Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Virol ; 93(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30944180

ABSTRACT

Equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) represent two members of the family Arteriviridae and pose major threats for the horse- and swine-breeding industries worldwide. A previous study suggested that PRRSV nsp4, a 3C-like protease, antagonizes interferon beta (IFN-ß) production by cleaving the NF-κB essential modulator (NEMO) at a single site, glutamate 349 (E349). Here, we demonstrated that EAV nsp4 also inhibited virus-induced IFN-ß production by targeting NEMO for proteolytic cleavage and that the scission occurred at four sites: E166, E171, glutamine 205 (Q205), and E349. Additionally, we found that, besides the previously reported cleavage site E349 in NEMO, scission by PRRSV nsp4 took place at two additional sites, E166 and E171. These results imply that while cleaving NEMO is a common strategy utilized by EAV and PRRSV nsp4 to antagonize IFN induction, EAV nsp4 adopts a more complex substrate recognition mechanism to target NEMO. By analyzing the abilities of the eight different NEMO fragments resulting from EAV or PRRSV nsp4 scission to induce IFN-ß production, we serendipitously found that a NEMO fragment (residues 1 to 349) could activate IFN-ß transcription more robustly than full-length NEMO, whereas all other NEMO cleavage products were abrogated for the IFN-ß-inducing capacity. Thus, NEMO cleavage at E349 alone may not be sufficient to completely inactivate the IFN response via this signaling adaptor. Altogether, our findings suggest that EAV and PRRSV nsp4 cleave NEMO at multiple sites and that this strategy is critical for disarming the innate immune response for viral survival.IMPORTANCE The arterivirus nsp4-encoded 3C-like protease (3CLpro) plays an important role in virus replication and immune evasion, making it an attractive target for antiviral therapeutics. Previous work suggested that PRRSV nsp4 suppresses type I IFN production by cleaving NEMO at a single site. In contrast, the present study demonstrates that both EAV and PRRSV nsp4 cleave NEMO at multiple sites and that this strategy is essential for disruption of type I IFN production. Moreover, we reveal that EAV nsp4 also cleaves NEMO at glutamine 205 (Q205), which is not targeted by PRRSV nsp4. Notably, targeting a glutamine in NEMO for cleavage has been observed only with picornavirus 3C proteases (3Cpro) and coronavirus 3CLpro In aggregate, our work expands knowledge of the innate immune evasion mechanisms associated with NEMO cleavage by arterivirus nsp4 and describes a novel substrate recognition characteristic of EAV nsp4.


Subject(s)
Equartevirus/metabolism , Interferon-beta/biosynthesis , Viral Nonstructural Proteins/metabolism , Animals , Arteriviridae/metabolism , Arterivirus/metabolism , Cell Line , Equartevirus/physiology , HEK293 Cells , Horses , Humans , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Immune Evasion , Immunity, Innate , Interferon-beta/metabolism , Porcine respiratory and reproductive syndrome virus/metabolism , Proteolysis , Signal Transduction , Swine , Virus Replication
2.
J Virol ; 92(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29444949

ABSTRACT

Equine arteritis virus (EAV) can establish long-term persistent infection in the reproductive tract of stallions and is shed in the semen. Previous studies showed that long-term persistence is associated with a specific allele of the CXCL16 gene (CXCL16S) and that persistent infection is maintained despite the presence of a local inflammatory and humoral and mucosal antibody responses. In this study, we demonstrated that equine seminal exosomes (SEs) are enriched in a small subset of microRNAs (miRNAs). Most importantly, we demonstrated that long-term EAV persistence is associated with the downregulation of an SE-associated miRNA (eca-mir-128) and with an enhanced expression of CXCL16 in the reproductive tract, a putative target of eca-mir-128. The findings presented here suggest that SE eca-mir-128 is implicated in the regulation of the CXCL16/CXCR6 axis in the reproductive tract of persistently infected stallions, a chemokine axis strongly implicated in EAV persistence. This is a novel finding and warrants further investigation to identify its specific mechanism in modulating the CXCL16/CXCR6 axis in the reproductive tract of the EAV long-term carrier stallion.IMPORTANCE Equine arteritis virus (EAV) has the ability to establish long-term persistent infection in the stallion reproductive tract and to be shed in semen, which jeopardizes its worldwide control. Currently, the molecular mechanisms of viral persistence are being unraveled, and these are essential for the development of effective therapeutics to eliminate persistent infection. Recently, it has been determined that long-term persistence is associated with a specific allele of the CXCL16 gene (CXCL16S) and is maintained despite induction of local inflammatory, humoral, and mucosal antibody responses. This study demonstrated that long-term persistence is associated with the downregulation of seminal exosome miRNA eca-mir-128 and enhanced expression of its putative target, CXCL16, in the reproductive tract. For the first time, this study suggests complex interactions between eca-mir-128 and cellular elements at the site of EAV persistence and implicates this miRNA in the regulation of the CXCL16/CXCR6 axis in the reproductive tract during long-term persistence.


Subject(s)
Arterivirus Infections/veterinary , Chemokine CXCL16/biosynthesis , Equartevirus/physiology , Exosomes/genetics , Horse Diseases/virology , MicroRNAs/biosynthesis , Receptors, CXCR6/biosynthesis , Semen/cytology , Animals , Arterivirus Infections/virology , Down-Regulation/genetics , Genitalia, Male/metabolism , Genitalia, Male/virology , Horses , Male , MicroRNAs/genetics
3.
J Virol ; 91(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28424285

ABSTRACT

Equine arteritis virus (EAV) has a global impact on the equine industry as the causative agent of equine viral arteritis (EVA), a respiratory, systemic, and reproductive disease of equids. A distinctive feature of EAV infection is that it establishes long-term persistent infection in 10 to 70% of infected stallions (carriers). In these stallions, EAV is detectable only in the reproductive tract, and viral persistence occurs despite the presence of high serum neutralizing antibody titers. Carrier stallions constitute the natural reservoir of the virus as they continuously shed EAV in their semen. Although the accessory sex glands have been implicated as the primary sites of EAV persistence, the viral host cell tropism and whether viral replication in carrier stallions occurs in the presence or absence of host inflammatory responses remain unknown. In this study, dual immunohistochemical and immunofluorescence techniques were employed to unequivocally demonstrate that the ampulla is the main EAV tissue reservoir rather than immunologically privileged tissues (i.e., testes). Furthermore, we demonstrate that EAV has specific tropism for stromal cells (fibrocytes and possibly tissue macrophages) and CD8+ T and CD21+ B lymphocytes but not glandular epithelium. Persistent EAV infection is associated with moderate, multifocal lymphoplasmacytic ampullitis comprising clusters of B (CD21+) lymphocytes and significant infiltration of T (CD3+, CD4+, CD8+, and CD25+) lymphocytes, tissue macrophages, and dendritic cells (Iba-1+ and CD83+), with a small number of tissue macrophages expressing CD163 and CD204 scavenger receptors. This study suggests that EAV employs complex immune evasion mechanisms that warrant further investigation.IMPORTANCE The major challenge for the worldwide control of EAV is that this virus has the distinctive ability to establish persistent infection in the stallion's reproductive tract as a mechanism to ensure its maintenance in equid populations. Therefore, the precise identification of tissue and cellular tropism of EAV is critical for understanding the molecular basis of viral persistence and for development of improved prophylactic or treatment strategies. This study significantly enhances our understanding of the EAV carrier state in stallions by unequivocally identifying the ampullae as the primary sites of viral persistence, combined with the fact that persistence involves continuous viral replication in fibrocytes (possibly including tissue macrophages) and T and B lymphocytes in the presence of detectable inflammatory responses, suggesting the involvement of complex viral mechanisms of immune evasion. Therefore, EAV persistence provides a powerful new natural animal model to study RNA virus persistence in the male reproductive tract.


Subject(s)
B-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Epithelium/virology , Equartevirus/physiology , Genitalia/virology , Stromal Cells/virology , Viral Tropism , Animals , Arterivirus Infections/veterinary , Arterivirus Infections/virology , Fluorescent Antibody Technique , Horse Diseases/virology , Horses , Immunohistochemistry , Male
4.
J Virol ; 90(7): 3366-84, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26764004

ABSTRACT

UNLABELLED: Previous studies in our laboratory have identified equine CXCL16 (EqCXCL16) to be a candidate molecule and possible cell entry receptor for equine arteritis virus (EAV). In horses, the CXCL16 gene is located on equine chromosome 11 (ECA11) and encodes a glycosylated, type I transmembrane protein with 247 amino acids. Stable transfection of HEK-293T cells with plasmid DNA carrying EqCXCL16 (HEK-EqCXCL16 cells) increased the proportion of the cell population permissive to EAV infection from <3% to almost 100%. The increase in permissiveness was blocked either by transfection of HEK-EqCXCL16 cells with small interfering RNAs (siRNAs) directed against EqCXCL16 or by pretreatment with guinea pig polyclonal antibody against EqCXCL16 protein (Gp anti-EqCXCL16 pAb). Furthermore, using a virus overlay protein-binding assay (VOPBA) in combination with far-Western blotting, gradient-purified EAV particles were shown to bind directly to the EqCXCL16 protein in vitro. The binding of biotinylated virulent EAV strain Bucyrus at 4°C was significantly higher in HEK-EqCXCL16 cells than nontransfected HEK-293T cells. Finally, the results demonstrated that EAV preferentially infects subpopulations of horse CD14(+) monocytes expressing EqCXCL16 and that infection of these cells is significantly reduced by pretreatment with Gp anti-EqCXCL16 pAb. The collective data from this study provide confirmatory evidence that the transmembrane form of EqCXCL16 likely plays a major role in EAV host cell entry processes, possibly acting as a primary receptor molecule for this virus. IMPORTANCE: Outbreaks of EVA can be a source of significant economic loss for the equine industry from high rates of abortion in pregnant mares, death in young foals, establishment of the carrier state in stallions, and trade restrictions imposed by various countries. Similar to other arteriviruses, EAV primarily targets cells of the monocyte/macrophage lineage, which, when infected, are believed to play a critical role in EVA pathogenesis. To this point, however, the host-specified molecules involved in EAV binding and entry into monocytes/macrophages have not been identified. Identification of the cellular receptors for EAV may provide insights to design antivirals and better prophylactic reagents. In this study, we have demonstrated that EqCXCL16 acts as an EAV entry receptor in EAV-susceptible cells, equine monocytes. These findings represent a significant advance in our understanding of the fundamental mechanisms associated with the entry of EAV into susceptible cells.


Subject(s)
Chemokines, CXC/physiology , Equartevirus/physiology , Host Specificity/genetics , Receptors, Virus/genetics , Virus Internalization , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Arterivirus Infections/virology , Base Sequence , Cell Line , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Cricetinae , Equartevirus/genetics , Guinea Pigs , HEK293 Cells , Horse Diseases/virology , Horses , Humans , RNA Interference , RNA, Small Interfering/genetics , Rabbits , Receptors, Virus/metabolism , Sequence Analysis, DNA , Virus Attachment
5.
Nucleic Acids Res ; 43(17): 8416-34, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26304538

ABSTRACT

RNA viruses encode an RNA-dependent RNA polymerase (RdRp) that catalyzes the synthesis of their RNA(s). In the case of positive-stranded RNA viruses belonging to the order Nidovirales, the RdRp resides in a replicase subunit that is unusually large. Bioinformatics analysis of this non-structural protein has now revealed a nidoviral signature domain (genetic marker) that is N-terminally adjacent to the RdRp and has no apparent homologs elsewhere. Based on its conservation profile, this domain is proposed to have nucleotidylation activity. We used recombinant non-structural protein 9 of the arterivirus equine arteritis virus (EAV) and different biochemical assays, including irreversible labeling with a GTP analog followed by a proteomics analysis, to demonstrate the manganese-dependent covalent binding of guanosine and uridine phosphates to a lysine/histidine residue. Most likely this was the invariant lysine of the newly identified domain, named nidovirus RdRp-associated nucleotidyltransferase (NiRAN), whose substitution with alanine severely diminished the described binding. Furthermore, this mutation crippled EAV and prevented the replication of severe acute respiratory syndrome coronavirus (SARS-CoV) in cell culture, indicating that NiRAN is essential for nidoviruses. Potential functions supported by NiRAN may include nucleic acid ligation, mRNA capping and protein-primed RNA synthesis, possibilities that remain to be explored in future studies.


Subject(s)
Nidovirales/enzymology , Nucleotidyltransferases/chemistry , RNA-Dependent RNA Polymerase/chemistry , Viral Proteins/chemistry , Binding Sites , Conserved Sequence , Equartevirus/enzymology , Equartevirus/physiology , Guanosine/chemistry , Guanosine Triphosphate/metabolism , Manganese/chemistry , Nidovirales/genetics , Nucleotides/metabolism , Nucleotidyltransferases/metabolism , Phosphates/chemistry , Polyproteins/chemistry , Polyproteins/metabolism , Protein Structure, Tertiary , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , Severe acute respiratory syndrome-related coronavirus/physiology , Uridine/chemistry , Uridine Triphosphate/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
6.
Proc Natl Acad Sci U S A ; 110(9): E838-47, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23401522

ABSTRACT

Protein ubiquitination regulates important innate immune responses. The discovery of viruses encoding deubiquitinating enzymes (DUBs) suggests they remove ubiquitin to evade ubiquitin-dependent antiviral responses; however, this has never been conclusively demonstrated in virus-infected cells. Arteriviruses are economically important positive-stranded RNA viruses that encode an ovarian tumor (OTU) domain DUB known as papain-like protease 2 (PLP2). This enzyme is essential for arterivirus replication by cleaving a site within the viral replicase polyproteins and also removes ubiquitin from cellular proteins. To dissect this dual specificity, which relies on a single catalytic site, we determined the crystal structure of equine arteritis virus PLP2 in complex with ubiquitin (1.45 Å). PLP2 binds ubiquitin using a zinc finger that is uniquely integrated into an exceptionally compact OTU-domain fold that represents a new subclass of zinc-dependent OTU DUBs. Notably, the ubiquitin-binding surface is distant from the catalytic site, which allowed us to mutate this surface to significantly reduce DUB activity without affecting polyprotein cleavage. Viruses harboring such mutations exhibited WT replication kinetics, confirming that PLP2-mediated polyprotein cleavage was intact, but the loss of DUB activity strikingly enhanced innate immune signaling. Compared with WT virus infection, IFN-ß mRNA levels in equine cells infected with PLP2 mutants were increased by nearly an order of magnitude. Our findings not only establish PLP2 DUB activity as a critical factor in arteriviral innate immune evasion, but the selective inactivation of DUB activity also opens unique possibilities for developing improved live attenuated vaccines against arteriviruses and other viruses encoding similar dual-specificity proteases.


Subject(s)
Endopeptidases/metabolism , Equartevirus/enzymology , Fibroblasts/immunology , Fibroblasts/virology , Host-Pathogen Interactions/immunology , Immunity, Innate , Papain/metabolism , Animals , Coronavirus Papain-Like Proteases , Endopeptidases/chemistry , Endopeptidases/genetics , Equartevirus/physiology , HEK293 Cells , Hemorrhagic Fever Virus, Crimean-Congo/enzymology , Horses , Humans , Interferon-beta/genetics , Models, Molecular , Mutation/genetics , Papain/chemistry , Papain/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Signal Transduction/immunology , Substrate Specificity , Ubiquitin/chemistry , Virus Replication , Zinc Fingers
7.
J Virol ; 87(3): 1454-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23152531

ABSTRACT

Virus replication strongly depends on cellular factors, in particular, on host proteins. Here we report that the replication of the arteriviruses equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) is strongly affected by low-micromolar concentrations of cyclosporine A (CsA), an inhibitor of members of the cyclophilin (Cyp) family. In infected cells, the expression of a green fluorescent protein (GFP) reporter gene inserted into the PRRSV genome was inhibited with a half-maximal inhibitory concentration (IC(50)) of 5.2 µM, whereas the GFP expression of an EAV-GFP reporter virus was inhibited with an IC(50) of 0.95 µM. Debio-064, a CsA analog that lacks its undesirable immunosuppressive properties, inhibited EAV replication with an IC(50) that was 3-fold lower than that of CsA, whereas PRRSV-GFP replication was inhibited with an IC(50) similar to that of CsA. The addition of 4 µM CsA after infection prevented viral RNA and protein synthesis in EAV-infected cells, and CsA treatment resulted in a 2.5- to 4-log-unit reduction of PRRSV or EAV infectious progeny. A complete block of EAV RNA synthesis was also observed in an in vitro assay using isolated viral replication structures. The small interfering RNA-mediated knockdown of Cyp family members revealed that EAV replication strongly depends on the expression of CypA but not CypB. Furthermore, upon fractionation of intracellular membranes in density gradients, CypA was found to cosediment with membranous EAV replication structures, which could be prevented by CsA treatment. This suggests that CypA is an essential component of the viral RNA-synthesizing machinery.


Subject(s)
Antiviral Agents/pharmacology , Cyclophilins/antagonists & inhibitors , Cyclosporine/pharmacology , Equartevirus/physiology , Porcine respiratory and reproductive syndrome virus/physiology , Virus Replication/drug effects , Animals , Cell Line , Gene Knockdown Techniques , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Inhibitory Concentration 50 , Lepidoptera , Microbial Sensitivity Tests , RNA, Viral/biosynthesis , Viral Proteins/biosynthesis
8.
J Virol ; 86(5): 2474-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190716

ABSTRACT

Virus-induced membrane structures support the assembly and function of positive-strand RNA virus replication complexes. The replicase proteins of arteriviruses are associated with double-membrane vesicles (DMVs), which were previously proposed to derive from the endoplasmic reticulum (ER). Using electron tomography, we performed an in-depth ultrastructural analysis of cells infected with the prototypic arterivirus equine arteritis virus (EAV). We established that the outer membranes of EAV-induced DMVs are interconnected with each other and with the ER, thus forming a reticulovesicular network (RVN) resembling that previously described for the distantly related severe acute respiratory syndrome (SARS) coronavirus. Despite significant morphological differences, a striking parallel between the two virus groups, and possibly all members of the order Nidovirales, is the accumulation in the DMV interior of double-stranded RNA, the presumed intermediate of viral RNA synthesis. In our electron tomograms, connections between the DMV interior and cytosol could not be unambiguously identified, suggesting that the double-stranded RNA is compartmentalized by the DMV membranes. As a novel approach to visualize and quantify the RNA content of viral replication structures, we explored electron spectroscopic imaging of DMVs, which revealed the presence of phosphorus in amounts equaling on average a few dozen copies of the EAV RNA genome. Finally, our electron tomograms revealed a network of nucleocapsid protein-containing protein tubules that appears to be intertwined with the RVN. This potential intermediate in nucleocapsid formation, which was not observed in coronavirus-infected cells, suggests that arterivirus RNA synthesis and assembly are coordinated in intracellular space.


Subject(s)
Arterivirus Infections/virology , Endoplasmic Reticulum/virology , Equartevirus/physiology , RNA, Viral/genetics , Virus Replication , Animals , Arterivirus/genetics , Arterivirus/physiology , Arterivirus/ultrastructure , Cell Line , Endoplasmic Reticulum/ultrastructure , Equartevirus/genetics , Equartevirus/ultrastructure , Intracellular Membranes/ultrastructure , Intracellular Membranes/virology , RNA, Viral/metabolism
9.
J Virol ; 86(7): 3701-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258262

ABSTRACT

Arteriviruses are enveloped positive-strand RNA viruses for which the attachment proteins and cellular receptors have remained largely controversial. Arterivirus particles contain at least eight envelope proteins, an unusually large number among RNA viruses. These appear to segregate into three groups: major structural components (major glycoprotein GP5 and membrane protein [M]), minor glycoproteins (GP2a, GP3, and GP4), and small hydrophobic proteins (E and the recently discovered ORF5a protein). Biochemical studies previously suggested that the GP5-M heterodimer of porcine reproductive and respiratory syndrome virus (PRRSV) interacts with porcine sialoadhesin (pSn) in porcine alveolar macrophages (PAM). However, another study proposed that minor protein GP4, along with GP2a, interacts with CD163, another reported cellular receptor for PRRSV. In this study, we provide genetic evidence that the minor envelope proteins are the major determinant of arterivirus entry into cultured cells. A PRRSV infectious cDNA clone was equipped with open reading frames (ORFs) encoding minor envelope and E proteins of equine arteritis virus (EAV), the only known arterivirus displaying a broad tropism in cultured cells. Although PRRSV and EAV are only distantly related and utilize diversified transcription-regulating sequences (TRSs), a viable chimeric progeny virus was rescued. Strikingly, this chimeric virus (vAPRRS-EAV2ab34) acquired the broad in vitro cell tropism of EAV, demonstrating that the minor envelope proteins play a critical role as viral attachment proteins. We believe that chimeric arteriviruses of this kind will be a powerful tool for further dissection of the arterivirus replicative cycle, including virus entry, subgenomic RNA synthesis, and virion assembly.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/physiology , Porcine respiratory and reproductive syndrome virus/physiology , Viral Envelope Proteins/metabolism , Viral Tropism , Animals , Arterivirus Infections/virology , Base Sequence , Cell Line , Chlorocebus aethiops , Equartevirus/genetics , Molecular Sequence Data , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Swine , Vero Cells , Viral Envelope Proteins/genetics
10.
Vet Res ; 44: 22, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23537375

ABSTRACT

The upper respiratory tract mucosa represents the first line of defense, which has to be overcome by pathogens before invading the host. Considering the economic and ethical aspects involved in using experimental animals for pathogenesis studies, respiratory mucosal explants, in which the tissue's three-dimensional architecture is preserved, may be ideal alternatives. Different respiratory mucosal explant cultures have been developed. However, none of them could be inoculated with pathogens solely at the epithelium side. In the present study, equine nasal and nasopharyngeal explants were embedded in agarose (3%), leaving the epithelium side exposed to allow apical inoculation. Morphometric analysis did not show degenerative changes during 72 h of cultivation. The number of apoptotic cells in the mucosa slightly increased over time. After validation, the system was used for apical infection with a European strain (08P178) of equine arteritis virus (EAV) (107.6TCID50/mL per explant). Impermeability of agarose to virus particles was demonstrated by the absence of labeled microspheres (40 nm) and a lack of EAV-antigens in RK13 cells seeded underneath the agarose layer in which inoculated explants were embedded. At 72 hpi, 27% of the EAV-positive cells were CD172a+ and 19% were CD3+ in nasal explants and 45% of the EAV-positive cells were CD172a+ and 15% were CD3+ in nasopharyngeal explants. Only a small percentage of EAV-positive cells were IgM+. This study validates the usefulness of a polarized mucosal explant system and shows that CD172a+ myeloid cells and CD3+ T lymphocytes represent important EAV-target cells in the respiratory mucosa.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/physiology , Horse Diseases/virology , Nasal Mucosa/virology , Virus Replication , Animals , Arterivirus Infections/virology , Cell Survival , Female , Horses , In Vitro Techniques , Male , Myeloid Cells/cytology , Myeloid Cells/virology , Sepharose/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/virology , Viral Proteins/genetics , Viral Proteins/physiology
11.
Arch Virol ; 158(3): 701-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23151818

ABSTRACT

NF-κB, a family of transcription factors involved in different cell functions and immune responses is targeted by viruses. The mechanism of NF-κB signalling and its role in replication of EAV have not been investigated. We demonstrate that EAV infection in BHK-21 cells activates NF-κB, and this activation was found to be mediated through the MyD88 pathway. Infection of IKKß(-/-) murine embryo fibroblasts (MEFs), which are deficient in NF-κB signalling, resulted in lower virus titre, less cytopathic effect, and reduced expression of viral proteins. These findings implicate the MyD88 pathway in EAV-induced NF-κB activation and suggest that NF-κB activation is essential for efficient replication of EAV.


Subject(s)
Equartevirus/genetics , Equartevirus/physiology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Virus Replication , Animals , Cell Line , Cricetinae , Cytopathogenic Effect, Viral , Equartevirus/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Signal Transduction , Viral Proteins/biosynthesis , Viral Proteins/genetics
12.
J Virol ; 84(10): 4898-911, 2010 May.
Article in English | MEDLINE | ID: mdl-20219931

ABSTRACT

Extensive cell culture passage of the virulent Bucyrus (VB) strain of equine arteritis virus (EAV) to produce the modified live virus (MLV) vaccine strain has altered its tropism for equine CD3(+) T lymphocytes and CD14(+) monocytes. The VB strain primarily infects CD14(+) monocytes and a small subpopulation of CD3(+) T lymphocytes (predominantly CD4(+) T lymphocytes), as determined by dual-color flow cytometry. In contrast, the MLV vaccine strain has a significantly reduced ability to infect CD14(+) monocytes and has lost its capability to infect CD3(+) T lymphocytes. Using a panel of five recombinant chimeric viruses, we demonstrated that interactions among the GP2, GP3, GP4, GP5, and M envelope proteins play a major role in determining the CD14(+) monocyte tropism while the tropism for CD3(+) T lymphocytes is determined by the GP2, GP4, GP5, and M envelope proteins but not the GP3 protein. The data clearly suggest that there are intricate interactions among these envelope proteins that affect the binding of EAV to different cell receptors on CD3(+) T lymphocytes and CD14(+) monocytes. This study shows, for the first time, that CD3(+) T lymphocytes may play an important role in the pathogenesis of equine viral arteritis when horses are infected with the virulent strains of EAV.


Subject(s)
Equartevirus/physiology , Monocytes/virology , Protein Interaction Mapping , T-Lymphocytes/virology , Viral Envelope Proteins/metabolism , Viral Tropism , Virus Attachment , Animals , CD3 Complex/analysis , Cells, Cultured , Endothelial Cells/virology , Horses , Lipopolysaccharide Receptors/analysis , Monocytes/chemistry , T-Lymphocytes/chemistry
13.
Sci Rep ; 10(1): 2909, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32076048

ABSTRACT

Equine arteritis virus (EAV) is maintained in the horse populations through persistently infected stallions. The aims of the study were to monitor the spread of EAV among Polish Hucul horses, to analyse the variability of circulating EAVs both between- and within-horses, and to identify allelic variants of the serving stallions EqCXCL16 gene that had been previously shown to strongly correlate with long-term EAV persistence in stallions. Serum samples (n = 221) from 62 horses including 46 mares and 16 stallions were collected on routine basis between December 2010 and May 2013 and tested for EAV antibodies. In addition, semen from 11 stallions was tested for EAV RNA. A full genomic sequence of EAV from selected breeding stallions was determined using next generation sequencing. The proportion of seropositive mares among the tested population increased from 7% to 92% during the study period, while the proportion of seropositive stallions remained similar (64 to 71%). The EAV genomes from different stallions were 94.7% to 99.6% identical to each other. A number (41 to 310) of single nucleotide variants were identified within EAV sequences from infected stallions. Four stallions possessed EqCXCL16S genotype correlated with development of long-term carrier status, three of which were persistent shedders and the shedder status of the remaining one was undetermined. None of the remaining 12 stallions with EqCXCL16R genotype was identified as a persistent shedder.


Subject(s)
Chemokine CXCL16/genetics , Equartevirus/physiology , Horses/genetics , Horses/virology , Quasispecies/genetics , Semen/virology , Alleles , Animals , Arterivirus Infections/blood , Arterivirus Infections/genetics , Arterivirus Infections/veterinary , Female , Genome, Viral , Genotype , Horse Diseases/genetics , Horse Diseases/virology , Horses/blood , Male , Phylogeny , Polymorphism, Single Nucleotide/genetics
14.
Curr Opin Virol ; 27: 57-70, 2017 12.
Article in English | MEDLINE | ID: mdl-29172072

ABSTRACT

Equine arteritis virus (EAV) and porcine reproductive and respiratory syndrome virus (PRRSV) are the most economically important members of the family Arteriviridae. EAV and PRRSV cause reproductive and respiratory disease in equids and swine, respectively and constitute a significant economic burden to equine and swine industries around the world. Furthermore, they both cause abortion in pregnant animals and establish persistent infection in their natural hosts, which fosters viral shedding in semen leading to sexual transmission. The primary focus of this article is to provide an update on the effects of these two viruses on the reproductive tract of their natural hosts and provide a comparative analysis of clinical signs, virus-host interactions, mechanisms of viral pathogenesis and viral persistence.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/pathogenicity , Host-Pathogen Interactions , Porcine Reproductive and Respiratory Syndrome/transmission , Porcine respiratory and reproductive syndrome virus/pathogenicity , Pregnancy Complications, Infectious/veterinary , Animals , Arterivirus Infections/transmission , Arterivirus Infections/virology , Equartevirus/physiology , Female , Horse Diseases/economics , Horse Diseases/transmission , Horse Diseases/virology , Horses , Male , Porcine Reproductive and Respiratory Syndrome/virology , Pregnancy , Pregnancy Complications, Infectious/virology , Swine , Swine Diseases/economics , Swine Diseases/transmission , Swine Diseases/virology
15.
Adv Exp Med Biol ; 581: 429-33, 2006.
Article in English | MEDLINE | ID: mdl-17037573

ABSTRACT

EVA is an important if uncommon disease of horses. Potential economic losses attributable to EVA include direct losses from abortion, pneumonia in neonates, and febrile disease in performance horses. Indirect losses are those associated with national and international trade/animal movement regulations, particularly those pertaining to persistently infected carrier stallions and their semen. However, EAV infection and EVA are readily prevented through serological and virological screening of horses, coupled with sound management practices that include appropriate quarantine and strategic vaccination.


Subject(s)
Arterivirus Infections/diagnosis , Arterivirus Infections/virology , Equartevirus/physiology , Animals , Arterivirus Infections/epidemiology , Equidae , Horses
16.
Virus Res ; 220: 104-11, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27117322

ABSTRACT

Equine herpesvirus 1 (EHV-1) and equine arteritis virus (EAV) induce respiratory problems and abortion in horses and are considered as two serious threats to equine industry. Both EHV-1 and EAV misuse patrolling leukocytes in the upper respiratory tract to breach the basement membrane (BM) and to migrate to blood vessels. So far, the behavior and impact of a double infection in the respiratory mucosa of a horse are unknown. In the present study, the outcome of double infections with EHV-1 and the low virulent EAV strain 08P187 (superinfection with an interval of 12h or co-infection) were compared with single infections in fully susceptible RK-13 cells and equine upper respiratory mucosa explants. When RK-13 cells were inoculated with either EHV-1 or EAV 12h prior to the subsequent EAV or EHV-1 inoculation, the latter EAV or EHV-1 infection was clearly suppressed at 24hpi or 36hpi, respectively, without EHV-1 and EAV co-infecting the same RK-13 cells. After simultaneous infection with EHV-1 and EAV, higher numbers of EAV infected cells but similar numbers of EHV-1 infected cells were found compared to the single infections, with a low number of EHV-1 and EAV co-infected RK-13 cells at 48hpi and 72hpi. In the upper respiratory mucosa exposed to EAV 12h prior to EHV-1, the number and size of the EHV-1-induced plaques were similar to those of the EHV-1 single infected mucosa explants. In nasal and nasopharyngeal mucosae, EAV and EHV-1 pre-infections slightly reduced the number of EHV-1 and EAV infected leukocytes compared to the single infections and co-infection. In double EAV and EHV-1 infected explants, no co-infected leukocytes were detected. From these results, it can be concluded that EAV and EHV-1 are only slightly influencing each other's infection and that they do not infect the same mucosal leukocytes.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/physiology , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/physiology , Horse Diseases/virology , Respiratory Mucosa/virology , Animals , Arterivirus Infections/virology , Cell Line , Coinfection , Epithelial Cells/virology , Equartevirus/pathogenicity , Herpesviridae Infections/virology , Herpesvirus 1, Equid/pathogenicity , Horses , Leukocytes/virology , Tissue Culture Techniques , Viral Load , Virus Replication
17.
J Comp Pathol ; 109(1): 29-46, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8408779

ABSTRACT

The relationship between stage of reproductive tract maturity and susceptibility to the experimental establishment of persistent infection with equine arteritis virus (EAV) was investigated in 21 prepubertal and 15 peripubertal colts. Five of six prepubertal colts inoculated intranasally remained infected in the reproductive tract from post-challenge day 28 to 93 and two of six from post-challenge day 120 to 180. No virus was detected in five of these animals killed on post-challenge day 210. Each of two peripubertal colts remained infected in the reproductive tract at post-challenge day 60 and one of nine was found to be persistently infected with EAV 15 months after challenge. These findings confirm that the virus can replicate in the reproductive tract of a significant proportion of colts for a variable period of time after clinical recovery in the absence of circulating concentrations of testosterone equivalent to those found in sexually mature stallions. Long-term persistent infection with EAV does not appear to occur in colts exposed to the virus before the onset of peripubertal development. We suggest that colts should be vaccinated at approximately 6 months of age, before peripubertal development but after the disappearance of maternally acquired antibodies.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/isolation & purification , Horse Diseases/epidemiology , Sexual Maturation/physiology , Age Factors , Age of Onset , Animals , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Arterivirus Infections/epidemiology , Arterivirus Infections/physiopathology , Disease Susceptibility , Epididymis/microbiology , Equartevirus/physiology , Horses , Male , Prostate/microbiology , Semen/microbiology , Testis/microbiology , Vas Deferens/microbiology , Virus Replication
18.
J Vet Med Sci ; 59(10): 943-5, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9362047

ABSTRACT

Equine arteritis virus (EAV) grown on RK13 cell cultures was tested for hemagglutination (HA) with erythrocytes from a variety of species at 4 degrees C, room temperature and 37 degrees C. HA was observed at all temperatures with erythrocytes from mouse and chicken but not with those of cattle, horse, rabbit, guinea pig, mongolian gerbil, goose or chick embryo. Chickens showed an individual variation in agglutinability of their erythrocytes, requiring selection of birds to obtain erythrocytes for HA. The HA activity was enhanced by treatment of virus materials with Tween 80 followed by treatment with ether. The HA reaction was inhibited by specific antiserum. Higher HA-inhibiting (HI) antibody titers were obtained by the incubation of serum-HA antigen mixture at 4 degrees C for 24 hr. HI antibody titers of individual horse sera showed a significant positive correlation with their neutralizing antibody titers.


Subject(s)
Equartevirus/physiology , Hemagglutination, Viral , Animals , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Cattle , Cell Line , Chick Embryo , Chickens , Equartevirus/drug effects , Equartevirus/immunology , Erythrocytes/cytology , Erythrocytes/physiology , Erythrocytes/virology , Geese , Gerbillinae , Guinea Pigs , Horses , Immune Sera/immunology , Immune Sera/pharmacology , Kidney/cytology , Kidney/virology , Mice , Rabbits , Species Specificity , Temperature , Time Factors
19.
J Vet Med Sci ; 60(4): 447-50, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9592716

ABSTRACT

Heparin inhibited hemagglutination (HA) by equine arteritis virus (EAV) as well as did HA by Aujeszky's disease virus (ADV), but failed to inhibit HA by parainfluenza virus type 3 (PIV-3). The minimal concentration of heparin required to inhibit 8 HA U of EAV was 0.1 U/ml. In addition, most EAV hemagglutinin was retained by heparin acrylic beads, as was ADV hemagglutinin, but was not PIV-3 hemagglutinin. Mouse erythrocytes failed to combine with the HA inhibitory factor of heparin. However, mouse erythrocytes treated with heparinase had greatly reduced agglutinability by EAV. All these findings suggest that a heparin-like molecule on the surface of mouse erythrocytes serves as the virus-cell receptor.


Subject(s)
Equartevirus/physiology , Hemagglutination/drug effects , Heparin/pharmacology , Animals , Cattle , Cell Line , Equartevirus/drug effects , Erythrocytes/drug effects , Erythrocytes/physiology , Erythrocytes/virology , Hemagglutination Inhibition Tests , Hemagglutination Tests , Hemagglutinins, Viral/physiology , Heparin Lyase/pharmacology , Herpesvirus 1, Suid/physiology , Kidney , Mice , Rabbits , Swine
20.
Virology ; 462-463: 388-403, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24913633

ABSTRACT

The advent of recombinant DNA technology, development of infectious cDNA clones of RNA viruses, and reverse genetic technologies have revolutionized how viruses are studied. Genetic manipulation of full-length cDNA clones has become an especially important and widely used tool to study the biology, pathogenesis, and virulence determinants of both positive and negative stranded RNA viruses. The first full-length infectious cDNA clone of equine arteritis virus (EAV) was developed in 1996 and was also the first full-length infectious cDNA clone constructed from a member of the order Nidovirales. This clone was extensively used to characterize the molecular biology of EAV and other Nidoviruses. The objective of this review is to summarize the characterization of the virulence (or attenuation) phenotype of the recombinant viruses derived from several infectious cDNA clones of EAV in horses, as well as their application for characterization of the molecular basis of viral neutralization, persistence, and cellular tropism.


Subject(s)
Cloning, Molecular , DNA, Complementary/genetics , Equartevirus/genetics , Equartevirus/physiology , Animals , Equartevirus/pathogenicity , Horses , Reverse Genetics , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL