Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 97(7): 1670-1675, 2016 07.
Article in English | MEDLINE | ID: mdl-27114233

ABSTRACT

African swine fever (ASF) is an emerging disease threat for the swine industry worldwide. No ASF vaccine is available and progress is hindered by lack of knowledge concerning the extent of ASFV strain diversity and the viral antigens conferring type-specific protective immunity in pigs. Available data from vaccination/challenge experiments in pigs indicate that ASF protective immunity may be haemadsorption inhibition (HAI) serotype-specific. Recently, we have shown that two ASFV proteins, CD2v (EP402R) and C-type lectin (EP153R), are necessary and sufficient for mediating HAI serological specificity (Malogolovkin et al., 2015).. Here, using ASFV inter-serotypic chimeric viruses and vaccination/challenge experiments in pigs, we demonstrate that serotype-specific CD2v and/or C-type lectin proteins are important for protection against homologous ASFV infection. Thus, these viral proteins represent significant protective antigens for ASFV that should be targeted in future vaccine design and development. Additionally, these data support the concept of HAI serotype-specific protective immunity.


Subject(s)
African Swine Fever Virus/immunology , African Swine Fever/immunology , African Swine Fever/prevention & control , Antibodies, Viral/immunology , Swine Diseases/prevention & control , Viral Proteins/immunology , Viral Vaccines/immunology , African Swine Fever/virology , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , Lectins, C-Type/immunology , Serogroup , Swine/virology , Swine Diseases/virology , Vaccination
2.
J Gen Virol ; 96(Pt 4): 866-873, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25524164

ABSTRACT

African swine fever (ASF) is an emerging disease threat for the swine industry worldwide. No ASF vaccine is available and progress is hindered by lack of knowledge concerning the extent of ASF virus (ASFV) strain diversity and the viral antigens responsible for protection in the pig. Available data from vaccination/challenge experiments in pigs indicate ASF protective immunity is haemadsorption inhibition (HAI) serotype-specific. A better understanding of ASFV HAI serological groups and their diversity in nature, as well as improved methods to serotype ASFV isolates, is needed. Here, we demonstrated that the genetic locus encoding ASFV CD2v and C-type lectin proteins mediates HAI serological specificity and that CD2v/C-type lectin genotyping provides a simple method to group ASFVs by serotype, thus facilitating study of ASFV strain diversity in nature, and providing information necessary for eventual vaccine design, development and efficacious use.


Subject(s)
African Swine Fever Virus/genetics , Genetic Loci , HLA-DR Serological Subtypes/genetics , Lectins, C-Type/genetics , Animals , Antigens, Viral/genetics , DNA, Viral/genetics , Genotype , Lectins, C-Type/immunology , Sensitivity and Specificity , Swine , Viral Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology
3.
J Gen Virol ; 95(Pt 8): 1790-1798, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24836671

ABSTRACT

The genus Macavirus of the subfamily Gammaherpesvirinae includes viruses that infect lymphoid cells of domestic and wild ruminants and swine, causing asymptomatic latent infections in reservoir hosts. Here, we describe the genome of bovine herpesvirus 6 (BoHV-6), a macavirus ubiquitous in healthy cattle populations. The BoHV-6 genome exhibited architecture conserved in macaviruses, including a repetitive H-DNA region and unique 141 kbp L-DNA region predicted to encode 77 genes. BoHV-6 encoded, in variable genomic regions, a novel complement of genes relative to other characterized macaviruses, probably contributing to distinctive aspects of BoHV-6 infection biology and host range. Most notably, BoHV-6 encoded the first herpesviral protein (Bov2.b2) similar to cellular ornithine decarboxylase, an enzyme that catalyses the first and rate-limiting step in the biosynthesis of polyamines. Bov2.b2 conceivably mediates a novel mechanism by which BoHV-6 promotes cell-cycle-dependent viral replication.


Subject(s)
DNA, Viral/chemistry , DNA, Viral/genetics , Gammaherpesvirinae/genetics , Genome, Viral , Animals , Cattle , Gammaherpesvirinae/isolation & purification , Gene Order , Molecular Sequence Data , Ornithine Decarboxylase/genetics , Sequence Analysis, DNA , Synteny , Viral Proteins/genetics
4.
J Virol ; 85(1): 264-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980501

ABSTRACT

Poxviruses have evolved various strategies to inhibit cytoplasmic events leading to activation of the nuclear factor κB (NF-κB) signaling pathway, with individual viruses often encoding multiple NF-κB inhibitors. Here, the novel orf virus (ORFV)-encoded protein ORFV002 was shown to inhibit nuclear events regulating NF-κB transcriptional activity. ORFV002 expression in cell cultures significantly decreased wild-type-virus-, tumor necrosis factor alpha (TNF-α)-, and lipopolysaccharide (LPS)-induced NF-κB-mediated gene expression. Expression of ORFV002 in cells, while not affecting phosphorylation or nuclear translocation of NF-κB-p65, markedly decreased TNF-α- and wild-type-virus-induced acetylation of NF-κB-p65, a p300-mediated nuclear modification of NF-κB-p65 that regulates its transactivating activity. ORFV002 was shown to colocalize and interact with NF-κB-p65, and expression of ORFV002 in cell cultures resulted in a reduced interaction of NF-κB-p65 with p300, suggesting that ORFV002 interferes with NF-κB-p65/p300 association. Deletion of ORFV002 from the OV-IA82 genome had no significant effect on ORFV pathogenesis in sheep, indicating that ORFV002 is nonessential for virus virulence in the natural host. This represents the first description of a nuclear inhibitor of NF-κB encoded by a poxvirus.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , NF-kappa B/antagonists & inhibitors , Orf virus/pathogenicity , Viral Proteins/metabolism , Animals , Cells, Cultured , Ecthyma, Contagious/pathology , Ecthyma, Contagious/virology , NF-kappa B/genetics , Orf virus/genetics , Orf virus/metabolism , Sheep , Signal Transduction , Viral Proteins/genetics
5.
J Virol ; 85(5): 2037-49, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21177808

ABSTRACT

Orf virus (ORFV), the type member of the genus Parapoxvirus of the Poxviridae, has evolved novel strategies (proteins and/or mechanisms of action) to modulate host cell responses regulated by the nuclear factor-κB (NF-κB) signaling pathway. Here, we present data indicating that ORFV ORFV121, a gene unique to parapoxviruses, encodes a novel viral NF-κB inhibitor that binds to and inhibits the phosphorylation and nuclear translocation of NF-κB-p65. The infection of cells with an ORFV121 deletion mutant virus (OV-IA82Δ121) resulted in increased NF-κB-mediated gene transcription, and the expression of ORFV121 in cell cultures significantly suppressed NF-κB-regulated reporter gene expression. ORFV ORFV121 physically interacts with NF-κB-p65 in the cell cytoplasm, thus providing a mechanism for the inhibition of NF-κB-p65 phosphorylation and nuclear translocation. Notably, the deletion of ORFV121 from the viral genome markedly decreased ORFV virulence and disease pathogenesis in sheep, indicating that ORFV121 is a virulence determinant for ORFV in the natural host.


Subject(s)
Ecthyma, Contagious/metabolism , Orf virus/metabolism , Orf virus/pathogenicity , Transcription Factor RelA/antagonists & inhibitors , Viral Proteins/metabolism , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Ecthyma, Contagious/genetics , Ecthyma, Contagious/virology , Gene Expression Regulation, Viral , HeLa Cells , Humans , Orf virus/genetics , Phosphorylation , Protein Binding , Protein Transport , Sheep , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Viral Proteins/genetics , Virulence
6.
J Virol ; 84(8): 3962-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20147406

ABSTRACT

The parapoxvirus orf virus (ORFV) is a pathogen of sheep and goats that has been used as a preventive and therapeutic immunomodulatory agent in several animal species. However, the functions (genes, proteins, and mechanisms of action) evolved by ORFV to modulate and manipulate immune responses are poorly understood. Here, the novel ORFV protein ORFV024 was shown to inhibit activation of the NF-kappaB signaling pathway, an important modulator of early immune responses against viral infections. Infection of primary ovine cells with an ORFV024 deletion mutant virus resulted in a marked increase in expression of NF-kappaB-regulated chemokines and other proinflammatory host genes. Expression of ORFV024 in cell cultures significantly decreased lipopolysaccharide (LPS)- and tumor necrosis factor alpha (TNF-alpha)-induced NF-kappaB-responsive reporter gene expression. Further, ORFV024 expression decreased TNF-alpha-induced phosphorylation and nuclear translocation of NF-kappaB-p65, phosphorylation, and degradation of IkappaBalpha, and phosphorylation of IkappaB kinase (IKK) subunits IKKalpha and IKKbeta, indicating that ORFV024 functions by inhibiting activation of IKKs, the bottleneck for most NF-kappaB activating stimuli. Although ORFV024 interferes with activation of the NF-kappaB signaling pathway, its deletion from the OV-IA82 genome had no significant effect on disease severity, progression, and time to resolution in sheep, indicating that ORFV024 is not essential for virus virulence in the natural host. This represents the first description of a NF-kappaB inhibitor encoded by a parapoxvirus.


Subject(s)
NF-kappa B/antagonists & inhibitors , Orf virus/immunology , Signal Transduction , Viral Proteins/physiology , Virulence Factors/physiology , Animals , Cells, Cultured , Chemokines/biosynthesis , Ecthyma, Contagious/pathology , Ecthyma, Contagious/virology , Gene Deletion , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation , Sheep , Viral Proteins/genetics , Virulence Factors/genetics
7.
Biotech Histochem ; 85(6): 355-63, 2010 Dec.
Article in English | MEDLINE | ID: mdl-19909216

ABSTRACT

The aim of this work was to determine the effects of cGnRH I pulse frequencies on FSH and LH release and the changes in features and number of cultured laying hen FSH-cells and LH-cells in vitro. Primary adenohypophyseal cell cultures taken from laying hens were stimulated by four 5 min pulses using 1 or 10 nM cGnRH, administered with interpulses between pulses at 15, 30 or 60 min. Pulse frequencies and dose dependent effects were examined in six separate experiments including two controls. After the last interpulse time, the supernatants were collected and stored at -70° C until the performance of an indirect enzyme-linked immunosorbent assay (ELISA) using chicken LH and chicken FSH antisera at 1:1000 and 1:2000 dilutions, respectively. Supernatants were coated in duplicate on the inner surface of Immulon 2 plates and later blocked with the optimal solutions. They were incubated with each antiserum and subsequently with isotype-specific peroxidase-labeled anti-rabbit antibodies. Hydrogen peroxide/o-phenylenediamine was added as substrate/chromogen and the optical density (OD) was determined at 492 nm. The ABC immunocytochemical method was performed to characterize and re-count the gonadotropes employing anti-chicken FSH and anti-chicken LH as primary antibodies. The number of FSH-LH cells was obtained using stereological analysis and the data were statistically processed. The ODs obtained for each anti-hormone were compared with the control groups and with each other. Significant differences were found in number of aggregated-positive LH cells, which decreased with 1 nM cGnRH-I, 15 vs. 30 min pulses, increased with 30 vs. 60 min pulses, and also with 10 nM cGnRH-I, 30 vs. 60 min pulses. Aggregated positive FSH cells, however, did not show significant differences in percentage at any GnRH dose or pulse frequencies, but did show activity at low pulse frequencies of 15 and 30 min. The results suggest that LH cells varied in percentage in a dose dependent manner at higher pulse frequency (15 min) and were dose independent at low pulse frequency (60 min) and showed inactive features; while FSH cell numbers were unaffected showing features of activity at low pulse frequencies. High and moderate pulse frequencies of cGnRH-I (15-30 min) increased the FSH release in dose independent manner without changes in features or percentage of FSH cells. Low pulse frequency (60 min) of cGnRH-I increased LH release dose independently disminished LH cell percentage and showed changes in cells' features. These results in avian cells showed differences in responses to GnRH pulse frequencies from those reported earlier in mammals.


Subject(s)
Gonadotrophs/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Chickens , Dose-Response Relationship, Drug , Follicle Stimulating Hormone/analysis , Follicle Stimulating Hormone/metabolism , Gonadotrophs/cytology , Gonadotrophs/metabolism , Luteinizing Hormone/analysis , Luteinizing Hormone/metabolism , Pituitary Gland, Anterior/drug effects , Time Factors
8.
Curr Top Microbiol Immunol ; 328: 43-87, 2009.
Article in English | MEDLINE | ID: mdl-19216435

ABSTRACT

African swine fever virus (ASFV) is a large, intracytoplasmically-replicating DNA arbovirus and the sole member of the family Asfarviridae. It is the etiologic agent of a highly lethal hemorrhagic disease of domestic swine and therefore extensively studied to elucidate the structures, genes, and mechanisms affecting viral replication in the host, virus-host interactions, and viral virulence. Increasingly apparent is the complexity with which ASFV replicates and interacts with the host cell during infection. ASFV encodes novel genes involved in host immune response modulation, viral virulence for domestic swine, and in the ability of ASFV to replicate and spread in its tick vector. The unique nature of ASFV has contributed to a broader understanding of DNA virus/host interactions.


Subject(s)
African Swine Fever Virus/physiology , African Swine Fever/virology , Animals , Swine
9.
Vet Rec ; 163(11): 331-5, 2008 Sep 13.
Article in English | MEDLINE | ID: mdl-18791208

ABSTRACT

Between 1998 and 2001, several cases of ataxia and paresis followed by recumbency and death were reported in cows from different farms in a restricted area of the Argentinian Patagonia. Five cases of this cluster were studied and a diagnosis of malignant schwannoma was established. Electron microscopy (em) of tumour samples from three of the animals revealed intracytoplasmic or interstitial structures resembling retroviral particles. Attempts to isolate a viral agent from the tumours were unsuccessful but the epidemiological data and the em findings suggest a viral aetiology.


Subject(s)
Cattle Diseases/pathology , Neurilemmoma/veterinary , Spinal Cord Neoplasms/veterinary , Animals , Argentina , Cattle , Cattle Diseases/virology , Female , Microscopy, Electron/veterinary , Neurilemmoma/pathology , Neurilemmoma/ultrastructure , Neurilemmoma/virology , Retroviridae/ultrastructure , Spinal Cord Neoplasms/pathology , Spinal Cord Neoplasms/ultrastructure , Spinal Cord Neoplasms/virology , Spinal Nerve Roots/pathology
10.
J Clin Microbiol ; 46(2): 438-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032617

ABSTRACT

Sheeppox virus (SPPV) is a member of the Capripoxvirus (CaPV) genus of the Poxviridae family. Members of this genus, which also include goatpox and lumpy skin disease viruses, cause economically significant disease in sheep, goats, and cattle. A rapid diagnostic assay for CaPV would be useful for disease surveillance as well as for detection of CaPV in clinical samples and for outbreak management. Here we describe a fluorogenic probe hydrolysis (TaqMan) PCR assay designed for rapid detection of CaPV and tested on sheep experimentally infected with a virulent strain of SPPV. This assay can detect SPPV in buffy coats, nasal swabs, oral swabs, scabs, and skin lesions as well as in lung and lymph nodes collected at necropsy. This single-tube diagnostic assay can be performed in 2 h or less and can detect viral DNA in preclinical, clinical, and postmortem samples.


Subject(s)
Capripoxvirus/isolation & purification , Polymerase Chain Reaction/methods , Poxviridae Infections/veterinary , Sheep Diseases/diagnosis , Virology/methods , Animals , Capripoxvirus/genetics , Disease Outbreaks/prevention & control , Fluorescence , Fluorescent Dyes/metabolism , Poxviridae Infections/diagnosis , Poxviridae Infections/virology , Sheep , Sheep Diseases/virology , Time Factors
11.
J Virol ; 81(20): 11392-401, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17686843

ABSTRACT

Sheeppox virus (SPPV), a member of the Capripoxvirus genus of the Poxviridae, is the etiologic agent of a significant disease of sheep in the developing world. Genomic analysis of pathogenic and vaccine capripoxviruses identified genes with potential roles in virulence and host range, including three genes with similarity to kelch-like genes of other poxviruses and eukaryotes. Here, a mutant SPPV with a deletion in the SPPV-019 kelch-like gene, DeltaKLP, was derived from the pathogenic strain SPPV-SA. DeltaKLP exhibited in vitro growth characteristics similar to those of SPPV-SA and revertant virus (RvKLP). DeltaKLP-infected cells exhibited a reduction in Ca(2+)-independent cell adhesion, suggesting that SPPV-019 may modulate cellular adhesion. When inoculated in sheep by the intranasal or intradermal routes, DeltaKLP was markedly attenuated, since all DeltaKLP-infected lambs survived infection. In contrast, SPPV-SA and RvKLP induced mortality approaching 100%. Lambs inoculated with DeltaKLP exhibited marked reduction or delay in fever response, gross lesions, viremia, and virus shedding compared to parental and revertant viruses. Together, these findings indicate that SPPV-019 is a significant SPPV virulence determinant in sheep.


Subject(s)
Capripoxvirus/genetics , Capripoxvirus/pathogenicity , Viral Proteins/physiology , Animals , Genes, Viral/physiology , Mutation , Sheep , Virulence/genetics
12.
Dev Biol (Basel) ; 126: 23-30; discussion 323, 2006.
Article in English | MEDLINE | ID: mdl-17058478

ABSTRACT

Despite a basic understanding of many aspects of FMD biology, much information regarding FMDV virulence, host range, and virus transmission remains poorly understood. Here we present how the use of high throughput sequencing for complete genome sequences of foot-and mouth disease virus (FMDV) led to a series of new insights into viral genome sequence conservation and variability, genetic diversity in nature and phylogenetic classification of isolates, including the first complete sequences of the South African Territories type 1 and 3 (SAT1 and SAT3) genomes. Comparative genomic analysis of full-length sequences of FMDV isolates did allow: (i) the identification of highly conserved regulatory or coding regions which are critical for aspects of virus biology as well as novel viral genomic motifs with likely biological relevance; (ii) characterization of the first complete sequences of the SAT1 and SAT3 genomes; (iii) identification of a novel SAT virus lineage genetically distinct from other SAT and Euro-Asiatic lineages; (iv) precise identification of strains circulating around the world for epidemiological and forensic attribution; (v) assessment of mutation and recombination processes as mechanisms equally involved in evolution; (vi) mutation rates, tolerance and constraints of genes and proteins during evolution of FMD viruses during in vivo replication and (vi) support for the hypothesis of a new evolutionary model.


Subject(s)
Foot-and-Mouth Disease Virus/genetics , Genome, Viral , Genomics/methods , Animals , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease Virus/physiology , Mutation/genetics , Phylogeny , Reassortant Viruses/genetics , Recombination, Genetic/genetics , Ruminants , Swine , Virus Replication
13.
J Virol ; 80(18): 9244-58, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16940536

ABSTRACT

Here we present the genomic sequence of horsepox virus (HSPV) isolate MNR-76, an orthopoxvirus (OPV) isolated in 1976 from diseased Mongolian horses. The 212-kbp genome contained 7.5-kbp inverted terminal repeats and lacked extensive terminal tandem repetition. HSPV contained 236 open reading frames (ORFs) with similarity to those in other OPVs, with those in the central 100-kbp region most conserved relative to other OPVs. Phylogenetic analysis of the conserved region indicated that HSPV is closely related to sequenced isolates of vaccinia virus (VACV) and rabbitpox virus, clearly grouping together these VACV-like viruses. Fifty-four HSPV ORFs likely represented fragments of 25 orthologous OPV genes, including in the central region the only known fragmented form of an OPV ribonucleotide reductase large subunit gene. In terminal genomic regions, HSPV lacked full-length homologues of genes variably fragmented in other VACV-like viruses but was unique in fragmentation of the homologue of VACV strain Copenhagen B6R, a gene intact in other known VACV-like viruses. Notably, HSPV contained in terminal genomic regions 17 kbp of OPV-like sequence absent in known VACV-like viruses, including fragments of genes intact in other OPVs and approximately 1.4 kb of sequence present only in cowpox virus (CPXV). HSPV also contained seven full-length genes fragmented or missing in other VACV-like viruses, including intact homologues of the CPXV strain GRI-90 D2L/I4R CrmB and D13L CD30-like tumor necrosis factor receptors, D3L/I3R and C1L ankyrin repeat proteins, B19R kelch-like protein, D7L BTB/POZ domain protein, and B22R variola virus B22R-like protein. These results indicated that HSPV contains unique genomic features likely contributing to a unique virulence/host range phenotype. They also indicated that while closely related to known VACV-like viruses, HSPV contains additional, potentially ancestral sequences absent in other VACV-like viruses.


Subject(s)
Genome, Viral , Vaccinia virus/genetics , Viruses/genetics , Animals , Cloning, Molecular , Cowpox virus/genetics , DNA, Viral , Horses , Models, Genetic , Molecular Sequence Data , Open Reading Frames , Phenotype , Phylogeny , Variola virus/genetics
14.
J Virol ; 80(10): 4978-91, 2006 May.
Article in English | MEDLINE | ID: mdl-16641289

ABSTRACT

Here, we present the genome sequence, with analysis, of a poxvirus infecting Nile crocodiles (Crocodylus niloticus) (crocodilepox virus; CRV). The genome is 190,054 bp (62% G+C) and predicted to contain 173 genes encoding proteins of 53 to 1,941 amino acids. The central genomic region contains genes conserved and generally colinear with those of other chordopoxviruses (ChPVs). CRV is distinct, as the terminal 33-kbp (left) and 13-kbp (right) genomic regions are largely CRV specific, containing 48 unique genes which lack similarity to other poxvirus genes. Notably, CRV also contains 14 unique genes which disrupt ChPV gene colinearity within the central genomic region, including 7 genes encoding GyrB-like ATPase domains similar to those in cellular type IIA DNA topoisomerases, suggestive of novel ATP-dependent functions. The presence of 10 CRV proteins with similarity to components of cellular multisubunit E3 ubiquitin-protein ligase complexes, including 9 proteins containing F-box motifs and F-box-associated regions and a homologue of cellular anaphase-promoting complex subunit 11 (Apc11), suggests that modification of host ubiquitination pathways may be significant for CRV-host cell interaction. CRV encodes a novel complement of proteins potentially involved in DNA replication, including a NAD(+)-dependent DNA ligase and a protein with similarity to both vaccinia virus F16L and prokaryotic serine site-specific resolvase-invertases. CRV lacks genes encoding proteins for nucleotide metabolism. CRV shares notable genomic similarities with molluscum contagiosum virus, including genes found only in these two viruses. Phylogenetic analysis indicates that CRV is quite distinct from other ChPVs, representing a new genus within the subfamily Chordopoxvirinae, and it lacks recognizable homologues of most ChPV genes involved in virulence and host range, including those involving interferon response, intracellular signaling, and host immune response modulation. These data reveal the unique nature of CRV and suggest mechanisms of virus-reptile host interaction.


Subject(s)
Alligators and Crocodiles/virology , Chordopoxvirinae/genetics , Genome, Viral , Multigene Family , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Chordopoxvirinae/enzymology , DNA Gyrase/chemistry , DNA Gyrase/genetics , Molecular Sequence Data , Phylogeny , Protein Structure, Tertiary , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Ubiquitin-Protein Ligases/genetics
15.
J Virol ; 79(10): 6487-504, 2005 May.
Article in English | MEDLINE | ID: mdl-15858032

ABSTRACT

Here we present complete genome sequences, including a comparative analysis, of 103 isolates of foot-and-mouth disease virus (FMDV) representing all seven serotypes and including the first complete sequences of the SAT1 and SAT3 genomes. The data reveal novel highly conserved genomic regions, indicating functional constraints for variability as well as novel viral genomic motifs with likely biological relevance. Previously undescribed invariant motifs were identified in the 5' and 3' untranslated regions (UTR), as was tolerance for insertions/deletions in the 5' UTR. Fifty-eight percent of the amino acids encoded by FMDV isolates are invariant, suggesting that these residues are critical for virus biology. Novel, conserved sequence motifs with likely functional significance were identified within proteins L(pro), 1B, 1D, and 3C. An analysis of the complete FMDV genomes indicated phylogenetic incongruities between different genomic regions which were suggestive of interserotypic recombination. Additionally, a novel SAT virus lineage containing nonstructural protein-encoding regions distinct from other SAT and Euroasiatic lineages was identified. Insights into viral RNA sequence conservation and variability and genetic diversity in nature will likely impact our understanding of FMDV infections, host range, and transmission.


Subject(s)
Foot-and-Mouth Disease Virus/genetics , Genome, Viral , Genomics , 3' Untranslated Regions , 5' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Capsid Proteins/genetics , Genetic Variation , Global Health , Molecular Sequence Data , Nucleic Acid Conformation , Phylogeny , Viral Nonstructural Proteins/genetics
16.
J Virol ; 79(2): 966-77, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15613325

ABSTRACT

Deerpox virus (DPV), an uncharacterized and unclassified member of the Poxviridae, has been isolated from North American free-ranging mule deer (Odocoileus hemionus) exhibiting mucocutaneous disease. Here we report the genomic sequence and comparative analysis of two pathogenic DPV isolates, W-848-83 (W83) and W-1170-84 (W84). The W83 and W84 genomes are 166 and 170 kbp, containing 169 and 170 putative genes, respectively. Nucleotide identity between DPVs is 95% over the central 157 kbp. W83 and W84 share similar gene orders and code for similar replicative, structural, virulence, and host range functions. DPV open reading frames (ORFs) with putative virulence and host range functions include those similar to cytokine receptors (R), including gamma interferon receptor (IFN-gammaR), interleukin 1 receptor (IL-1R), and type 8 CC-chemokine receptors; cytokine binding proteins (BP), including IL-18BP, IFN-alpha/betaBP, and tumor necrosis factor binding protein (TNFBP); serpins; and homologues of vaccinia virus (VACV) E3L, K3L, and A52R proteins. DPVs also encode distinct forms of major histocompatibility complex class I, C-type lectin-like protein, and transforming growth factor beta1 (TGF-beta1), a protein not previously described in a mammalian chordopoxvirus. Notably, DPV encodes homologues of cellular endothelin 2 and IL-1R antagonist, novel poxviral genes also likely involved in the manipulation of host responses. W83 and W84 differ from each other by the presence or absence of five ORFs. Specifically, homologues of a CD30 TNFR family protein, swinepox virus SPV019, and VACV E11L core protein are absent in W83, and homologues of TGF-beta1 and lumpy skin disease virus LSDV023 are absent in W84. Phylogenetic analysis indicates that DPVs are genetically distinct from viruses of other characterized poxviral genera and that they likely comprise a new genus within the subfamily Chordopoxvirinae.


Subject(s)
Chordopoxvirinae/genetics , Deer/virology , Genome, Viral , Amino Acid Sequence , Animals , Base Sequence , Chordopoxvirinae/classification , Endothelins/physiology , Interleukin-1/physiology , Molecular Sequence Data , Open Reading Frames , Receptors, Interleukin-1/physiology
17.
Vet Res Commun ; 28(2): 159-70, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14992245

ABSTRACT

An outbreak of pulmonary adenomatosis (OPA) occurred in sheep in Patagonia, Argentina's southernmost region. On the affected farm, nine animals died over a 6-month period with pulmonary lesions of OPA. In all cases, the histology of the lungs was characterized by proliferation of cuboideal and prismatic cells lining the alveoli. Inflammatory exudates and accumulation of alveolar macrophages were marked in most cases, but in six of the cases there was no excess fluid in the airways. The presence of the Jaagsiekte retrovirus was demonstrated in the lungs by immunocytochemistry and PCR. To the best of our knowledge, this is the first report of OPA in Patagonia.


Subject(s)
DNA, Viral/analysis , Disease Outbreaks/veterinary , Jaagsiekte sheep retrovirus/isolation & purification , Lung Neoplasms/veterinary , Pulmonary Adenomatosis, Ovine/epidemiology , Animals , Argentina/epidemiology , Female , Immunohistochemistry/veterinary , Jaagsiekte sheep retrovirus/genetics , Lung/pathology , Lung Neoplasms/epidemiology , Lung Neoplasms/virology , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Male , Polymerase Chain Reaction/veterinary , Pulmonary Adenomatosis, Ovine/virology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Sheep
18.
J Virol ; 78(1): 168-77, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14671098

ABSTRACT

Bovine papular stomatitis virus (BPSV) and orf virus (ORFV), members of the genus Parapoxvirus of the Poxviridae, are etiologic agents of worldwide diseases affecting cattle and small ruminants, respectively. Here we report the genomic sequences and comparative analysis of BPSV strain BV-AR02 and ORFV strains OV-SA00, isolated from a goat, and OV-IA82, isolated from a sheep. Parapoxvirus (PPV) BV-AR02, OV-SA00, and OV-IA82 genomes range in size from 134 to 139 kbp, with an average nucleotide composition of 64% G+C. BPSV and ORFV genomes contain 131 and 130 putative genes, respectively, and share colinearity over 127 genes, 88 of which are conserved in all characterized chordopoxviruses. BPSV and ORFV contain 15 and 16 open reading frames (ORFs), respectively, which lack similarity to other poxvirus or cellular proteins. All genes with putative roles in pathogenesis, including a vascular endothelial growth factor (VEGF)-like gene, are present in both viruses; however, BPSV contains two extra ankyrin repeat genes absent in ORFV. Interspecies sequence variability is observed in all functional classes of genes but is highest in putative virulence/host range genes, including genes unique to PPV. At the amino acid level, OV-SA00 is 94% identical to OV-IA82 and 71% identical to BV-AR02. Notably, ORFV 006/132, 103, 109, 110, and 116 genes (VEGF, homologues of vaccinia virus A26L, A33R, and A34R, and a novel PPV ORF) show an unusual degree of intraspecies variability. These genomic differences are consistent with the classification of BPSV and ORFV as two PPV species. Compared to other mammalian chordopoxviruses, PPV shares unique genomic features with molluscum contagiosum virus, including a G+C-rich nucleotide composition, three orthologous genes, and a paucity of nucleotide metabolism genes. Together, these data provide a comparative view of PPV genomics.


Subject(s)
Genome, Viral , Orf virus/genetics , Parapoxvirus/genetics , Poxviridae Infections/veterinary , Sequence Analysis, DNA , Animals , Cattle , Cell Line , Goats , Molecular Sequence Data , Open Reading Frames , Orf virus/isolation & purification , Parapoxvirus/isolation & purification , Poxviridae Infections/virology , Sheep , Viral Proteins/genetics
19.
J Virol ; 77(19): 10339-47, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12970418

ABSTRACT

Here we present the complete genomic sequence of bovine herpesvirus 5 (BHV-5), an alphaherpesvirus responsible for fatal meningoencephalitis in cattle. The 138390-bp genome encodes 70 putative proteins and resembles the alpha2 subgroup of herpesviruses in genomic organization and gene content. BHV-5 is very similar to BHV-1, the etiological agent of infectious bovine rhinotracheitis, as reflected by the high level of amino acid identity in their protein repertoires (average, 82%). The highest similarity to BHV-1 products (>or=95% amino acid identity) is found in proteins involved in viral DNA replication and processing (UL5, UL15, UL29, and UL39) and in virion proteins (UL14, UL19, UL48, and US6). Among the least conserved (

Subject(s)
Genome, Viral , Herpesvirus 5, Bovine/genetics , Amino Acid Sequence , Base Sequence , Genes, Immediate-Early , Molecular Sequence Data , Promoter Regions, Genetic , Repetitive Sequences, Nucleic Acid , Virus Latency/genetics
20.
Virus Res ; 51(1): 93-103, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9381799

ABSTRACT

Bovine herpes virus 1 establishes a latent infection in sensory ganglionic neurons of cattle. During a latent infection latency related (LR) transcripts are the only detectable viral RNAs. DNA sequences in the LR promoter are positively regulated by neural factors. The 5' terminus of LR RNA in productively infected bovine cells is 20-30 nucleotides downstream from two overlapping TATA like elements. In contrast, the major start sites of LR transcription in trigeminal ganglia of latently infected cattle was 200-300 nucleotides upstream. Electrophoretic mobility shift assays (EMSA) were utilized to identify regions of the LR promoter that specifically bind factors present in dorsal root ganglia of cattle. Nuclear extracts from dorsal root ganglia of cattle or rat pheochromocytoma cells (PC12) contain abundant factors which specifically bind to a 72 bp XhoI-XbaI fragment. The 72 bp fragment is adjacent to the major start sites of LR transcriptional in trigeminal ganglia of latently infected cattle. In contrast, nuclear extracts from non-neural cells, bovine turbinate or Rat-2, did not exhibit similar binding patterns suggesting these factors were not abundant, had reduced binding affinity, or were absent in non-neural cells. Binding was localized to a 20 bp region of the XhoI-XbaI fragment by EMSA and Exonuclease III footprinting. When the XhoI-XbaI fragment was deleted, LR promoter activity was repressed in PC12 cells. Taken together, we conclude the XhoI-XbaI fragment is important for LR-RNA expression in neurons.


Subject(s)
Base Sequence/genetics , Herpesvirus 1, Bovine/genetics , Herpesvirus 1, Bovine/physiology , Nerve Growth Factors/pharmacology , PC12 Cells/physiology , Animals , Base Sequence/physiology , Cattle , Cells, Cultured , DNA Footprinting , DNA, Viral/analysis , DNA, Viral/metabolism , Deoxyribonucleases, Type II Site-Specific/genetics , Deoxyribonucleases, Type II Site-Specific/metabolism , Electrophoresis, Polyacrylamide Gel , Herpesvirus 1, Bovine/drug effects , Molecular Sequence Data , Nuclear Proteins/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Rats , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...