Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 96(Pt 7): 1746-56, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25809915

ABSTRACT

The viral protein Npro is unique to the genus Pestivirus within the family Flaviviridae. After autocatalytic cleavage from the nascent polyprotein, Npro suppresses type I IFN (IFN-α/ß) induction by mediating proteasomal degradation of IFN regulatory factor 3 (IRF-3). Previous studies found that the Npro-mediated IRF-3 degradation was dependent of a TRASH domain in the C-terminal half of Npro coordinating zinc by means of the amino acid residues C112, C134, D136 and C138. Interestingly, four classical swine fever virus (CSFV) isolates obtained from diseased pigs in Thailand in 1993 and 1998 did not suppress IFN-α/ß induction despite the presence of an intact TRASH domain. Through systematic analyses, it was found that an amino acid mutation at position 40 or mutations at positions 17 and 61 in the N-terminal half of Npro of these four isolates were related to the lack of IRF-3-degrading activity. Restoring a histidine at position 40 or both a proline at position 17 and a lysine at position 61 based on the sequence of a functional Npro contributed to higher stability of the reconstructed Npro compared with the Npro from the Thai isolate. This led to enhanced interaction of Npro with IRF-3 along with its degradation by the proteasome. The results of the present study revealed that amino acid residues in the N-terminal domain of Npro are involved in the stability of Npro, in interaction of Npro with IRF-3 and subsequent degradation of IRF-3, leading to downregulation of IFN-α/ß production.


Subject(s)
Classical Swine Fever Virus/immunology , Endopeptidases/chemistry , Endopeptidases/immunology , Host-Pathogen Interactions , Interferon Regulatory Factors/antagonists & inhibitors , Interferon Type I/antagonists & inhibitors , Viral Proteins/chemistry , Viral Proteins/immunology , Amino Acid Substitution , Animals , Classical Swine Fever/virology , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/isolation & purification , DNA Mutational Analysis , Down-Regulation , Endopeptidases/genetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/immunology , Mutation, Missense , Protein Binding , Protein Interaction Mapping , Protein Stability , Protein Structure, Tertiary , Swine , Thailand , Viral Proteins/genetics
2.
PLoS Pathog ; 9(10): e1003704, 2013.
Article in English | MEDLINE | ID: mdl-24146623

ABSTRACT

Pestiviruses express their genome as a single polypeptide that is subsequently cleaved into individual proteins by host- and virus-encoded proteases. The pestivirus N-terminal protease (N(pro)) is a cysteine autoprotease that cleaves between its own C-terminus and the N-terminus of the core protein. Due to its unique sequence and catalytic site, it forms its own cysteine protease family C53. After self-cleavage, N(pro) is no longer active as a protease. The released N(pro) suppresses the induction of the host's type-I interferon-α/ß (IFN-α/ß) response. N(pro) binds interferon regulatory factor-3 (IRF3), the key transcriptional activator of IFN-α/ß genes, and promotes degradation of IRF3 by the proteasome, thus preventing induction of the IFN-α/ß response to pestivirus infection. Here we report the crystal structures of pestivirus N(pro). N(pro) is structurally distinct from other known cysteine proteases and has a novel "clam shell" fold consisting of a protease domain and a zinc-binding domain. The unique fold of N(pro) allows auto-catalysis at its C-terminus and subsequently conceals the cleavage site in the active site of the protease. Although many viruses interfere with type I IFN induction by targeting the IRF3 pathway, little information is available regarding structure or mechanism of action of viral proteins that interact with IRF3. The distribution of amino acids on the surface of N(pro) involved in targeting IRF3 for proteasomal degradation provides insight into the nature of N(pro)'s interaction with IRF3. The structures thus establish the mechanism of auto-catalysis and subsequent auto-inhibition of trans-activity of N(pro), and its role in subversion of host immune response.


Subject(s)
Classical Swine Fever Virus/enzymology , Cysteine Proteases/chemistry , Interferon Type I , Protein Folding , Animals , Catalysis , Catalytic Domain , Classical Swine Fever Virus/genetics , Crystallography, X-Ray , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Structure-Activity Relationship , Swine
3.
Vaccine ; 29(7): 1491-503, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21184857

ABSTRACT

Virus replicon particles (VRP) are genetically engineered infectious virions incapable of generating progeny virus due to partial or complete deletion of at least one structural gene. VRP fulfil the criteria of a safe vaccine and gene delivery system. With VRP derived from classical swine fever virus (CSF-VRP), a single intradermal vaccination protects from disease. Spreading of the challenge virus in the host is however not completely abolished. Parameters that are critical for immunogenicity of CSF-VRP are not well characterized. Considering the importance of type I interferon (IFN-α/ß) to immune defence development, we generated IFN-α/ß-inducing VRP to determine how this would influence vaccine efficacy. We also evaluated the effect of co-expressing granulocyte macrophage colony-stimulating factor (GM-CSF) in the vaccine context. The VRP were capable of long-term replication in cell culture despite the presence of IFN-α/ß. In vivo, RNA replication was essential for the induction of an immune response. IFN-α/ß-inducing and GM-CSF-expressing CSF-VRP were similar to unmodified VRP in terms of antibody and peripheral T-cell responses, and in reducing the blood levels of challenge virus RNA. Importantly, the IFN-α/ß-inducing VRP did show increased efficacy over the unmodified VRP in terms of B-cell and T-cell responses, when tested with secondary immune responses by in vitro restimulation assay.


Subject(s)
Classical Swine Fever Virus/immunology , Classical Swine Fever/prevention & control , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antibody Formation , Cell Line , Classical Swine Fever/immunology , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interferon-alpha/genetics , Interferon-beta/genetics , RNA, Viral/biosynthesis , RNA, Viral/blood , Replicon , Swine , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/genetics , Virus Replication
4.
J Mol Biol ; 391(2): 438-49, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19540847

ABSTRACT

Pestiviruses, such as bovine viral diarrhea virus and classical swine fever virus (CSFV), use the viral protein N(pro) to subvert host cell antiviral responses. N(pro) is the first protein encoded by the single large open reading frame of the pestivirus positive-sense RNA genome and has an autoproteolytic activity, cleaving itself off from the polyprotein. N(pro) also targets interferon regulatory factor 3 (IRF3), a transcription factor for alpha/beta interferon genes, and promotes its proteasomal degradation, a process that is independent of the proteolytic activity of N(pro). We determined that N(pro) contains a novel metal-binding TRASH motif consisting of Cys-X(21)-Cys-X(3)-Cys (where X is any amino acid) at its C-terminus. We also found that N(pro) coordinates a single zinc atom as determined by graphite furnace-atomic absorption spectrophotometry and inductively coupled plasma-mass spectrometry. Mutational and biochemical analyses show that the cysteine residues in the TRASH motif are required for zinc binding and protein stability. Individual substitutions of the cysteines in the TRASH motif of CSFV N(pro) abolished the interaction of N(pro) with IRF3 and resulted in the loss of virus-mediated IRF3 degradation in CSFV-infected cells. Thus, the zinc-binding ability of N(pro) in pestiviruses appears to be essential for the virus-mediated degradation of IRF3.


Subject(s)
Classical Swine Fever Virus/metabolism , Diarrhea Viruses, Bovine Viral/metabolism , Interferon Regulatory Factor-3/metabolism , Metalloproteins/metabolism , Nucleocapsid Proteins/metabolism , Zinc/metabolism , Amino Acid Sequence , Animals , Aspartic Acid/chemistry , Aspartic Acid/genetics , Aspartic Acid/metabolism , Binding Sites , Cell Line , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Genes, Reporter , Interferon Regulatory Factor-3/chemistry , Interferon Regulatory Factor-3/genetics , Metalloproteins/chemistry , Metalloproteins/genetics , Molecular Sequence Data , Mutation , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Protein Stability , Protein Structure, Tertiary , Zinc/chemistry
5.
J Virol ; 83(2): 817-29, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18987150

ABSTRACT

Pestiviruses prevent alpha/beta interferon (IFN-alpha/beta) production by promoting proteasomal degradation of interferon regulatory factor 3 (IRF3) by means of the viral N(pro) nonstructural protein. N(pro) is also an autoprotease, and its amino-terminal coding sequence is involved in translation initiation. We previously showed with classical swine fever virus (CSFV) that deletion of the entire N(pro) gene resulted in attenuation in pigs. In order to elaborate on the role of the N(pro)-mediated IRF3 degradation in classical swine fever pathogenesis, we searched for minimal amino acid substitutions in N(pro) that would specifically abrogate this function. Our mutational analyses showed that degradation of IRF3 and autoprotease activity are two independent but structurally overlapping functions of N(pro). We describe two mutations in N(pro) that eliminate N(pro)-mediated IRF3 degradation without affecting the autoprotease activity. We also show that the conserved standard sequence at these particular positions is essential for N(pro) to interact with IRF3. Surprisingly, when these two mutations are introduced independently in the backbones of highly and moderately virulent CSFV, the resulting viruses are not attenuated, or are only partially attenuated, in 8- to 10-week-old pigs. This contrasts with the fact that these mutant viruses have lost the capacity to degrade IRF3 and to prevent IFN-alpha/beta induction in porcine cell lines and monocyte-derived dendritic cells. Taken together, these results demonstrate that contrary to previous assumptions and to the case for other viral systems, impairment of IRF3-dependent IFN-alpha/beta induction is not a prerequisite for CSFV virulence.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/pathogenicity , Endopeptidases/genetics , Endopeptidases/metabolism , Interferon Regulatory Factor-3/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Amino Acid Substitution/genetics , Animals , Cell Line , Mutagenesis, Site-Directed , Mutation, Missense , Swine , Virulence
6.
Virology ; 365(2): 376-89, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17482232

ABSTRACT

The nonstructural protein NS2-3 of pestiviruses undergoes tightly regulated processing. For bovine viral diarrhea virus it was shown that uncleaved NS2-3 is required for infectious particle formation while cleaved NS3 is essential for genome replication. To further investigate the functions of NS2-3 and NS4A in the pestivirus life cycle, we established T7 RNA polymerase-dependent trans-complementation for p7-NS2-3-4A of classical swine fever virus (CSFV). Expression of NS2-3 and NS4A in trans restored the production of infectious particles from genomes lacking NS2-3 expression. Co-expression of cleaved NS4A was essential. None of the enzymatic activities harbored by NS2-3 were required for infectious particle formation. Importantly, expression of uncleavable NS2-3 together with NS4A rescued infectious particles from a genome lacking NS2, demonstrating that cleaved NS2 per se has no additional essential function. These data indicate that NS2-3 and NS3, each in association with NS4A, have independent functions in the CSFV life cycle.


Subject(s)
Classical Swine Fever Virus/physiology , Viral Nonstructural Proteins/physiology , Virus Replication/physiology , Animals , Cell Line , Classical Swine Fever Virus/genetics , Genetic Complementation Test , Immunohistochemistry , Models, Biological , Swine , Viral Nonstructural Proteins/analysis , Viral Nonstructural Proteins/genetics , Virion/chemistry , Virus Replication/genetics
7.
J Virol ; 81(7): 3087-96, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17215286

ABSTRACT

Viruses have evolved a multitude of strategies to subvert the innate immune system by interfering with components of the alpha/beta interferon (IFN-alpha/beta) induction and signaling pathway. It is well established that the pestiviruses prevent IFN-alpha/beta induction in their primary target cells, such as epitheloidal and endothelial cells, macrophages, and conventional dendritic cells, a phenotype mediated by the viral protein N(pro). Central players in the IFN-alpha/beta induction cascade are interferon regulatory factor 3 (IRF3) and IRF7. Recently, it was proposed that classical swine fever virus (CSFV), the porcine pestivirus, induced the loss of IRF3 by inhibiting the transcription of IRF3 mRNA. In the present study, we show that endogenous IRF3 and IRF3 expressed from a cytomegalovirus (CMV) promoter are depleted in the presence of CSFV by means of N(pro), while CSFV does not inhibit CMV promoter-driven protein expression. We also demonstrate that CSFV does not reduce the transcriptional activity of the IRF3 promoter and does not affect the stability of IRF3 mRNA. In fact, CSFV N(pro) induces proteasomal degradation of IRF3, as demonstrated by proteasome inhibition studies. Furthermore, N(pro) coprecipitates with IRF3, suggesting that the proteasomal degradation of IRF3 is induced by a direct or indirect interaction with N(pro). Finally, we show that N(pro) does not downregulate IRF7 expression.


Subject(s)
Classical Swine Fever Virus/metabolism , Endopeptidases/metabolism , Interferon Regulatory Factor-3/metabolism , Proteasome Endopeptidase Complex/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Classical Swine Fever Virus/genetics , Cytomegalovirus/genetics , Endopeptidases/genetics , Gene Expression Regulation , Humans , Interferon Regulatory Factor-3/genetics , Molecular Sequence Data , Phosphorylation , Promoter Regions, Genetic/genetics , Proteasome Inhibitors , Protein Binding , RNA, Messenger/genetics , Signal Transduction , Swine , Viral Proteins/genetics
8.
Vet Res ; 37(5): 655-70, 2006.
Article in English | MEDLINE | ID: mdl-16777037

ABSTRACT

Classical swine fever virus replicon particles (CSF-VRP) deficient for E(rns) were evaluated as a non-transmissible marker vaccine. A cDNA clone of CSFV strain Alfort/187 was used to obtain a replication-competent mutant genome (replicon) lacking the sequence encoding the 227 amino acids of the glycoprotein E(rns) (A187delE(rns)). For packaging of A187delE(rns) into virus particles, porcine kidney cell lines constitutively expressing E(rns) of CSFV were established. The rescued VRP were infectious in cell culture but did not yield infectious progeny virus. Single intradermal vaccination of two pigs with 10(7) TCID(50) of VRP A187delE(rns) elicited neutralizing antibodies, anti-E2 antibodies, and cellular immune responses determined by an increase of IFN-gamma producing cells. No anti-E(rns) antibodies were detected in the vaccinees confirming that this vaccine represents a negative marker vaccine allowing differentiation between infected and vaccinated animals. The two pigs were protected against lethal challenge with the highly virulent CSFV strain Eystrup. In contrast, oral immunization resulted in only partial protection, and neither CSFV-specific antibodies nor stimulated T-cells were found before challenge. These data represent a good basis for more extended vaccination/challenge trials including larger numbers of animals as well as more thorough analysis of virus shedding using sentinel animals to monitor horizontal spread of the challenge virus.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/immunology , Classical Swine Fever/prevention & control , Classical Swine Fever/virology , Gene Deletion , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Cell Line , Classical Swine Fever/immunology , Genes, Viral , Genetic Markers , Injections, Intradermal/veterinary , Swine , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
9.
Virology ; 340(2): 265-76, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16043207

ABSTRACT

Classical swine fever virus (CSFV) protects cells from double-stranded (ds) RNA-mediated apoptosis and IFN-alpha/beta induction. This phenotype is lost when CSFV lacks N(pro) (DeltaN(pro) CSFV). In the present study, we demonstrate that N(pro) counteracts dsRNA-mediated apoptosis and IFN-alpha/beta induction independently of other CSFV elements. For this purpose, we generated porcine SK-6 and PK-15 cell lines constitutively expressing N(pro) fused to the enhanced green fluorescent protein (EGFP). The survival of the SK6-EGFP-N(pro) cell line after polyinosinic polycytidylic acid [poly(IC)] treatment was comparable to that of CSFV-infected SK-6 cells and was significantly higher than the survival of the parent cell line. In PK-15 cells, the presence of EGFP-N(pro) prevented the DeltaN(pro) CSFV- and poly(IC)-mediated IFN-alpha/beta production. Importantly, N(pro) also inhibited IFN-alpha and IFN-beta promoter-driven luciferase expression in human cells and blocked IFN-alpha/beta induction mediated by Newcastle disease virus. This establishes a novel function for N(pro) in counteraction of the IFN-alpha/beta induction pathway.


Subject(s)
Apoptosis/physiology , Classical Swine Fever Virus/physiology , Endopeptidases/pharmacology , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , RNA, Double-Stranded/pharmacology , Viral Proteins/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , DNA Primers , Genes, Reporter , Humans , Kidney , Plasmids , Poly I-C/genetics , Poly I-C/metabolism , Poly I-C/pharmacology , RNA, Double-Stranded/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Viral/pharmacology
10.
Vaccine ; 23(25): 3318-28, 2005 May 09.
Article in English | MEDLINE | ID: mdl-15837238

ABSTRACT

A cDNA clone of the classical swine fever virus (CSFV) strain Alfort/187 [Ruggli N, Tratschin JD, Mittelholzer C, Hofmann MA. Nucleotide sequence of classical swine fever virus strain Alfort/187 and transcription of infectious RNA from stably cloned full-length cDNA. J Virol 1996;70(6):3478-87] was used to construct two E2 deletion mutants lacking either the complete E2 gene or, alternatively, a stretch of 204 nucleotides encoding 68 amino acids located in the C-terminal region of the E2 glycoprotein. The respective in vitro synthesized mutant RNAs replicated in SK-6 cells but no infectious virus was generated. Both replicons could be packaged into virus particles in SK-6 cells constitutively expressing E2 of CSFV. For the resulting CSF virus replicon particles (CSF-VRP) A187-E2del373 and A187-E2del68 titers of 10(6) and 10(7) TCID(50)/ml, respectively, were obtained. Oronasal vaccination with 10(7) TCID(50) of either of the two CSF-VRP protected pigs against a challenge with a lethal dose of CSFV strain Eystrup. In contrast, after intradermal vaccination VRP A187-E2del68 but not VRP A187-E2del373 lacking the complete E2 gene induced a protective immune response. We conclude that E2-complemented CSF-VRP have the potential to be used as live-attenuated non-transmissible oral vaccines for pigs. In addition, our data suggest that E2 of CSFV is dispensable for the induction of mucosal but not of parenteral immunity.


Subject(s)
Classical Swine Fever Virus/immunology , Classical Swine Fever/immunology , Classical Swine Fever/prevention & control , Viral Envelope Proteins/genetics , Viral Vaccines/immunology , Administration, Intranasal , Administration, Oral , Animals , Cells, Cultured , Gene Deletion , Injections, Intradermal , RNA, Viral/analysis , RNA, Viral/biosynthesis , Swine , Vaccines, Synthetic/immunology
11.
Vaccine ; 22(3-4): 317-28, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14670312

ABSTRACT

We have reported earlier that replacement of the N(pro) gene of classical swine fever virus (CSFV) by the murine ubiquitin gene only slightly affects the characteristics of virus replication in the porcine kidney cell line SK-6 [J. Virol. 72 (1998) 7681]. Here, for the moderately virulent CSFV strain Alfort/187 as well as for the highly virulent strain Eystrup we show that the respective N(pro)-deleted viruses are attenuated. Vaccination of pigs with either of the two deletion mutants resulted in the induction of a strong antibody response. Animals were protected against challenge with a lethal dose of highly virulent CSFV indicating that N(pro) deletion mutants are excellent candidates for a modified live virus vaccine. A chimeric virus obtained by replacement of the N(pro) gene in the Eystrup virus by the corresponding sequence of the avirulent CSFV vaccine strain Riems resulted in a virus that was highly virulent. This indicates that the virulence of CSFV correlates with the presence of N(pro) and also suggests that N(pro) is not responsible for the varying virulence observed between individual strains of CSFV.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/immunology , Genes, Viral/genetics , Genes, Viral/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology , Animals , Cell Line , Classical Swine Fever/prevention & control , Classical Swine Fever/virology , Classical Swine Fever Virus/pathogenicity , Cloning, Molecular , Kinetics , Mice , Mutation/genetics , RNA/biosynthesis , Swine , Transcription, Genetic/genetics , Transfection , Ubiquitin/genetics , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virulence Factors/biosynthesis , Virulence Factors/genetics , Virus Replication/genetics
12.
Virus Res ; 98(2): 105-16, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14659557

ABSTRACT

The virulence of classical swine fever virus (CSFV) strains including established laboratory strains as well as field isolates ranges from avirulent to highly virulent. Here, we describe the construction and characterisation of two cDNA-derived CSFV strains, each corresponding to one of these extremes. The recombinant virus vEy-37 caused acute disease indistinguishable from that provoked by infection with the highly virulent parent strain Eystrup. In contrast, vRiems-3, a molecular clone of the CSFV vaccine strain Riems, was avirulent and induced protective immunity in pigs. After repeated passage of vEy-37 in porcine kidney SK-6 cells adaptive mutations in the E(rns) gene were observed. The respective reconstructed mutant virus grew to titres that were almost 4log units higher when compared to vEy-37. The mutation in the E(rns) gene had only a minor effect on the virulence of the virus. The complete genomic sequences of the two CSFV strains, Eystrup and Riems, have been deposited in GenBank (accession number AF326963 for CSFV Eystrup, AY259122 for CSFV Riems/IVI).


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever/virology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Classical Swine Fever/epidemiology , Classical Swine Fever/etiology , Classical Swine Fever/pathology , Classical Swine Fever Virus/classification , Classical Swine Fever Virus/pathogenicity , Classical Swine Fever Virus/physiology , Molecular Sequence Data , Phylogeny , Swine , Virulence , Virus Replication/physiology
13.
J Virol ; 77(13): 7645-54, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12805464

ABSTRACT

Classical swine fever virus (CSFV) replicates efficiently in cell lines and monocytic cells, including macrophages (MPhi), without causing a cytopathic effect or inducing interferon (IFN) secretion. In the present study, the capacity of CSFV to interfere with cellular antiviral activity was investigated. When the porcine kidney cell line SK-6 was infected with CSFV, there was a 100-fold increased capacity to resist to apoptosis induced by polyinosinic-polycytidylic acid [poly(IC)], a synthetic double-stranded RNA. In MPhi, the virus infection inhibited poly(IC)-induced alpha/beta IFN (type I IFN) synthesis. This interference with cellular antiviral defense correlated with the presence of the viral N(pro) gene. Mutants lacking the N(pro) gene (DeltaN(pro) CSFV) did not protect SK-6 cells from poly(IC)-induced apoptosis, despite growth properties and protein expression levels similar to those of the wild-type virus. Furthermore, DeltaN(pro) CSFV did not prevent poly(IC)-induced type I IFN production in MPhi but rather induced type I IFN in the absence of poly(IC) in both MPhi and the porcine kidney cell line PK-15, but not in SK-6 cells. With MPhi and PK-15, an impaired replication of the DeltaN(pro) CSFV compared with wild-type virus was noted. In addition, DeltaN(pro) CSFV, but not wild-type CSFV, could interfere with vesicular stomatitis virus replication in PK-15 cells. Taken together, these results provide evidence for a novel function associated with CSFV N(pro) with respect to the inhibition of the cellular innate immune system.


Subject(s)
Antiviral Agents/physiology , Classical Swine Fever Virus/physiology , Animals , Apoptosis/physiology , Cell Line , Immunohistochemistry , Swine , Virus Replication
14.
Clin Diagn Lab Immunol ; 9(6): 1183-91, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12414748

ABSTRACT

Two types of porcine reproductive and respiratory syndrome virus (PRRSV) have been reported, the European type (EU PRRSV) and the North American type (US PRRSV). We developed a dual enzyme-linked immunosorbent assay (ELISA) for the simultaneous detection and differentiation of serum antibodies directed against either of the two PRRSV types. This tandem PRRS ELISA is based on affinity-purified recombinant nucleocapsid protein expressed in Escherichia coli. Sensitivity and specificity were assessed by using the IDEXX HerdChek PRRS ELISA and the indirect immunofluorescence assay as reference tests. A total of 1571 sera originating from the United States, Europe, and two PRRS-free countries, i.e., Switzerland and New Zealand, were used for validation of the tandem PRRS ELISA. The new test performed at least as well as the reference tests in regard to sensitivity (0.94 for the US PRRS ELISA and 0.93 for the EU PRRS ELISA) and specificity (0.96 for the US PRRS ELISA and 0.99 for the EU PRRS ELISA). Positive sera were correctly differentiated in 582 of 591 cases, indicating a high differentiation capability of this dual ELISA. The robustness and repeatability of the test were assessed and found to be appropriate for diagnostic applications. Taken together, the data indicate that the tandem PRRS ELISA described here is the first differentiation ELISA for PRRSV serology based on recombinant antigen. It is convenient with respect to antigen production, and it is reliable, economical, and highly sensitive and specific. Thus, it is considered to be a powerful tool for routine diagnostics, epidemiological surveys, and outbreak investigations.


Subject(s)
Antibodies, Viral/blood , Enzyme-Linked Immunosorbent Assay/methods , Nucleocapsid Proteins/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Amino Acid Sequence , Animals , Cloning, Molecular , Molecular Sequence Data , Nucleocapsid Proteins/isolation & purification , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Reproducibility of Results , Swine
15.
Virus Res ; 85(2): 173-85, 2002 May 10.
Article in English | MEDLINE | ID: mdl-12034484

ABSTRACT

Bicistronic genomes of the classical swine fever virus (CSFV) strain Alfort/187 (A187) were established by insertion of a second cistron consisting of an internal ribosome entry site of the encephalomyocarditis virus and a coding sequence in the 3' untranslated region of the genome. Introduction of the selectable marker gene for neomycin phosphotransferase into the second cistron of the CSFV replicon A187 Delta E2-CAT allowed the establishment of porcine SK-6 cell lines constitutively expressing the respective bicistronic replicon RNA. In cells transfected with RNA representing the full-length viral genome and containing the gene coding for bacterial enhanced green fluorescence protein (EGFP) in the second cistron infectious bicistronic virus was synthesized. Expression of EGFP in cells infected with this virus indicated the potential of CSFV as a viral vector. Finally, after insertion of the sequence encoding the signal peptide of the CSFV E2 protein followed either by the E2 or the E2-p7 sequence into the replicon A187 Delta E2 which carries an in frame deletion of 465 nucleotides in the E2 gene, infectious viruses vA187 Delta E2-IRES-sigE2 and vA187 Delta E2-IRES-sigE2p7, respectively, were obtained. This shows that E2 deletion mutants can be complemented by expression of E2 from a separate cistron.


Subject(s)
Classical Swine Fever Virus/genetics , Gene Expression , Genes, Viral , Genetic Vectors/genetics , Viral Envelope Proteins/genetics , Viral Structural Proteins/genetics , Animals , Cell Line , Genes, Reporter , Genetic Complementation Test , Genome, Viral , Green Fluorescent Proteins , Luminescent Proteins/genetics , RNA, Viral , Replicon , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...