Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Osteoarthritis Cartilage ; 30(1): 52-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34626797

ABSTRACT

OBJECTIVE: Inflammatory hand arthritis (IHA) results in impaired function. Local gene therapy with ART-I02, a recombinant adeno-associated virus (AAV) serotype 5 vector expressing interferon (IFN)-ß, under the transcriptional control of nuclear factor κ-B responsive promoter, was preclinically shown to have favorable effects. This study aimed to investigate the safety and tolerability of local gene therapy with ART-I02 in patients with IHA. METHODS: In this first-in-human, dose-escalating, cohort study, 12 IHA patients were to receive a single intra-articular (IA) injection of ART-I02 ranging 0.3 × 1012-1.2 × 1013 genome copies in an affected hand joint. Adverse events (AEs), routine safety laboratory and the clinical course of disease were periodically evaluated. Baseline- and follow-up contrast enhanced magnetic resonance images (MRIs), shedding of viral vectors in bodily fluids, and AAV5 and IFN-ß immune responses were evaluated. A data review committee provided safety recommendations. RESULTS: Four patients were enrolled. Long-lasting local AEs were observed in 3 patients upon IA injection of ART-I02. The AEs were moderate in severity and could be treated conservative. Given the duration of the AEs and their possible or probable relation to ART-I02, no additional patients were enrolled. No systemic treatment emergent AEs were observed. The MRIs reflected the AEs by (peri)arthritis. No T-cell response against AAV5 or IFN-ß, nor IFN-ß antibodies could be detected. Neutralizing antibody titers against AAV5 raised post-dose. CONCLUSION: Single IA doses of 0.6 × 1012 or 1.2 × 1012 ART-I02 vector genomes were administered without systemic side effects or serious AEs. However, local tolerability was insufficient for continuation. TRIAL REGISTRATION: NCT02727764.


Subject(s)
Arthritis/therapy , Dependovirus , Genetic Therapy/methods , Genetic Vectors , Hand Joints , Interferon-beta/administration & dosage , Aged , Cohort Studies , Dependovirus/metabolism , Female , Genetic Therapy/adverse effects , Humans , Interferon-beta/biosynthesis , Middle Aged
2.
J Virol ; 79(19): 12495-506, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16160177

ABSTRACT

Virions of porcine reproductive and respiratory syndrome virus (PRRSV) contain six membrane proteins: the major proteins GP5 and M and the minor proteins GP2a, E, GP3, and GP4. Here, we studied the envelope protein requirements for PRRSV particle formation and infectivity using full-length cDNA clones in which the genes encoding the membrane proteins were disrupted by site-directed mutagenesis. By transfection of RNAs transcribed from these cDNAs into BHK-21 cells and analysis of the culture medium using ultracentrifugation, radioimmunoprecipitation, and real-time reverse transcription-PCR, we observed that the production of viral particles is dependent on both major envelope proteins; no particles were released when either the GP5 or the M protein was absent. In contrast, particle production was not dependent on the minor envelope proteins. Remarkably, in the absence of any one of the latter proteins, the incorporation of all other minor envelope proteins was affected, indicating that these proteins interact with each other and are assembled into virions as a multimeric complex. Independent evidence for such complexes was obtained by coexpression of the minor envelope proteins in BHK-21 cells using a Semliki Forest virus expression system. By analyzing the maturation of their N-linked oligosaccharides, we found that the glycoproteins were each retained in the endoplasmic reticulum unless expressed together, in which case they were collectively transported through the Golgi complex to the plasma membrane and were even detected in the extracellular medium. As the PRRSV particles lacking the minor envelope proteins are not infectious, we hypothesize that the virion surface structures formed by these proteins function in viral entry by mediating receptor binding and/or virus-cell fusion.


Subject(s)
Porcine respiratory and reproductive syndrome virus/physiology , Viral Envelope Proteins/physiology , Virus Assembly , Animals , Cell Line , Cricetinae , Immunoprecipitation , Membrane Glycoproteins/analysis , Mutagenesis, Site-Directed , Nucleocapsid/chemistry , Polymerase Chain Reaction , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/growth & development , Ultracentrifugation , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
3.
Neth J Med ; 63(1): 14-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15719847

ABSTRACT

Lipoprotein lipase (LPL) deficiency is a rare, hereditary disorder of lipoprotein metabolism characterised by severely increased triglyceride levels, and associated with an increased risk for pancreatitis. Since no adequate treatment modality is available for this disorder, we set out to develop an LPL gene therapy protocol. This paper focuses on the clinical presentation of LPL deficiency, summarises the preclinical investigations in animal models and describes the rationale to evaluate gene therapy for this monogenetic disorder of lipid metabolism in humans.


Subject(s)
Genetic Therapy , Hyperlipoproteinemia Type I/therapy , Animals , Diet , Genetic Therapy/methods , Genetic Vectors , Humans , Hyperlipoproteinemia Type I/genetics , Hypertriglyceridemia/genetics , Lipoprotein Lipase/genetics , Mutation , Pancreatitis/genetics , Prevalence , Risk Factors , Xanthomatosis/genetics
4.
J Gen Virol ; 85(Pt 12): 3715-3723, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15557245

ABSTRACT

The arterivirus porcine reproductive and respiratory syndrome virus (PRRSV) contains four glycoproteins, GP(2a), GP(3), GP(4) and GP(5), the functions of which are still largely unresolved. In this study, the significance of the N-glycosylation of the GP(2a) and GP(5) proteins of PRRSV strain LV was investigated. Both glycoproteins contain two predicted N-glycosylation sites that are highly conserved between North American-type and European-type PRRSV. Using site-directed mutagenesis, single and double mutant full-length PRRSV cDNA clones were generated. After analysing the expression of the mutant proteins and the actual use of the four putative glycosylation sites in the wild-type proteins, the production of mutant virus particles and their infectivities were investigated. The results showed that the N-linked glycans normally present on the GP(2a) protein are not essential for particle formation, as is the oligosaccharide attached to N53 of the GP(5) protein. In contrast, the oligosaccharide linked to N46 of the GP(5) protein is strongly required for virus particle production. The specific infectivities of the mutant viruses were investigated by comparing their infectivity-per-particle ratios with that of wild-type virus. The results showed that the lack of either one or both of the N-linked oligosaccharides on GP(2a) or of the oligosaccharide attached to N53 of GP(5) did not significantly affect the infectivities of the viruses. In contrast, the two recombinant viruses lacking the oligosaccharide bound to N46 exhibited a significantly reduced specific infectivity compared with the wild-type virus. The implications of the differential requirements of the modifications of GP(2a) and GP(5) for PRRSV assembly and infectivity are discussed.


Subject(s)
Oligosaccharides/physiology , Porcine respiratory and reproductive syndrome virus/physiology , Viral Envelope Proteins/physiology , Animals , Cricetinae , Glycosylation , Mutagenesis , Mutation , Viral Envelope Proteins/chemistry , Virion/physiology
5.
J Gen Virol ; 84(Pt 6): 1535-1543, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12771423

ABSTRACT

A set of neutralizing monoclonal antibodies (mAbs) directed against the GP(5) protein of European type porcine reproductive and respiratory syndrome virus (PRRSV) has been produced previously (Weiland et al., 1999). This set reacted with a plaque-purified virus (PPV) subpopulation of Dutch isolate Intervet-10 (I-10), but not with the European prototype PRRSV LV. In order to map the neutralization epitope in the GP(5) protein of the PPV strain, the ORF5 nucleotide sequence of PPV was determined. When the amino acid sequence derived from this nucleotide sequence was compared with that of PRRSV LV, four amino acid differences were found. Using site-directed mutagenesis, we showed that a proline residue at position 24 of the GP(5) sequence of the PPV strain enabled recognition by the neutralizing mAbs. Pepscan analysis demonstrated that the epitope recognized by the neutralizing mAbs stretched from residues 29 to 35. Surprisingly, the reactivity of the mAbs in the Pepscan system was independent of the presence of a proline in position 24. Moreover, residue 24 is located within the predicted signal peptide, implying that either the signal peptide is not cleaved or is cleaved due to the presence of Pro(24) such that the epitope remains intact. Our results demonstrate the presence of a neutralization epitope in the N-terminal ectodomain of the GP(5) protein of PRRSV and imply a role for the ectodomain of GP(5) in the infection of PRRSV.


Subject(s)
Porcine respiratory and reproductive syndrome virus/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antibodies, Viral , Antigens, Viral/chemistry , Antigens, Viral/genetics , Base Sequence , Binding Sites/genetics , Cell Line , Cricetinae , DNA, Viral/genetics , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , In Vitro Techniques , Molecular Sequence Data , Mutation , Neutralization Tests , Porcine respiratory and reproductive syndrome virus/genetics , Proline/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sus scrofa , Transfection , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Viral Proteins/immunology
6.
Vaccine ; 21(19-20): 2556-63, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12744891

ABSTRACT

Three porcine reproductive and respiratory syndrome virus (PRRSV) recombinants, generated by mutagenesis of an infectious cDNA clone of the Lelystad virus (LV) isolate, were tested for their safety and protective efficacy as potential PRRSV vaccines in pigs. Recombinant vABV688 contains two amino acid substitutions in the minor structural protein GP(2) resulting in improved growth on cell line CL2621; in recombinant vABV707 the region encoding the ectodomain of the major unglycosylated membrane protein M has been replaced by that of the murine lactate dehydrogenase-elevating arterivirus; recombinant vABV746 lacks the six C-terminal amino acids of the nucleocapsid protein N. First, we determined the safety of these recombinant viruses by monitoring the stability of the introduced mutations in 8-week-old pigs. We showed that the introduced genomic mutations were maintained throughout the viraemic period. Second, the protective efficacy of immunization with the recombinant viruses against challenge with a homologous and a heterologous PRRSV strain was determined in two pigs and compared with the efficacy of vABV437, a virus derived from the parental LV cDNA. The viraemia in pigs immunized with the recombinant viruses was reduced compared to pigs immunized with vABV437. In addition, the length of viraemia was reduced in the sentinel pigs that were introduced into the groups immunized with vABV746, vABV688, and vABV707, however, all of the sentinel pigs became infected. Pigs immunized with vABV707 and vABV437 were protected against challenge with homologous virus LV-Ter Huurne and transmission of the latter virus. None of the immunized pigs were protected against heterologous challenge with the virulent US isolate SDSU#73, but the vABV707- and vABV746-immunized pigs were protected against transmission of this virus from challenged pigs. In conclusion, the obtained viral recombinants are interesting candidates to be further explored for their use as vaccines against PRRSV.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Vaccines, Synthetic/therapeutic use , Viral Vaccines/therapeutic use , Amino Acid Substitution , Animals , Base Sequence , Cells, Cultured , DNA Primers , Macrophages, Alveolar/cytology , Macrophages, Alveolar/virology , Reverse Transcriptase Polymerase Chain Reaction , Safety , Swine , Time Factors
7.
Arch Virol ; 148(1): 177-87, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12536303

ABSTRACT

The aim of this study was to identify the receptor(s) for PRRSV on porcine alveolar macrophages (PAMs) by producing monoclonal antibodies (MAbs) against these cells. Hybridoma supernatants were selected for their ability to block PRRSV infection. Four MAbs, 1-8D2, 9.4C7, 9.9F2, and 3-3H2 inhibited infection and recognised cell surface, PAM-specific antigens as shown by immunofluorescence and immunoperoxidase monolayer assay. These MAbs were then used to identify cellular proteins involved in PRRSV infection by radioimmunoprecipitation assays (RIPAs). MAbs 1-8D2 and 9.9F2 each recognised a 150 kDa-polypeptide doublet, while MAbs 9.4C7 and 3-3H2 both recognised a 220 kDa-polypeptide. Glycosidase treatment demonstrated all these polypeptides to be N-glycosylated. Thus, multiple glycoproteins appear to be involved in infection of PAMs by PRRSV.


Subject(s)
Glycoproteins/metabolism , Macrophages, Alveolar/chemistry , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Receptors, Virus/metabolism , Animals , Antibodies, Monoclonal/immunology , Hybridomas/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Porcine Reproductive and Respiratory Syndrome/immunology , Receptors, Virus/immunology , Swine
8.
Virology ; 303(2): 364-73, 2002 Nov 25.
Article in English | MEDLINE | ID: mdl-12490397

ABSTRACT

Arteriviruses are enveloped, positive-strand RNA viruses for which the two major envelope proteins GP(5) and M occur as disulfide-linked heterodimers. These were assumed to serve the viral targeting functions, but recent ectodomain swapping studies with equine arteritis virus (EAV) indicate that the GP(5) protein does not determine arteriviral tropism. Here, we focused on the short, 13- to 18-residue ectodomain of the M protein. Using an infectious cDNA clone of the Lelystad virus isolate of porcine reproductive and respiratory syndrome virus (PRRSV), we substituted the genomic sequence encoding the M ectodomain by that of murine lactate dehydrogenase-elevating virus, EAV, and the US PRRSV-isolate, VR2332. Viable viruses with a chimeric M protein were obtained in all three cases, but for the latter two only after removal of the genomic overlap between the M and GP(5) genes. Characterization of the chimeric viruses revealed that they could be distinguished immunologically from wild-type virus, that they were genetically stable in vitro, but that they were impaired in their growth, reaching lower titers than the parental virus. The latter appeared to be due to an increased particle-to-infectivity ratio of the chimeric virus particles. Interestingly, the chimeric viruses had retained their ability to infect porcine cells and had not acquired tropism for cells susceptible to the viruses from which the foreign ectodomains were derived. We conclude that the surface structures composed by the arterivirus M and GP(5) ectodomains do not determine viral tropism.


Subject(s)
Arterivirus/physiology , Recombinant Fusion Proteins/physiology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Animals , Arterivirus/genetics , Arterivirus/immunology , Base Sequence , Equartevirus/physiology , Lactate dehydrogenase-elevating virus/physiology , Molecular Sequence Data , Open Reading Frames , Porcine respiratory and reproductive syndrome virus/physiology , Swine , Transfection , Viral Envelope Proteins/physiology , Viral Matrix Proteins/chemistry
9.
J Virol ; 76(3): 1521-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11773426

ABSTRACT

We used an infectious cDNA clone of porcine reproductive and respiratory syndrome virus (PRRSV) to investigate the presence of essential replication elements in the region of the genome encoding the structural proteins. Deletion analysis showed that a stretch of 34 nucleotides (14653 to 14686) within ORF7, which encodes the nucleocapsid protein, is essential for RNA replication. Strand-specific reverse transcription-PCR analysis of viral RNA isolated from transfected BHK-21 cells revealed that this region is required for negative-strand genomic RNA synthesis. The 34-nucleotide stretch is highly conserved among PRRSV isolates and folds into a putative hairpin. A 7-base sequence within the loop of this structure was suggested to base-pair with a sequence present in the loop of a hairpin located in the 3' noncoding region, resulting in a kissing interaction. Mutational analyses confirmed that this kissing interaction is required for RNA replication.


Subject(s)
3' Untranslated Regions/physiology , Porcine respiratory and reproductive syndrome virus/genetics , RNA, Viral/physiology , Virus Replication/genetics , Animals , Cell Line , Cricetinae , Nucleic Acid Conformation , Nucleocapsid/genetics , Open Reading Frames , Porcine respiratory and reproductive syndrome virus/physiology , Swine , Viral Matrix Proteins/genetics , Viral Nonstructural Proteins/genetics
10.
J Gen Virol ; 82(Pt 11): 2607-2614, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602771

ABSTRACT

In order to obtain attenuated live vaccine candidates of porcine reproductive and respiratory syndrome virus (PRRSV), a series of deletions was introduced at the 3' end of the viral genome using an infectious cDNA clone of the Lelystad virus isolate. RNA transcripts from the full-length cDNA clones were transfected into BHK-21 cells. The culture supernatant of these cells was subsequently used to infect porcine alveolar macrophages to detect the production of progeny virus. It is shown that C-terminal truncation of the nucleocapsid (N) protein, encoded by ORF7, was tolerated for up to six amino acids without blocking the production of infectious virus. Mutants containing larger deletions produced neither virus nor virus-like particles containing viral RNA. Deletion analysis of the 3' UTR immediately downstream of ORF7 showed that infectious virus was still produced after removal of seven nucleotides behind the stop codon of ORF7. Deletion of 32 nucleotides in this region abolished RNA replication and, consequently, no infectious virus was formed. Serial passage on porcine alveolar macrophages demonstrated that the viable deletion mutants were genetically stable at the site of mutation. In addition, the deletions did not affect the growth properties of the recombinant viruses in vitro, while their antigenic profiles were similar to that of wild-type virus. Immunoprecipitation experiments with the six-residue N protein-deletion mutant confirmed that the truncated protein was indeed smaller than the wild-type N protein. The deletion mutants produced in this study are interesting candidate vaccines to prevent PRRS disease in pigs.


Subject(s)
Genome, Viral , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/genetics , Sequence Deletion , 3' Untranslated Regions , Animals , Base Sequence , Cells, Cultured , Cloning, Molecular , DNA, Complementary/genetics , Macrophages, Alveolar/virology , Molecular Sequence Data , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Open Reading Frames , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Sequence Analysis, DNA , Swine , Transfection
11.
Virology ; 278(2): 380-9, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11118361

ABSTRACT

The potential of porcine reproductive and respiratory syndrome virus (PRRSV) as a viral vector was explored by the insertion of a sequence encoding a foreign antigen into the infectious cDNA clone of the Lelystad virus isolate. An epitope of the hemagglutinin (HA) protein of human influenza A virus was introduced at the 5' end and at the 3' end of ORF7, in each case resulting in a fusion protein between the HA epitope and the nucleocapsid (N) protein. Furthermore, in the construct carrying the HA sequences at the 5' end of ORF7, additional in-frame insertions encoding the autoprotease 2A of foot-and-mouth disease virus were made between the HA and ORF7 sequences to ensure the generation of a functional N protein from its hybrid precursor. When RNA transcripts from these full-length cDNA clones were transfected into BHK-21 cells, they were each found to replicate, to express the HA epitope, and to produce progeny virus. However, fusion of the HA epitope directly to the nucleocapsid protein either at the N terminus or at the C terminus adversely affected both the viability and the genetic stability of the recombinant PRRS viruses. Serial passage of the recombinant viruses on porcine alveolar macrophages demonstrated that these viruses had lost (part of) the HA epitope at passage four. In contrast, in the PRRS viruses expressing the HA epitope from a precursor cleavable by the autoprotease 2A peptide, the HA epitope was still intact after four passages, and no effect on the viability of these viruses was observed. Immunoprecipitation and pulse chase experiments revealed the efficient and presumably cotranslational cleavage of the HA epitope from the N protein by the 2A protease. Our results demonstrate the feasibility of using PRRSV as a viral vector that might be suitable for the delivery of antigens from other pathogens to the immune system of the pig.


Subject(s)
Epitopes/genetics , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A virus/genetics , Porcine respiratory and reproductive syndrome virus/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cricetinae , DNA Primers , Epitopes/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleocapsid/genetics , Open Reading Frames , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Swine , Transcription, Genetic , Transfection
12.
Vet Res ; 31(1): 11-21, 2000.
Article in English | MEDLINE | ID: mdl-10726635

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is a positive-strand RNA virus that belongs to the Arteriviridae family. PRRSV grows in primary alveolar macrophages and in monkey kidney cell lines. The genomic RNA is approximately 15 kb. The genome encodes the RNA replicase (ORF1a and ORF1b), the glycoproteins GP2 to GP5, the integral membrane protein M, and the nucleocapsid protein N (ORFs 2 to 7). A comparison of nucleotide sequences of different strains indicates that European and North American strains represent two distinct antigenic types. Various PRRSV-specific monoclonal antibodies and recombinant structural proteins have been produced. Well-defined PRRSV mutants can be generated with the recently developed infectious cDNA clone of PRRSV.


Subject(s)
Porcine respiratory and reproductive syndrome virus , Animals , Genome, Viral , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , Viral Structural Proteins/immunology
13.
J Gen Virol ; 81(Pt 2): 497-505, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644849

ABSTRACT

Porcine reproductive and respiratory virus (PRRSV) primarily infects and destroys alveolar macrophages of the pig. The aim of the present study was to characterize the changes of leukocyte populations in the broncho-alveolar lavage fluid (BALF) of PRRSV-infected pigs. Piglets were inoculated intranasally with PRRSV strain LV ter Huurne. On various days post-infection the piglets were sacrificed and the lungs removed, washed semi-quantitatively and analysed by flow cytometry. The total number of recovered BALF cells increased approximately 10 times between day 10 and day 21 of infection and decreased thereafter. The number of small low-autofluorescent cells (SLAC), i.e. lymphocytic and monocytic cells, increased very strongly from day 2 until day 21 of infection; in contrast, the number of large highly autofluorescent cells (LHAC), i.e. mostly macrophages, remained constant until day 14 of infection, increased slightly on day 21 and then decreased. On day 21 of infection in specific-pathogen-free piglets approximately 60% of the SLAC consisted of CD2(+)CD8(+)CD4(-)gammadeltaTCR(-) cells, which were partly CD8(+)CD6(+) and partly CD8(+)CD6(-). These phenotypes correspond to that of cytotoxic T-cells and natural killer cells respectively. From these results we can conclude that during a PRRSV infection the total number of BALF cells increases mainly due to an influx of lymphocytic cells with a cytolytic phenotype.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , CD8-Positive T-Lymphocytes/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , CD8-Positive T-Lymphocytes/pathology , Cell Count , Flow Cytometry , Germ-Free Life , Immunophenotyping , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lung/immunology , Lung/pathology , Porcine Reproductive and Respiratory Syndrome/pathology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Specific Pathogen-Free Organisms , Swine
14.
Adv Exp Med Biol ; 440: 199-206, 1998.
Article in English | MEDLINE | ID: mdl-9782281

ABSTRACT

A plasmid containing a full-length cDNA copy of the Lelystad virus isolate (LV) of porcine reproductive and respiratory syndrome virus was constructed. When RNA that was transcribed in vitro from this full-length cDNA clone was transfected to BHK-21 cells, infectious LV was produced and secreted. The virus was rescued by passage to porcine alveolar lung macrophages or CL2621 cells. When infectious transcripts were transfected to porcine alveolar lung macrophages or CL2621 cells no infectious virus was produced due to the poor transfection efficiency of these cells. The growth properties of the viruses produced by BHK-21 cells transfected with infectious transcripts of LV cDNA resembled the growth properties of the parental virus from which the cDNA was derived. The infectious clone of LV enables us to mutagenize the viral genome at specific sites and thus will be useful for detailed molecular characterization of the virus, as well as for the development of a safe and effective live vaccine for use in pigs.


Subject(s)
DNA, Viral/biosynthesis , Porcine respiratory and reproductive syndrome virus/genetics , Animals , Cell Line , Cloning, Molecular , Cricetinae , DNA, Complementary , Genome, Viral , Porcine respiratory and reproductive syndrome virus/pathogenicity , RNA, Viral , Swine
16.
J Virol ; 72(1): 380-7, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9420236

ABSTRACT

The 5'-terminal end of the genomic RNA of the Lelystad virus isolate (LV) of porcine reproductive and respiratory syndrome virus was determined. To construct full-length cDNA clones, the 5'-terminal sequence was ligated to cDNA clones covering the complete genome of LV. When RNA that was transcribed in vitro from these full-length cDNA clones was transfected into BHK-21 cells, infectious LV was produced and secreted. The virus was rescued by passage to porcine alveolar lung macrophages or CL2621 cells. When infectious transcripts were transfected to porcine alveolar lung macrophages or CL2621 cells, no infectious virus was produced due to the poor transfection efficiency of these cells. The growth properties of the viruses produced by BHK-21 cells transfected with infectious transcripts of LV cDNA resembled the growth properties of the parental virus from which the cDNA was derived. Two nucleotide changes leading to a unique PacI restriction site directly downstream of the ORF7 gene were introduced in the genome-length cDNA clone. The virus recovered from this mutated cDNA clone retained the PacI site, which confirmed the de novo generation of infectious LV from cloned cDNA. These results indicate that the infectious clone of LV enables us to mutagenize the viral genome at specific sites and that it will therefore be useful for detailed molecular characterization of the virus, as well as for the development of a safe and effective live vaccine for use in pigs.


Subject(s)
DNA, Complementary/genetics , DNA, Viral/genetics , Porcine respiratory and reproductive syndrome virus/genetics , Animals , Base Sequence , Cell Line , Cloning, Molecular , Cricetinae , DNA Primers/genetics , Genetic Markers , Genome, Viral , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/pathogenicity , RNA, Viral/biosynthesis , RNA, Viral/genetics , Swine , Transcription, Genetic , Transfection
17.
Virology ; 252(1): 106-14, 1998 Dec 05.
Article in English | MEDLINE | ID: mdl-9875321

ABSTRACT

The purpose of this study was to analyze the antigenic structure of the nucleocapsid protein N of the Lelystad virus isolate of porcine reproductive and respiratory syndrome virus (PRRSV) and to identify antigenic differences between this prototype European isolate and other North American isolates. To do this, we generated a panel of monoclonal antibodies (mAbs) directed against the N protein of Lelystad virus and tested them in competition assays with other N-specific mAbs described previously (Drew et al., 1995; Nelson et al., 1993; van Nieuwstadt et al., 1996). Four different competition groups of mAbs were identified. Pepscan analysis with solid-phase dodecapeptides was used to identify specific antigenic regions in the N protein that were bound by the mAbs. In this pepscan analysis, we found that the mAb of the first competition group reacted with linear peptides whose core sequences consisted of amino acids 2-12 (site A), the mAbs of the second group reacted with peptides whose core sequences consisted of amino acids 25-30 (site B), and the mAb of the third group reacted with peptides whose core sequences consisted of amino acids 40-46 (site C). However, the fourth group of mAbs binding to an antigenic region, provisionally designated as domain D, reacted very weakly or did not react at all with solid-phase dodecapeptides. To further characterize the structure of the epitopes in domain D, we produced chimeric constructs composed of the N protein sequences of Lelystad virus and another arterivirus lactate dehydrogenase-elevating virus, which was used because its N protein has similarity in amino acid sequence and hydropathicity profile but does not react with our mAbs. When the mAbs specific to domain D were tested for binding to the chimeric N proteins expressed by Semliki Forest virus, we found that the regions between amino acids 51-67 and amino acids 80-90 are involved in the formation or are part of the epitopes in domain D. Therefore, we conclude that the N protein contains four distinct antigenic regions. The epitopes mapped to sites A-C are linear, whereas the epitopes mapped to domain D are more conformation dependent or discontinuous. Sites A and C contain epitopes that are conserved in European but not in North American isolates; site B contains epitopes that are conserved in European and North American isolates; and site D contains epitopes that are either conserved or not conserved in European and North American isolates. The antigenic regions identified here might be important for the development of diagnostic test for PRRSV in particular tests that discriminate between different antigenic types of PRRSV.


Subject(s)
Antibodies, Monoclonal/immunology , Nucleocapsid Proteins/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Amino Acid Sequence , Animals , Antibody Specificity , Binding Sites , Binding, Competitive , Cell Line , Cloning, Molecular , Cricetinae , DNA, Viral/chemistry , Epitope Mapping/methods , Mice , Molecular Sequence Data , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/genetics , Peptide Mapping/methods , Porcine respiratory and reproductive syndrome virus/chemistry , Porcine respiratory and reproductive syndrome virus/genetics , Recombinant Fusion Proteins/chemistry
18.
J Virol ; 71(8): 6061-7, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9223499

ABSTRACT

GP4 is a minor structural glycoprotein encoded by ORF4 of Lelystad virus (LV). When it was immunoprecipitated from cell lysates and extracellular virus of CL2621 cells infected with LV, it was shown to have an apparent molecular mass of approximately 28 and 31 kDa, respectively. This difference in size occurred because its core N-glycans were modified to complex type N-glycans during the transport of the protein through the endoplasmic reticulum and Golgi compartment. A panel of 15 neutralizing monoclonal antibodies (MAbs) reacted with the native GP4 protein expressed by LV and the recombinant GP4 protein expressed in a Semliki Forest virus expression system. However, these MAbs did not react with the GP4 protein of U.S. isolate VR2332. To map the binding site of the MAbs, chimeric constructs composed of ORF4 of LV and VR2332 were generated. The reactivity of these constructs indicated that all the MAbs were directed against a region spanning amino acids 40 to 79 of the GP4 protein of LV. Six MAbs reacted with solid-phase synthetic dodecapeptides. The core of this site consists of amino acids 59 to 67 (SAAQEKISF). Comparison of the amino acid sequences of GP4 proteins from various European and North American isolates indicated that the neutralization domain spanning amino acids 40 to 79 is the most variable region of GP4. The neutralization domain of GP4, described here, is the first identified for LV.


Subject(s)
Open Reading Frames , Porcine respiratory and reproductive syndrome virus/genetics , Protein Processing, Post-Translational , Viral Structural Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Molecular Sequence Data , Neutralization Tests , Rabbits , Swine , Viral Structural Proteins/biosynthesis , Viral Structural Proteins/chemistry
19.
Vet Microbiol ; 55(1-4): 197-202, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9220614

ABSTRACT

Lelystad virus (LV), the prototype of porcine reproductive respiratory syndrome virus, is a small enveloped virus, containing a positive strand RNA genome of 15 kb. LV is tentatively classified in the family Arteriviridae, which consists of lactate dehydrogenase-elevating virus (LDV), equine arteritis virus (EAV) and simian hemorrhagic fever virus (SHFV). These viruses have a similar genome organization and replication strategy as coronaviruses, but the size of the genome is much smaller (12-15 kb) and they have different morphological and physicochemical properties. The genome of LV contains eight open reading frames (ORFs) that encode the replicase genes (ORFs 1a and 1b), envelope proteins (ORFs 2 to 6) and the nucleocapsid protein (ORF7). Genomic comparison of European and North American isolates has shown that the structural proteins encoded by ORFs 2 to 7 vary widely. The amino acid sequences of ORFs 2 to 7 of North American strains share only 55 to 79% identical amino acids with those of European strains. Using polyvalent porcine anti-LV serum, gene-specific anti-peptide sera and monoclonal antibodies, we have identified six structural proteins of LV and their corresponding genes. These are: the 15 kDa unglycosylated nucleocapsid protein (N) encoded by ORF7, an 18 kDa unglycosylated integral membrane protein M encoded by ORF6, a 25 kDa N-glycosylated protein encoded by ORF5, a 31-35 kDa N-glycosylated protein encoded by ORF4, a 45-50 kDa N-glycosylated protein encoded by ORF3 and a 29-30 kDa N-glycosylated protein encoded by ORF2. A nomenclature for these structural proteins is proposed.


Subject(s)
Genome, Viral , Porcine respiratory and reproductive syndrome virus/genetics , Animals , Arterivirus/classification , Arterivirus/genetics , Europe , North America , Nucleocapsid/genetics , Open Reading Frames , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/isolation & purification , Swine , Viral Envelope Proteins/genetics , Viral Structural Proteins/genetics
20.
Virology ; 225(1): 44-51, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8918532

ABSTRACT

Previously we have shown that Lelystad virus (LV) contains five structural proteins, a nucleocapsid protein N, an integral membrane protein M, and three glycoproteins GP3, GP4, and GP5. In this study we identified a sixth structural protein of Lelystad virus. The protein has an apparent molecular weight of 29 to 30 kDa, was recognized by an ORF2-specific antipeptide serum in Western immunoblotting using sucrose gradient purified LV virions, and was shown to be N-glycosylated. It was therefore designated GP2. The GP2 protein was also immunoprecipitated by ORF2-specific antipeptide serum from lysates and extracellular virus of CL2621 cells infected with LV. A fraction of the GP2 protein present in these lysates contained an intrachain disulfide bond. Endoglycosidase H treatment of immunoprecipitates indicated that the endoglycosidase H-sensitive N-glycans of the GP2 protein become endoglycosidase H-resistant during passage through the Golgi compartment in cells infected with LV. In contrast, the N-glycans of the GP2 protein expressed individually in a recombinant Semliki Forest virus remained endoglycosidase H-sensitive, indicating that the GP2 protein was retained in the endoplasmic reticulum. LV is the first arterivirus which has been demonstrated to contain six virion-associated proteins, one nucleocapsid protein, one integral membrane protein, and four glycoproteins.


Subject(s)
Glycoproteins/chemistry , Open Reading Frames/genetics , Porcine respiratory and reproductive syndrome virus/chemistry , Viral Structural Proteins/chemistry , Alkylating Agents/pharmacology , Amidohydrolases , Amino Acid Sequence , Animals , Cell Line , Dimerization , Disulfides , Ethylmaleimide/pharmacology , Glycoproteins/analysis , Glycoproteins/genetics , Glycosylation , Hexosaminidases , Iodoacetamide/pharmacology , Molecular Sequence Data , Molecular Weight , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Porcine respiratory and reproductive syndrome virus/genetics , Precipitin Tests , Viral Structural Proteins/analysis , Viral Structural Proteins/genetics , Virion/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...