1,299
Views
3
CrossRef citations to date
0
Altmetric
ORIGINAL ARTICLES

An in vivo experimental model to determine antigenic variations among infectious bursal disease viruses

, , , , &
Pages 309-315 | Received 08 Oct 2012, Accepted 06 Mar 2013, Published online: 13 May 2013

Abstract

Infectious bursal disease virus (IBDV) is a double-stranded RNA virus causing infectious bursal disease in chickens. IBDV undergoes antigenic drift, so characterizing the antigenicity of IBDV plays an important role for identification and selection of vaccine candidates. In this study, an in vivo experimental model was developed to differentiate a new antigenic variant of IBDV. To this end, a hyper-immune serum to IBDV E/Del-type virus was generated in specific pathogen-free chickens and a standard volume of the hyper-immune serum was serially diluted and injected in specific pathogen-free birds via intravenous, subcutaneous, or intramuscular routes. The chickens were bled at different time points in order to evaluate the dynamics of virus neutralization titres. Based on the results, chickens were injected with different serum dilutions by the subcutaneous route. Twenty-four hours later, chickens were bled and then challenged with 100 median chicken infectious doses of the E/Del virus and a new IBDV variant. Chickens were euthanized at 7 days post infection and the bursa of Fabricius was removed for microscopic evaluation to determine the bursal lesion score. The determined virus neutralization titre along with the bursal lesion score was used to determine the breakthrough titre in the in vivo chicken model. Based on the data obtained, an antigenic subtype of IBDV was identified and determined to be different from E/Del. This model is a sensitive model for determination of IBDV antigenicity of non-tissue culture adapted IBDV.

Introduction

Infectious bursal disease (IBD) is an immunosuppressive disease of young chickens and is a threat to the poultry industry worldwide (Allan et al., Citation1972; Faragher et al., Citation1972; Hirai et al., Citation1974d; Rosenberger & Gelb, Citation1976; Giambrone et al., Citation1977; Saif, Citation1991). The causative agent, infectious bursal disease virus (IBDV), affects the bursa of Fabricius (BF) due to a lytic infection of proliferating lymphocytes of the B-cell lineage (Ivanyi & Morris, Citation1976; Hirai & Calnek, Citation1979; Käufer & Weiss, Citation1980; Müller, Citation1986). The disease was first described by Cosgrove (Citation1962) as avian nephrosis and was later recognized as IBD due to the pathologic/histologic changes in the BF (Hitchner, Citation1963; Helmboldt & Garner, Citation1964). Chickens between 3 and 6 weeks of age are highly susceptible for clinical IBD (Cosgrove, Citation1962; Hirai et al., Citation1974d; Ley et al., Citation1983) when protective levels of neutralizing antibody levels are absent. IBDV is a double-stranded, non-enveloped, icosahedral virus (Delmas et al., Citation2005) with diameter ranging from 55 to 65 nm (Hirai et al., Citation1974b; Nick et al., Citation1976). IBDV belongs to the family Birnaviridae, genus Avibirnavirus. IBDV is a bisegmented virus containing segments A and B (Dobos et al., Citation1979; Müller et al., Citation1979; Kibenge et al., Citation1988). Segment A encodes on two open reading frames (ORFs) for viral proteins (VPs). ORF1 encodes for the viral polyprotein, which is autoproteolitically cleaved by its viral protease to three VPs, the premature preVP2, VP4 and VP3 (Birghan et al., Citation2000; Lejal et al., Citation2000). A second ORF (Spies et al., Citation1989, Bayliss et al., Citation1990) precedes and partially overlaps the first ORF and encodes for VP5 (Mundt & Müller, Citation1995), which is non-essential for virus replication in cell culture (Mundt et al., Citation1997) but attenuated IBDV when the expression was omitted (Yao et al., Citation1998). Segment B encodes for VP1, which represents the viral RNA-dependent RNA polymerase (Spies et al., Citation1987; von Einem et al., Citation2004).

Based on virulence in susceptible chickens, IBDV serotype I strains were broadly classified into three pathotypes: classical, very virulent (vv), and variant strains. Classical IBDV strains cause a strong inflammatory response in the BF, and chickens infected with virulent classical IBDV show clinical signs (Cosgrove, Citation1962; Käufer & Weiss, Citation1980). In contrast, variant IBDV strains result in atrophy of the BF in the absence of any inflammatory changes (Rosenberger & Cloud, Citation1986; Sharma et al., Citation1989). The subclinical disease caused by variant IBDVs may barely be noticed and sometimes only an increase in the incidence of respiratory diseases might be observed (Saif Citation1984; Rosenberger & Cloud, Citation1986). More dramatic are vvIBDV, which cause a fast depletion of the B lymphocytes in the BF that is also associated with petechial haemorrhage in the muscles and a haemorrhagic BF (van den Berg & Meulemans, Citation1991). IBDV strains described in the early 1960s from the USA belong to what is now referenced as the classical subtype. IBDV strains isolated in the USA in the mid-1980s were described as variant strains due to change in their pathogenic phenotype and were later determined to be antigenically different from earlier viruses (Saif, Citation1984; Rosenberger et al., Citation1985; Snyder et al., Citation1988a, 1988b; Snyder, Citation1990). All three pathotypes of IBDV induce immunosuppression, leading to an increase in the opportunity for secondary pathogens to invade the host and cause multisystemic diseases (Cho, Citation1970; Fadly et al., Citation1976; Rosenberger & Gelb, Citation1978; Pejkovski et al., Citation1979; Yuasa et al., Citation1980; Moradian et al., Citation1990). There are several variant strains of IBDV described—such as Md (Saif, Citation1984), E/Del (Rosenberger et al., Citation1985), variant A (Rosenberger et al., Citation1987), IN (Ismail et al., Citation1990), GLS and DS 326 (Snyder et al., Citation1992), and AL2 (Toro et al., Citation2009)—circulating in US commercial poultry operations. Based on the variability of IBDV strains in the field, immunization with vaccines that are antigenically similar to the circulating IBDV strains plays a key role in IBDV control. Breeder hens are commonly vaccinated with a series of live and killed vaccines in an effort to induce antibodies that are transferred to the offspring via the eggs (Wyeth & Cullen, Citation1978, Citation1979; Wyeth Citation1980). Vaccination programmes differ between poultry operations and are highly dependent upon the virus challenge circulating in the field.

Identification of circulating field strains of IBDV plays an important role in prevention and control. Field surveillance studies are necessary in the poultry flocks within commercial operations to select the most appropriate vaccines for use in vaccination programmes as well as to identify possible vaccine candidates (Dormitorio et al., Citation2007). Various diagnostic tools were developed for identification of IBDV in poultry flocks. Initially, isolation and identification of IBDV by electron microscopy was used to confirm the presence of the virus (Hirai et al., Citation1974a, Citation1974c). Diagnosis of IBDV was later based on the agar gel diffusion test (Hirai & Shimakura, Citation1974b) and also by virus neutralization (VN) assays (Weisman & Hitchner, Citation1978). Advanced molecular methods, such as reverse transcription-polymerase chain reaction (RT-PCR), supported rapid diagnosis of IBDV (Lee et al., Citation1992; Clementi et al., Citation1995; Moody et al., Citation2000). RT-PCR followed by restriction fragment length polymorphism (RFLP) was used for genotyping IBDV (Jackwood & Jackwood, Citation1994; Lin et al., Citation1994; Jackwood & Nielsen, Citation1997; Jackwood & Sommer, Citation1998) and was intended to diagnose IBDV based on comparison of RFLP patterns from unknown IBDVs with patterns of known viruses. However, these methods did not provide the most important information for determining the antigenicity of the viruses. Antigenic characterization of the IBDVs was necessary to determine whether changes in field viruses would result in a diminished protection from current vaccines. For this purpose, an antigen-capture enzyme-linked immunosorbent assay was developed that provided a system for differentiation between different antigenic subtypes of IBDV based on certain reaction patterns using a panel of neutralizing monoclonal antibodies (mAbs; Van der Marel et al., Citation1990). In-depth analysis of amino acid exchanges causing differences in the antigenicity of IBDV lead to the conclusion that IBDV antigenicity is more complex than expected (Letzel et al., Citation2007; Icard et al., Citation2008; Durairaj et al., Citation2011). It also became clear that amino acid exchanges outside the projection domain might result in antigenic differences (Durairaj et al., Citation2011). Using this approach, several IBDVs lacking reactivity with any neutralizing mAb and indicating a different antigenic make-up (Icard et al., Citation2008; Durairaj et al., Citation2011) were detected and isolated (IBDVn-Var). Attempts to characterize the viruses in cross-neutralization assays in embryonated eggs were subjective since the viruses were not lethal in embryos, as observed with some IBDV strains, and embryo lesions were variable. We therefore developed an in vivo model to evaluate the antigenicity of IBDVn-var strains. Since E/Del-like IBDV are the most common variant strains circulating in the USA, a system based on E/Del neutralizing antibodies was developed with a read-out system focused on lesions in the BF. Using this system, we were able to demonstrate that one virus field isolate was antigenically different from E/Del IBDV. The in vivo experimental model described will serve as a defined platform for biological evaluation of IBDV field isolates, lacking an antigenic signature in our in vitro characterization model, with known vaccine strains.

Materials and Methods

Cells

The VN assay was performed in the chicken embryo fibroblast cell line DF1 (Himly et al., Citation1998) grown in Dulbecco's modified Eagle's medium with 4.5 g/l glucose (DMEM-4.5; Thermo Scientific, Waltham, Massachusetts, USA) supplemented with 10% foetal bovine serum (Mediatech, Manassas, Virginia, USA), penicillin (100 IU/ml) and streptomycin (100 µg/ml). Cells were incubated at 37°C with 5% CO2.

Virus propagation in specific pathogen-free chickens

The viruses used in this experiment were E/Del (kindly provided by Ruud Hein, Intervet, Millsboro, Delaware, USA) and one IBDV field isolate from Alabama (IBDVn-var, Genbank accession number JF748992) previously described (Durairaj et al., Citation2011). For virus propagation, 3-week-old specific pathogen-free (SPF) chickens (Merial, Gainesville, Georgia, USA) were orally infected with E/Del and IBDVn-var in separate Horsfall–Bauer-type isolation units in a forced-air positive-pressure system (Poultry Diagnostic and Research Center, The University of Georgia, Athens, Georgia, USA). The SPF chickens were maintained in isolators and given feed and water ad libitum for the duration of the study. All chicken experiments were approved by the University of Georgia animal care and use committee (AUP number A2010 04-064-Y2-A1). The chickens were humanely euthanized and necropsied 96 h after infection. The BFs were harvested and homogenized in viral transport medium as previously described (Durairaj et al., Citation2011). The virus stocks were aliquoted and stored at −80oC until further use.

Virus titrations

Both viruses were titrated in 9-day-old embryonated SPF eggs (Merial) and 3-week-old SPF chickens to determine the median embryo infectious dose (EID50) and the median chicken infectious dose (CID50), respectively. The determination of the EID50 was performed by inoculating 100 µl per dilution into five embryonated eggs via the chorio-allantoic membrane route (Hitchner, Citation1970). The inoculated embryonated eggs were maintained in an incubator at 37°C with 55% relative humidity and candled daily to check for viability. Eggs with dead embryos were removed and stored at 4°C until 7 days after inoculation when the remaining embryonated eggs were evaluated for the presence of lesions characteristic of IBDV (retarded growth, green-spotted liver, enlarged spleen). For the determination of the CID50, the SPF chickens were divided into groups of five chickens each. Chickens were bled prior to inoculation and the sera were tested by VN assay to confirm the absence of pre-existing IBDV antibodies. Seven groups per virus were infected by the ocular route with serially diluted viral inoculum of E/Del or IBDVn-var. One group was not inoculated and served as the negative control. The chickens were maintained in Horsfall–Bauer isolation units and given feed and water ad libitum. Seven days after infection, the chickens were humanely euthanized, and BFs were collected and preserved in 10% neutral buffered formalin for microscopic evaluation. The lesions in the BF were scored based on the degree of B-lymphocyte depletion using a bursal lesion score (BLS) from 1 to 4: score of 1, up to 10% of the bursal follicles show depletion; score of 2, 10 to 30% of the bursal follicles show depletion; score of 3, 31 to 70% of the bursal follicles show depletion; and score of 4, >70% of the bursal follicles show depletion. Any BLS >1 was regarded as positive. Calculations of both the EID50 and CID50 were performed according to the method of Reed & Muench (Citation1938).

Generation of hyperimmune serum in chickens

Three-week-old SPF chickens were orally infected with 100 µl E/Del-like virus 8903 (kindly provided by Ruud Hein, Intervet), with a titre of 101.5 median tissue culture infective dose (TCID50). Three weeks later, 100 µl E/Del-like 8903 strain (102.0 TCID50) was administered by the intramuscular route. Chickens were intramuscularly injected 3 weeks later with an oil-emulsion vaccine containing one volume part β-propiolactone inactivated 8903 virus containing 107 TCID50/ml prior to inactivation and one part incomplete Freund's adjuvant. The inactivated virus was passaged twice in DF1 cells to confirm successful inactivation due to absence of any cytopathic effect. Five weeks later, chickens were exsanguinated under aseptic conditions. The clotted blood samples were centrifuged by 700×g at 4°C, and serum individually harvested and heat inactivated at 56°C for 30 min. The virus neutralizing antibody titre was determined (see below) and serum samples with a virus neutralizing antibody titre > 213 were pooled, aliquoted and the final virus neutralizing antibody titre was determined for the pool. The serum samples were stored at −80oC.

Determination of virus neutralizing antibody titre

The VN assay was performed to determine levels of E/Del-specific neutralizing antibodies in the chicken sera obtained. First, the TCID50 for the E/Del like strain 8903 was determined following standard methods. For the VN test, 50 µl foetal bovine serum-free DMEM-4.5 was added to all wells of a 96-well tissue culture plate. Next 50 µl of the serum was added to the first column (2–1) and serially two-fold diluted through the 12th column of the plate (2–12). In case the VN titre was greater than 212, a second plate was used to continue the dilutions. Next, 50 µl of virus containing 100 TCID50 was added to each serum dilution and incubated for 1 h at 37°C. During that time, DF1 cells were trypsinized, resuspended in 10% foetal bovine serum-containing DMEM-4.5 and adjusted to a cell density of 5×105 cells/ml. One hundred microlitres of DF1 cells were added to all wells of the virus–serum suspension and incubated for 5 days at 37°C and 5% CO2. After incubation, cells were scored for the presence of a CPE. The end-point of the VN test for a serum sample was determined to be the reciprocal of the highest dilution, expressed in log2, in which there was no visible CPE. During each test, the diluted virus was back-titrated to determine the true TCID50. If the TCID50 was greater than a 0.25 log10 difference from 100 TCID50, the VN test was regarded as invalid and was repeated.

Administration of hyperimmune serum

Two 3-week-old SPF chickens per group were used for this study. Three hundred microlitres of the hyperimmune serum were administered to each chicken via either the intramuscular (i.m.), intravenous (i.v.) or subcutaneous (s.c.) route and two chickens were used as negative control. Each group was kept in a Horsefall–Bauer isolation unit and given water and feed ad libitum. Chickens were bled at 24, 48, 72, and 168 h post injection via the brachial vein on the side opposite of the inoculation. The clotted blood samples were centrifuged at 700×g for 5 min and the serum was harvested. The serum samples were heat inactivated at 56°C for 30 min and the VN test was performed as described above to study the dynamics of VN antibody titre.

Differentiation of IBDV strains in vivo

Three-week-old SPF chickens were divided into 21 groups of five chickens per group. Each chicken was wing-banded for identification. Serum was diluted up to a dilution of 1:128. Ten groups of five chickens per group were used for each virus during the experiments: Group 1 (serum undiluted), Group 2 (1:2 diluted), Group 3 (1:4 diluted), Group 4 (1:6 diluted), Group 5 (1:8 diluted), Group 6 (1:16 diluted), Group 7 (1:32 diluted), Group 8 (1:64 diluted), Group 9 (1:128 diluted), and Group 10 (phosphate-buffered saline). Twenty-four hours after injection of the appropriate serum dilution, the chickens were bled via the brachial vein on the opposite wing and blood samples were processed for the VN test as described above. One hour later, one group from each serum dilution level was challenged with either 100 CID50 of E/Del or 100 CID50 IBDVn-var. One group of SPF chickens did not receive serum and was left unchallenged. Seven days after challenge, chickens were humanely euthanized and BF collected for microscopic evaluation to determine the BLS.

Histopathology

Bursal tissues were collected at the time of necropsy on an individual basis to allow the matching of each serum sample to the appropriate chicken. The bursal sample was placed in 10% neutral buffered formalin, and was paraffin embedded. Sections of the paraffin-embedded BF were stained with haematoxylin and eosin following standard histologic procedures. The stained sections were microscopically examined for the presence of bursal lesions. Based on the depletion of the B lymphocytes, the BLS was determined for each BF based on the scoring system described above.

Results

Determination of viral titres

Initially, cross-neutralization assays in SPF embryos inoculated via the chorio-allantoic membrane were performed to determine the antigenic relatedness of IBDVn-var with E/Del. However, IBDVn-var was not embryo lethal and identification of embryo lesions was minimal and somewhat subjective. Although results from the initial cross-neutralization studies in embryos indicated IBDVn-var was antigenically different from E/Del, additional testing was needed to provide more definitive and reliable data. Determination of the CID50 was thus undertaken to determine the titre of the viruses. The read-out system was the presence of lesions in the BF, thus providing the most sensitive system for determining infection in each dilution. The infectious titre was determined using material generated from the BF. This would provide a standard and comparable system for evaluating infection at later time points, during titration and challenge. Although the EID50 was not used during the study, the calculated titres for the E/Del bursa material was 104.1/100 µl for EID50 and 104.5/100 µl for CID50. Interestingly, titration of IBDVn-var resulted in a titre of 103.5/100 µl (EID50) and 104.5/100 µl (CID50). Thus if the calculation for the infectious dose would have been based on 100 EID50, the infectivity for the BF would have been underestimated by a factor of 10 for IBDVn-var and a higher dose would have been used for infection in comparison with the E/Del virus.

Generation and administration of hyperimmune serum

The final VN titre of the pooled hyperimmune serum from chickens vaccinated with the E/Del-like strain 8903 as described above was 214. This serum was used for all subsequent experiments. Three routes of application (i.m., s.c., i.v.) were tested for their practicality and reliability. From the user's point of view, the easiest route was the i.m. application. The s.c. route of application had the same practicality and reliability. The i.v. route was not easy to perform and failed several times during experiments (data not shown). Based on the VN titres measured at several time points after serum administration, it was observed following i.m. application that the VN titre decreased relatively fast (). As expected, the VN titre was highest after i.v. application in comparison with both i.m. and s.c. application. The s.c. administration resulted in a higher titre than the i.m., but lower than the i.v. route of administration; however, the decline of the s.c. VN titre was comparable with the i.v. application. After taking into consideration all aspects of the serum administration, the s.c. route was chosen since it was easy to perform, reliable and resulted in VN titres that lasted for a relatively long period of time.

Table 1.  Virus neutralization titre after different applications methods and time points.

Comparison of E/Del and IBDVn-var in the in vivo model

The experiments were performed using 3-week-old SPF chickens. The chickens were s.c. injected with E/Del-specific antiserum at different dilutions. The VN titre prior to dilution was 214 when 100 TCID50 of the E/Del-like strain 8903 were used for the VN test. In a separate experiment, the identity of the tissue-culture-adapted 8903 was confirmed by indirect immunofluorescence using the panel of mAbs (10, 57, R63, 67, B69), which resulted in a positive reaction with only R63 and 67 as previously described (Icard et al., Citation2008). This confirmed the E/Del-like antigenic subtype. Twenty-four hours later the chickens were bled and the VN titres were determined using the E/Del-like strain 8903 ( and ). As expected the VN titres declined in parallel with the dilution of the serum. Furthermore, the VN titres ranged from one titre (Group 9) to five different titres (Group 5) and indicated variable uptake of the antibodies into the bloodstream when administered by the s.c. route. The highest VN titre observed was 1024 in three chickens injected with either the undiluted or the 1:2 diluted serum. The control chickens were free of any IBDV strain 8903 neutralizing antibodies. The result of the challenge infection with IBDV strain E/Del and the IBDVn-Var was evaluated using the BLS at day 7 following challenge. In the group of the chickens infected with only virus and no serum, a BLS of 4 was detected in every chicken indicating a sufficient infectious dose of virus. Chickens that did not receive serum or virus had a BLS of 1. Chickens that received serum preparations and were challenged were regarded as protected if the BLS was between 1 and 2. The results showed that E/Del-challenged chickens were fully protected up to a VN titre of 128 (see ). A few chickens were protected when the VN titre was ≤64. Even at a VN titre of 8, two out of six chickens were fully protected—as indicated by a BLS of 1.

Table 2.  Virus neutralization titre at day of challenge infection.

Table 3.  Protection of chickens after challenge infection with two different IBDV strains.

Results from the challenge infection with IBDVn-var were different. Only chickens with a VN titre ≥512 were fully protected. Fifty to 60% (VN titres of 256 and 128, respectively) of birds were protected from challenge infection, which indicated that IBDVn-var was able to break through VN titres at levels where chickens were still fully protected when challenged with the homologue E/Del strain. The data clearly showed that only one chicken out of 30 was protected from IBDVn-var challenge at VN titre levels ≤32, while six out of 30 chickens were still protected at these titres after challenge with E/Del. It needs to be mentioned that no clinical signs of disease were observed in any chickens, also indicating that the IBDVn-var clinically belongs to the large group of variant IBDV strains.

Discussion

IBDV is ubiquitous in poultry operations worldwide. Mutations in the viral progeny can arise during replication as the viral replicase probably lacks a proofreading mechanism, responsible for repair of misincorporated nucleotides. In addition, changes in the antigenic make-up can occur by selection of escape mutants due to presence of neutralizing antibodies and may result in IBDVs that differ from vaccines in either their antigenicity or virulence or both. The results of this selection process are not predictable. Consequently, this requires the continuous characterization of field isolates for their pathogenic potential and antigenic make-up. The development of neutralizing mAbs that were used to classify different IBDV antigenic subtypes (Snyder et al., Citation1988a, 1988b, 1992; Snyder, Citation1990) and their subsequent use in an antigen-capture enzyme-linked immunosorbent assay was an important step for antigenic characterization of field isolates (van der Marel et al., Citation1990) and served as an effective surveillance tool. More rapid techniques were developed and the use of RT-PCR followed by restriction enzyme digestion (RFLP) of the amplified cDNA fragment (Jackwood & Jackwood, Citation1994) became mainstream for typing IBDV. Using this approach it was possible to distinguish between different IBDV subtypes, but did not provide essential information regarding the virulence or antigenicity of viruses (Jackwood & Nielsen, Citation1997; Jackwood & Sommer, Citation1997). In some cases, the data generated by RT-PCR/RFLP led to the incorrect designation of IBDV subtype. One example was the detection of two IBDV isolates that were grouped into the vvIBDV group based on RFLP. Surprisingly, one isolate caused 70 to 80% mortality, typical for vvIBDV, while the other isolate caused only 10% mortality (Hoque et al., Citation2001).

Various antigenic strains of IBDV have been described in the USA based on their reactivity with mAbs: E/Del-like IBDV reacted with mAbs R63 and 67, GLS-like IBDV reacted with mAbs 10 and 57, and classical IBDV reacted with mAbs 10, R63, and B69 (Snyder et al., Citation1988a, Citation1988b, Citation1992; Snyder, Citation1990). In addition, an IBDV isolated in Belgium reacted with mAbs 10, R63, 67, and B69, a combination previously not described (Letzel et al., Citation2007). Furthermore, a new group of viruses was described that did not react with any of the mAbs using a diagnostic reverse genetics system for IBDV (Icard et al., Citation2008; Durairaj et al., Citation2011). It is important to keep in mind that virus isolates represent only a snapshot of IBDVs evolving in the field. Based on an evolutionary advantage they might become established in an environment, or in the case of an evolutionary disadvantage they may go unnoticed. Surveillance is thus a necessary tool for monitoring IBDV evolution in the field. The recently described IBDVs that did not react with any mAbs have also been identified in South America and Europe (unpublished data). Consequently these IBDV isolates need to be characterized either in vitro or in vivo. One virus isolate from Alabama, belonging to the group of IBDVs that do not react with any of the mAbs, was evaluated in vitro and in vivo (Durairaj et al., Citation2011). Virus titrations in SPF chicken embryos repeatedly resulted in no embryo mortality and minimal embryo lesions that were inconsistent between virus dilutions, making it difficult to quantify the virus after infection of chicken embryos (data not shown). This particular virus appeared to be less embryo adapted than observed for other IBDV field isolates. Ultimately the virus was titrated in chickens and provided the read-out system for subsequent in vivo VN assays. The use of susceptible IBDV antibody-free chickens represents the most sensitive and reliable system for detection and quantification of IBDV. In the in vivo model described, the IBDV antibody titres detected in the serum after administration of IBDV hyperimmune serum using the s.c. route were comparable with those obtained following i.v. administration, although only two chickens were used for the experiments. The results showed that, independent of the serum dilution, IBDV antibody titres were present in a gradient in the chicken serum. The VN titre levels were not always in agreement with the dilution of the serum used in a particular group. This may be a result in differences in the uptake of IBDV antibodies into the bloodstream. Owing to this finding, the data were arranged by chicken and corresponding VN titre followed by the addition of the BLS score. The results obtained showed that the E/Del was neutralized, as expected, at E/Del-specific VN titre levels, whereas IBDVn-var was able to break through even higher VN titres and induced bursal lesions. These results confirmed the assumption that a virus lacking reactivity with any of the neutralizing mAbs in the in vitro assay was also antigenically different from E/Del since the same IBDV titres for infection were used. The neutralizing epitopes that have been recognized by the immune system of the mice are thus also important for VN in chickens. Besides this, other, so far unidentified, neutralizing epitopes must also be present on the viral capsid that may be more cross-reactive. This assumption is based on the observation that at least partial VN of IBDVn-var was observed. These epitopes are shared between both viruses investigated here and requires the analysis of such cross-neutralizing epitopes in more detail for a better understanding of the biology of the virus. This might help to improve virus vaccines by molecular techniques that are available. The findings described here do not show that IBDVn-var was also different from classical as well as GLS-like IBDVs, but allows the assumption. Hyperimmune sera specific for classical IBDV (e.g. D78) or GLS-like IBDV (GLS-05) are being prepared as described in this paper and will be used similarly in this in vivo system for a more in-depth analysis of newly emerging IBDV field isolates.

In summary, an in vivo experimental model was developed that can be used for the biological evaluation of IBDV field isolates in unvaccinated susceptible chickens. The use of IBDV subtype-specific antisera for passive immunization will allow the determination of the break-through titres between IBDV isolates in a titre-dependent manner. The in vivo experimental model is a highly sensitive method for evaluating IBDV infections since the results are based on lesions in the BF that represents the target organ for IBDV. It is clear that this study provides data to support the use of an in vivo neutralization model to identify relevant antigenic differences between IBDVs and further refinement of the model is needed to decrease the numbers of chickens necessary for the analysis and to finally establish an in vitro method.

References

  • Allan , W.H. , Faragher , J.T. and Cullen , G.A. 1972 . Immunosuppression by the infectious bursal agent in chickens immunised against Newcastle disease . Veterinary Record , 90 : 511 – 512 . doi: 10.1136/vr.90.18.511
  • Bayliss , C.D. , Spies , U. , Shaw , K. , Peters , R.W. , Papageorgiou , A. , Müller , H. and Boursnell , M.E. 1990 . A comparison of the sequences of segment A of four infectious bursal disease virus strains and identification of a variable region in VP2 . Journal of General Virology , 71 : 1303 – 1312 . doi: 10.1099/0022-1317-71-6-1303
  • Birghan , C. , Mundt , E. and Gorbalenya , A.E. 2000 . A non-canonical Lon proteinase deficient of the ATPase domain employs the Ser-Lys catalytic dyad to impose broad control over the life cycle of a double-stranded RNA virus . The EMBO Journal , 19 : 114 – 123 . doi: 10.1093/emboj/19.1.114
  • Cho , B.R. 1970 . Experimental dual infections of chickens with infectious bursal and Marek's disease agents I. Preliminary observation on the effect of infectious bursal agent on Marek's disease . Avian Diseases , 14 : 665 – 675 . doi: 10.2307/1588638
  • Clementi , M. , Menzo , S. , Manzin , A. and Bagnarelli , P. 1995 . IBDV detection by real-time RT-PCR quantitative molecular methods in virology . Archives of Virology , 40 : 1523 – 1539 . doi: 10.1007/BF01322527
  • Cosgrove , A.S. 1962 . An apparently new disease of chickens—avian nephrosis . Avian Diseases , 6 : 385 – 389 . doi: 10.2307/1587909
  • Delmas , B. , Kibenge , F.S.B. , Leong , J.C. , Mundt , E. , Vakharia , V.N. and Wu , J.L. 2005 . “ Birnaviridae ” . In Virus Taxonomy , Edited by: Fauquet , C.M. , Mayo , M.A. , Maniloff , J. , Desselberger, , U. and Ball , A.L. 561 – 569 . London : Academic Press .
  • Dobos , P. , Hill , B.J. , Hallett , R. , Kells , D.T. , Becht , H. and Teninges , D. 1979 . Biophysical and biochemical characterization of five animal viruses with bisegmented double-stranded RNA genomes . Journal of Virology , 32 : 593 – 605 .
  • Dormitorio , T.V. , Giambrone , J.J. , Guo , K. and Jackwood , D.J. 2007 . Molecular and phenotypic characterization of infectious bursal disease virus isolates . Avian Diseases , 51 : 597 – 600 . doi: 10.1637/0005-2086(2007)51[597:MAPCOI]2.0.CO;2
  • Durairaj , V. , Sellers , H.S. , Linnemann , E.G. , Icard , A.H. and Mundt , E. 2011 . Investigation of the antigenic evolution of field isolates using the reverse genetics system of infectious bursal disease virus (IBDV) . Archives of Virology , 156 : 1717 – 1728 . doi: 10.1007/s00705-011-1040-x
  • Fadly , A.M. , Winterfield , R.W. and Olander , H.J. 1976 . Role of the bursa of Fabricius in the pathogenicity of inclusion body hepatitis and infectious bursal disease viruses . Avian Diseases , 20 : 467 – 477 . doi: 10.2307/1589379
  • Faragher , J.T. , Allan , W.H. and Cullen , G.A. 1972 . Immunosuppressive effect of the infectious bursal agent in the chicken . Nature New Biology , 237 : 118 – 119 .
  • Giambrone , J.J. , Donahoe , J.P. , Dawe , D.L. and Eidson , C.S. 1977 . Specific suppression of the bursa-dependent immune system of chicks with infectious bursal disease virus . American Journal of Veterinary Research , 38 : 581 – 583 .
  • Helmboldt , C.F. and Garner , E. 1964 . Experimentally induced Gumboro disease (IBA) . Avian Diseases , 8 : 561 – 575 . doi: 10.2307/1587944
  • Himly , M. , Foster , D.N. , Bottoli , I. , Iacovoni , J.S. and Vogt , P.K. 1998 . The DF-1 chicken fibroblast cell line: transformation induced by diverse oncogenes and cell death resulting from infection by avian leukosis viruses . Virology , 248 : 295 – 304 . doi: 10.1006/viro.1998.9290
  • Hirai , K. and Calnek , B.W. 1979 . In vitro replication of infectious bursal disease virus in stablished lymphoid cell lines and chicken B lymphocytes . Infection and Immunity , 25 : 964 – 970 .
  • Hirai , K. , Kawamoto , E. and Shimakura , S. 1974a . Some properties of precipitating antigens associated with infectious bursal disease virus . Infection and Immunity , 10 : 1235 – 1240 .
  • Hirai , K. and Shimakura , S. 1974b . Structure of infectious bursal disease virus . Journal of Virology , 14 : 957 – 964 .
  • Hirai , K. , Shimakura , S. and Kawamoto , E. 1974c . Electron-microscope characterization of infectious bursal disease virus . Avian Diseases , 18 : 467 – 471 . doi: 10.2307/1589116
  • Hirai , K. , Shimakura , S. , Kawamoto , E. , Taguchi , F. , Kim , S.T. , Chang , C.N. and Iritani , Y. 1974d . The immunodepressive effect of infectious bursal disease virus in chickens . Avian Diseases , 18 : 50 – 57 . doi: 10.2307/1589241
  • Hitchner , S.B. 1963 . Discussion on Gumboro disease . University of Delware Poultry Pathology Letter No. 46 .
  • Hitchner , S.B. 1970 . Infectivity of infectious bursal disease virus for embryonating eggs . Poultry Science , 49 : 511 – 516 . doi: 10.3382/ps.0490511
  • Hoque , M.M. , Omar , A.R. , Chong , L.K. , Hair-Bejo , M. and Aini , I. 2001 . Pathogenicity of SspI-positive infectious bursal disease virus and molecular characterization of the VP2 hypervariable region . Avian Pathology , 30 : 369 – 380 . doi: 10.1080/03079450120066377
  • Icard , A.H. , Sellers , H.S. and Mundt , E. 2008 . Detection of infectious bursal disease virus isolates with unknown antigenic properties by reverse genetics . Avian Diseases , 52 : 590 – 598 . doi: 10.1637/8302-040408-Reg.1
  • Ismail , N.M. , Saif , Y.M. , Wigle , W.L. , Havenstein , G.B. and Jackson , C. 1990 . Infectious bursal disease virus variant from commercial leghorn pullets . Avian Diseases , 34 : 141 – 145 . doi: 10.2307/1591345
  • Ivanyi , J. and Morris , R. 1976 . Immunodeficiency in the chicken. IV. An immunological study of infectious bursal disease . Clinical and Experimental Immunology , 23 : 154 – 165 .
  • Jackwood , D.J. and Jackwood , R.J. 1994 . Infectious bursal disease viruses: molecular differentiation of antigenic subtypes among serotype 1 viruses . Avian Diseases , 38 : 531 – 537 . doi: 10.2307/1592075
  • Jackwood , D.J. and Nielsen , C.K. 1997 . Detection of infectious bursal disease viruses in commercially reared chickens using the reverse transcriptase/polymerase chain reaction–restriction endonuclease assay . Avian Diseases , 41 : 137 – 143 . doi: 10.2307/1592453
  • Jackwood , D.J. and Sommer , S.E. 1997 . Restriction fragment length polymorphisms in the VP2 gene of infectious bursal disease viruses . Avian Diseases , 41 : 627 – 637 . doi: 10.2307/1592154
  • Jackwood , D.J. and Sommer , S.E. 1998 . Genetic heterogeneity in the VP2 gene of infectious bursal disease viruses detected in commercially reared chickens . Avian Diseases , 42 : 321 – 339 . doi: 10.2307/1592483
  • Käufer , I. and Weiss , E. 1980 . Significance of bursa of Fabricius as target organ in infectious bursal disease of chickens . Infection and Immunity , 27 : 364 – 367 .
  • Kibenge , F.S. , Dhillon , A.S. and Russell , R.G. 1988 . Biochemistry and immunology of infectious bursal disease virus . Journal of General Virology , 69 : 1757 – 1775 . doi: 10.1099/0022-1317-69-8-1757
  • Lee , L.H. , Yu , S.L. and Shieh , H.K. 1992 . Detection of infectious bursal disease virus infection using the polymerase chain reaction . Journal of Virological Methods , 40 : 243 – 253 . doi: 10.1016/0166-0934(92)90083-P
  • Lejal , N. , Da Costa , B. , Huet , J.C. and Delmas , B.J. 2000 . Role of Ser-652 and Lys-692 in the protease activity of infectious bursal disease virus VP4 and identification of its substrate cleavage sites . Journal of General Virology , 81 : 983 – 992 .
  • Letzel , T. , Coulibaly , F. , Rey , F.A. , Delmas , B. , Jagt , E. , van Loon , A.A. and Mundt , E. 2007 . Molecular and structural bases for the antigenicity of VP2 of infectious bursal disease virus . Journal of Virology , 81 : 12827 – 12835 . doi: 10.1128/JVI.01501-07
  • Ley , D.H. , Yamamoto , R. and Bickford , A.A. 1983 . The pathogenesis of infectious bursal disease: serologic, histopathologic, and clinical chemical observations . Avian Diseases , 27 : 1060 – 1085 . doi: 10.2307/1590207
  • Lin , T.L. , Wu , C.C. , Rosenberger , J.K. and Saif , Y.M. 1994 . Rapid differentiation of infectious bursal disease virus serotypes by polymerase chain reaction . Journal of Veterinary Diagnostic Investigation , 6 : 100 – 102 . doi: 10.1177/104063879400600119
  • Moody , A. , Sellers , S. and Bumstead , N. 2000 . Measuring infectious bursal disease virus RNA in blood by multiplex real-time quantitative RT-PCR . Journal of Virological Methods , 85 : 55 – 64 . doi: 10.1016/S0166-0934(99)00156-1
  • Moradian , A. , Thorsen , J. and Julian , R.J. 1990 . Single and combined infections of specific-pathogen-free chickens with infectious bursal disease virus and an intestinal isolate of reovirus . Avian Diseases , 34 : 63 – 72 . doi: 10.2307/1591335
  • Müller , H. 1986 . Replication of infectious bursal disease virus in lymphoid cells . Archives of Virology , 87 : 191 – 203 . doi: 10.1007/BF01315299
  • Müller , H. , Scholtissek , C. and Becht , H. 1979 . The genome of infectious bursal disease virus consists of two segments of double-stranded RNA . Journal of Virology , 31 : 584 – 589 .
  • Mundt , E. , Köllner , B. and Kretzschmar , D. 1997 . VP5 of infectious bursal disease virus is not essential for viral replication in cell culture . Journal of Virology , 71 : 5647 – 5651 .
  • Mundt , E. and Müller , H. 1995 . Complete nucleotide sequences of 5′- and 3′-noncoding regions of both genome segments of different strains of infectious bursal disease virus . Virology , 209 : 10 – 18 . doi: 10.1006/viro.1995.1226
  • Nick , H. , Cursiefen , D. and Becht , H. 1976 . Structural and growth characteristics of infectious bursal disease virus . Journal of Virology , 18 : 227 – 234 .
  • Pejkovski , C. , Davelaar , F.G. and Kouwenhoven , B. 1979 . Immunosuppressive effect of infectious bursal disease virus on vaccination against infectious bronchitis . Avian Pathology , 8 : 95 – 106 . doi: 10.1080/03079457908418330
  • Reed , L.J. and Muench , H. 1938 . A simple method of estimating fifty percent endpoints . American Journal of Epidemiology , 27 : 493 – 497 .
  • Rosenberger , J.K. and Cloud , S.S. 1986 . Isolation and characterization of variant infectious bursal disease viruses (Abstract) . Journal of the American Veterinary Medical Association , 189 : 357
  • Rosenberger , J.K. , Cloud , S.S. , Gelb , J. , Odor , E. & Dohms , S.E. 1985 . Sentinel birds survey of Delmarva broiler flocks . In Proceedings of the 20th National Meeting on Poultry Health and Condemnation Ocean City (pp. 94 – 101 ). Maryland .
  • Rosenberger , J.K. , Cloud , S.S. and Metz , A. 1987 . Use of infectious bursal disease virus variant vaccines in broilers and broiler breeders . Proceedings of the Western Poultry Disease Conference , 36 : 105 – 109 .
  • Rosenberger , J.K. and Gelb , J. 1976 . Immunosuppressive effects of the infectious bursal agent and relationships to other poultry diseases . Proceedings in Annual Meeting of the United States Animal Health Association , 80 : 283 – 289 .
  • Rosenberger , J.K. and Gelb , J. Jr . 1978 . Response to several avian respiratory viruses as affected by infectious bursal disease virus . Avian Diseases , 22 : 95 – 105 . doi: 10.2307/1589512
  • Saif , Y.M. 1984 . Infectious bursal disease virus type . In Proceedings of the 19th National Meeting On Poultry Health and Condemnations Ocean City (pp. 105 – 107 ). Maryland .
  • Saif , Y.M. 1991 . Immunosuppression induced by infectious bursal disease virus . Veterinary Immunology and Immunopathology , 30 : 45 – 50 . doi: 10.1016/0165-2427(91)90007-Y
  • Sharma , J.M. , Dohms , J.E. and Metz , A.L. 1989 . Comparative pathogenesis of serotype 1 and variant serotype 1 isolates of infectious bursal disease virus and their effect on humoral and cellular immune competence of specific-pathogen-free chickens . Avian Diseases , 33 : 112 – 124 . doi: 10.2307/1591076
  • Snyder , D.B. 1990 . Changes in the field status of infectious bursal disease virus . Avian Pathology , 19 : 419 – 423 . doi: 10.1080/03079459008418695
  • Snyder , D.B. , Lana , D.P. , Cho , B.R. and Marquardt , W.W. 1988a . Group and strain-specific neutralization sites of infectious bursal disease virus defined with monoclonal antibodies . Avian Diseases , 32 : 527 – 534 . doi: 10.2307/1590923
  • Snyder , D.B. , Lana , D.P. , Savage , P.K. , Yancey , F.S. , Mengel , S.A. and Marquardt , W.W. 1988b . Differentiation of infectious bursal disease virus directly from infected tissues with neutralizing monoclonal antibodies: evidence of a major antigenic shift in recent field isolates . Avian Diseases , 32 : 535 – 539 . doi: 10.2307/1590924
  • Snyder , D.B. , Vakharia , V.N. and Savage , P.K. 1992 . Naturally occurring-neutralizing monoclonal antibody escape variants define the epidemiology of infectious bursal disease viruses in the United States . Archives of Virology , 127 : 89 – 101 . doi: 10.1007/BF01309577
  • Spies , U. , Müller , H. and Becht , H. 1987 . Properties of RNA polymerase activity associated with infectious bursal disease virus and characterization of its reaction products . Virus Research , 8 : 127 – 140 . doi: 10.1016/0168-1702(87)90024-4
  • Spies , U. , Müller , H. and Becht , H. 1989 . Nucleotide sequence of infectious bursal disease virus genome segment A delineates two major open reading frames . Nucleic Acids Research , 17 : 7982 doi: 10.1093/nar/17.19.7982
  • Toro , H. , Effler , J.C. , Hoerr , F.J. and van Ginkel , F.W. 2009 . Pathogenicity of infectious bursal disease virus variant AL2 in young chickens . Avian Diseases , 53 : 78 – 82 . doi: 10.1637/8455-082208-Reg.1
  • Van den Berg , T.P. and Meulemans , G. 1991 . Acute infectious bursal disease in poultry: protection afforded by maternally derived antibodies and interference with live vaccination . Avian Pathology , 20 : 409 – 421 . doi: 10.1080/03079459108418779
  • Van der Marel , P. , Snyder , D. and Lütticken , D. 1990 . Antigenic characterization of IBDV field isolates by their reactivity with a panel of monoclonal antibodies . Deutsche Tierärztliche Wochenschrift , 97 : 81 – 83 .
  • Von Einem , U.I. , Gorbalenya , A.E. , Schirrmeier , H. , Behrens , S.E. , Letzel , T. and Mundt , E. 2004 . VP1 of infectious bursal disease virus is an RNA-dependent RNA polymerase . Journal of General Virology , 85 : 2221 – 2229 . doi: 10.1099/vir.0.19772-0
  • Weisman , J. and Hitchner , S.B. 1978 . Virus-neutralization versus agar-gel precipitin tests for detecting serological response to infectious bursal disease virus . Avian Diseases , 22 : 598 – 603 . doi: 10.2307/1589634
  • Wyeth , P.J. and Cullen , G.A. 1978 . Transmission of immunity from inactivated infectious bursal disease oil-emulsion vaccinated parent chickens to their chicks . Veterinary Record , 102 : 362 – 363 . doi: 10.1136/vr.102.16.362
  • Wyeth , P.J. and Cullen , G.A. 1979 . The use of an inactivated infectious bursal disease oil emulsion vaccine in commercial broiler parent chickens . Veterinary Record , 104 : 188 – 193 . doi: 10.1136/vr.104.9.188
  • Wyeth , P.J. 1980 . Passively transferred immunity to IBD following live vaccination of parent chickens by two different routes . Veterinary Record , 106 : 289 – 290 . doi: 10.1136/vr.106.13.289
  • Yao , K. , Goodwin , M.A. and Vakharia , V.N. 1998 . Generation of a mutant infectious bursal disease virus that does not cause bursal lesions . Journal of Virology , 72 : 2647 – 2654 .
  • Yuasa , N. , Taniguchi , T. , Noguchi , T. and Yoshida , I. 1980 . Effect of infectious bursal disease virus infection on incidence of anemia by chicken anemia agent . Avian Diseases , 24 : 202 – 209 . doi: 10.2307/1589780

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.