forest virus Nsp2-C (residues 471-799) had no RNA triphosphatase activity. Replacement of Lys-192 by Asn in the nucleotide-binding site completely abolished RNA triphosphatase and nucleoside triphosphatase activities of Semliki Forest virus Nsp2 and Nsp2-N. Here we provide biochemical characterization of the newly found function of Nsp2 and discuss the unique properties of the entire Alphavirus-capping apparatus.


Subject(s)
Acid Anhydride Hydrolases/metabolism , Alphavirus/enzymology , Cysteine Endopeptidases/metabolism , GTP Phosphohydrolases/metabolism , RNA Caps/metabolism , Semliki forest virus/enzymology , Amino Acid Sequence , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Guanosine Triphosphate/metabolism , Kinetics , Molecular Sequence Data , Recombinant Proteins/metabolism , Substrate Specificity
19.
Nat Med ; 5(7): 823-7, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10395329

ABSTRACT

'Naked' nucleic acid vaccines are potentially useful candidates for the treatment of patients with cancer, but their clinical efficacy has yet to be demonstrated. We sought to enhance the immunogenicity of a nucleic acid vaccine by making it 'self-replicating'. We accomplished this by using a gene encoding an RNA replicase polyprotein derived from the Semliki forest virus, in combination with a model antigen. A single intramuscular injection of a self-replicating RNA immunogen elicited antigen-specific antibody and CD8+ T-cell responses at doses as low as 0.1 microg. Pre-immunization with a self-replicating RNA vector protected mice from tumor challenge, and therapeutic immunization prolonged the survival of mice with established tumors. The self-replicating RNA vectors did not mediate the production of substantially more model antigen than a conventional DNA vaccine did in vitro. However, the enhanced efficacy in vivo correlated with a caspase-dependent apoptotic death in transfected cells. This death facilitated the uptake of apoptotic cells by dendritic cells, providing a potential mechanism for enhanced immunogenicity. Naked, non-infectious, self-replicating RNA may be an excellent candidate for the development of new cancer vaccines.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Colonic Neoplasms/prevention & control , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/therapeutic use , Animals , Antibody Formation , Apoptosis , Colonic Neoplasms/immunology , Dendritic Cells/immunology , Enhancer Elements, Genetic , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Plasmids , Promoter Regions, Genetic , RNA-Dependent RNA Polymerase/biosynthesis , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Semliki forest virus/enzymology , Semliki forest virus/genetics , Transfection , Tumor Cells, Cultured
20.
EMBO J ; 18(11): 3164-72, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10357827

ABSTRACT

The replication complexes of all positive strand RNA viruses of eukaryotes are associated with membranes. In the case of Semliki Forest virus (SFV), the main determinant of membrane attachment seems to be the virus-encoded non-structural protein NSP1, the capping enzyme of the viral mRNAs, which has guanine-7-methyltransferase and guanylyltransferase activities. We show here that both enzymatic activities of SFV NSP1 are inactivated by detergents and reactivated by anionic phospholipids, especially phosphatidylserine. The region of NSP1 responsible for binding to membranes as well as to liposomes was mapped to a short segment, which is conserved in the large alphavirus-like superfamily of viruses. A synthetic peptide of 20 amino acids from the putative binding site competed with in vitro synthesized NSP1 for binding to liposomes containing phosphatidylserine. These findings suggest a molecular mechanism by which RNA virus replicases attach to intracellular membranes and why they depend on the membranous environment.


Subject(s)
Cell Membrane/metabolism , Nucleotidyltransferases/metabolism , Phospholipids/metabolism , Semliki forest virus/enzymology , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Anions/metabolism , Binding, Competitive , Cell Membrane/chemistry , Conserved Sequence/genetics , Detergents/pharmacology , Enzyme Activation/drug effects , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Liposomes/antagonists & inhibitors , Liposomes/metabolism , Molecular Sequence Data , Mutation , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Phosphatidylserines/metabolism , Phosphatidylserines/pharmacology , Phospholipids/pharmacology , Protein Binding/drug effects , Protein Biosynthesis , Protein Conformation/drug effects , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , S-Adenosylmethionine/antagonists & inhibitors , S-Adenosylmethionine/metabolism , Semliki forest virus/genetics , Solubility/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...