Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Med Microbiol ; 73(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38935078

ABSTRACT

Introduction. Avian reovirus (ARV) is associated with arthritis/tenosynovitis and malabsorption syndrome in chickens. The σC and σB proteins, both exposed to the virus capsid, are highly immunogenic and could form the basis for diagnostic devices designed to assess the immunological status of the flock.Gap Statement. Commercial ARV ELISAs cannot distinguish between vaccinated and infected animals and might not detect circulating ARV strains.Aim. We aimed to develop a customized test to detect the circulating field ARV strains as well as distinguish between vaccinated and unvaccinated animals.Methodology. We developed ELISA assays based on recombinant (r) σB, σC and the nonstructural protein σNS and tested them using antisera of vaccinated and unvaccinated chickens as well as negative controls. Fragments of σB and σC proteins were also used to study regions that could be further exploited in diagnostic tests.Results. Vaccinated and unvaccinated birds were positive by commercial ELISA, with no difference in optical density values. In contrast, samples of unvaccinated animals showed lower absorbance in the rσB and rσC ELISA tests and higher absorbance in the rσNS ELISA test than the vaccinated animals. Negative control samples were negative in all tests. Fragmentation of σB and σC proteins showed that some regions can differentiate between vaccinated and unvaccinated animals. For example, σB amino acids 128-179 (σB-F4) and σC amino acids 121-165 (σC-F4) exhibited 85 and 95% positivity among samples of vaccinated animals but only 5% and zero positivity among samples of unvaccinated animals, respectively.Conclusion. These data suggest that unvaccinated birds might have been exposed to field strains of ARV. The reduction in absorbance in the recombinant tests possibly reflects an increased specificity of our test since unvaccinated samples showed less cross-reactivity with the vaccine proteins immobilized on ELISAs. The discrepant results obtained with the protein fragment tests between vaccinated and unvaccinated animals are discussed in light of the diversity between ARV strains.


Subject(s)
Chickens , Enzyme-Linked Immunosorbent Assay , Orthoreovirus, Avian , Poultry Diseases , Recombinant Proteins , Reoviridae Infections , Animals , Orthoreovirus, Avian/immunology , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/isolation & purification , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/veterinary , Reoviridae Infections/veterinary , Reoviridae Infections/diagnosis , Poultry Diseases/virology , Poultry Diseases/diagnosis , Recombinant Proteins/immunology , Antibodies, Viral/blood , Capsid Proteins/immunology , Capsid Proteins/genetics , Viral Proteins/immunology , Viral Proteins/genetics
2.
Vet Microbiol ; 261: 109214, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34461358

ABSTRACT

Novel duck reovirus (NDRV) causes high morbidity in ducklings, and recovered ducklings are often remarkably stunted in growth. In this study, four NDRV strains were isolated from the NDRV outbreaks that occurred in different regions of Shandong province, China. The biological characteristics and pathogenicity of the four NDRV strains were elucidated, and the N20 was identified as a naturally attenuated strain. Three-day-old ducklings were immunized with live N20 strain (100 ELD50/duck), and challenged with 104.52 ELD50 of virulent N19 strain at 7 days post immunization. The vaccinated ducklings showed no evidence of clinical signs, gross and histopathological lesions, or loss of body weight, and 100 % protection against the virulent NDRV N19 infection. The NDRV-specific antibodies were generated in the immunized ducklings and could neutralize different NDRV strains. These results indicated that the N20 strain was a promising live attenuated vaccine candidate against highly pathogenic NDRV infection.


Subject(s)
Orthoreovirus, Avian/immunology , Poultry Diseases/prevention & control , Reoviridae Infections/veterinary , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/blood , China , Ducks , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/isolation & purification , Poultry Diseases/virology , Reoviridae Infections/prevention & control , Vaccines, Attenuated/immunology
3.
Trop Anim Health Prod ; 52(3): 1049-1054, 2020 May.
Article in English | MEDLINE | ID: mdl-31705354

ABSTRACT

Recently, the Egyptian broiler industry has experienced an increased incidence of avian reovirus (ARV) infections. However, to date, no studies have been carried out to investigate the epidemiologic status of ARV infections as well as the genetic characteristics of the currently circulating ARV strains. The present study estimates the seroprevalence of ARV infections in Alexandria, El-Behera, Giza, Kafr El-Sheikh, and Gharbia governorates, Egypt, during the period 2017-2018. A total of 150 serum samples from 15 unvaccinated broiler flocks with suspicious ARV infection were screened using a commercial enzyme-linked immunosorbent assay kit. All the tested flocks were found to be positive for ARV-specific antibodies, and the overall seropositivity rate was 80.6%. Meanwhile, 5 (33.3%) flocks were confirmed for the presence of ARV through a reverse transcription-polymerase chain reaction (RT-PCR) assay based on the σA-encoding gene. Phylogenetic analysis based on the nucleotide sequences of the σA-encoding gene revealed that the obtained ARV isolate, designated EGY1, was grouped in the S1113-like cluster of ARV and displayed 100% and 98.7% nucleotide identity with the Chinese MSO1 isolate and the S1133 vaccine strain, respectively. In addition, amino acid alignments with the S1133 vaccine strain revealed that the σA protein of the EGY1 isolate carried the substitutions G81S and A118V. In conclusion, the present study provides the evidence for a ubiquitous distribution of ARV infection in Egypt as well as represents a starting point for genetic characterization of the currently circulating ARV strains.


Subject(s)
Chickens , Orthoreovirus, Avian/genetics , Phylogeny , Poultry Diseases/epidemiology , Poultry Diseases/virology , Reoviridae Infections/veterinary , Animals , Antibodies, Viral/blood , Base Sequence , Egypt/epidemiology , Orthoreovirus, Avian/immunology , Reoviridae Infections/epidemiology , Reoviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Seroepidemiologic Studies
4.
Vaccine ; 38(4): 882-889, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31708178

ABSTRACT

African horse sickness virus (AHSV) is an insect-borne pathogen that causes acute disease in horses and other equids. In an effort to improve the safety of currently available vaccines and to acquire new knowledge about the determinants of AHSV immunogenicity, new generation vaccines are being developed. In this work we have generated and tested a novel immunization approach comprised of nonstructural protein 1 (NS1) of AHSV serotype 4 (AHSV-4) incorporated into avian reovirus muNS protein microspheres (MS-NS1) and/or expressed using recombinant modified vaccinia virus Ankara vector (MVA-NS1). The protection conferred against AHSV by a homologous MS-NS1 or heterologous MS-NS1 and MVA-NS1 prime/boost was evaluated in IFNAR (-/-) mice. Our results indicate that immunization based on MS-NS1 and MVA-NS1 afforded complete protection against the infection with homologous AHSV-4. Moreover, priming with MS-NS1 and boost vaccination with MVA-NS1 (MS-MVA-NS1) triggered NS1 specific cytotoxic CD8 + T cells and prevented AHSV disease in IFNAR (-/-) mice after challenge with heterologous serotype AHSV-9. Cross-protective immune responses are highly important since AHS can be caused by nine different serotypes, which means that a universal polyvalent vaccination would need to induce protective immunity against all serotypes.


Subject(s)
African Horse Sickness Virus/immunology , African Horse Sickness/prevention & control , Immunization , Viral Vaccines/administration & dosage , African Horse Sickness/immunology , Animals , Female , Horses , Immunity/immunology , Mice , Mice, Knockout , Microspheres , Orthoreovirus, Avian/immunology , Receptor, Interferon alpha-beta/genetics , Serogroup , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology
6.
Vet Microbiol ; 233: 85-92, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31176417

ABSTRACT

Muscovy duck reovirus (MDRV) causes serious immunodeficiency in the intestinal mucosa, although the underlying histopathological mechanisms remain unclear. Thus, we investigated the impact of MDRV infection on intestinal morphology using hematoxylin and eosin staining. Immune-related cells were also quantified by staining with hematoxylin and eosin, toluidine blue, and periodic acid-Schiff stain, or by immunohistochemistry and cytochemistry for lectin. Similarly, CD4+ and CD8+ cells were quantified by flow cytometry, and the expression of several immune-related molecules was quantified by radioimmunoassay. We found that MDRV clearly damaged the intestinal mucosa, based on tissue morphology, villus length, villus width, intestinal thickness, villus height/crypt depth ratio, and villus surface area. MDRV also altered the density or distribution of lymphocytes, mastocytes, and goblet cells in the small intestinal mucosa, as well as microfold cells in Peyer's patches. In addition, MDRV markedly depleted CD4+ cells from the intestinal mucosa and lowered the CD4+:CD8+ ratio in peripheral blood. Moreover, MDRV diminished the levels of secretory IgA and mucosal addressin cell adhesion molecule-1 (p < 0.01), but elevated those of histamine and nitric oxide (p < 0.01 or p < 0.05). Finally, MDRV significantly suppressed IL-1ß, IL-4, IL-5, and IL-8 levels (p < 0.01 or p < 0.05) mid-infection. Collectively, our data suggest that MDRV severely damages the structure and function of the intestinal mucosa by modulating immune cells and immune-related factors, thus leading to local immunodeficiency. Our findings lay the foundation for further research on the pathogenesis of MDRV.


Subject(s)
Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/virology , Orthoreovirus, Avian/immunology , Reoviridae Infections/immunology , Age Factors , Animals , CD4 Lymphocyte Count , Cytokines/immunology , Ducks/virology , Duodenum , Fibroblasts/virology , Histamine/analysis , Immunoglobulin A, Secretory/analysis , Intestine, Small/immunology , Nitric Oxide/analysis , Orthoreovirus, Avian/pathogenicity , Poultry Diseases/virology , Reoviridae Infections/pathology , Viral Load
7.
Vaccine ; 36(52): 8001-8007, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30420117

ABSTRACT

The Muscovy duck reovirus (MDRV) is a highly pathogenic virus that causes substantial economic losses in the Muscovy duck industry. While MDRV poses a significant threat to Muscovy ducklings, no vaccine candidates are available to date to alleviate MDRV infection throughout the world. The present study presents efforts toward establishing an attenuated vaccine for MDRV. For this purpose, a live attenuated vaccine strain named CA was obtained via alternate propagation of the MDRV isolate MW9710 in both Muscovy duck embryo fibroblasts (MDEFs) and chicken embryo fibroblasts (CEFs) for 90 passages. The CA strain achieved an adaptive growth capacity in CEFs with a viral titer that ranged between 105.0-105.5 TCID50/100 µL and lost its pathogenicity in 1-day-old Muscovy ducklings. Compared to the parent strain MW9710, the CA strain has 42 scattered amino acid substitutions, most of which are located in the λB, λC, µB, σB, and σC protein. The CA strain maintained its attenuation and showed no gene mutation or virulence reversion after back propagation into 1-day-old ducklings for five rounds. The minimum protective dose was calculated to be 300 TCID50 of the CA strain. Furthermore, a single dose of CA vaccine protected immunized ducklings against lethal challenge by the virulent MDRV strain MW9710 and significantly decreased viral loads. In summary, the CA strain exhibited striking genetic stability, excellent safety, and effective immunogenicity. This CA strain of MDRV is a promising vaccine candidate for the prevention and control of MDRV infection.


Subject(s)
Orthoreovirus, Avian/immunology , Poultry Diseases/prevention & control , Reoviridae Infections/veterinary , Viral Vaccines/immunology , Animals , Chickens/immunology , Ducks/immunology , Fibroblasts/virology , Immunogenicity, Vaccine , Orthoreovirus, Avian/genetics , Poultry Diseases/immunology , Reoviridae Infections/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Load
8.
J Virol Methods ; 257: 69-72, 2018 07.
Article in English | MEDLINE | ID: mdl-29660384

ABSTRACT

Avian reovirus (ARV) causes significant economic losses to the poultry industry worldwide. The ARV proteins fall into three different classes based on their sizes:λ (large); µ (medium) and σ (small). σB, an outer capsid protein of the ARV contains group specific neutralizing epitopes and induces strong immune response in naturally infected chickens. This study describes the development of a rapid dot-enzyme linked immunosorbent assay (dot-ELISA) using recombinant σB protein antigen of 54 kDa (approx). The assay is rapid (4-5 h) and results can be read by the naked eye. Sixteen ARV positive serum samples (group A) produced strong reaction in the dot-ELISA while twenty of the ARV negative serum samples (group B) collected from SPF chickens showed no reaction. Seventy six randomly collected serum samples were tested with a commercial indirect ELISA kit and the in-house developed dot-ELISA. A total of sixty eight serum samples were found to be positive by indirect ELISA and sixty five serum samples were found to be positive by dot-ELISA. Therefore, using the commercial ELISA as the reference test, the dot-ELISA had a diagnostic sensitivity of 83.8% and specificity of 88.6%. This dot-ELISA can be used as a simple, reliable and inexpensive alternative to commercial ELISA kits for serodiagnosis of ARV where the facilities for standard ELISA are not available.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Enzyme-Linked Immunosorbent Assay/methods , Orthoreovirus, Avian/isolation & purification , Poultry Diseases/diagnosis , Reoviridae Infections/veterinary , Animals , Chickens , Orthoreovirus, Avian/immunology , Poultry Diseases/virology , Reoviridae Infections/diagnosis , Sensitivity and Specificity
9.
Sci Rep ; 7(1): 3565, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28620186

ABSTRACT

In recent years, emerging strains of pathogenic arthrogenic avian reovirus (ARV) have become a challenge to the chicken industry across USA and Canada causing significant economic impact. In this study, we characterized emerging variant ARV strains and examined their genetic and antigenic relationship with reference strains. We isolated 37 emerging variant ARV strains from tendons of broiler chickens with clinical cases of arthritis/tenosynovitis at commercial farms in Saskatchewan, Canada. Viral characterization using immunocytochemistry, gold-immunolabeling and electron microscopy revealed distinct features characteristic of ARV. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analyses of the viral Sigma C gene revealed genetic heterogeneity between the field isolates. On phylogenetic analyses, the Sigma C amino acid sequences of the isolates were clustered into four distinct genotypic groups. These ARV field strains were genetically diverse and quite distant from the vaccine and vaccine related field strains. Antibodies produced against a commercial Reo 2177 ® vaccine did not neutralize these variants. Moreover, structure based analysis of the Sigma C protein revealed significant antigenic variability between the cluster groups and the vaccine strains. To the best of our knowledge, this is the first report on the genetic, phenotypic and antigenic characterization of emerging ARVs in Canada.


Subject(s)
Arthritis, Infectious/veterinary , Cattle Diseases/immunology , Cattle Diseases/virology , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/immunology , Reoviridae Infections/veterinary , Animals , Biopsy , Canada/epidemiology , Cattle , Cattle Diseases/diagnosis , Cattle Diseases/epidemiology , Models, Molecular , Phenotype , Phylogeny , Polymorphism, Restriction Fragment Length , Protein Conformation , Seroepidemiologic Studies , Symptom Assessment , Viral Proteins/chemistry , Viral Proteins/immunology
10.
Antiviral Res ; 142: 55-62, 2017 06.
Article in English | MEDLINE | ID: mdl-28322923

ABSTRACT

Bluetongue virus (BTV) is the causative agent of bluetongue disease (BT), which affects domestic and wild ruminants. At the present, 27 different serotypes have been documented. Vaccination has been demonstrated as one of the most effective methods to avoid viral dissemination. To overcome the drawbacks associated with the use of inactivated and attenuated vaccines we engineered a new recombinant BTV vaccine candidate based on proteins VP2, VP7, and NS1 of BTV-4 that were incorporated into avian reovirus muNS-Mi microspheres (MS-VP2/VP7/NS1) and recombinant modified vaccinia virus Ankara (rMVA). The combination of these two antigen delivery systems in a heterologous prime-boost vaccination strategy generated significant levels of neutralizing antibodies in IFNAR(-/-) mice. Furthermore, this immunization strategy increased the ratio of IgG2a/IgG1 in sera, indicating an induction of a Th1 response, and elicited a CD8 T cell response. Immunized mice were protected against lethal challenges with the homologous serotype 4 and the heterologous serotype 1 of BTV. All these results support the strategy based on microspheres in combination with rMVAs as a promising multiserotype vaccine candidate against BTV.


Subject(s)
Bluetongue virus/immunology , Bluetongue/prevention & control , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Microspheres , Receptor, Interferon alpha-beta/immunology , Vaccination , Vaccinia virus/immunology , Animals , Antibodies, Neutralizing , Bluetongue virus/classification , CD8-Positive T-Lymphocytes/metabolism , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Line , Chlorocebus aethiops , Immunization , Immunization, Secondary/methods , Immunoglobulin G/blood , Lysosomal-Associated Membrane Protein 1/metabolism , Male , Mice , Mice, 129 Strain , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/immunology , Serogroup , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology , Vero Cells , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology
11.
Vet Microbiol ; 206: 152-156, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28024855

ABSTRACT

Avian reoviruses are the causative agent of viral arthritis/tenosynovitis in chickens and turkeys. Clinical signs of disease include swelling of the hock joints accompanied by lesions in the gastrocnemius and digital flexor tendons causing lameness in addition to hydropericardium. The economic impact is significant as it results in poor weight gain, increased feed conversion ratios and condemnations at the processing plant. Vaccination with both live attenuated and inactivated oil emulsion vaccines have been used successfully for decades to control the disease. Current commercial vaccine strains belong to the same serotype and are antigenically and serologically distinct from circulating variant field viruses isolated from clinical cases of tenosynovitis. Since 2012, there has been a dramatic increase in the number of clinical cases of tenosynovitis in commercial poultry and commercial vaccines are unable to provide adequate levels of protection against disease. Producers have elected to use custom inactivated vaccines in the absence of any commercially available homologous vaccines. Identification and selection of field isolates for use in autogenous vaccines can be difficult especially when multiple reoviruses are co-circulating among flocks. In addition, field data suggests that in some cases the custom vaccines are providing adequate protection against disease but as new genetic variants emerge, new vaccines are needed.


Subject(s)
Chickens/immunology , Orthoreovirus, Avian/immunology , Poultry Diseases/prevention & control , Reoviridae Infections/veterinary , Turkeys/immunology , Animals , Arthritis, Infectious/epidemiology , Arthritis, Infectious/prevention & control , Arthritis, Infectious/veterinary , Arthritis, Infectious/virology , Chickens/virology , Immunity , Poultry , Poultry Diseases/epidemiology , Poultry Diseases/virology , Reoviridae Infections/epidemiology , Reoviridae Infections/prevention & control , Tenosynovitis/epidemiology , Tenosynovitis/prevention & control , Tenosynovitis/veterinary , Tenosynovitis/virology , Turkeys/virology , Vaccination/veterinary , Viral Vaccines/administration & dosage
12.
Sci Rep ; 6: 39092, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27974824

ABSTRACT

Novel duck reovirus (NDRV) disease emerged in China in 2011 and continues to cause high morbidity and about 5.0 to 50% mortality in ducklings. Currently there are no approved vaccines for the virus. This study aimed to assess the efficacy of a new vaccine created from the baculovirus and sigma C gene against NDRV. In this study, a recombinant baculovirus containing the sigma C gene was constructed, and the purified protein was used as a vaccine candidate in ducklings. The efficacy of sigma C vaccine was estimated according to humoral immune responses, cellular immune response and protection against NDRV challenge. The results showed that sigma C was highly expressed in Sf9 cells. Robust humoral and cellular immune responses were induced in all ducklings immunized with the recombinant sigma C protein. Moreover, 100% protection against lethal challenge with NDRV TH11 strain was observed. Summary, the recombinant sigma C protein could be utilized as a good candidate against NDRV infection.


Subject(s)
Orthoreovirus, Avian/immunology , Poultry Diseases/prevention & control , Reoviridae Infections/prevention & control , Viral Proteins/immunology , Animals , Ducks , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , Poultry Diseases/immunology , Recombinant Proteins/immunology , Reoviridae Infections/immunology , Sf9 Cells , Vaccines, Subunit/immunology , Viral Proteins/genetics , Viral Proteins/metabolism
13.
Virology ; 487: 104-11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517397

ABSTRACT

We have previously shown that the replication of avian reovirus (ARV) in chicken embryo fibroblasts (CEF) is more resistant to the antiviral action of interferon (IFN) than the replication of vesicular stomatitis virus (VSV) or vaccinia virus (VV). In this study we examined the capacity of these three viruses to induce the expression of IFN when infecting avian cells. Efficient expression of both type-α and type-ß IFNs, as well as of the double-stranded RNA (dsRNA)-activated protein kinase (PKR), takes place in ARV-infected CEF, but not in cells infected with VSV or VV. PKR expression is not directly induced by ARV infection, but by the IFN secreted by ARV-infected cells. IFN induction in ARV-infected cells requires viral uncoating, but not viral gene expression, a situation similar to that reported for apoptosis induction by ARV-infected cells. However, our results demonstrate that IFN induction by ARV-infected CEF occurs by a caspase-independent mechanism.


Subject(s)
Interferon-alpha/immunology , Interferon-beta/immunology , Orthoreovirus, Avian/immunology , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/immunology , eIF-2 Kinase/immunology , Animals , Apoptosis/immunology , Caspases/metabolism , Cell Proliferation , Cells, Cultured , Chick Embryo , Chickens , Cricetinae , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Orthoreovirus, Avian/growth & development , Primary Cell Culture , Vaccinia virus/growth & development , Vesicular stomatitis Indiana virus/growth & development , Virus Replication , eIF-2 Kinase/biosynthesis
14.
J Virol Methods ; 220: 55-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25907470

ABSTRACT

A novel duck reovirus (N-DRV) disease emerged in China in 2000 and it has become an epidemic genotype. A test for detection of virus-specific antibodies in serum samples would be useful for epidemiological investigations. Currently, Currently, serological assays for N-DRV diagnosis are not available. A test for detection of virus-specific antibodies in serum samples would be useful for epidemiological investigations. In this study, a highly sensitive and specific indirect enzyme-linked immunosorbent assay (ELISA) for the detection of antibodies to N-DRV was developed. The outer capsid (σC) of N-DRV was cloned and expressed in Escherichia coli as a coating antigen. The antigen concentration and serum dilution were optimized using a checkerboard titration. Furthermore, the specificity of σC-ELISA assay was confirmed by cross checking with other duck viral pathogens. In comparison with the western blot, the sensitivity and specificity of the σC-ELISA was 92.6% and 88.9%, respectively, and agreement of two tests was excellent with κ value of 0.786 (p < 0.05). A serological survey was performed using the assay on serum samples from different age and species of duck flocks in the Zhejiang and Jiangsu Province, China. The seropositive rate of the 1209 serum samples was 57.7%. In conclusion, the developed σC-ELISA assay is a very specific and sensitive test that will be useful for large-scale serological survey in N-DRV infection and monitoring antibodies titers against N-DRV.


Subject(s)
Antibodies, Viral/blood , Bird Diseases/diagnosis , Ducks/virology , Enzyme-Linked Immunosorbent Assay/methods , Orthoreovirus, Avian/immunology , Reoviridae Infections/veterinary , Veterinary Medicine/methods , Animals , Antigens, Viral/immunology , Bird Diseases/virology , Capsid Proteins/immunology , China/epidemiology , Reoviridae Infections/diagnosis , Reoviridae Infections/virology , Sensitivity and Specificity , Seroepidemiologic Studies
15.
Antiviral Res ; 110: 42-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25057758

ABSTRACT

Vaccination is critical for controlling the spread of bluetongue virus (BTV). The inactivated BTV vaccines that are now being used in Europe are effective in preventing outbreaks of BTV but secondary effects associated to repetitive inoculation of aluminum-containing adjuvants and the need to develop safer, cross-reactive, and more efficacious vaccines with differential diagnostic capability have re-stimulated the interest in developing improved vaccination strategies against BTV. We have engineered a subunit BTV vaccine candidate based on proteins VP2, VP7, and NS1 of BTV-4 incorporated into avian reovirus (ARV) muNS-Mi microspheres (MS-VP2/MS-VP7/MS-NS1). IFNAR(-/-) mice immunized with MS-VP2/MS-VP7/MS-NS1 without adjuvant generated significant levels of neutralizing antibodies specific to BTV-4. In addition, vaccination stimulated specific T cell responses, predominantly CD4+, against the virus. Immunized mice were fully protected against a homologous challenge with a lethal dose of BTV-4 and partially cross-protected against a heterologous challenge with a lethal dose of BTV-1. These results support MS-VP2/MS-VP7/MS-NS1 as a promising subunit vaccine candidate against multiple serotypes of BTV as well as the use of microspheres as an alternative delivery method with potent intrinsic adjuvant activity.


Subject(s)
Bluetongue virus/immunology , Capsid Proteins/immunology , Orthoreovirus, Avian/immunology , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Baculoviridae/genetics , Bluetongue/immunology , Bluetongue/prevention & control , CD4-Positive T-Lymphocytes/immunology , Capsid Proteins/genetics , Cell Line , Chick Embryo , Chlorocebus aethiops , Cricetinae , Drug Carriers/therapeutic use , Male , Mice , Mice, Knockout , Microspheres , Orthoreovirus, Avian/genetics , Sf9 Cells , Sheep/virology , Spodoptera , Vaccination , Vaccines, Subunit/immunology , Vero Cells , Viral Nonstructural Proteins/genetics
16.
Avian Pathol ; 43(4): 371-8, 2014.
Article in English | MEDLINE | ID: mdl-24980420

ABSTRACT

Turkey arthritis reovirus (TARV) has been isolated from the gastrocnemius tendons and tibiotarsal joint fluid of lame male turkeys >12 weeks old in the Midwest. Two experiments were conducted to compare the pathogenicity in turkeys of three TARVs (TARV-MN2, TARV-MN4 and TARV-O'Neil), one turkey enteric reovirus (TERV strain MN1) and one chicken arthritis reovirus (CARV strain MN1). Two hundred microlitres of virus were inoculated by the oral, intratracheal, or footpad route into 6-day-old poults placed in isolator units. Poults were necropsied at 1 and 4 weeks post infection in Experiment 1, and at 2 and 4 weeks post infection in Experiment 2. Reovirus was detected by reverse transcription-polymerase chain reaction and virus isolation in tendons of TARV-inoculated poults at 1, 2 and 4 weeks post infection. TARV-O'Neil and TARV-MN2 were detected in tendons of sentinal birds at 1 and 4 weeks and 1 week p.i., respectively. In general, TARVs produced lymphocytic tenosynovitis of the gastrocnemius and digital flexor tendon sheaths without inflammation of the tendons proper. In Experiment 1, poults inoculated with TARV-MN2 and TARV-O'Neil had significantly higher gastrocnemius tendon inflammation scores, as determined by histology, than those inoculated with TERV-MN1 or CARV-MN1. In Experiment 2, poults inoculated with TARV-MN2 and TARV-O'Neil had significantly higher gastrocnemius tendon inflammation scores than those inoculated with TARV-MN4 and virus-free medium (negative control group). Koch's postulates was fulfilled when TARV-MN2 and TARV-O'Neil were re-isolated from tendons of poults that had originally been challenged with either of these viruses. Results of these experiments indicate that TARVs have a unique ability to induce gastrocnemius tenosynovitis in turkeys and that administration of TARV-O'Neil through the oral or intratracheal route is a reproducible model to study pathogenesis of TARV infection.


Subject(s)
Antibodies, Viral/blood , Chickens , Orthoreovirus, Avian/pathogenicity , Poultry Diseases/pathology , Reoviridae Infections/veterinary , Turkeys , Animals , Arthritis, Experimental/mortality , Arthritis, Experimental/pathology , Arthritis, Experimental/veterinary , Arthritis, Experimental/virology , Disease Models, Animal , Joints/pathology , Male , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/immunology , Orthoreovirus, Avian/isolation & purification , Poultry Diseases/mortality , Poultry Diseases/virology , RNA, Viral/genetics , Reoviridae Infections/mortality , Reoviridae Infections/pathology , Reoviridae Infections/virology , Tendons/pathology , Tenosynovitis/mortality , Tenosynovitis/pathology , Tenosynovitis/veterinary , Tenosynovitis/virology
17.
Acta Virol ; 58(2): 108-13, 2014.
Article in English | MEDLINE | ID: mdl-24957714

ABSTRACT

Avian reovirus (ARV) is an important agent of several diseases causing considerable losses in poultry farming. An outer capsid protein (σC) of ARV, is known as a virus-cell attachment protein essential for virus infectivity. In this study, the σC gene of ARV was cloned and expressed in Escherichia coli. The expressed recombinant protein was used as immunogen for raising a specific IgY antibody in laying hens. At 14 weeks post immunization, the antibody titers in serum and egg yolk reached 302,000 and 355,000, respectively. The IgY antibody was capable to neutralize ARV in BHK-21 cells and it strongly reacted in ELISA with ARV but not with heterologous viruses. The IgY antibody detected ARV in field samples of infected animal tissues in dot blot assay. These results suggest that an efficient, economic and rapid diagnostics of ARV can be performed routinely using the IgY antibody against a recombinant ARV σC protein.


Subject(s)
Antibodies, Viral , Immunoglobulins , Orthoreovirus, Avian/immunology , Poultry Diseases/diagnosis , Reoviridae Infections/veterinary , Viral Proteins/immunology , Animals , Antibodies, Viral/immunology , Chickens , Immunoblotting/instrumentation , Immunoblotting/methods , Immunoglobulins/immunology , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/isolation & purification , Poultry Diseases/virology , Reoviridae Infections/diagnosis , Reoviridae Infections/virology , Viral Proteins/genetics
18.
PLoS One ; 8(11): e81533, 2013.
Article in English | MEDLINE | ID: mdl-24312314

ABSTRACT

BACKGROUND: Avian reovirus (ARV) causes arthritis, tenosynovitis, runting-stunting syndrome (RSS), malabsorption syndrome (MAS) and immunosuppression in chickens. σB is one of the major structural proteins of ARV, which is able to induce group-specific antibodies against the virus. METHODS AND RESULTS: The present study described the identification of two linear B-cell epitopes in ARV σB through expressing a set of partially overlapping and consecutive truncated peptides spanning σB screened with two monoclonal antibodies (mAbs) 1F4 and 1H3-1.The data indicated that (21)KTPACW(26) (epitope A) and (32)WDTVTFH(38) (epitope B) were minimal determinants of the linear B cell epitopes. Antibodies present in the serum of ARV-positive chickens recognized the minimal linear epitopes in Western blot analyses. By sequence alignment analysis, we determined that the epitopes A and B were not conserved among ARV, duck reovirus (DRV) and turkey reovirus (TRV) strains. Western blot assays, confirmed that epitopes A and B were ARV-specific epitopes, and they could not react with the corresponding peptides of DRV and TRV. CONCLUSIONS AND SIGNIFICANCE: We identified (21)KTPACW(26) and (32)WDTVTFH(38) as σB -specific epitopes recognized by mAbs 1F4 and 1H3-1, respectively. The results in this study may have potential applications in development of diagnostic techniques and epitope-based marker vaccines against ARV groups.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes, B-Lymphocyte/immunology , Orthoreovirus, Avian/immunology , Amino Acid Sequence , Animals , Cross Reactions , Epitopes, B-Lymphocyte/chemistry , Female , Mice
19.
Virus Res ; 178(2): 530-4, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24076298

ABSTRACT

SigmaC (σC) protein, which mediates virus attachment to target cells, is the most variable proteins of avian reovirus (ARV). It is responsible for inducing protective antibody immune responses in animals. To understand the antigenic determinants of σC protein, a set of partially overlapping and consecutive peptides spanning σC were expressed and then screened with the monoclonal antibody (mAb) 2B5 directed against σC. The mAb 2B5 recognized peptides with the σC motif (45)ELLHRSISDISTTV(58). Further identification of the displayed B-cell epitope was conducted with a set of truncated peptides expressed as GST fusion proteins. The Western blot and ELISA results indicated that (45)ELLHRSISDI(54) was the minimal determinant of the linear B-cell epitope. Using sequences analysis, we found that this epitope was not a common motif shared among the other members of the ARV and DRV groups. Furthermore, cross reactivity analysis showed that the associated coding motif of other ARV and DRV groups was not recognized by 2B5. These data suggested that (45)ELLHRSISDI(54) was a type-specific linear B-cell epitope of avian reovirus. The results in this study may have potential applications in the development of diagnostic techniques and epitope-based marker vaccines against ARV, which is prevalent in China.


Subject(s)
Antigens, Viral/immunology , Epitopes, B-Lymphocyte/immunology , Orthoreovirus, Avian/immunology , Viral Structural Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Blotting, Western , China , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Female , Mice , Mice, Inbred BALB C
20.
Vet Immunol Immunopathol ; 147(3-4): 154-60, 2012 Jun 30.
Article in English | MEDLINE | ID: mdl-22575372

ABSTRACT

Avian reovirus (ARV) is an important pathogen in poultry industry and causes great economic losses. As attenuated Salmonella typhimurium is already being used as an effective vehicle for the transfer of DNA vaccines, so in this study we evaluated two DNA vaccines mediated by S. typhimurium on their ability of eliciting antibody production. SPF chickens were respectively immunized with SL7207 (pVAX-σB), SL7207 (pVAX-σC) and SL7027 (pVAX-σB-σC) three times. The results showed that the antibody production was highly dependent on the immunizing times, detectable antibodies of serum antibody IgG and small intestinal mucosal antibody IgA were generated at week 4 and were further improved at week 6 and antibody titers in group SL7207 (pVAX-σC) were higher than that in group SL7207 (pVAX-σB), demonstrating that SL7207 (pVAX-σC) was more powerful than SL7207 (pVAX-σB) in antibody production. The higher antibody titer in SL7027 (pVAX-σB-σC) than that in SL7207 (pVAX-σC) group showed that co-expressing σB and σC could improve antibody production. IFN-γ detection showed that significant higher IFN-γ was generated both in groups SL7027 (pVAX-σB-σC) and SL7207 (pVAX-σC). Subsequent challenge showed that SL7207 (pVAX-σB), SL7207 (pVAX-σC) and SL7027 (pVAX-σB-σC) conferred 50%, 75% and 87.5% respectively.


Subject(s)
Bird Diseases/prevention & control , Chickens/immunology , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/immunology , Reoviridae Infections/veterinary , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Base Sequence , Bird Diseases/immunology , Chickens/virology , DNA, Viral/genetics , Immunity, Mucosal , Interferon-gamma/biosynthesis , Intestinal Mucosa/immunology , Orthoreovirus, Avian/pathogenicity , Reoviridae Infections/immunology , Reoviridae Infections/prevention & control , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Vaccination/veterinary , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/genetics , Viral Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...