Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Avian Dis ; 68(2): 89-98, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885050

ABSTRACT

Outbreaks of infectious bronchitis (IB) continue to occur from novel variants of IB virus (IBV) emerging from selection of vaccine subpopulations and/or naturally occurring recombination events. S1 sequencing of Arkansas (Ark) -type viruses obtained from clinical cases in Alabama broilers and backyard chickens shows both Ark Delmarva Poultry Industry (ArkDPI) vaccine subpopulations as well as Ark vaccine viruses showing recombination with other IB vaccine viruses. IB Ark-type isolates AL5, most similar to an ArkDPI vaccine subpopulation selected in chickens, AL4, showing a cluster of three nonsynonymous changes from ArkDPI subpopulations selected in chickens, and AL9, showing recombination with Massachusetts (Mass) -type IBV, were examined for pathogenicity and ability to break through immunity elicited by vaccination with a commercial ArkDPI vaccine. Analysis of predicted S1 protein structures indicated the changes were in regions previously shown to comprise neutralizing epitopes. Thus, they were expected to contribute to immune escape and possibly virulence. Based on clinical signs, viral load, and histopathology, all three isolates caused disease in naïve chickens, although AL9 and AL5 viral loads in trachea were statistically significantly higher (30- and 40-fold) than AL4. S1 gene sequencing confirmed the stability of the relevant changes in the inoculated viruses in the chickens, although virus in some individual chickens exhibited additional S1 changes. A single amino acid deletion in the S1 NTD was identified in some individual chickens. The location of this deletion in the predicted structure of S1 suggested the possibility that it was a compensatory change for the reduced ability of AL4 to replicate in the trachea of naïve chickens. Chickens vaccinated with a commercial ArkDPI vaccine at day of hatch and challenged at 21 days of age showed that vaccination provided incomplete protection against challenge with these viruses. Moreover, based on viral RNA copy numbers in trachea, differences were detected in the ability of the vaccine to protect against these IBV isolates, with the vaccine protecting the most poorly against AL4. These results provide additional evidence supporting that IBV attenuated vaccines, especially ArkDPI vaccines, contribute to perpetuating the problem of IB in commercial chickens.


Protección contra los virus de la bronquitis infecciosa vacunales recombinantes y las subpoblaciones de vacunas seleccionadas en pollos. Los brotes de la bronquitis infecciosa aviar continúan presentándose a partir de nuevas variantes de dicho virus, que surgen de la selección de subpoblaciones de vacunas y/o eventos de recombinación que ocurren naturalmente. La secuenciación del gene S1 de virus tipo Arkansas (Ark) obtenidos de casos clínicos en pollos de engorde y de traspatio de Alabama muestra que tanto las subpoblaciones de la cepa vacunal Arkansas Delmarva Poultry Industry (ArkDPI) así como los virus de la vacuna Arkansas muestran recombinación con otros virus vacunales de la bronquitis infecciosa. Los aislamientos del virus de la bronquitis infecciosa Arkansas tipo "AL5", más similares a una subpoblación de vacuna ArkDPI seleccionada en pollos, "AL4", que muestra un grupo de tres cambios no sinónimos de subpoblaciones de ArkDPI seleccionadas en pollos y el tipo "AL9", que muestra recombinación con el serotipo Massachusetts, se examinaron para determinar su patogenicidad y capacidad para traspasar la inmunidad generada por la vacunación con una vacuna comercial ArkDPI. El análisis de las estructuras predichas de la proteína S1 indicó que los cambios se produjeron en regiones que previamente se había demostrado comprendían epítopos neutralizantes. Por lo tanto, se esperaba que contribuyeran al escape inmunológico y posiblemente a la virulencia. Con base en los signos clínicos, la carga viral y la histopatología, los tres aislados causaron enfermedad en pollos sin exposición previa, aunque las cargas virales de AL9 y AL5 en la tráquea fueron estadísticamente significativamente mayores (30 y 40 veces) en comparación con AL4. La secuenciación del gene S1 confirmó la estabilidad de los cambios relevantes en los virus inoculados en los pollos, aunque el virus en algunos pollos individuales exhibió cambios adicionales en el gene S1. Se identificó una deleción de un solo aminoácido en el dominio terminal N del gene S1 (NTD S1) en algunos pollos individuales. La ubicación de esta eliminación en la estructura predicha del gene S1 sugirió la posibilidad de que se tratara de un cambio compensatorio por la capacidad reducida de AL4 para replicarse en la tráquea de pollos sin exposición previa. Los pollos vacunados con una vacuna comercial ArkDPI el día de la eclosión y desafiados a los 21 días de edad mostraron que la vacunación proporcionó una protección incompleta contra el desafío con estos virus. Además, basándose en el número de copias del ARN viral en la tráquea, se detectaron diferencias en la capacidad de la vacuna para proteger contra estos aislados del virus de la bronquitis infecciosa, siendo la vacuna con la protección más deficiente contra AL4. Estos resultados proporcionan evidencia adicional que respalda que las vacunas atenuadas contra el virus de la bronquitis infecciosa, especialmente las vacunas ArkDPI, contribuyen a perpetuar esta enfermedad en los pollos comerciales.


Subject(s)
Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Viral Vaccines , Animals , Infectious bronchitis virus/immunology , Infectious bronchitis virus/genetics , Infectious bronchitis virus/pathogenicity , Poultry Diseases/prevention & control , Poultry Diseases/virology , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Viral Vaccines/immunology , Recombination, Genetic
2.
Avian Dis ; 66(1): 101-105, 2022 03.
Article in English | MEDLINE | ID: mdl-35308010

ABSTRACT

Resistance to infectious bronchitis (IB) is a polygenic trait, but little is known about how resistance distributes in the host population. In this study, a relatively large number (n = 369) of specific-pathogen-free white leghorn chickens (Gallus gallus) were challenged with an Arkansas -type virulent IB virus (IBV), and resistance was evaluated 5 days after challenge by viral load (IBV RNA) in the trachea and cecal tonsils, as well as by tracheal histomorphometry (mucosal thickness and lymphocyte infiltration). Contrary to expectations, results showed a non-Gaussian distribution of resistance of the whole population against challenge. Indeed, most chickens accumulated toward higher resistance, i.e., lower viral loads and less tracheal damage. The current results also indicated limited differences in resistance to IBV between sexes. Tracheal viral load was significantly higher in males than that in females, but tracheal damage did not significantly differ between sexes. The difference in tracheal viral load found in males and females could have implications for viral spread in commercial chicken populations.


Nota de investigación­Distribución de la resistencia al virus de la bronquitis infecciosa en una población de pollos susceptibles. La resistencia a la bronquitis infecciosa es un rasgo poligénico, pero se sabe poco acerca de cómo se distribuye la resistencia en la población huésped. En este estudio, varios (n=369) pollos White Leghorn libres de patógenos específicos fueron desafiados con un virus de la bronquitis infecciosa virulento de tipo Arkansas y la resistencia se evaluó cinco días después del desafío mediante la carga viral en la tráquea y las tonsilas cecales, así como por histomorfometría traqueal (grosor de la mucosa e infiltración de linfocitos). Contrariamente a lo esperado, los resultados mostraron una distribución no gaussiana de la resistencia de toda la población frente al desafío. De hecho, la mayoría de los pollos se distribuyeron hacia una mayor resistencia, es decir, cargas virales más bajas y menos daño traqueal. Los resultados actuales también indicaron diferencias limitadas en la resistencia al virus de la bronquitis infecciosa entre sexos. La carga viral traqueal fue significativamente mayor en los machos en comparación con las hembras, pero el daño traqueal no fue significativamente diferente entre sexos. La diferencia en la carga viral traqueal encontrada en machos y hembras podría tener implicaciones para la propagación viral en las poblaciones de pollos comerciales.


Subject(s)
Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Viral Vaccines , Animals , Chickens , Coronavirus Infections/veterinary , Male , Specific Pathogen-Free Organisms
3.
Avian Dis ; 65(4): 637-642, 2021 12.
Article in English | MEDLINE | ID: mdl-35068109

ABSTRACT

Despite continuous and extensive efforts to control infectious bronchitis (IB) throughout the century, the disease continues to be one of the most economically relevant diseases affecting the poultry production worldwide. Since the early 1990s, numerous scientists have explicitly warned about the role of attenuated vaccines on IB virus (IBV) evolution and the detrimental consequences of their use to the poultry industry. Herein, we review evidence indicating that the use of live vaccines increases genetic/phenotypic diversity of IBV, enhances their fitness in the environment, and ultimately aggravates and perpetuates the problem for the poultry industry. The available evidence leads to the unequivocal conclusion that attenuated IBV vaccines should be replaced by vaccines using alternative technologies if IBV is to be controlled effectively.


Estudio recapitulativo- El control global de la bronquitis infecciosa requiere reemplazar las vacunas atenuadas por tecnologías alternativas. A pesar de los continuos y extensos esfuerzos para controlar el virus de la bronquitis infecciosa a lo largo del siglo, la bronquitis infecciosa sigue siendo una de las enfermedades de mayor relevancia económica que afecta a la producción avícola en todo el mundo. Desde principios de la década de los 1990s, numerosos científicos han advertido explícitamente sobre el papel de las vacunas atenuadas en la evolución del virus de la bronquitis infecciosa y las consecuencias perjudiciales de su uso para la industria avícola. En este artículo, se revisa la evidencia que indica que el uso de vacunas vivas aumenta la diversidad genética y fenotípica del virus de la bronquitis infecciosa, mejora su aptitud en el medio ambiente y en última instancia, agrava y perpetúa el problema para la industria avícola. La evidencia disponible lleva a la conclusión inequívoca de que las vacunas atenuadas contra este virus deben ser reemplazadas por vacunas que utilicen tecnologías alternativas si se quiere controlar eficazmente a este virus.


Subject(s)
Bronchitis , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Viral Vaccines , Animals , Bronchitis/veterinary , Chickens , Coronavirus Infections/prevention & control , Coronavirus Infections/veterinary , Infectious bronchitis virus/genetics , Poultry Diseases/prevention & control , Vaccines, Attenuated
4.
Vaccine ; 35(43): 5864-5871, 2017 10 13.
Article in English | MEDLINE | ID: mdl-28899630

ABSTRACT

The avian coronavirus infectious bronchitis virus (IBV) S1 subunit of the spike (S) glycoprotein mediates viral attachment to host cells and the S2 subunit is responsible for membrane fusion. Using IBV Arkansas-type (Ark) S protein histochemistry, we show that extension of S1 with the S2 ectodomain improves binding to chicken tissues. Although the S1 subunit is the major inducer of neutralizing antibodies, vaccination with S1 protein has been shown to confer inadequate protection against challenge. The demonstrated contribution of S2 ectodomain to binding to chicken tissues suggests that vaccination with the ectodomain might improve protection compared to vaccination with S1 alone. Therefore, we immunized chickens with recombinant trimeric soluble IBV Ark-type S1 or S-ectodomain protein produced from codon-optimized constructs in mammalian cells. Chickens were primed at 12days of age with water-in-oil emulsified S1 or S-ectodomain proteins, and then boosted 21days later. Challenge was performed with virulent Ark IBV 21days after boost. Chickens immunized with recombinant S-ectodomain protein showed statistically significantly (P<0.05) reduced viral loads 5days post-challenge in both tears and tracheas compared to chickens immunized with recombinant S1 protein. Consistent with viral loads, significantly reduced (P<0.05) tracheal mucosal thickness and tracheal lesion scores revealed that recombinant S-ectodomain protein provided improved protection of tracheal integrity compared to S1 protein. These results indicate that the S2 domain has an important role in inducing protective immunity. Thus, including the S2 domain with S1 might be promising for better viral vectored and/or subunit vaccine strategies.


Subject(s)
Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Infectious bronchitis virus/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Subunit/immunology , Animals , Antibodies, Viral/immunology , Cell Line , Chickens/immunology , Genetic Vectors/immunology , HEK293 Cells , Humans , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Vaccination/methods , Vaccines, Attenuated/immunology , Viral Load/methods , Viral Vaccines/immunology , Virus Attachment/drug effects
5.
Avian Dis ; 61(3): 397-401, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28957002

ABSTRACT

We previously demonstrated that chickens primed with a recombinant Newcastle disease virus LaSota (rLS) expressing the S2 gene of infectious bronchitis virus (IBV) and boosted with an attenuated IBV Massachusetts (Mass)-type vaccine were protected against IBV Arkansas (Ark)-type virulent challenge. A possible basis for the reported ability of IBV 4/91 (serotype 793/B) vaccine to protect against divergent IBV strains (e.g., QX, Q1, and D1466) in a prime-boost approach with an IBV Mass vaccine is that an immune response against the S2 protein of IBV 4/91 is cross-protective. Therefore, we evaluated the protective capabilities of the S2 protein of IBV 4/91 expressed from rLS. The level of S2 amino acid sequence identity between 4/91 and the Ark challenge strain used in this study (90.7%) is within the range of S2 amino acid sequence identities between 4/91 and Q1 (91%-94%) and QX (89%-94%) strains. Chickens primed with attenuated Mass IBV at 1 day of age and boosted with rLS/IBV.S2-4/91 at 14 days of age were challenged with a virulent Ark IBV strain at 28 days of age. Protection (reduction of clinical signs and viral loads) assessed 5 days postchallenge showed nonsignificant differences between chickens primed with Mass vaccine and boosted with rLS/IBV.S2-4/91 and chickens vaccinated with Mass only. Thus, the observed level of protection is attributable only to the effect of the Mass vaccine, indicating that the S2 of IBV 4/91 does not induce broad cross-protective immunity.


Subject(s)
Coronavirus Infections/veterinary , Infectious bronchitis virus/immunology , Poultry Diseases/prevention & control , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Attenuated/immunology , Animals , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Newcastle disease virus/immunology , Poultry Diseases/immunology , Poultry Diseases/virology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology
6.
Avian Pathol ; 44(5): 408-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26223977

ABSTRACT

Polyvalent infectious bronchitis virus vaccination is common worldwide. The possibility of vaccine interference after simultaneous combined vaccination with Arkansas (Ark) and Massachusetts (Mass)-type vaccines was evaluated in an effort to explain the high prevalence of Ark-type infectious bronchitis virus in vaccinated chickens. Chickens ocularly vaccinated with combinations of Ark and Mass showed predominance of Mass vaccine virus before 9 days post-vaccination (DPV) in tears. Even when Mass and Ark vaccines were inoculated into separate eyes, Mass vaccine virus was able to outcompete Ark vaccine virus. Although Mass vaccine virus apparently had a replication advantage over Ark vaccine in ocular tissues, Ark vaccine virus appeared to have an advantage in spreading to and/or replicating in the trachea. When chickens vaccinated with Ark or Mass vaccine were housed together, Mass vaccine virus was able to spread to Ark-vaccinated chickens, but the Ark vaccine was not detected in Mass-vaccinated chickens. Only Mass vaccine was detected in tears of sentinel birds introduced into groups receiving both vaccines. Furthermore, Ark vaccine virus RNA was not detectable until 10 DPV in most tear samples from chickens vaccinated with both Ark and Mass vaccines at varying Ark vaccine doses, while high concentrations of Mass virus RNA were detectable at 3-7 DPV. In contrast, Ark vaccine virus replicated effectively early after vaccination in chickens vaccinated with Ark vaccine alone. The different replication dynamics of Ark and Mass viruses in chickens vaccinated with combined vaccines did not result in reduced protection against Ark challenge at 21 DPV. Further studies are needed to clarify if the viral interference detected determines differences in protection against challenge at other time points after vaccination.


Subject(s)
Coronavirus Infections/veterinary , Infectious bronchitis virus/immunology , Poultry Diseases/prevention & control , Vaccination , Viral Vaccines/immunology , Animals , Arkansas , Chickens , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Disease Models, Animal , Infectious bronchitis virus/genetics , Infectious bronchitis virus/isolation & purification , Infectious bronchitis virus/physiology , Massachusetts , Poultry Diseases/virology , RNA, Viral/isolation & purification , Sequence Analysis, DNA , Serogroup , Specific Pathogen-Free Organisms , Vaccines, Combined , Virus Replication
7.
Avian Dis ; 58(2): 279-86, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25055633

ABSTRACT

Infectious bronchitis virus (IBV) is a highly contagious coronavirus prevalent in all countries with an extensive poultry industry and continues to cause economic losses. IBV strains of the Ark serotype are highly prevalent in the Southeastern United States despite extensive vaccination. One explanation for this observation is the high genetic variability of IBV. In addition, IBV Ark-type vaccines may induce suboptimal mucosal immune responses, contributing to the prevalence and persistence of the Ark serotype. To test this hypothesis, chickens were ocularly vaccinated with a commercially available live attenuated IBV Ark-Delmarva Poultry Industry vaccine strain and both mucosal and systemic antibody responses were measured. The highest immunoglobulin A (IgA) spot-forming cell (SFC) response was observed in the Harderian glands (HG) and to a lesser extent in the spleen and conjunctiva-associated lymphoid tissues, while a limited IgG SFC response was observed in either the mucosal or systemic immune compartment. Interestingly, the peak IgA SFC response occurred 2 days earlier in spleen than in the head-associated lymphoid tissues despite ocular vaccination. Furthermore, IgA IBV-specific antibody levels significantly increased over controls 3 days earlier in tears and 4 days earlier in plasma than did IgG antibodies. IgA antibody levels were higher than IgG antibody levels throughout the primary response in tears and were similar in magnitude in plasma. In addition, a very early increase in IgA antibodies on day 3 postvaccination was observed in tears; such a response was not observed in plasma. This early increase is consistent with a mucosal T-independent IgA response to IBV. In the secondary response the IBV antibody levels significantly increased over controls starting on day 1 after boosting, and the IgG antibody levels were higher than the IgA antibody levels in both tears and plasma. In summary, ocular vaccination induced higher IgA antibodies in the primary IBV response, while the memory response is dominated by IgG antibodies. Thus, lower mucosal IgA antibody levels are observed upon secondary exposure to IBV, which may contribute to vulnerability of host epithelial cells to infection by IBV and persistence of the Ark serotype.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/immunology , Poultry Diseases/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/metabolism , Avian Proteins/metabolism , Coronavirus Infections/immunology , Enzyme-Linked Immunospot Assay/veterinary , Immunity, Humoral , Immunity, Mucosal , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Lymphoid Tissue/immunology , Specific Pathogen-Free Organisms , Spleen/immunology , Vaccination/veterinary , Vaccines, Attenuated/immunology
8.
Avian Dis ; 58(1): 83-9, 2014 03.
Article in English | MEDLINE | ID: mdl-24758118

ABSTRACT

We developed a recombinant Newcastle disease virus (NDV) LaSota (rLS) expressing the infectious bronchitis virus (IBV) S2 gene (rLS/IBV.S2). The recombinant virus showed somewhat-reduced pathogenicity compared to the parental lentogenic LaSota strain but effectively elicited hemagglutination inhibition antibodies against NDV and protected chickens against lethal challenge with virulent NDV/CA02. IBV heterotypic protection was assessed using a prime-boost approach with a commercially available attenuated IBV Massachusetts (Mass)-type vaccine. Specific-pathogen-free chickens primed ocularly with rLS/IBV.S2 at 4 days of age and boosted with Mass at 18 days of age were completely protected against challenge at 41 days of age with a virulent Ark-type strain. In a second experiment, we compared protection conferred by priming with rLS/IBV.S2 and boosting with Mass (rLS/IBV.S2+Mass) versus priming and boosting with Mass (Mass+Mass). We also modified the timing of vaccination to prime at 1 day of age and boost at 12 days of age. Challenge with virulent Ark was performed at 21 days of age. Based on clinical signs, both vaccinated groups appeared equally protected against challenge compared to unvaccinated challenged chickens. Viral loads in lachrymal fluids of birds receiving rLS/IBV.S2+Mass showed a clear tendency of improved protection compared to Mass+Mass; however, the difference did not achieve statistical significance. A significant difference (P < 0.05) was determined between these groups regarding incidence of detection of challenge IBV RNA in the trachea; viral RNA was detected in 50% of rLS/IBV.S2+Mass-vaccinated chickens while chickens vaccinated with Mass+Mass and unvaccinated challenged controls showed 84 and 90% incidence of IBV RNA detection in the trachea, respectively. These results demonstrate that overexposing the IBV S2 to the chicken immune system by means of a vectored vaccine, followed by boost with whole virus, protects chickens against IBV showing dissimilar S1.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/immunology , Newcastle disease virus/immunology , Poultry Diseases/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Hemagglutination Inhibition Tests/veterinary , Infectious bronchitis virus/pathogenicity , Newcastle disease virus/genetics , Polymerase Chain Reaction/veterinary , Poultry Diseases/immunology , Specific Pathogen-Free Organisms , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Attenuated/immunology , Viral Load/veterinary
9.
Avian Dis ; 58(1): 102-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24758121

ABSTRACT

Factors responsible for the persistence of Arkansas Delmarva Poultry Industry (ArkDPI)-derived infectious bronchitis vaccines in commercial flocks and the high frequency of isolation of ArkDPI-type infectious bronchitis viruses in respiratory cases are still unclear. We compared dynamics of vaccine viral subpopulations, viral loads, persistence in trachea and cloaca, and the magnitude of infectious bronchitis virus (1BV)-specific antibody induction after vaccination with two commercial ArkDPI-derived Arkansas (Ark) serotype vaccines. One of the vaccines (coded vaccine B) produced significantly higher vaccine virus heterogeneity in vaccinated chickens than the other vaccine (coded A). Chickens vaccinated with vaccine B had significantly higher viral loads in tears at 5 days postvaccination (DPV) than those vaccinated with vaccine A. Vaccine B also induced a significantly higher lachrymal immunoglobulin M response at 11 DPV, an earlier peak of IBV-specific lachrymal immunoglobulin A, and higher serum antibodies than vaccine A. In addition, a significantly higher proportion of birds vaccinated with vaccine B had vaccine virus detected in the trachea at 20 DPV than those vaccinated with vaccine A. Furthermore, the virus detected at 20 DPV in most of the chickens vaccinated with vaccine B was a single specific subpopulation (subpopulation 4) selected from multiple vaccine subpopulations detected earlier at 5 and 7 DPV in the same chickens. On the other hand, a higher proportion of chickens vaccinated with vaccine A had virus detected in cloacal swabs at 20 DPV. Thus we found differences in mucosal antibody induction and selection and persistence of vaccine viruses between two ArkDPI-derived vaccines from different manufacturers. The higher vaccine virus heterogeneity observed in chickens vaccinated with vaccine B compared with those vaccinated with vaccine A may be responsible for these differences. Thus the high frequency of Ark IBV viruses in the field may be due to the inherent ability of some ArkDPI-derived vaccine viruses to be selected and persist in vaccinated chickens. Vaccine virus persistence may offer genetic material for recombination or may undergo mutations with the potential to result in increased virulence.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/genetics , Poultry Diseases/immunology , Poultry Diseases/virology , Viral Vaccines/immunology , Animals , Antibodies, Viral/metabolism , Cloaca/immunology , Cloaca/pathology , Cloaca/virology , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Enzyme-Linked Immunosorbent Assay/veterinary , Immunoglobulin A/metabolism , Immunoglobulin M/metabolism , Infectious bronchitis virus/classification , Infectious bronchitis virus/isolation & purification , Male , Molecular Sequence Data , Poultry Diseases/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Analysis, DNA/veterinary , Sequence Analysis, Protein/veterinary , Specific Pathogen-Free Organisms , Spike Glycoprotein, Coronavirus/genetics , Tears/immunology , Tears/virology , Trachea/immunology , Trachea/pathology , Trachea/virology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Load/veterinary , Viral Vaccines/genetics
10.
Avian Dis ; 56(3): 449-55, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23050459

ABSTRACT

Conventional and molecular epidemiologic studies have confirmed the ability of infectious bronchitis virus (IBV) to rapidly evolve and successfully circumvent extensive vaccination programs implemented since the early 1950s. IBV evolution has often been explained as variation in gene frequencies as if evolution were driven by genetic drift alone. However, the mechanisms regulating the evolution of IBV include both the generation of genetic diversity and the selection process. IBV's generation of genetic diversity has been extensively investigated and ultimately involves mutations and recombination events occurring during viral replication. The relevance of the selection process has been further understood more recently by identifying genetic and phenotypic differences between IBV populations prior to, and during, replication in the natural host. Accumulating evidence suggests that multiple environmental forces within the host, including immune responses (or lack thereof) and affinity for cell receptors, as well as physical and biochemical conditions, are responsible for the selection process. Some scientists have used or adopted the related quasispecies frame to explain IBV evolution. The quasispecies frame, while providing a distinct explanation of the dynamics of populations in which mutation is a frequent event, exhibits relevant limitations which are discussed herein. Instead, it seems that IBV populations evolving by the generation of genetic variability and selection on replicons follow the evolutionary mechanisms originally proposed by Darwin. Understanding the mechanisms underlying the evolution of IBV is of basic relevance and, without doubt, essential to appropriately control and prevent the disease.


Subject(s)
Biological Evolution , Genetic Variation , Infectious bronchitis virus/genetics , Selection, Genetic
11.
Avian Dis ; 56(3): 501-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23050466

ABSTRACT

Infectious bronchitis coronavirus (IBV) shows extensive genotypic and phenotypic variability. The evolutionary process involves generation of genetic diversity by mutations and recombination followed by replication of those phenotypes favored by selection. In the current study, we examined changes occurring in a wild Arkansas (Ark) challenge strain in chickens that were vaccinated either ocularly with commercially available attenuated ArkDPI-derived vaccines or in ovo with a replication-defective recombinant adenovirus expressing a codon-optimized IBV Ark S1 gene (AdArkIBV.S1(ck)). Commercial IBV Ark vaccines A, B, and C provided slightly differing levels of protection against homologous challenge. Most importantly for the current study, chickens vaccinated with the different vaccines displayed significant differences in specific B-lymphocyte responses in the Harderian gland (i.e., the challenge virus encountered differing immune selective pressure during invasion among host groups). Based on S1 sequences, five predominant populations were found in different individual vaccinated/challenged chickens. Chickens with the strongest immune response (vaccine A) were able to successfully impede replication of the challenge virus in most chickens, and only the population predominant in the challenge strain was detected in a few IBV-positive birds. In contrast, in chickens showing less than optimal specific immune responses (vaccines B and C) IBV was detected in most chickens, and populations different from the predominant one in the challenge strain were selected and became predominant. These results provide scientific evidence for the assumption that poor vaccination contributes to the emergence of new IBV strains via mutation and/or selection. In ovo vaccination with a low dose of AdArkIBV.S1(ck) resulted in a mild increase of systemic antibody and reduced viral shedding but no protection against IBV signs and lesions. Under these conditions we detected only virus populations identical to the challenge virus. Possible explanations are discussed. From a broad perspective, these results indicate that selection is an important force driving IBV evolution.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/classification , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Immunoglobulin G/blood , Infectious bronchitis virus/genetics , Trachea/pathology
12.
Avian Pathol ; 41(5): 451-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22897690

ABSTRACT

We followed changes in a portion of the S1 gene sequence of the dominant populations of an infectious bronchitis virus (IBV) Arkansas (Ark) vaccine strain during serial passage in chickens infected with the immunosuppressive chicken anaemia virus (CAV) and/or infectious bursal disease virus (IBDV) as well as in immunocompetent chickens. The IBV-Ark vaccine was applied ocularly and tears were collected from infected chickens for subsequent ocular inoculation in later passages. The experiment was performed twice. In both experiments the dominant S1 genotype of the vaccine strain was rapidly and negatively selected in all chicken groups (CAV, IBDV, CAV+IBDV and immunocompetent). Based on the S1 genotype, the same IBV subpopulations previously reported in immunocompetent chickens and named component (C) 1 to C5 emerged both in immunocompetent and immunodeficient chickens. During the first passage different subpopulations emerged, followed by the establishment of one or two predominant populations after further passages. Only when the subpopulation designated C2 became established in either CAV-infected or IBDV-infected chickens was IBV maintained for more than four passages. These results indicate that selection does not cease in immunodeficient chickens and that phenotype C2 may show a distinct adaptation to this environment. Subpopulations C1 or C4 initially became established in immunocompetent birds but became extinct after only a few succeeding passages. A similar result was observed in chickens co-infected with CAV+IBDV. These results suggest that the generation of genetic diversity in IBV is constrained. This finding constitutes further evidence for phenotypic drift occurring mainly as a result of selection.


Subject(s)
Chicken anemia virus/physiology , Genetic Drift , Infectious bronchitis virus/physiology , Infectious bursal disease virus/physiology , Poultry Diseases/virology , Virus Replication , Animals , Antibodies, Viral/blood , Birnaviridae Infections/immunology , Birnaviridae Infections/veterinary , Birnaviridae Infections/virology , Chicken anemia virus/immunology , Chickens , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Genetic Variation , Genotype , Host-Pathogen Interactions , Immunocompromised Host , Infectious bronchitis virus/genetics , Infectious bronchitis virus/isolation & purification , Infectious bursal disease virus/immunology , Poultry Diseases/immunology , Poultry Diseases/prevention & control , RNA, Viral/genetics , Selection, Genetic , Sequence Analysis, DNA , Specific Pathogen-Free Organisms , Spike Glycoprotein, Coronavirus/genetics , Tears/virology , Viral Vaccines/immunology
13.
Avian Dis ; 56(4): 642-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23397834

ABSTRACT

We investigated the significance of differing proportions of specific subpopulations among commercial Arkansas (Ark) Delmarva poultry industry (DPI) vaccines with regard to vaccination outcome. Two ArkDPI-derived vaccines that contain a higher proportion of viruses with S1 genes that become selected during replication in chickens exhibited more rapid establishment of those selected subpopulations in chickens, produced significantly higher viral loads in tears, and induced higher antibody responses compared with two other ArkDPI vaccines with lower proportions of viruses that become selected in chickens. The presence of higher proportions of selected subpopulations was also associated with a significantly higher incidence of respiratory signs early after vaccination and in some cases more severe tracheal lesions. However, one of the ArkDPI-derived vaccines with a lower proportion of selected subpopulations, despite producing a lower viral load in tears, also induced a higher incidence of respiratory signs later after vaccination and more severe tracheal lesions. Furthermore, one of the ArkDPI-derived vaccines with a higher proportion of selected subpopulations, despite producing a higher viral loads in tears, resulted in less severe tracheal damage. These discrepancies suggest that infectious bronchitis virus (IBV) load in tears may not always predict degree of tracheal damage and that phenotypic characteristics other than S1 may also be involved in severity of vaccine reactions following ArkDPI vaccine administration. We observed lower antibody responses to the vaccines that produced lower viral loads, which might contribute to the persistence of Ark serotype IBV vaccines observed in commercial flocks.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/genetics , Poultry Diseases/immunology , Poultry Diseases/virology , Viral Vaccines/immunology , Animals , Antibodies, Viral/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Harderian Gland/virology , Immunoglobulin G/metabolism , Infectious bronchitis virus/classification , Infectious bronchitis virus/isolation & purification , Interferon-gamma/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Molecular Sequence Data , Poultry Diseases/pathology , RNA, Messenger/genetics , Respiratory System/immunology , Respiratory System/pathology , Respiratory System/virology , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Analysis, DNA/veterinary , Sequence Analysis, Protein/veterinary , Specific Pathogen-Free Organisms , Spike Glycoprotein, Coronavirus , Tears/virology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Load/veterinary , Viral Vaccines/genetics
14.
Expert Rev Vaccines ; 10(11): 1539-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22043954

ABSTRACT

The disease-fighting power of vaccines has been a public health bonanza credited with the worldwide reduction of mortality and morbidity. The goal to further amplify its power by boosting vaccine coverage requires the development of a new generation of rapid-response vaccines that can be mass produced at low costs and mass administered by nonmedical personnel. The new vaccines also have to be endowed with a higher safety margin than that of conventional vaccines. The nonreplicating adenovirus-vectored vaccine holds promise in boosting vaccine coverage because the vector can be rapidly manufactured in serum-free suspension cells in response to a surge in demand, and noninvasively administered by nasal spray into human subjects in compliance with evolutionary medicine. In contrast to parenteral injection, noninvasive mucosal vaccination minimizes systemic inflammation. Moreover, pre-existing adenovirus immunity does not interfere appreciably with the potency of an adenovirus-vectored nasal vaccine. Nasal administration of adenovirus vectors encoding pathogen antigens is not only fear-free and painless, but also confers rapid and sustained protection against mucosal pathogens as a drug-vaccine duo since adenovirus particles alone without transgene expression can induce an anti-influenza state in the airway. In addition to human vaccination, animals can also be mass immunized by this class of vectored vaccines.


Subject(s)
Adenoviruses, Human/genetics , Drug Carriers/administration & dosage , Drug Delivery Systems/methods , Genetic Vectors , Vaccination/methods , Vaccines/administration & dosage , Vaccines/immunology , Administration, Intranasal , Humans
15.
Avian Dis ; 55(2): 255-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21793442

ABSTRACT

Even though males represent only 8%-12% of the birds of a breeder flock, their role in infectious bronchitis virus (IBV) dissemination is largely unknown. We first assessed the effect of IBV replication in the chicken testes. Ten-week-old males were inoculated with Arkansas (Ark) or Massachusetts (Mass) IBV virulent strains. Seven days postinoculation (DPI) IBV RNA was detected in testicles of 100% of M41- and in 96% of Ark-infected males. Marginal nonsynonymous variation was detected in spike (S) gene of the predominant population of IBV replicating in the testes compared to the S gene of the predominant population of viruses prior to inoculation. IBV M41 and Ark were detected in spermatogonia and Sertoli cells of testicles of infected roosters by immunofluorescence, without evident histopathological changes. We next assessed venereal transmission of IBV by artificially inseminating 54-wk-old hens either with semen from IBV-infected roosters or with IBV suspended in naïve semen. IBV RNA was detected in the trachea of all hens inseminated with IBV-spiked semen and in 50% of hens inseminated with semen from IBV-infected males. The egg internal and external quality was negatively affected in hens inseminated with semen containing IBV. These results provide experimental evidence for IBV venereal transmission.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/isolation & purification , Poultry Diseases/virology , Sexually Transmitted Diseases, Viral/veterinary , Testis/virology , Animals , Coronavirus Infections/transmission , Coronavirus Infections/virology , Female , Male , Ovum/virology , Poultry Diseases/transmission , RNA, Viral/isolation & purification , Sexually Transmitted Diseases, Viral/transmission , Spermatogonia/virology , Trachea/virology
16.
Avian Dis ; 55(2): 285-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21793447

ABSTRACT

Protective immunity against avian influenza (AI) can be elicited in chickens in a single-dose regimen by in ovo vaccination with a replication-competent adenovirus (RCA)-free human adenovirus serotype 5 (Ad)-vector encoding the AI virus (AIV) hemagglutinin (HA). We evaluated vaccine potency, antibody persistence, transfer of maternal antibodies (MtAb), and interference between MtAb and active in ovo or mucosal immunization with RCA-free recombinant Ad expressing a codon-optimized AIV H5 HA gene from A/turkey/WI/68 (AdTW68.H5(ck)). Vaccine coverage and intrapotency test repeatability were based on anti-H5 hemagglutination inhibition (HI) antibody levels detected in in ovo vaccinated chickens. Even though egg inoculation of each replicate was performed by individuals with varying expertise and with different vaccine batches, the average vaccine coverage of three replicates was 85%. The intrapotency test repeatability, which considers both positive as well as negative values, varied between 0.69 and 0.71, indicating effective vaccination. Highly pathogenic (HP) AIV challenge of chicken groups vaccinated with increasing vaccine doses showed 90% protection in chickens receiving > or = 10(8) ifu (infectious units)/bird. The protective dose 50% (PD50) was determined to be 10(6.5) ifu. Even vaccinated chickens that did not develop detectable antibody levels were effectively protected against HP AIV challenge. This result is consistent with previous findings ofAd-vector eliciting T lymphocyte responses. Higher vaccine doses significantly reduced viral shedding as determined by AIV RNA concentration in oropharyngeal swabs. Assessment of antibody persistence showed that antibody levels of in ovo immunized chickens continued to increase until 12 wk and started to decline after 18 wk of age. Intramuscular (IM) booster vaccination with the same vaccine at 16 wk of age significantly increased the antibody responses in breeder hens, and these responses were maintained at high levels throughout the experimental period (34 wk of age). AdTW68.H5(ch)-immunized breeder hens effectively transferred MtAb to progeny chickens. The level of MtAb in the progenies was consistent with the levels detected in the breeders, i.e., intramuscularly boosted breeders transferred higher concentrations of antibodies to the offspring. Maternal antibodies declined with time in the progenies and achieved marginal levels by 34 days of age. Chickens with high maternal antibody levels that were vaccinated either in ovo or via mucosal routes (ocular or spray) did not seroconvert. In contrast, chickens without MtAb successfully developed specific antibody levels after either in ovo or mucosal vaccination. These results indicate that high levels of MtAb interfered with active Ad-vectored vaccination.


Subject(s)
Adenoviridae/genetics , Antibodies, Viral/blood , Chickens , Immunity, Maternally-Acquired , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Aging , Animals , Influenza Vaccines/administration & dosage , Ovum , Vaccines, Synthetic/immunology , Virus Shedding
17.
Avian Dis ; 55(1): 43-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21500634

ABSTRACT

We evaluated protection conferred by mucosal vaccination with replication-competent adenovirus-free recombinant adenovirus expressing a codon-optimized avian influenza (AI) H5 gene from A/turkey/WI/68 (AdTW68.H5ck). Commercial, layer-type chicken groups were either singly vaccinated ocularly at 5 days of age, singly vaccinated via spray at 5 days of age, or ocularly primed at 5 days and ocularly boosted at 15 days of age. Only chickens primed and boosted via the ocular route developed AI systemic antibodies with maximum hemagglutination inhibition mean titers of 3.9 log2 at 32 days of age. In contrast, single vaccination via the ocular or spray routes maintained an antibody status similar to unvaccinated controls. All chickens (16/16) subjected to ocular priming and boosting with AdTW68.H5ck survived challenge with highly pathogenic AI virus A/chicken/Queretaro/14588-19/95 (H5N2). Single ocular vaccination resulted in 63% (10/16) of birds surviving the challenge followed by a 44% (7/16) survival of single-sprayed vaccinated birds. Birds vaccinated twice via the ocular route also showed significantly lower (P < 0.05) AI virus RNA concentrations in oropharyngeal swabs compared to unvaccinated-challenged controls.


Subject(s)
Adenoviridae/genetics , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Vaccines, DNA/immunology , Aging , Animals , Antibodies, Viral/blood , Drug Administration Routes , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Immunization, Secondary , Influenza Vaccines/administration & dosage , Vaccines, DNA/administration & dosage , Virus Replication/genetics
19.
Avian Dis ; 54(2): 807-13, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20608523

ABSTRACT

Arkansas (Ark)-type infectious bronchitis virus (IBV) subpopulations with an S gene sequence distinct from the vaccine predominant consensus were previously found in the upper respiratory tract of chickens within 3 days after inoculation. This finding indicated that a distinct virus subpopulation was rapidly positively selected by the chicken upper respiratory tract. We hypothesized that during host invasion, the replicating IBV population further changes as it confronts the distinct environments of different tissues, leading to selection of the most fit population. We inoculated 15-day-old chickens with 10(4) 50% embryo infective doses of an Ark-type IBV commercial vaccine via the ocular and nasal routes and characterized the sequences of the S1 gene of IBV contained in tear fluid, trachea, and reproductive tract of individual chickens at different times postinoculation. The predominant IBV phenotype contained in the vaccine (before inoculation) became a minor or nondetectable population at all times in all tissues after replication in the majority of the chickens, corroborating our previous findings. Five new predominant populations designated component (C) 1 through C5, showing distinct nonsynonymous changes, i.e., nucleotide changes resulting in different amino acids encoded and thus in a phenotypic change of the predominant virus population, were detected in the tissues or fluids of individual vaccinated chickens. Due to the different biochemical properties of some amino acids that changed in the S1 glycoprotein, we anticipate that phenotypic shift occurred during the invasion process. Significant differences were detected in the incidence of some distinct IBV predominant populations in tissues and fluids; e.g., phenotype C1 showed the highest incidence in the reproductive tract of the chickens, achieving a significant difference versus its incidence in the trachea (P < 0.05). These results indicate for the first time that IBV undergoes intraspatial variation during host invasion, i.e., the dominant genotype/phenotype further changes during host invasion as the microenvironment of distinct tissues exerts selective pressure on the replicating virus population.


Subject(s)
Chickens , Coronavirus Infections/veterinary , Infectious bronchitis virus/genetics , Poultry Diseases/virology , Selection, Genetic , Animals , Coronavirus Infections/virology , Female , Host-Pathogen Interactions , Male , Oviducts/virology , Specific Pathogen-Free Organisms , Tears/virology , Testis/virology , Time Factors , Trachea/virology
20.
Avian Dis ; 54(1 Suppl): 224-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20521636

ABSTRACT

Protective immunity to avian influenza (AI) virus can be elicited in chickens by in ovo or intramuscular vaccination with replication-competent adenovirus (RCA)-free human recombinant adenovirus serotype 5 (Ad5) encoding AI virus H5 (AdTW68.H5) or H7 (AdCN94.H7) hemagglutinins. We evaluated bivalent in ovo vaccination with AdTW68.H5 and AdCN94.H7 and determined that vaccinated chickens developed robust hemagglutination inhibition (HI) antibody levels to both H5 and H7 AI strains. Additionally, we evaluated immune responses of 1-day-old chickens vaccinated via spray with AdCN94.H7. These birds showed increased immunoglobulin A responses in lachrymal fluids and increased interleukin-6 expression in Harderian gland-derived lymphocytes. However, specific HI antibodies were not detected in the sera of these birds. Because pigs might play a role as a "mixing vessel" for the generation of pandemic influenza viruses we explored the use of RCA-free adenovirus technology to immunize pigs against AI virus. Weanling piglets vaccinated intramuscularly with a single dose of RCA-free AdTW68.H5 developed strong systemic antibody responses 3 wk postvaccination. Intranasal application of AdTW68.H5 in piglets resulted in reduced vaccine coverage, i.e., 33% of pigs (2/6) developed an antibody response, but serum antibody levels in those successfully immunized animals were similar to intramuscularly vaccinated animals.


Subject(s)
Adenoviridae , Chickens , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Poultry Diseases/prevention & control , Swine Diseases/prevention & control , Animals , Antibodies, Viral , Humans , Influenza A Virus, H5N1 Subtype , Influenza Vaccines/administration & dosage , Ovum/immunology , Recombinant Proteins/immunology , Specific Pathogen-Free Organisms , Swine , Vaccines, Synthetic/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...