Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
1.
Viruses ; 15(1)2023 01 16.
Article in English | MEDLINE | ID: mdl-36680295

ABSTRACT

Equine viral arteritis is an infectious disease of equids caused by equine arteritis virus (EAV), an RNA virus of the family Arteriviridae. Dendritic cells (DC) are important modulators of the immune response with the ability to present antigen to naïve T cells and can be generated in vitro from monocytes (MoDC). DC are important targets for many viruses and this interaction is crucial for the establishment-or rather not-of an anti-viral immunity. Little is known of the effect EAV has on host immune cells, particularly DC. To study the interaction of eqDC with EAV in vitro, an optimized eqMoDC system was used, which was established in a previous study. MoDC were infected with strains of different genotypes and pathogenicity. Virus replication was determined through titration and qPCR. The effect of the virus on morphology, phenotype and function of cells was assessed using light microscopy, flow cytometry and in vitro assays. This study confirms that EAV replicates in monocytes and MoDC. The replication was most efficient in mature MoDC, but variable between strains. Only the virulent strain caused a significant down-regulation of certain proteins such as CD14 and CD163 on monocytes and of CD83 on mature MoDC. Functional studies conducted after infection showed that EAV inhibited the endocytic and phagocytic capacity of Mo and mature MoDC with minimal effect on immature MoDC. Infected MoDC showed a reduced ability to stimulate T cells. Ultimately, EAV replication resulted in an apoptosis-mediated cell death. Thus, EAV evades the host anti-viral immunity both by inhibition of antigen presentation early after infection and through killing infected DC during replication.


Subject(s)
Equartevirus , Animals , Horses , Equartevirus/genetics , Monocytes , Virulence , Dendritic Cells , Cell Differentiation
2.
Viruses ; 14(4)2022 04 01.
Article in English | MEDLINE | ID: mdl-35458479

ABSTRACT

Equine arteritis virus (EAV), an enveloped positive-strand RNA virus, is an important pathogen of horses and the prototype member of the Arteiviridae family. Unlike many other enveloped viruses, which possess homotrimeric spikes, the spike responsible for cellular tropism in Arteriviruses is a heterotrimer composed of 3 glycoproteins: GP2, GP3, and GP4. Together with the hydrophobic protein E they are the minor components of virus particles. We describe the expression of all 3 minor glycoproteins, each equipped with a different tag, from a multi-cassette system in mammalian BHK-21 cells. Coprecipitation studies suggest that a rather small faction of GP2, GP3, and GP4 form dimeric or trimeric complexes. GP2, GP3, and GP4 co-localize with each other and also, albeit weaker, with the E-protein. The co-localization of GP3-HA and GP2-myc was tested with markers for ER, ERGIC, and cis-Golgi. The co-localization of GP3-HA was the same regardless of whether it was expressed alone or as a complex, whereas the transport of GP2-myc to cis-Golgi was higher when this protein was expressed as a complex. The glycosylation pattern was also independent of whether the proteins were expressed alone or together. The recombinant spike might be a tool for basic research but might also be used as a subunit vaccine for horses.


Subject(s)
Arterivirus , Equartevirus , Animals , Equartevirus/genetics , Equartevirus/metabolism , Glycoproteins/genetics , Guanidines , Horses , Mammals , Piperazines , Viral Envelope Proteins/metabolism
3.
Infect Genet Evol ; 85: 104455, 2020 11.
Article in English | MEDLINE | ID: mdl-32668365

ABSTRACT

Susceptibility to long-term persistent infection with Equine Arteritis Virus (EAV) in stallions is related with EqCXCL16 gene alleles of the host. In our study EqCXCL16 gene alleles were determined for 63 EAV shedders and 126 non-shedders of various horse breeds. In total, 60 (31.7%) out of 189 tested stallions were identified as carriers of susceptible variants of EqCXCL16 by real time PCR and Sanger sequencing. The presence of susceptible genotype was related to horse breed with the highest percentage in Wielkopolska breed, Polish coldblood and Silesian breed horses. Strong correlation between EqCXCL16 susceptible genotypes and EAV shedding in semen (p < .0001) was observed.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus Infections/virology , Chemokine CXCL16/genetics , Equartevirus/genetics , Horse Diseases/virology , Horses/virology , Alleles , Amino Acid Sequence , Animals , Genotype , Phylogeny , Poland/epidemiology , RNA, Viral , Semen/virology , Sequence Analysis
4.
Viruses ; 12(2)2020 02 14.
Article in English | MEDLINE | ID: mdl-32075207

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is prevalent throughout the world and has caused great economic losses to the swine industry. Nonstructural protein 10 (nsp10) is a superfamily 1 helicase participating in multiple processes of virus replication and one of the three most conserved proteins in nidoviruses. Here we report three high resolution crystal structures of highly pathogenic PRRSV nsp10. PRRSV nsp10 has multiple domains, including an N-terminal zinc-binding domain (ZBD), a ß-barrel domain, a helicase core with two RecA-like domains, and a C-terminal domain (CTD). The CTD adopts a novel fold and is required for the overall structure and enzymatic activities. Although each domain except the CTD aligns well with its homologs, PRRSV nsp10 adopts an unexpected extended overall structure in crystals and solution. Moreover, structural and functional analyses of PRRSV nsp10 versus its closest homolog, equine arteritis virus nsp10, suggest that DNA binding might induce a profound conformational change of PRRSV nsp10 to exert functions, thus shedding light on the mechanisms of activity regulation of this helicase.


Subject(s)
DNA Helicases/chemistry , Porcine respiratory and reproductive syndrome virus/enzymology , Viral Nonstructural Proteins/chemistry , Crystallization , DNA Helicases/genetics , Equartevirus/genetics , Porcine respiratory and reproductive syndrome virus/pathogenicity , Protein Structure, Secondary , Viral Nonstructural Proteins/genetics , Virus Replication
5.
Viruses ; 11(8)2019 08 09.
Article in English | MEDLINE | ID: mdl-31404947

ABSTRACT

Equine arteritis virus (EAV) is a prototype member of the Arterivirus family, comprising important pathogens of domestic animals. Minor glycoproteins of Arteriviruses are responsible for virus entry and cellular tropism. The experimental methods for studying minor Arterivirus proteins are limited because of the lack of antibodies and nested open reading frames (ORFs). In this study, we generated recombinant EAV with separated ORFs 3 and 4, and Gp3 carrying HA-tag (Gp3-HA). The recombinant viruses were stable on passaging and replicated in titers similar to the wild-type EAV. Gp3-HA was incorporated into the virion particles as monomers and as a Gp2/Gp3-HA/Gp4 trimer. Gp3-HA localized in ER and, to a lesser extent, in the Golgi, it also co-localized with the E protein but not with the N protein. The co-localization of Gp3-HA and the E protein with ERGIC was reduced. Moreover, EAV with Gp3-HA could become a valuable research tool for identifying host cell factors during infection and the role of Gp3 in virus attachment and entry.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/genetics , Equartevirus/metabolism , Horse Diseases/virology , Host-Pathogen Interactions , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Animals , Cell Line , Genetic Engineering , Genome, Viral , Golgi Apparatus/metabolism , Horses , Intracellular Space , Mutation , Open Reading Frames , Protein Transport , Virus Replication
6.
J Virol ; 93(18)2019 09 15.
Article in English | MEDLINE | ID: mdl-31243130

ABSTRACT

Previously, the cyclophilin inhibitors cyclosporine (CsA) and alisporivir (ALV) were shown to inhibit the replication of diverse RNA viruses, including arteriviruses and coronaviruses, which both belong to the order Nidovirales In this study, we aimed to identify arterivirus proteins involved in the mode of action of cyclophilin inhibitors and to investigate how these compounds inhibit arterivirus RNA synthesis in the infected cell. Repeated passaging of the arterivirus prototype equine arteritis virus (EAV) in the presence of CsA revealed that reduced drug sensitivity is associated with the emergence of adaptive mutations in nonstructural protein 5 (nsp5), one of the transmembrane subunits of the arterivirus replicase polyprotein. Introduction of singular nsp5 mutations (nsp5 Q21R, Y113H, or A134V) led to an ∼2-fold decrease in sensitivity to CsA treatment, whereas combinations of mutations further increased EAV's CsA resistance. The detailed experimental characterization of engineered EAV mutants harboring CsA resistance mutations implicated nsp5 in arterivirus RNA synthesis. Particularly, in an in vitro assay, EAV RNA synthesis was far less sensitive to CsA treatment when nsp5 contained the adaptive mutations mentioned above. Interestingly, for increased sensitivity to the closely related drug ALV, CsA-resistant nsp5 mutants required the incorporation of an additional adaptive mutation, which resided in nsp2 (H114R), another transmembrane subunit of the arterivirus replicase. Our study provides the first evidence for the involvement of nsp2 and nsp5 in the mechanism underlying the inhibition of arterivirus replication by cyclophilin inhibitors.IMPORTANCE Currently, no approved treatments are available to combat infections with nidoviruses, a group of positive-stranded RNA viruses, including important zoonotic and veterinary pathogens. Previously, the cyclophilin inhibitors cyclosporine (CsA) and alisporivir (ALV) were shown to inhibit the replication of diverse nidoviruses (both arteriviruses and coronaviruses), and they may thus represent a class of pan-nidovirus inhibitors. In this study, using the arterivirus prototype equine arteritis virus, we have established that resistance to CsA and ALV treatment is associated with adaptive mutations in two transmembrane subunits of the viral replication machinery, nonstructural proteins 2 and 5. This is the first evidence for the involvement of specific replicase subunits of arteriviruses in the mechanism underlying the inhibition of their replication by cyclophilin inhibitors. Understanding this mechanism of action is of major importance to guide future drug design, both for nidoviruses and for other RNA viruses inhibited by these compounds.


Subject(s)
Equartevirus/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Nonstructural Proteins/metabolism , Arterivirus/genetics , Cell Line , Cyclophilins/metabolism , Cyclosporine/antagonists & inhibitors , Equartevirus/metabolism , HEK293 Cells , Humans , Mutation , Nidovirales/genetics , Nidovirales/metabolism , Nucleic Acid Synthesis Inhibitors/metabolism , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/genetics , Virus Replication
7.
J Virol ; 93(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30918077

ABSTRACT

Equine arteritis virus (EAV) is the causative agent of equine viral arteritis (EVA), a reproductive and respiratory disease of horses. Following natural infection, 10 to 70% of infected stallions can become carriers of EAV and continue to shed virus in the semen. In this study, sequential viruses isolated from nasal secretions, buffy coat cells, and semen of seven experimentally infected and two naturally infected EAV carrier stallions were deep sequenced to elucidate the intrahost microevolutionary process after a single transmission event. Analysis of variants from nasal secretions and buffy coat cells lacked extensive positive selection; however, characteristics of the mutant spectra were different in the two sample types. In contrast, the initial semen virus populations during acute infection have undergone a selective bottleneck, as reflected by the reduction in population size and diversifying selection at multiple sites in the viral genome. Furthermore, during persistent infection, extensive genome-wide purifying selection shaped variant diversity in the stallion reproductive tract. Overall, the nonstochastic nature of EAV evolution during persistent infection was driven by active intrahost selection pressure. Among the open reading frames within the viral genome, ORF3, ORF5, and the nsp2-coding region of ORF1a accumulated the majority of nucleotide substitutions during persistence, with ORF3 and ORF5 having the highest intrahost evolutionary rates. The findings presented here provide a novel insight into the evolutionary mechanisms of EAV and identified critical regions of the viral genome likely associated with the establishment and maintenance of persistent infection in the stallion reproductive tract.IMPORTANCE EAV can persist in the reproductive tract of infected stallions, and consequently, long-term carrier stallions constitute its sole natural reservoir. Previous studies demonstrated that the ampullae of the vas deferens are the primary site of viral persistence in the stallion reproductive tract and the persistence is associated with a significant inflammatory response that is unable to clear the infection. This is the first study that describes EAV full-length genomic evolution during acute and long-term persistent infection in the stallion reproductive tract using next-generation sequencing and contemporary sequence analysis techniques. The data provide novel insight into the intrahost evolution of EAV during acute and persistent infection and demonstrate that persistent infection is characterized by extensive genome-wide purifying selection and a nonstochastic evolutionary pattern mediated by intrahost selective pressure, with important nucleotide substitutions occurring in ORF1a (region encoding nsp2), ORF3, and ORF5.


Subject(s)
Arterivirus Infections/genetics , Equartevirus/genetics , Host-Pathogen Interactions/genetics , Amino Acid Sequence/genetics , Animals , Arterivirus Infections/virology , Base Sequence/genetics , Carrier State/virology , Equartevirus/metabolism , Equartevirus/pathogenicity , Evolution, Molecular , Genome, Viral/genetics , Horse Diseases/virology , Horses/genetics , Male , Open Reading Frames/genetics , Phylogeny , Semen/virology , Sequence Analysis/methods
8.
Arch Virol ; 163(6): 1469-1478, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29435711

ABSTRACT

Quantitation of virions is one of the important indexes in virological studies. To establish a sensitive and rapid quantitative detection method for equine arteritis virus (EAV), an antigen-capture enzyme-linked immunosorbent assay (AC-ELISA) was developed by using two EAV nucleoprotein monoclonal antibodies (mAbs), 2B9 and 2B3, prepared in this study. After condition optimization, mAb 2B9 was used as the capture antibody, and HRP-labeled 2B3 was chosen as the detecting antibody. The AC-ELISA had a good standard curve when viral particles of the Bucyrus EAV strain were used as a reference standard. The detection limit for the Bucyrus EAV strain was 36 PFU, and the method had a good linear relationship between 72-2297 PFU. The AC-ELISA could specifically detect the Bucyrus EAV strain and had no cross-reaction with other equine viruses. The sensitivity of the AC-ELISA was much higher than that of a western blotting assay but lower than that of a real-time PCR method. However, as a quantitative antigen detection method, the sensitivity of the AC-ELISA was approximately 300 times than the western blotting assay. Furthermore, the AC-ELISA assay could be successfully used in quantification of viral content in an in vitro infection assay, such as a one-step growth curve of EAV, as well as in a transfection assay, such as virus rescue from an infectious cDNA clone of EAV. These results show that the AC-ELISA established in this study is a good alternative for antigen detection of EAV, being a simple, convenient and quantitative detection method for EAV antigens.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Viral/chemistry , Antigens, Viral/analysis , Arterivirus Infections/veterinary , Enzyme-Linked Immunosorbent Assay/methods , Equartevirus/isolation & purification , Horse Diseases/diagnosis , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/biosynthesis , Antibodies, Viral/isolation & purification , Antigens, Viral/genetics , Antigens, Viral/immunology , Arterivirus Infections/diagnosis , Arterivirus Infections/virology , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay/standards , Enzyme-Linked Immunosorbent Assay/veterinary , Epithelial Cells , Equartevirus/genetics , Equartevirus/immunology , Female , HEK293 Cells , Horse Diseases/virology , Horseradish Peroxidase/chemistry , Horses , Humans , Immunization , Limit of Detection , Mice , Mice, Inbred BALB C , Virion/genetics , Virion/immunology
9.
Virus Res ; 246: 28-34, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29337162

ABSTRACT

Recently, a novel antiviral compound (K22) that inhibits replication of a broad range of animal and human coronaviruses was reported to interfere with viral RNA synthesis by impairing double-membrane vesicle (DMV) formation (Lundin et al., 2014). Here we assessed potential antiviral activities of K22 against a range of viruses representing two (sub)families of the order Nidovirales, the Arteriviridae (porcine reproductive and respiratory syndrome virus [PRRSV], equine arteritis virus [EAV] and simian hemorrhagic fever virus [SHFV]), and the Torovirinae (equine torovirus [EToV] and White Bream virus [WBV]). Possible effects of K22 on nidovirus replication were studied in suitable cell lines. K22 concentrations significantly decreasing infectious titres of the viruses included in this study ranged from 25 to 50 µM. Reduction of double-stranded RNA intermediates of viral replication in nidovirus-infected cells treated with K22 confirmed the anti-viral potential of K22. Collectively, the data show that K22 has antiviral activity against diverse lineages of nidoviruses, suggesting that the inhibitor targets a critical and conserved step during nidovirus replication.


Subject(s)
Antiviral Agents/pharmacology , Arterivirus/drug effects , Benzamides/pharmacology , Coronaviridae/drug effects , Equartevirus/drug effects , Piperidines/pharmacology , Porcine respiratory and reproductive syndrome virus/drug effects , Torovirus/drug effects , Animals , Arterivirus/genetics , Arterivirus/growth & development , Arterivirus/metabolism , Carps , Cell Line , Chlorocebus aethiops , Coronaviridae/genetics , Coronaviridae/growth & development , Coronaviridae/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , Equartevirus/genetics , Equartevirus/growth & development , Equartevirus/metabolism , Mesocricetus , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/metabolism , RNA, Double-Stranded/antagonists & inhibitors , RNA, Double-Stranded/biosynthesis , RNA, Double-Stranded/genetics , RNA, Viral/antagonists & inhibitors , RNA, Viral/biosynthesis , RNA, Viral/genetics , Torovirus/genetics , Torovirus/growth & development , Torovirus/metabolism , Virus Replication/drug effects
10.
Appl Microbiol Biotechnol ; 102(1): 413-423, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29134331

ABSTRACT

Reverse genetics is one of the most powerful tools in modern virology. Equine arteritis virus (EAV) is the prototype member of the Equartevirus. In this study, a new reverse genetics system for the recovery of equine arteritis virus from a cDNA plasmid, which contains viral cDNA sequence flanked by hammerhead ribozyme (HamRz) and hepatitis delta virus ribozyme (HdvRz) sequences in both terminals of the viral genome, was developed by optimization of the promoter and terminator regions. Cellular RNA polymerase II drove the transcription of the viral genome. The results showed that the rescued virus (ic-EAV) shared similar morphological and growth characteristics with the wild-type (WT) virus, and could be distinguished from the WT virus via an engineered BspEI restriction site in the nsp3 gene. By using the reverse genetics method established in this study, a G-to-C silent mutation at site 12642 resulted in a significant change in the plaque size of the rescued virus. Moreover, an eGFP-labeled EAV was constructed by introducing the eGFP gene into the infectious clone of EAV, which facilitated the observation of the infection of EAV in target cells. Hence, the newly reverse genetics method of EAV established in this study can be easily manipulated and would be helpful for studying the pathogenic mechanism of EAV.


Subject(s)
DNA, Complementary/genetics , DNA, Viral/genetics , Equartevirus/genetics , Genome, Viral , Reverse Genetics/methods , Animals , Cell Line , Cells, Cultured , Cloning, Molecular/methods , Equartevirus/growth & development , Equartevirus/isolation & purification , Genetic Vectors , Horses , Plasmids/genetics , RNA Polymerase II/genetics , RNA, Viral/genetics , Virion/genetics
11.
BMC Vet Res ; 13(1): 316, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29115996

ABSTRACT

BACKGROUND: Equine arteritis virus (EAV) is responsible for infections in equids. It can spread easily within the horse population and has a major impact on the horse breeding industry. No EAV outbreak has ever been reported in Serbia. To determine whether EAV is nonetheless circulating there, especially in the Vojvodina region, 340 horse serum samples were subjected to serology testing to detect EAV antibodies. In parallel, semen samples from three seropositive stallions were collected to evaluate their EAV status, using RT-qPCR and virus isolation on cell culture. RESULTS: Horse sera with EAV antibodies represented 15.88% (54/340) of the tested samples, 83.23% (283/340) being negative, and just three samples (0.89%) being uninterpretable due to cytotoxicity. Only 7.2% (10/138) of horses kept by private owners on their own property were seropositive for EAV, whereas 21.8% (44/202) of horses kept on stud farms had EAV antibodies. Phylogenetic analysis showed that the Serbian EAV isolate was most closely related to isolates from the neighbouring Hungary. CONCLUSIONS: EAV is circulating in the Serbian horse population, especially among the breeding population certainly due to the use of EAV shedder stallions since there is no surveillance programme in Serbia and only limited checks on racehorses. Moreover, phylogenetic analysis indicates that the EAV isolated from a Lipizzaner stallion in Serbia is closely related to isolates from Hungary, and together form a new cluster.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/isolation & purification , Horse Diseases/epidemiology , Horse Diseases/virology , Animal Husbandry , Animals , Antibodies, Viral , Arterivirus Infections/epidemiology , Equartevirus/genetics , Female , Horses , Male , Phylogeny , Semen/virology , Serbia/epidemiology , Seroepidemiologic Studies
12.
Transbound Emerg Dis ; 64(6): 1655-1660, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28921885

ABSTRACT

A novel equine arteritis virus (EAV) was isolated and sequenced from feral donkeys in Chile. Phylogenetic analysis indicates that the new virus and South African asinine strains diverged at least 100 years from equine EAV strains. The results indicate that asinine strains belonged to a different EAV genotype.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/isolation & purification , Equidae , Animals , Arterivirus Infections/virology , Chile , Equartevirus/classification , Equartevirus/genetics , Male , Phylogeny , Polymerase Chain Reaction/veterinary , Viral Proteins/analysis
13.
Methods Mol Biol ; 1602: 11-28, 2017.
Article in English | MEDLINE | ID: mdl-28508211

ABSTRACT

Development and characterization of several infectious cDNA clones of equine arteritis virus (EAV) have been described in the literature. Here we describe the assembly of the full-length infectious cDNA clone of the virulent Bucyrus strain (VBS; ATCC VR-796) of EAV in a plasmid vector. This system allows generation of infectious in vitro-transcribed (IVT) RNA from the linearized plasmid that can be transfected or electroporated into mammalian cells to produce infectious recombinant progeny virus. This is an efficient reverse genetics system that allows easy manipulation of EAV genomes to study molecular biology of the virus and pathogenesis of equine viral arteritis.


Subject(s)
DNA, Complementary , Equartevirus/genetics , Genome, Viral , Animals , Arterivirus Infections/veterinary , Cell Line , Horse Diseases/virology , Horses , Mutagenesis, Site-Directed , Plasmids/genetics , RNA, Viral , Recombination, Genetic , Transfection , Virus Replication
14.
PLoS Genet ; 12(12): e1006467, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27930647

ABSTRACT

Equine arteritis virus (EAV) is the causative agent of equine viral arteritis (EVA), a respiratory, systemic, and reproductive disease of horses and other equid species. Following natural infection, 10-70% of the infected stallions can become persistently infected and continue to shed EAV in their semen for periods ranging from several months to life. Recently, we reported that some stallions possess a subpopulation(s) of CD3+ T lymphocytes that are susceptible to in vitro EAV infection and that this phenotypic trait is associated with long-term carrier status following exposure to the virus. In contrast, stallions not possessing the CD3+ T lymphocyte susceptible phenotype are at less risk of becoming long-term virus carriers. A genome wide association study (GWAS) using the Illumina Equine SNP50 chip revealed that the ability of EAV to infect CD3+ T lymphocytes and establish long-term carrier status in stallions correlated with a region within equine chromosome 11. Here we identified the gene and mutations responsible for these phenotypes. Specifically, the work implicated three allelic variants of the equine orthologue of CXCL16 (EqCXCL16) that differ by four non-synonymous nucleotide substitutions (XM_00154756; c.715 A → T, c.801 G → C, c.804 T → A/G, c.810 G → A) within exon 1. This resulted in four amino acid changes with EqCXCL16S (XP_001504806.1) having Phe, His, Ile and Lys as compared to EqCXL16R having Tyr, Asp, Phe, and Glu at 40, 49, 50, and 52, respectively. Two alleles (EqCXCL16Sa, EqCXCL16Sb) encoded identical protein products that correlated strongly with long-term EAV persistence in stallions (P<0.000001) and are required for in vitro CD3+ T lymphocyte susceptibility to EAV infection. The third (EqCXCL16R) was associated with in vitro CD3+ T lymphocyte resistance to EAV infection and a significantly lower probability for establishment of the long-term carrier state (viral persistence) in the male reproductive tract. EqCXCL16Sa and EqCXCL16Sb exert a dominant mode of inheritance. Most importantly, the protein isoform EqCXCL16S but not EqCXCL16R can function as an EAV cellular receptor. Although both molecules have equal chemoattractant potential, EqCXCL16S has significantly higher scavenger receptor and adhesion properties compared to EqCXCL16R.


Subject(s)
Arterivirus Infections/genetics , Chemokines, CXC/genetics , Equartevirus/genetics , Horse Diseases/genetics , Alleles , Amino Acid Sequence/genetics , Animals , Arterivirus Infections/veterinary , Arterivirus Infections/virology , CD3 Complex/genetics , CD3 Complex/immunology , Equartevirus/pathogenicity , Genetic Predisposition to Disease , Genome-Wide Association Study , Horse Diseases/virology , Horses/genetics , Horses/virology , Male , Phylogeny , Semen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology
15.
Arch Virol ; 161(11): 3125-36, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27541817

ABSTRACT

Equine arteritis virus (EAV) is the causative agent of equine viral arteritis, a respiratory and reproductive disease of equids. EAV infection can induce abortion in pregnant mares, fulminant bronchointerstitial pneumonia in foals, and persistent infection in stallions. Here, we developed two RNA in situ hybridization (ISH) assays (conventional and RNAscope(®) ISH) for the detection of viral RNA in formalin-fixed paraffin-embedded (FFPE) tissues and evaluated and compared their performance with nucleocapsid-specific immunohistochemistry (IHC) and virus isolation (VI; gold standard) techniques. The distribution and cellular localization of EAV RNA and antigen were similar in tissues from aborted equine fetuses. Evaluation of 80 FFPE tissues collected from 16 aborted fetuses showed that the conventional RNA ISH assay had a significantly lower sensitivity than the RNAscope(®) and IHC assays, whereas there was no difference between the latter two assays. The use of oligonucleotide probes along with a signal amplification system (RNAscope(®)) can enhance detection of EAV RNA in FFPE tissues, with sensitivity comparable to that of IHC. Most importantly, these assays provide important tools with which to investigate the mechanisms of EAV pathogenesis.


Subject(s)
Arterivirus Infections/diagnosis , Equartevirus/isolation & purification , Fetus/virology , Horse Diseases/diagnosis , In Situ Hybridization/methods , Molecular Diagnostic Techniques/methods , Virology/methods , Animals , Equartevirus/genetics , Female , Horses , Immunohistochemistry , RNA, Viral/analysis , RNA, Viral/genetics , Sensitivity and Specificity
16.
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
17.
Virology ; 487: 68-74, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26499043

ABSTRACT

All RNA viruses encode an RNA-dependent RNA polymerase (RdRp), which in arteriviruses is expressed as the C-terminal domain of nonstructural protein 9 (nsp9). Previously, potent primer-dependent RdRp activity has been demonstrated for the homologous polymerase subunit (nsp12) of the distantly related coronaviruses. The only previous study focusing on the in vitro activity of nsp9 of an arterivirus (equine arteritis virus; EAV) reported weak de novo polymerase activity on homopolymeric RNA templates. However, this activity was not retained when Mn(2+) ions were omitted from the assay or when biologically relevant templates were supplied, which prompted us to revisit the biochemical properties of this polymerase. Based on the properties of active-site mutants, we conclude that the RNA-synthesizing activities observed in de novo and primer-dependent polymerase and terminal transferase assays cannot be attributed to recombinant EAV nsp9-RdRp. Our results illustrate the potential pitfalls of characterizing polymerases using highly sensitive biochemical assays.


Subject(s)
Equartevirus/enzymology , Equartevirus/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Nonstructural Proteins/genetics , Arterivirus Infections , Coronavirus/enzymology , Coronavirus/genetics , RNA, Viral/genetics
18.
Arch Virol ; 161(4): 821-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26711457

ABSTRACT

Strains of equine arteritis virus (EAV) differ in their virulence phenotypes, causing anywhere from subclinical infections to severe disease in horses. Here, we describe the in silico design and de novo synthesis of a full-length infectious cDNA clone of the horse-adapted virulent Bucyrus strain (VBS) of EAV encoding mCherry along with in vitro characterization of the progeny virions (EAV sVBSmCherry) in terms of host-cell tropism, replicative capacity and stability of the mCherry coding sequences following sequential passage in cell culture. The relative stability of the mCherry sequence during sequential cell culture passage coupled with a comparable host-cell range phenotype (equine endothelial cells, CD3(+) T cells and CD14(+) monocytes) to parental EAV VBS suggest that EAV-sVBSmCherry-derived virus could become a valuable research tool for identification of host-cell tropism determinants and for characterization of the viral proteins involved in virus attachment and entry into different subpopulations of peripheral blood mononuclear cells. Furthermore, this study demonstrates that advances in nucleic acid synthesis technology permit synthesis of complex viral genomes with overlapping genes like those of arteriviruses, thereby circumventing the need for complicated molecular cloning techniques. In summary, de novo nucleic acid synthesis technology facilitates innovative viral vector design without the tedium and risks posed by more-conventional laboratory techniques.


Subject(s)
DNA, Complementary/genetics , Equartevirus/genetics , Equartevirus/pathogenicity , Luminescent Proteins/metabolism , Animals , Antibodies, Monoclonal , Antigens, Viral , Cell Line , Cloning, Molecular , Cricetinae , Flow Cytometry , Gene Expression Regulation, Viral/physiology , Horses , Luminescent Proteins/genetics , Microscopy, Fluorescence , Rabbits , Virulence , Red Fluorescent Protein
19.
Viral Immunol ; 28(7): 391-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26331346

ABSTRACT

The nucleocapsid (N) protein is the most conserved structural protein in equine arteritis virus (EAV). This study aimed to identify the minimal conserved B cell epitope on the EAV N protein. The purified N protein was used to immunize mice for preparing monoclonal antibody (mAb). The reactivity of mAb was evaluated by Western blot and immunofluorescence assay. Moreover, 11 overlapping peptides (named MBP-N1 to MBP-N11) were designed to localize the linear antigenic epitope within the N protein. The peptides were identified by indirect enzyme-linked immunosorbent assay (ELISA) and Western blot. The minimal conserved B cell epitope on the EAV N protein was identified. The homology analysis was also performed. An EAV N-reactive mAb was selected and designated as 1C11. Indirect ELISA results showed that overlapping domain between MBP-N10 and MBP-N11 was recognized by the mAb 1C11. Furthermore, the indirect ELISA and Western blot showed that (101)QRKVAP(106) was the minimal linear epitope of the EAV N protein. The homology analysis showed that the identified epitope was conserved among all EAV strains analyzed in this work, with the exception of the ARVAC. One EAV N-specific mAb (1C11) was developed, and a minimal linear peptide epitope ((101)QRKVAP(106)) within the N protein was identified.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Equartevirus/immunology , Nucleocapsid Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Blotting, Western , Conserved Sequence , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte/genetics , Equartevirus/genetics , Female , Fluorescent Antibody Technique , Mice, Inbred BALB C , Nucleocapsid Proteins/genetics , Sequence Homology, Amino Acid
20.
Intervirology ; 58(4): 197-204, 2015.
Article in English | MEDLINE | ID: mdl-26279281

ABSTRACT

Outbreaks of hemorrhagic syndrome-like disease with high mortality rates have frequently occurred in Pelodiscus sinensis farms. The purpose of this study was to investigate the pathogen through challenge infection assays and partial sequencing of the genome of the pathogen. A 453-bp amplicon was obtained by random PCR using the nucleic acid extracted from the tissue homogenate filtrate and showed 32% identity at the amino acid level with the replicase polyprotein of the porcine reproductive and respiratory syndrome virus by Blastx. Multiple alignments indicated the putative protein sequence has some similarities to the replicase polyprotein of Arteriviridae, and the phylogenetic tree showed it was closely related to equine arteritis virus. This sequence was found in the lung of the diseased P. sinensis by in situ hybridization. Dot blot hybridization and quantitative RT-PCR showed that the lung had the highest content of virus. The peak replication of P. sinensis hemorrhagic syndrome virus (TSHSV) in the lung occurred 4 days after infection. The ribonucleic nature of the viral genome was confirmed by RNase A or DNase I treatments. We named the virus TSHSV in this study as P. sinensis is also known as Trionyx sinensis. These results provide a fundamental basis for further understanding the biology and the molecular mechanisms of TSHSV.


Subject(s)
RNA Virus Infections/veterinary , RNA Viruses/genetics , RNA Viruses/isolation & purification , RNA, Viral/genetics , Turtles/virology , Amino Acid Sequence , Animals , Animals, Domestic/virology , Base Sequence , Equartevirus/genetics , Genome, Viral , Hemorrhage/veterinary , Hemorrhage/virology , Lung/virology , RNA Virus Infections/virology , RNA Viruses/classification , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...