Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 102(8)2021 08.
Article in English | MEDLINE | ID: mdl-34356005

ABSTRACT

The family Arteriviridae comprises enveloped RNA viruses with a linear, positive-sense genome of approximately 12.7 to 15.7 kb. The spherical, pleomorphic virions have a median diameter of 50-74 nm and include eight to eleven viral proteins. Arteriviruses infect non-human mammals in a vector-independent manner. Infections are often persistent and can either be asymptomatic or produce overt disease. Some arteriviruses are important veterinary pathogens while others infect particular species of wild rodents or African non-human primates. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Arteriviridae, which is available at ictv.global/report/arteriviridae.


Subject(s)
Arteriviridae/classification , Arteriviridae/genetics , Phylogeny , Animals , Arteriviridae/ultrastructure , Arterivirus/classification , Arterivirus/genetics , Endocytosis , Genome, Viral , Primates , RNA Virus Infections , Viral Proteins/genetics , Virion/classification , Virion/genetics , Virion/ultrastructure , Virus Attachment , Virus Replication
2.
Arch Virol ; 164(10): 2593-2597, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31270606

ABSTRACT

Trionyx sinensis hemorrhagic syndrome virus (TSHSV) is a newly discovered lethal arterivirus that causes serious disease in Trionyx sinensis in China. In this study, the complete genome sequence of TSHSV was determined by RACE cloning, and the functions of the predicted proteins were predicted. The complete genome of TSHSV was found to be 17,875 bp in length, and a 3'-end poly(A) tail was detected. Eight TSHSV hypothetical proteins (TSHSV-HPs) were predicted by gene model identification. TSHSV-HP2, 3 and 4 were associated with replicase activity, since papain-like protease (PLPs), serine-type endopeptidase, P-loop-containing nucleoside triphosphate hydrolase, and EndoU-like endoribonuclease motifs were detected. Phylogenetic analysis showed that TSHSV clusters with an arterivirus from a Chinese broad-headed pond turtle.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/classification , Arterivirus/isolation & purification , Phylogeny , Turtles/virology , Animals , Arterivirus/genetics , Arterivirus Infections/virology , China , Genome, Viral , RNA, Messenger , Sequence Analysis, DNA , Viral Proteins/genetics
3.
J Biol Chem ; 293(31): 12054-12067, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29887523

ABSTRACT

Nidovirus endoribonucleases (NendoUs) include nonstructural protein 15 (nsp15) from coronaviruses and nsp11 from arteriviruses, both of which have been reported to participate in the viral replication process and in the evasion of the host immune system. Results from a previous study of coronaviruses SARS-CoV, HCoV-229E, and MHV nsp15 indicate that it mainly forms a functional hexamer, whereas nsp11 from the arterivirus PRRSV is a dimer. Here, we found that porcine Deltacoronavirus (PDCoV) nsp15 primarily exists as dimers and monomers in vitro Biological experiments reveal that a PDCoV nsp15 mutant lacking the first 27 amino acids of the N-terminal domain (Asn-1-Asn-27) forms more monomers and displays decreased enzymatic activity, indicating that this region is important for its dimerization. Moreover, multiple sequence alignments and three-dimensional structural analysis indicated that the C-terminal region (His-251-Val-261) of PDCoV nsp15 is 10 amino acids shorter and forms a shorter loop than that formed by the equivalent sequence (Gln-259-Phe-279) of SARS-CoV nsp15. This result may explain why PDCoV nsp15 failed to form hexamers. We speculate that NendoUs may have originated from XendoU endoribonucleases (XendoUs) forming monomers in eukaryotic cells, that NendoU from arterivirus gained the ability to form dimers, and that the coronavirus variants then evolved the capacity to assemble into hexamers. We further propose that PDCoV nsp15 may be an intermediate in this evolutionary process. Our findings provide a theoretical basis for improving our understanding of NendoU evolution and offer useful clues for designing drugs and vaccines against nidoviruses.


Subject(s)
Coronavirus/chemistry , Endoribonucleases/chemistry , Nidovirales/chemistry , Protein Subunits/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Arterivirus/chemistry , Arterivirus/classification , Arterivirus/genetics , Arterivirus/metabolism , Binding Sites , Cloning, Molecular , Coronavirus/classification , Coronavirus/genetics , Coronavirus/metabolism , Crystallography, X-Ray , Endoribonucleases/genetics , Endoribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Evolution, Molecular , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Models, Molecular , Nidovirales/classification , Nidovirales/genetics , Nidovirales/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/chemistry , Severe acute respiratory syndrome-related coronavirus/classification , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics
4.
J Virol ; 91(6)2017 03 15.
Article in English | MEDLINE | ID: mdl-28053107

ABSTRACT

In five experimentally characterized arterivirus species, the 5'-end genome coding region encodes the most divergent nonstructural proteins (nsp's), nsp1 and nsp2, which include papain-like proteases (PLPs) and other poorly characterized domains. These are involved in regulation of transcription, polyprotein processing, and virus-host interaction. Here we present results of a bioinformatics analysis of this region of 14 arterivirus species, including that of the most distantly related virus, wobbly possum disease virus (WPDV), determined by a modified 5' rapid amplification of cDNA ends (RACE) protocol. By combining profile-profile comparisons and phylogeny reconstruction, we identified an association of the four distinct domain layouts of nsp1-nsp2 with major phylogenetic lineages, implicating domain gain, including duplication, and loss in the early nsp1 evolution. Specifically, WPDV encodes highly divergent homologs of PLP1a, PLP1b, PLP1c, and PLP2, with PLP1a lacking the catalytic Cys residue, but does not encode nsp1 Zn finger (ZnF) and "nuclease" domains, which are conserved in other arteriviruses. Unexpectedly, our analysis revealed that the only catalytically active nsp1 PLP of equine arteritis virus (EAV), known as PLP1b, is most similar to PLP1c and thus is likely to be a PLP1b paralog. In all non-WPDV arteriviruses, PLP1b/c and PLP1a show contrasting patterns of conservation, with the N- and C-terminal subdomains, respectively, being enriched with conserved residues, which is indicative of different functional specializations. The least conserved domain of nsp2, the hypervariable region (HVR), has its size varied 5-fold and includes up to four copies of a novel PxPxPR motif that is potentially recognized by SH3 domain-containing proteins. Apparently, only EAV lacks the signal that directs -2 ribosomal frameshifting in the nsp2 coding region.IMPORTANCE Arteriviruses comprise a family of mammalian enveloped positive-strand RNA viruses that include some of the most economically important pathogens of swine. Most of our knowledge about this family has been obtained through characterization of viruses from five species: Equine arteritis virus, Simian hemorrhagic fever virus, Lactate dehydrogenase-elevating virus, Porcine respiratory and reproductive syndrome virus 1, and Porcine respiratory and reproductive syndrome virus 2 Here we present the results of comparative genomics analyses of viruses from all known 14 arterivirus species, including the most distantly related virus, WPDV, whose genome sequence was completed in this study. Our analysis focused on the multifunctional 5'-end genome coding region that encodes multidomain nonstructural proteins 1 and 2. Using diverse bioinformatics techniques, we identified many patterns of evolutionary conservation that are specific to members of distinct arterivirus species, both characterized and novel, or their groups. They are likely associated with structural and functional determinants important for virus replication and virus-host interaction.


Subject(s)
Arterivirus/classification , Arterivirus/genetics , Evolution, Molecular , Genes, Viral , Genome, Viral , Protein Domains , Viral Nonstructural Proteins/genetics , Computational Biology , Genetic Variation , Phylogeny
5.
J Virol ; 89(15): 8082-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25972539

ABSTRACT

Simian hemorrhagic fever (SHF) is lethal for macaques. Based on clinical presentation and serological diagnosis, all reported SHF outbreaks were thought to be caused by different strains of the same virus, simian hemorrhagic fever virus (SHFV; Arteriviridae). Here we show that the SHF outbreaks in Sukhumi in 1964 and in Alamogordo in 1989 were caused not by SHFV but by two novel divergent arteriviruses. Our results indicate that multiple divergent simian arteriviruses can cause SHF.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/isolation & purification , Hemorrhagic Fevers, Viral/veterinary , Macaca/virology , Primate Diseases/virology , Amino Acid Sequence , Animals , Arterivirus/classification , Arterivirus/genetics , Arterivirus/physiology , Arterivirus Infections/history , Arterivirus Infections/virology , Evolution, Molecular , Hemorrhagic Fevers, Viral/history , Hemorrhagic Fevers, Viral/virology , History, 20th Century , Humans , Molecular Sequence Data , Phylogeny , Primate Diseases/history , Sequence Homology, Amino Acid , Viral Proteins/chemistry , Viral Proteins/genetics
6.
J Gen Virol ; 94(Pt 10): 2141-2163, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23939974

ABSTRACT

Arteriviruses are positive-stranded RNA viruses that infect mammals. They can cause persistent or asymptomatic infections, but also acute disease associated with a respiratory syndrome, abortion or lethal haemorrhagic fever. During the past two decades, porcine reproductive and respiratory syndrome virus (PRRSV) and, to a lesser extent, equine arteritis virus (EAV) have attracted attention as veterinary pathogens with significant economic impact. Particularly noteworthy were the 'porcine high fever disease' outbreaks in South-East Asia and the emergence of new virulent PRRSV strains in the USA. Recently, the family was expanded with several previously unknown arteriviruses isolated from different African monkey species. At the molecular level, arteriviruses share an intriguing but distant evolutionary relationship with coronaviruses and other members of the order Nidovirales. Nevertheless, several of their characteristics are unique, including virion composition and structure, and the conservation of only a subset of the replicase domains encountered in nidoviruses with larger genomes. During the past 15 years, the advent of reverse genetics systems for EAV and PRRSV has changed and accelerated the structure-function analysis of arterivirus RNA and protein sequences. These systems now also facilitate studies into host immune responses and arterivirus immune evasion and pathogenesis. In this review, we have summarized recent advances in the areas of arterivirus genome expression, RNA and protein functions, virion architecture, virus-host interactions, immunity, and pathogenesis. We have also briefly reviewed the impact of these advances on disease management, the engineering of novel candidate live vaccines and the diagnosis of arterivirus infection.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/genetics , Arterivirus/pathogenicity , Animals , Arterivirus/classification , Arterivirus/physiology , Arterivirus Infections/epidemiology , Arterivirus Infections/virology , Genome, Viral/genetics , Genome, Viral/physiology , Global Health , Mammals , Phylogeny
7.
J Virol ; 87(1): 688-91, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23077302

ABSTRACT

Simian hemorrhagic fever virus (SHFV) is an arterivirus that causes severe disease in captive macaques. We describe two new SHFV variants subclinically infecting wild African red-tailed guenons (Cercopithecus ascanius). Both variants are highly divergent from the prototype virus and variants infecting sympatric red colobus (Procolobus rufomitratus). All known SHFV variants are monophyletic and share three open reading frames not present in other arteriviruses. Our data suggest a need to modify the current arterivirus classification.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/classification , Arterivirus/isolation & purification , Genetic Variation , Primate Diseases/virology , RNA, Viral/genetics , Africa , Animals , Arterivirus/genetics , Arterivirus Infections/virology , Asymptomatic Infections , Cercopithecus , Cluster Analysis , Genotype , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA
8.
PLoS One ; 6(4): e19056, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21544192

ABSTRACT

BACKGROUND: Simian hemorrhagic fever virus (SHFV) has caused lethal outbreaks of hemorrhagic disease in captive primates, but its distribution in wild primates has remained obscure. Here, we describe the discovery and genetic characterization by direct pyrosequencing of two novel, divergent SHFV variants co-infecting a single male red colobus monkey from Kibale National Park, Uganda. METHODOLOGY/PRINCIPAL FINDINGS: The viruses were detected directly from blood plasma using pyrosequencing, without prior virus isolation and with minimal PCR amplification. The two new SHFV variants, SHFV-krc1 and SHFV-krc2 are highly divergent from each other (51.9% nucleotide sequence identity) and from the SHFV type strain LVR 42-0/M6941 (52.0% and 51.8% nucleotide sequence identity, respectively) and demonstrate greater phylogenetic diversity within SHFV than has been documented within any other arterivirus. Both new variants nevertheless have the same 3' genomic architecture as the type strain, containing three open reading frames not present in the other arteriviruses. CONCLUSIONS/SIGNIFICANCE: These results represent the first documentation of SHFV in a wild primate and confirm the unusual 3' genetic architecture of SHFV relative to the other arteriviruses. They also demonstrate a degree of evolutionary divergence within SHFV that is roughly equivalent to the degree of divergence between other arterivirus species. The presence of two such highly divergent SHFV variants co-infecting a single individual represents a degree of within-host viral diversity that exceeds what has previously been reported for any arterivirus. These results expand our knowledge of the natural history and diversity of the arteriviruses and underscore the importance of wild primates as reservoirs for novel pathogens.


Subject(s)
Arterivirus/genetics , Colobus/virology , Animals , Arterivirus/classification , Male , Polymerase Chain Reaction
9.
J Gen Virol ; 81(Pt 6): 1473-84, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10811931

ABSTRACT

A 20089 nucleotide (nt) sequence was determined for the 5' end of the (+)-ssRNA genome of gill-associated virus (GAV), a yellow head-like virus infecting Penaeus monodon prawns. Clones were generated from a approximately 22 kb dsRNA purified from lymphoid organ total RNA of GAV-infected prawns. The region contains a single gene comprising two long overlapping open reading frames, ORF1a and ORF1b, of 4060 and 2646 amino acids, respectively. The ORFs are structurally related to the ORF1a and ORF1ab polyproteins of coronaviruses and arteriviruses. The 99 nt overlap between ORF1a and ORF1b contains a putative AAAUUUU 'slippery' sequence associated with -1 ribosomal frameshifting. A 131 nt stem-loop with the potential to form a complex pseudoknot resides 3 nt downstream of this sequence. Although different to the G/UUUAAAC frameshift sites and 'H-type' pseudoknots of nidoviruses, in vitro transcription/translation analysis demonstrated that the GAV element also facilitates read-through of the ORF1a/1b junction. As in coronaviruses, GAV ORF1a encodes a 3C-like cysteine protease domain located between two hydrophobic regions. However, its sequence suggests some structural relationship to the chymotrypsin-like serine proteases of arteriviruses. ORF1b encodes homologues of the 'SDD' polymerase, which among (+)-RNA viruses is unique to nidoviruses, as well as metal-ion-binding and helicase domains. The presence of a dsRNA replicative intermediate and ORF1a and ORF1ab polyproteins translated by a-1 frameshift suggests that GAV represents the first invertebrate member of the Order NIDOVIRALES:


Subject(s)
Arterivirus/genetics , Coronavirus/genetics , Open Reading Frames , Penaeidae/virology , Amino Acid Sequence , Animals , Arterivirus/classification , Base Sequence , Binding Sites , Cloning, Molecular , Coronavirus/classification , Frameshift Mutation , Genes, Overlapping , Gills/virology , Molecular Sequence Data , Nucleic Acid Conformation , RNA, Viral/analysis , Ribosomes , Sequence Analysis, RNA
10.
Vet Res ; 28(4): 305-52, 1997.
Article in French | MEDLINE | ID: mdl-9312328

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is an infectious disease in swine that emerged 10 years ago. Today, PRRS is endemic in many if not all the pig-producing countries. The syndrome is due to a small enveloped RNA virus which belongs to the new Arteriviridae group. This group also includes the equine arterivirus and the simian hemorrhagic fever virus. The disease produces many clinical symptoms in pigs of any age but the two major features of the syndrome are respiratory and reproductive failure. An influenza-like illness is often observed consisting of a transient loss of appetite, slight hyperthermia and respiratory distress. The reproductive problems include late-term abortion, stillbirth and young piglet mortality. The virus is remarkably well adapted to its natural host. It infects almost exclusively pig monocytes or macrophages. For unexplained reasons, the virus may persist in the body for extended periods (several weeks) after an antibody immune response has been mounted. Within herds, the virus may persist for several years. In the acute phase of the infection, the immune functions of the pig may be impaired, however, a chronic immunosuppressive effect does not seem to occur thereafter. Several vaccines are available, which all have protective value against PRRS clinical disorders. None of them, however, has been tested for its ability to prevent or shorten the persistent infection in individual pigs or the long-term effect of virus circulation within herds. In the future, PRRS virus may be only an additional contaminant of farms with limited effects on profitability; however the possibility for this persistent infection to supply swine or eventually other species with new pathogenic variants must be also considered. This review provides a detailed description of the aetiology, pathology, immunology, diagnostic methods and epidemiology of PRRS.


Subject(s)
Porcine Reproductive and Respiratory Syndrome/physiopathology , Porcine respiratory and reproductive syndrome virus/classification , Abortion, Veterinary/virology , Animals , Arterivirus/classification , Female , Fetal Death/veterinary , Lung/pathology , Porcine Reproductive and Respiratory Syndrome/diagnosis , Porcine Reproductive and Respiratory Syndrome/epidemiology , Porcine respiratory and reproductive syndrome virus/isolation & purification , Porcine respiratory and reproductive syndrome virus/physiology , Pregnancy , Swine , Virus Replication
11.
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
13.
Am J Vet Res ; 57(6): 834-9, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8725809

ABSTRACT

OBJECTIVE: To compare the virulence of selected strains of porcine reproductive and respiratory syndrome virus (PRRSV) relative to reproductive performance of pregnant gilts. DESIGN: 16 pregnant gilts (principals) were exposed oronasally to 4 strains (vaccine strain RespPRRS, field strains VR-2385, VR-2431, and NADC-8, 4 gilts/strain) of PRRSV on or about day 90 of gestation, 4 pregnant gilts (controls) were kept under similar conditions, except for exposure to PRRSV. Samples and specimens obtained from gilts, pigs (before ingestion of colostrum), and fetuses were tested for PRRSV and homologous antibody. ANIMALS: 20 pregnant gilts. PROCEDURE: The virulence of each strain of PRRSV was evaluated mainly on the clinical status of the corresponding litters at farrowing. RESULTS: Most gilts remained clinically normal throughout the study and farrowed normally at or near the expected farrowing time. All virus strains crossed the placenta of principal gilts to infect fetuses in utero. The number of late-term dead fetuses (which appeared to be the best measure of relative virulence) ranged from 0 for litters of control gilts and gilts exposed to strain RespPRRS, to 38 for gilts exposed to strain NADC-8. All principal gilts became viremic and developed antibody against PRRSV. All strains persisted in alveolar macrophages of at least some principal gilts for at least 7 weeks after exposure. CONCLUSION: Strains of PRRSV vary in virulence. CLINICAL RELEVANCE: The effects of PRRSV on reproductive performance are strain dependent and this should be considered in making a tentative diagnosis on the basis of clinical observations.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/physiology , Fetal Death/veterinary , Pregnancy Complications, Infectious/veterinary , Swine Diseases/physiopathology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Arterivirus/classification , Arterivirus/pathogenicity , Arterivirus Infections/epidemiology , Arterivirus Infections/physiopathology , Body Temperature/physiology , Female , Fetal Death/physiopathology , Fetal Death/virology , Fetus/physiology , Fetus/virology , Incidence , Palatine Tonsil/virology , Pregnancy , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/physiopathology , Swine , Swine Diseases/epidemiology , Swine Diseases/virology , Viremia/physiopathology , Viremia/veterinary , Viremia/virology , Virulence
14.
J Vet Med Sci ; 58(4): 385-8, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8741277

ABSTRACT

Four cytopathogenic viruses were isolated in CPK cells derived from porcine kidneys from tonsils and lungs of 3 of 15 pigs affected with porcine reproductive and respiratory syndrome virus. Physicochemically and morphologically, the isolates were similar to a coronavirus. The isolates were not distinguished from transmissible gastroenteritis virus (TGEV) by a neutralization test using polyclonal antibodies, but differentiated from TGEV by monoclonal antibodies capable of discriminating between TGEV and porcine respiratory coronavirus (PRCV), indicating that the isolates were PRCV. In a serological survey of 30 serum samples each collected from about 50 days old pigs in the 2 affected farms, 29 (97%) and 15 (50%) sera were positive for neutralizing antibody against the isolate with the titers ranging from 2 to 64, respectively.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/isolation & purification , Lung/virology , Palatine Tonsil/virology , Swine Diseases , Transmissible gastroenteritis virus/isolation & purification , Animals , Antibodies, Monoclonal , Arterivirus/classification , Arterivirus Infections/pathology , Arterivirus Infections/virology , Cells, Cultured , Fluorescent Antibody Technique , Kidney/virology , Microscopy, Electron , Respiratory Tract Diseases/pathology , Respiratory Tract Diseases/veterinary , Respiratory Tract Diseases/virology , Swine , Syndrome , Transmissible gastroenteritis virus/classification , Transmissible gastroenteritis virus/ultrastructure
16.
Virology ; 207(2): 543-8, 1995 Mar 10.
Article in English | MEDLINE | ID: mdl-7886957

ABSTRACT

Full-length simian hemorrhagic fever virus (SHFV) genome RNA (about 15 kb in length) and six subgenomic RNAs, ranging in size from 0.65 to 4.7 kb, were detected by Northern blot hybridization in MA104 cytoplasmic extracts with a 3' genomic antisense probe. The 5' regions of the two smallest subgenomic RNAs (RNAs 6 and 7) were cloned and sequenced. Sequence analysis indicated that these two RNAs contained a common 5' leader sequence joined to the subgenomic RNA bodies via a highly conserved junction sequence; the junction sequence of RNA 7 was 5'-TTAACC-3', while that of RNA 6 was 5'-TCAACC-3'. The complete 5' leader sequence (208 nt) was obtained from genomic RNA. The genomic 5' junction sequence is identical to that of RNA 7. Northern blot hybridization with an antisense 5' leader probe confirmed the presence of the complete leader sequence in all six species of subgenomic RNA. In its virion morphology, genome size, gene order, and replication strategy, SHFV is most similar to viruses such as equine arteritis virus, lactate dehydrogenase-elevating virus, and Lelystad virus/porcine respiratory and reproductive syndrome virus.


Subject(s)
Arterivirus/genetics , RNA, Viral/genetics , Animals , Arterivirus/classification , Arterivirus/isolation & purification , Base Sequence , Cloning, Molecular , Conserved Sequence , DNA Primers/genetics , DNA, Complementary/genetics , Equartevirus/genetics , Genome, Viral , Lactate dehydrogenase-elevating virus/genetics , Macaca , Molecular Sequence Data , Open Reading Frames , RNA, Messenger/genetics
17.
Arch Virol ; 140(8): 1405-18, 1995.
Article in English | MEDLINE | ID: mdl-7661693

ABSTRACT

The cDNA sequence of the 3'-terminal genomic region of the Québec IAF-exp91 strain of porcine reproductive and respiratory syndrome virus (PRRSV) was determined and compared to those of other reference strains from Europe (Lelystad virus) and US (ATCC VR2385, MN-1b). The sequence (2834 nucleotides) which encompassed ORFs 3 to 7 revealed extensive genomic variations between the Québec strain and Lelystad virus (LV), resulting from high number of base substitutions, additions and deletions. The ORFs 5, 3, and 7 seemed to be relatively the most variable; the predicted encoding products of the Québec and LV strains displayed only 52%, 54%, and 59% amino acid identities, respectively. Nevertheless, in vitro translation experiments of the structural genes (ORFs 5, 6, and 7) and radioimmunoprecipitation assays with extracellular virions gave results similar to those previously reported for LV. In contrast, close genomic relationships were demonstrated between Québec and US strains. Taking together, these results indicate that, although structurally similar, North American PRRSV strains belong to a genotype distinct from that of the LV, thus supporting previous findings that allowed to divide PRRSV isolates into two antigenic subgroups (U.S. and European).


Subject(s)
Arterivirus/genetics , Genes, Viral , Genome, Viral , Open Reading Frames/genetics , Viral Structural Proteins/genetics , Amino Acid Sequence , Animals , Arterivirus/chemistry , Arterivirus/classification , Base Sequence , Conserved Sequence , DNA, Complementary/genetics , Europe , Molecular Sequence Data , Quebec , Swine/virology
18.
Arch Virol ; 140(8): 1451-60, 1995.
Article in English | MEDLINE | ID: mdl-7661696

ABSTRACT

The 3'-portion of the genome of a U.S. isolate of the porcine reproductive and respiratory syndrome (PRRS) virus, ATCCVR-2332, was cloned and sequenced. The resultant 3358 nucleotides contain 6 open reading frames (ORFs) with homologies to ORFs 2 through 7 of the European strain of the PRRS virus and other members of the free-standing genus of arteriviruses. Both VR-2332 and the European isolate (called the Lelystad virus) have been identified as infectious agents responsible for the swine disease called PRRS. Comparative sequence analysis indicates that there are degrees of amino acid identity to the Lelystad virus open reading frames ranging from 55% in ORF 5 to 79% in ORF 6. Hydropathy profiles indicate that the ORFs of VR-2332 and Lelystad virus correspond structurally despite significant sequence differences. These results are consistent with the biological similarities but distinct serological properties of North American and European isolates of the virus.


Subject(s)
Arterivirus/genetics , Genome, Viral , Viral Structural Proteins/genetics , Amino Acid Sequence , Animals , Arterivirus/classification , Arterivirus/isolation & purification , Cloning, Molecular , Equartevirus/genetics , Europe , Glycosylation , Isoelectric Point , Lactate dehydrogenase-elevating virus/genetics , Molecular Sequence Data , Molecular Weight , Open Reading Frames/genetics , Swine/virology , United States , Viral Structural Proteins/chemistry
19.
Arch Virol ; 140(4): 745-55, 1995.
Article in English | MEDLINE | ID: mdl-7794115

ABSTRACT

The putative membrane (M) protein (ORF 6) and nucleocapsid (N) protein (ORF 7) genes of five U.S. isolates of porcine reproductive and respiratory syndrome virus (PRRSV) with differing virulence were cloned and sequenced. To determine the genetic variation and the phylogenetic relationship of PRRSV, the deduced amino acid sequences of the putative M and N proteins from these isolates were aligned, to the extent known, with other PRRSV isolates, and also other members of the proposed arterivirus group including lactate dehydrogenase-elevating virus (LDV) and equine arteritis virus (EAV). There was 96-100% amino acid sequence identity in the putative M and N genes among U.S. and Canadian PRRSV isolates with differing virulence. However, their amino acid sequences varied extensively from those of European PRRSV isolates, and displayed only 57-59% and 78-81% identity, respectively. The phylogenetic trees constructed on the basis of the putative M and N genes of the proposed arterivirus group were similar and indicated that both U.S. and European PRRSV isolates were related to LDV and were distantly related to EAV. The U.S. and European PRRSV isolates fell into two distinct groups, suggesting that U.S. and European PRRSV isolates represent two distinct genotypes.


Subject(s)
Arterivirus/classification , Capsid/genetics , Viral Core Proteins/genetics , Viral Matrix Proteins/genetics , Amino Acid Sequence , Animals , Arterivirus/genetics , Base Sequence , DNA Primers , DNA, Viral , Europe , Genes, Viral , Genotype , Molecular Sequence Data , Open Reading Frames , Phylogeny , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Swine , United States
20.
Arch Virol Suppl ; 9: 441-8, 1994.
Article in English | MEDLINE | ID: mdl-8032274

ABSTRACT

Lelystad virus (LV) is an enveloped positive-stranded RNA virus, which causes abortions and respiratory disease in pigs. The complete nucleotide sequence of the genome of LV has been determined. This sequence is 15.1 kb in length and contains a poly(A) tail at the 3' end. Open reading frames that might encode the viral replicases (ORFs 1a and 1b), membrane-associated proteins (ORFs 2 to 6) and the nucleocapsid protein (ORF7) have been identified. Sequence comparisons have indicated that LV is distantly related to the coronaviruses and toroviruses and closely related to lactate dehydrogenase-elevating virus (LDV) and equine arteritis virus (EAV). A 3' nested set of six subgenomic RNAs is produced in LV-infected alveolar lung macrophages. These subgenomic RNAs contain a leader sequence, which is derived from the 5' end of the viral genome. Altogether, these data show that LV is closely related evolutionarily to LDV and EAV, both members of a recently proposed family of positive-stranded RNA viruses, the Arteriviridae.


Subject(s)
Arterivirus/genetics , Genome, Viral , RNA Viruses/classification , Animals , Arterivirus/classification , Arterivirus/growth & development , Equartevirus/classification , Equartevirus/genetics , Gene Expression , Lactate dehydrogenase-elevating virus/classification , Lactate dehydrogenase-elevating virus/genetics , RNA Viruses/genetics , RNA, Viral/genetics , Sequence Homology, Amino Acid , Swine , Swine Diseases/microbiology , Viral Proteins/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...