Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Viruses ; 13(11)2021 10 24.
Article in English | MEDLINE | ID: mdl-34834949

ABSTRACT

(1) Background: Equine arteritis virus (EAV) infection causes reproductive losses and systemic vasculitis in susceptible equidae. The intact male becomes the virus' reservoir upon EAV infection, as it causes a chronic-persistent infection of the accessory sex glands. Infected semen is the main source of virus transmission. (2) Here, we describe acute EAV infection and spread in a stallion population after introduction of new members to the group. (3) Conclusions: acute clinical signs, acute phase detection of antigen via (PCR) nasal swabs or (EDTA) blood, and seroconversion support the idea of transmission via seminal fluids into the respiratory tract(s) of others. This outbreak highlights EAV's horizontal transmission via the respiratory tract. This route should be considered in a chronic-persistently infected herd, when seronegative animals are added to the group.


Subject(s)
Arterivirus Infections/epidemiology , Arterivirus Infections/veterinary , Disease Outbreaks , Equartevirus , Horse Diseases/epidemiology , Animals , Arterivirus Infections/transmission , Arterivirus Infections/virology , Disease Transmission, Infectious , Horse Diseases/virology , Horses , Male , Masturbation , Persistent Infection , Respiratory System/virology , Semen/virology
2.
Int J Mol Sci ; 22(16)2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34445732

ABSTRACT

Infection with viruses, such as the lactate dehydrogenase-elevating virus (LDV), is known to trigger the onset of autoimmune anemia through the enhancement of the phagocytosis of autoantibody-opsonized erythrocytes by activated macrophages. Type I interferon receptor-deficient mice show enhanced anemia, which suggests a protective effect of these cytokines, partly through the control of type II interferon production. The development of anemia requires the expression of Fcγ receptors (FcγR) I, III, and IV. Whereas LDV infection decreases FcγR III expression, it enhances FcγR I and IV expression in wild-type animals. The LDV-associated increase in the expression of FcγR I and IV is largely reduced in type I interferon receptor-deficient mice, through both type II interferon-dependent and -independent mechanisms. Thus, the regulation of the expression of FcγR I and IV, but not III, by interferons may partly explain the exacerbating effect of LDV infection on anemia that results from the enhanced phagocytosis of IgG autoantibody-opsonized erythrocytes.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Arterivirus Infections/immunology , Interferons/metabolism , Lactate dehydrogenase-elevating virus/immunology , Receptors, IgG/metabolism , Anemia, Hemolytic, Autoimmune/virology , Animals , Arterivirus Infections/virology , Host-Pathogen Interactions , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis
3.
PLoS One ; 15(8): e0237091, 2020.
Article in English | MEDLINE | ID: mdl-32750064

ABSTRACT

Wobbly possum disease virus (WPDV) is an arterivirus that was originally identified in common brushtail possums (Trichosurus vulpecula) in New Zealand, where it causes severe neurological disease. In this study, serum samples (n = 188) from Australian common brushtail, mountain brushtail (Trichosurus cunninghami) and common ringtail (Pseudocheirus peregrinus) possums were tested for antibodies to WPDV using ELISA. Antibodies to WPDV were detected in possums from all three species that were sampled in the states of Victoria and South Australia. Overall, 16% (30/188; 95% CI 11.0-22.0) of possums were seropositive for WPDV and 11.7% (22/188; 95% CI 7.5-17.2) were equivocal. The frequency of WPDV antibody detection was the highest in possums from the two brushtail species. This is the first reported serological evidence of infection with WPDV, or an antigenically similar virus, in Australian possums, and the first study to find antibodies in species other than common brushtail possums. Attempts to detect viral RNA in spleens by PCR were unsuccessful. Further research is needed to characterise the virus in Australian possums and to determine its impact on the ecology of Australian marsupials.


Subject(s)
Arterivirus Infections/epidemiology , Arterivirus/pathogenicity , Trichosurus/virology , Animals , Antibodies, Viral/blood , Arterivirus/immunology , Arterivirus Infections/blood , Arterivirus Infections/virology , Australia , Serologic Tests , Trichosurus/immunology
4.
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
5.
J Am Assoc Lab Anim Sci ; 59(3): 328-333, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32079556

ABSTRACT

Lactate dehydrogenase elevating virus (LDV) continues to be one of the most common contaminants of cells and cell byproducts. As such, many institutions require that tumor cell lines, blood products, and products derived or passaged in rodent tissues are free of LDV as well as other pathogens that are on institutional exclusion lists prior to their use in rodents. LDV is difficult to detect by using a live-animal sentinel health monitoring program because the virus does not reliably pass to sentinel animals. After switching to an exhaust air dust health monitoring system, our animal resources center was able to detect a presumably long-standing LDV infection in a mouse colony. This health monitoring system uses IVC rack exhaust air dust collection media in conjunction with PCR analysis. Ultimately, the source of the contamination was identified as multiple LDV-positive patient-derived xenografts and multiple LDV-positive breeding animals. This case study is the first to demonstrate the use of environmental PCR testing as a method for detecting LDV infection in a mouse vivarium.


Subject(s)
Arterivirus Infections/veterinary , Environmental Microbiology , Housing, Animal , Lactate dehydrogenase-elevating virus/isolation & purification , Mice , Rodent Diseases/virology , Animals , Arterivirus Infections/virology , Cell Line, Tumor/virology , Dust , Heterografts , Humans , Polymerase Chain Reaction , Tumor Cells, Cultured/virology
6.
Arch Virol ; 164(10): 2593-2597, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31270606

ABSTRACT

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


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/classification , Arterivirus/isolation & purification , Phylogeny , Turtles/virology , Animals , Arterivirus/genetics , Arterivirus Infections/virology , China , Genome, Viral , RNA, Messenger , Sequence Analysis, DNA , Viral Proteins/genetics
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.
Virol Sin ; 33(4): 335-344, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30069823

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) causes substantial economic losses to the global pig industry. Alternative polyadenylation (APA) is a mechanism that diversifies gene expression, which is important for tumorigenesis, development, and cell differentiation. However, it is unclear whether APA plays a role in the course of PRRSV infection. To address this issue, in this study we carried out a whole-genome transcriptome analysis of PRRSV-infected Marc-145 African green monkey kidney cells and identified 185 APA switching genes and 393 differentially expressed genes (DEGs). Most of these genes were involved in cellular process, metabolism, and biological regulation, and there was some overlap between the two gene sets. DEGs were found to be more directly involved in the antiviral response than APA genes. These findings provide insight into the dynamics of host gene regulation during PRRSV infection and a basis for elucidating the pathogenesis of PRRSV.


Subject(s)
3' Untranslated Regions/genetics , Arterivirus Infections/genetics , Porcine respiratory and reproductive syndrome virus/physiology , Transcriptome/genetics , Animals , Arterivirus Infections/virology , Cell Line , Chlorocebus aethiops , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions , Porcine respiratory and reproductive syndrome virus/genetics , Reproducibility of Results , Virus Replication
9.
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
10.
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
11.
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
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.
Clin Vaccine Immunol ; 24(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28814389

ABSTRACT

Equine arteritis virus (EAV) has the ability to establish persistent infection in the reproductive tract of the stallion (carrier) and is continuously shed in its semen. We have recently demonstrated that EAV persists within stromal cells and a subset of lymphocytes in the stallion accessory sex glands in the presence of a significant local inflammatory response. In the present study, we demonstrated that EAV elicits a mucosal antibody response in the reproductive tract during persistent infection with homing of plasma cells into accessory sex glands. The EAV-specific immunoglobulin isotypes in seminal plasma included IgA, IgG1, IgG3/5, and IgG4/7. Interestingly, seminal plasma IgG1 and IgG4/7 possessed virus-neutralizing activity, while seminal plasma IgA and IgG3/5 did not. However, virus-neutralizing IgG1 and IgG4/7 in seminal plasma were not effective in preventing viral infectivity. In addition, the serological response was primarily mediated by virus-specific IgM and IgG1, while virus-specific serum IgA, IgG3/5, IgG4/7, and IgG6 isotype responses were not detected. This is the first report characterizing the immunoglobulin isotypes in equine serum and seminal plasma in response to EAV infection. The findings presented herein suggest that while a broader immunoglobulin isotype diversity is elicited in seminal plasma, EAV has the ability to persist in the reproductive tract, in spite of local mucosal antibody and inflammatory responses. This study provides further evidence that EAV employs complex immune evasion mechanisms during persistence in the reproductive tract that warrant further investigation.


Subject(s)
Antibodies, Viral/analysis , Arterivirus Infections/veterinary , Equartevirus/immunology , Horse Diseases/immunology , Immunity, Mucosal , Reproductive Tract Infections/veterinary , Semen/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Arterivirus Infections/immunology , Arterivirus Infections/virology , Horse Diseases/virology , Horses , Immune Evasion , Immunity, Humoral , Immunoglobulin G/analysis , Immunoglobulin G/immunology , Immunoglobulin M/analysis , Immunoglobulin M/immunology , Male , Reproductive Tract Infections/immunology , Reproductive Tract Infections/virology , Viremia
14.
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
15.
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
16.
J Virol ; 90(15): 6724-6737, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27170760

ABSTRACT

UNLABELLED: Nonhuman primates (NHPs) are a historically important source of zoonotic viruses and are a gold-standard model for research on many human pathogens. However, with the exception of simian immunodeficiency virus (SIV) (family Retroviridae), the blood-borne viruses harbored by these animals in the wild remain incompletely characterized. Here, we report the discovery and characterization of two novel simian pegiviruses (family Flaviviridae) and two novel simian arteriviruses (family Arteriviridae) in wild African green monkeys from Zambia (malbroucks [Chlorocebus cynosuros]) and South Africa (vervet monkeys [Chlorocebus pygerythrus]). We examine several aspects of infection, including viral load, genetic diversity, evolution, and geographic distribution, as well as host factors such as age, sex, and plasma cytokines. In combination with previous efforts to characterize blood-borne RNA viruses in wild primates across sub-Saharan Africa, these discoveries demonstrate that in addition to SIV, simian pegiviruses and simian arteriviruses are widespread and prevalent among many African cercopithecoid (i.e., Old World) monkeys. IMPORTANCE: Primates are an important source of viruses that infect humans and serve as an important laboratory model of human virus infection. Here, we discover two new viruses in African green monkeys from Zambia and South Africa. In combination with previous virus discovery efforts, this finding suggests that these virus types are widespread among African monkeys. Our analysis suggests that one of these virus types, the simian arteriviruses, may have the potential to jump between different primate species and cause disease. In contrast, the other virus type, the pegiviruses, are thought to reduce the disease caused by human immunodeficiency virus (HIV) in humans. However, we did not observe a similar protective effect in SIV-infected African monkeys coinfected with pegiviruses, possibly because SIV causes little to no disease in these hosts.


Subject(s)
Arterivirus Infections/epidemiology , Biological Evolution , Flaviviridae Infections/epidemiology , Genetic Variation , Lentivirus Infections/epidemiology , Viral Load , Africa/epidemiology , Animals , Animals, Wild , Arterivirus/genetics , Arterivirus/pathogenicity , Arterivirus Infections/genetics , Arterivirus Infections/virology , Flaviviridae/genetics , Flaviviridae/pathogenicity , Flaviviridae Infections/genetics , Flaviviridae Infections/virology , Genome, Viral , Haplorhini , Humans , Lentivirus/genetics , Lentivirus/pathogenicity , Lentivirus Infections/genetics , Lentivirus Infections/virology , Phylogeny , Prevalence
17.
J Virol Methods ; 234: 7-15, 2016 08.
Article in English | MEDLINE | ID: mdl-27036504

ABSTRACT

Equine arteritis virus (EAV) is the causative agent of equine viral arteritis (EVA), a respiratory and reproductive disease of horses. Most importantly, EAV induces abortion in pregnant mares and can establish persistent infection in up to 10-70% of the infected stallions, which will continue to shed the virus in their semen. The objective of this study was to develop and evaluate a reverse transcription insulated isothermal polymerase chain reaction (RT-iiPCR) for the detection of EAV in semen and tissue samples. The newly developed assay had a limit of detection of 10 RNA copies and a 10-fold higher sensitivity than a previously described real-time RT-PCR (RT-qPCR). Evaluation of 125 semen samples revealed a sensitivity and specificity of 98.46% and 100.00%, respectively for the RT-qPCR assay, and 100.00% and 98.33%, respectively for the RT-iiPCR assay. Both assays had the same accuracy (99.2%, k=0.98) compared to virus isolation. Corresponding values derived from testing various tissue samples (n=122) collected from aborted fetuses, foals, and EAV carrier stallions are as follows: relative sensitivity, specificity, and accuracy of 88.14%, 96.83%, and 92.62% (k=0.85), respectively for the RT-qPCR assay, and 98.31%, 92.06%, and 95.08% (k=0.90), respectively for the RT-iiPCR assay. These results indicate that RT-iiPCR is a sensitive, specific, and a robust test enabling detection of EAV in semen and tissue samples with very considerable accuracy. Even though the RT-qPCR assay showed a sensitivity and specificity equal to virus isolation for semen samples, its diagnostic performance was somewhat limited for tissue samples. Thus, this new RT-iiPCR could be considered as an alternative tool in the implementation of EAV control and prevention strategies.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus/isolation & purification , Horse Diseases/diagnosis , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Semen/virology , Animals , Arterivirus Infections/diagnosis , Arterivirus Infections/prevention & control , Arterivirus Infections/virology , Female , Horse Diseases/virology , Horses , Male , Open Reading Frames , Pregnancy , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Temperature
18.
Virus Res ; 220: 70-90, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27071852

ABSTRACT

All eukaryotic positive-stranded RNA (+RNA) viruses appropriate host cell membranes and transform them into replication organelles, specialized micro-environments that are thought to support viral RNA synthesis. Arteriviruses (order Nidovirales) belong to the subset of +RNA viruses that induce double-membrane vesicles (DMVs), similar to the structures induced by e.g. coronaviruses, picornaviruses and hepatitis C virus. In the last years, electron tomography has revealed substantial differences between the structures induced by these different virus groups. Arterivirus-induced DMVs appear to be closed compartments that are continuous with endoplasmic reticulum membranes, thus forming an extensive reticulovesicular network (RVN) of intriguing complexity. This RVN is remarkably similar to that described for the distantly related coronaviruses (also order Nidovirales) and sets them apart from other DMV-inducing viruses analysed to date. We review here the current knowledge and open questions on arterivirus replication organelles and discuss them in the light of the latest studies on other DMV-inducing viruses, particularly coronaviruses. Using the equine arteritis virus (EAV) model system and electron tomography, we present new data regarding the biogenesis of arterivirus-induced DMVs and uncover numerous putative intermediates in DMV formation. We generated cell lines that can be induced to express specific EAV replicase proteins and showed that DMVs induced by the transmembrane proteins nsp2 and nsp3 form an RVN and are comparable in topology and architecture to those formed during viral infection. Co-expression of the third EAV transmembrane protein (nsp5), expressed as part of a self-cleaving polypeptide that mimics viral polyprotein processing in infected cells, led to the formation of DMVs whose size was more homogenous and closer to what is observed upon EAV infection, suggesting a regulatory role for nsp5 in modulating membrane curvature and DMV formation.


Subject(s)
Arterivirus/ultrastructure , Cell Membrane/ultrastructure , Endoplasmic Reticulum/ultrastructure , Organelles/ultrastructure , Organelles/virology , Viral Nonstructural Proteins/genetics , Animals , Arterivirus/genetics , Arterivirus/metabolism , Arterivirus Infections/veterinary , Arterivirus Infections/virology , Cell Line , Cell Membrane/virology , Coronavirus/genetics , Coronavirus/metabolism , Coronavirus/ultrastructure , Electron Microscope Tomography , Endoplasmic Reticulum/virology , Gene Expression , Host-Pathogen Interactions , Viral Nonstructural Proteins/metabolism
19.
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
20.
PLoS One ; 11(3): e0151313, 2016.
Article in English | MEDLINE | ID: mdl-26963736

ABSTRACT

Simian hemorrhagic fever (SHF) is an often lethal disease of Asian macaques. Simian hemorrhagic fever virus (SHFV) is one of at least three distinct simian arteriviruses that can cause SHF, but pathogenesis studies using modern methods have been scarce. Even seemingly straightforward studies, such as examining viral tissue and cell tropism in vivo, have been difficult to conduct due to the absence of standardized SHFV-specific reagents. Here we report the establishment of an in situ hybridization assay for the detection of SHFV and distantly related Kibale red colobus virus 1 (KRCV-1) RNA in cell culture. In addition, we detected SHFV RNA in formalin-fixed, paraffin-embedded tissues from an infected rhesus monkey (Macaca mulatta). The assay is easily performed and can clearly distinguish between SHFV and KRCV-1. Thus, if further developed, this assay may be useful during future studies evaluating the mechanisms by which a simian arterivirus with a restricted cell tropism can cause a lethal nonhuman primate disease similar in clinical presentation to human viral hemorrhagic fevers.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/genetics , Arterivirus/isolation & purification , Macaca mulatta/virology , RNA, Viral/genetics , Animals , Arterivirus Infections/pathology , Arterivirus Infections/virology , Humans , In Situ Hybridization , RNA, Viral/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...