Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Am J Vet Res ; 85(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38422620

ABSTRACT

OBJECTIVE: To determine the efficacy of primary or booster intranasal vaccination of beef steers on clinical protection and pathogen detection following simultaneous challenge with bovine respiratory syncytial virus and bovine herpes virus 1. METHODS: 30 beef steers were randomly allocated to 3 different treatment groups starting at 2 months of age. Group A (n = 10) was administered a single dose of a parenteral modified-live vaccine and was moved to a separate pasture. Groups B (n = 10) and C (10) remained unvaccinated. At 6 months of age, all steers were weaned and transported. Subsequently, groups A and B received a single dose of an intranasal modified-live vaccine vaccine while group C remained unvaccinated. Group C was housed separately until challenge. Two days following vaccination, all steers were challenged with bovine respiratory syncytial virus and bovine herpes virus 1 and housed in a single pen. Clinical and antibody response outcomes and the presence of nasal pathogens were evaluated. RESULTS: The odds of clinical disease were lower in group A compared with group C on day 7 postchallenge; however, antibody responses and pathogen detection were not significantly different between groups before and following viral challenge. All calves remained negative for Histophilus somni and Mycoplasma bovis; however, significantly greater loads of Mannheimia haemolytica and Pasteurella multocida were detected on day 7 postchallenge compared with day -2 prechallenge. CLINICAL RELEVANCE: Intranasal booster vaccination of beef steers at 6 months of age reduced clinical disease early after viral challenge. Weaning, transport, and viral infection promoted increased detection rates of M haemolytica and P multocida regardless of vaccination status.


Subject(s)
Administration, Intranasal , Coinfection , Herpesvirus 1, Bovine , Immunization, Secondary , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Bovine , Animals , Cattle , Herpesvirus 1, Bovine/immunology , Male , Administration, Intranasal/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Immunization, Secondary/veterinary , Coinfection/veterinary , Coinfection/prevention & control , Coinfection/microbiology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Infections/prevention & control , Infectious Bovine Rhinotracheitis/prevention & control , Infectious Bovine Rhinotracheitis/immunology , Cattle Diseases/prevention & control , Cattle Diseases/microbiology , Cattle Diseases/virology , Cattle Diseases/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Bacterial Shedding , Antibodies, Viral/blood , Herpesviridae Infections/veterinary , Herpesviridae Infections/prevention & control , Random Allocation , Vaccination/veterinary
2.
Microbiol Resour Announc ; 12(10): e0040523, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37750692

ABSTRACT

We report the complete genome sequences of seven virulent Newcastle disease viruses (NDVs) that were isolated from chickens from live bird markets in the Arusha, Iringa, Mbeya, and Tanga regions of Tanzania in 2012. Phylogenetic analysis revealed that all isolates belong to sub-genotype XIII.1.1.

3.
Front Vet Sci ; 10: 1026296, 2023.
Article in English | MEDLINE | ID: mdl-36742982

ABSTRACT

Newcastle disease virus (NDV) infects a wide range of bird species worldwide and is of importance to the poultry industry. Although certain virus genotypes are clearly associated with wild bird species, the role of those species in the movement of viruses and the migratory routes they follow is still unclear. In this study, we performed a phylogenetic analysis of nineteen NDV sequences that were identified among 21,924 samples collected from wild and synanthropic birds from different regions of Ukraine from 2006 to 2015 and compared them with isolates from other continents. In synanthropic birds, NDV strains of genotype II, VI, VII, and XXI of class II were detected. The fusion gene sequences of these strains were similar to strains detected in birds from different geographical regions of Europe and Asia. However, it is noteworthy to mention the isolation of vaccine viruses from synanthropic birds, suggesting the possibility of their role in viral transmission from vaccinated poultry to wild birds, which may lead to the further spreading of vaccine viruses into other regions during wild bird migration. Moreover, here we present the first publicly available complete NDV F gene from a crow (genus Corvus). Additionally, our phylogenetic results indicated a possible connection of Ukrainian NDV isolates with genotype XXI strains circulating in Kazakhstan. Among strains from wild birds, NDVs of genotype 1 of class I and genotype I of class II were detected. The phylogenetic analysis highlighted the possible exchange of these NDV strains between wild waterfowl from the Azov-Black Sea region of Ukraine and waterfowl from different continents, including Europe, Asia, and Africa.

4.
Viruses ; 14(12)2022 11 25.
Article in English | MEDLINE | ID: mdl-36560631

ABSTRACT

Newcastle disease (ND) is endemic in poultry in Bangladesh. We performed genotypic and pathotypic characterization of four ND virus (NDV) isolates from recent outbreaks in broiler chickens in Bangladesh during the period of 2020-2021. Phylogenetic analysis based on the complete fusion protein gene coding sequences classified the viruses into NDV class II genotype VII.2 together with viruses from Indonesia isolated between 2014 and 2021 and a single 2020 Indian isolate. Pathogenicity testing using the intracerebral pathogenicity index in day-old chickens and mean embryo death time in embryonating chicken eggs revealed that the Bangladeshi isolates are velogenic. Inoculation of 35-day-old chickens with two NDV isolates (LT67 and N5) resulted in 100% morbidity by 3 days post inoculation (DPI), and all birds succumbed to infection by 7 DPI. Massive hemorrhages, congestion and necrotic lesions were observed in different visceral organs, which were typical for infection with a velogenic viscerotropic pathotype of NDV. At microscopic examination, tracheitis, severe pneumonia, focal proventriculitis, transmural enteritis, focal myocarditis, severe congestion and necrosis in kidneys, and lymphoid depletion in lymphoid tissues were found. Our study reports the first outbreak of the panzootic genotype VII.2 NDV in poultry in Bangladesh and documents a possible recent re-introduction of this NDV genotype from Southeast or East Asia. This study further provides viral distribution and epidemiological data that can facilitate the effective control of NDV.


Subject(s)
Newcastle Disease , Poultry Diseases , Animals , Newcastle disease virus , Chickens , Phylogeny , Bangladesh/epidemiology , Poultry Diseases/epidemiology , Genotype , Disease Outbreaks/veterinary
5.
Appl Environ Microbiol ; 88(11): e0046622, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35612300

ABSTRACT

Avian paramyxoviruses (APMVs) (subfamily Avulavirinae) have been isolated from over 200 species of wild and domestic birds around the world. The International Committee on Taxonomy of Viruses (ICTV) currently defines 22 different APMV species, with Avian orthoavulavirus 1 (whose viruses are designated APMV-1) being the most frequently studied due to its economic burden to the poultry industry. Less is known about other APMV species, including limited knowledge on the genetic diversity in wild birds, and there is a paucity of public whole-genome sequences for APMV-2 to -22. The goal of this study was to use MinION sequencing to genetically characterize APMVs isolated from wild bird swab samples collected during 2016 to 2018 in the United States. Multiplexed MinION libraries were prepared using a random strand-switching approach using 37 egg-cultured, influenza-negative, hemagglutination-positive samples. Forty-one APMVs were detected, with 37 APMVs having complete polymerase coding sequences allowing for species identification using ICTV's current Paramyxoviridae phylogenetic methodology. APMV-1, -4, -6, and -8 viruses were classified, one putative novel species (Avian orthoavulavirus 23) was identified from viruses isolated in this study, two putative new APMV species (Avian metaavulavirus 24 and 27) were identified from viruses isolated in this study and from retrospective GenBank sequences, and two putative new APMV species (Avian metaavulavirus 25 and 26) were identified solely from retrospective GenBank sequences. Furthermore, coinfections of APMVs were identified in four samples. The potential limitations of the branch length being the only species identification criterion and the potential benefit of a group pairwise distance analysis are discussed. IMPORTANCE Most species of APMVs are understudied and/or underreported, and many species were incidentally identified from asymptomatic wild birds; however, the disease significance of APMVs in wild birds is not fully determined. The rapid rise in high-throughput sequencing coupled with avian influenza surveillance programs have identified 12 different APMV species in the last decade and have challenged the resolution of classical serological methods to identify new viral species. Currently, ICTV's only criterion for Paramyxoviridae species classification is the requirement of a branch length of >0.03 using a phylogenetic tree constructed from polymerase (L) amino acid sequences. The results from this study identify one new APMV species, propose four additional new APMV species, and highlight that the criterion may have insufficient resolution for APMV species demarcation and that refinement or expansion of this criterion may need to be established for Paramyxoviridae species identification.


Subject(s)
Animals, Wild , Avulavirus Infections , Avulavirus , Bird Diseases , Animals , Animals, Wild/virology , Avulavirus/genetics , Avulavirus/isolation & purification , Avulavirus Infections/epidemiology , Avulavirus Infections/veterinary , Avulavirus Infections/virology , Bird Diseases/epidemiology , Bird Diseases/virology , Birds , Phylogeny , Retrospective Studies , Sentinel Surveillance/veterinary , United States/epidemiology
6.
Vaccines (Basel) ; 9(10)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34696297

ABSTRACT

Newcastle disease (ND) is one of the most economically important poultry diseases. Despite intensive efforts with current vaccination programs, this disease still occurs worldwide, causing significant mortality even in vaccinated flocks. This has been partially attributed to a gap in immunity during the post-hatch period due to the presence of maternal antibodies that negatively impact the replication of the commonly used live vaccines. In ovo vaccines have multiple advantages and present an opportunity to address this problem. Currently employed in ovo ND vaccines are recombinant herpesvirus of turkeys (HVT)-vectored vaccines expressing Newcastle disease virus (NDV) antigens. Although proven efficient, these vaccines have some limitations, such as delayed immunogenicity and the inability to administer a second HVT vaccine post-hatch. The use of live ND vaccines for in ovo vaccination is currently not applicable, as these are associated with high embryo mortality. In this study, recombinant NDV-vectored experimental vaccines containing an antisense sequence of avian interleukin 4 (IL4R) and their backbones were administered in ovo at different doses in 18-day-old commercial eggs possessing high maternal antibodies titers. The hatched birds were challenged with virulent NDV at 2 weeks-of-age. Post-hatch vaccine shedding, post-challenge survival, challenge virus shedding, and humoral immune responses were evaluated at multiple timepoints. Recombinant NDV (rNDV) vaccinated birds had significantly reduced post-hatch mortality compared with the wild-type LaSota vaccine. All rNDV vaccines were able to penetrate maternal immunity and induce a strong early humoral immune response. Further, the rNDV vaccines provided protection from clinical disease and significantly decreased virus shedding after early virulent NDV challenge at two weeks post-hatch. The post-challenge hemagglutination-inhibition antibody titers in the vaccinated groups remained comparable with the pre-challenge titers, suggesting the capacity of the studied vaccines to prevent efficient replication of the challenge virus. Post-hatch survival after vaccination with the rNDV-IL4R vaccines was dose-dependent, with an increase in survival as the dose decreased. This improved survival and the dose-dependency data suggest that novel attenuated in ovo rNDV-based vaccines that are able to penetrate maternal immunity to elicit a strong immune response as early as 14 days post-hatch, resulting in high or full protection from virulent challenge, show promise as a contributor to the control of Newcastle disease.

7.
Vaccines (Basel) ; 9(9)2021 Aug 26.
Article in English | MEDLINE | ID: mdl-34579191

ABSTRACT

In ovo vaccination has been employed by the poultry industry for over 20 years to control numerous avian diseases. Unfortunately, in ovo live vaccines against Newcastle disease have significant limitations, including high embryo mortality and the inability to induce full protection during the first two weeks of life. In this study, a recombinant live attenuated Newcastle disease virus vaccine containing the antisense sequence of chicken interleukin 4 (IL-4), rZJ1*L-IL4R, was used. The rZJ1*L-IL4R vaccine was administered in ovo to naïve specific pathogen free embryonated chicken eggs (ECEs) and evaluated against a homologous challenge. Controls included a live attenuated recombinant genotype VII vaccine based on the virus ZJ1 (rZJ1*L) backbone, the LaSota vaccine and diluent alone. In the first of two experiments, ECEs were vaccinated at 18 days of embryonation (DOE) with either 104.5 or 103.5 50% embryo infectious dose (EID50/egg) and chickens were challenged at 21 days post-hatch (DPH). In the second experiment, 103.5 EID50/egg of each vaccine was administered at 19 DOE, and chickens were challenged at 14 DPH. Chickens vaccinated with 103.5 EID50/egg of rZJ1*L-IL4R had hatch rates comparable to the group that received diluent alone, whereas other groups had significantly lower hatch rates. All vaccinated chickens survived challenge without displaying clinical disease, had protective hemagglutination inhibition titers, and shed comparable levels of challenge virus. The recombinant rZJ1*L-IL4R vaccine yielded lower post-vaccination mortality rates compared with the other in ovo NDV live vaccine candidates as well as provided strong protection post-challenge.

8.
Viruses ; 13(8)2021 08 01.
Article in English | MEDLINE | ID: mdl-34452385

ABSTRACT

Newcastle disease virus (NDV) is a significant pathogen of poultry; however, variants also affect other species, including pigeons. While NDV is endemic in Bangladesh, and poultry isolates have been recently characterized, information about viruses infecting pigeons is limited. Worldwide, pigeon-derived isolates are commonly of low to moderate virulence for chickens. Here, we studied a pigeon-derived NDV isolated in Bangladesh in 2010. To molecularly characterize the isolate, we sequenced its complete fusion gene and performed a comprehensive phylogenetic analysis. We further studied the biological properties of the virus by estimating mean death time (MDT) and by experimentally infecting 5-week-old naïve Sonali chickens. The studied virus clustered in sub-genotype XXI.1.2 with NDV from pigeons from Pakistan isolated during 2014-2018. Deduced amino acid sequence analysis showed a polybasic fusion protein cleavage site motif, typical for virulent NDV. The performed in vivo pathogenicity testing showed a MDT of 40.8 h, and along with previously established intracerebral pathogenicity index of 1.51, these indicated a velogenic pathotype for chickens, which is not typical for pigeon-derived viruses. The experimental infection of chickens resulted in marked neurological signs and high mortality starting at 7 days post infection (dpi). Mild congestion in the thymus and necrosis in the spleen were observed at an advanced stage of infection. Microscopically, lymphoid depletion in the thymus, spleen, and bursa of Fabricius were found at 5 dpi, which progressed to severe in the following days. Mild to moderate proliferation of glial cells was noticed in the brain starting at 2 dpi, which gradually progressed with time, leading to focal nodular aggregation. This study reports the velogenic nature for domestic chickens of a pigeon-derived NDV isolate of sub-genotype XXI.1.2. Our findings show that not all pigeon-derived viruses are of low virulence for chickens and highlight the importance of biologically evaluating the pathogenicity of NDV isolated from pigeons.


Subject(s)
Chickens/virology , Columbidae/virology , Newcastle Disease/mortality , Newcastle disease virus/genetics , Newcastle disease virus/pathogenicity , Poultry Diseases/mortality , Animals , Bangladesh , Eggs/virology , Genome, Viral , Genotype , Newcastle disease virus/classification , Newcastle disease virus/isolation & purification , Phylogeny , Poultry Diseases/virology , Sequence Analysis, DNA , Virulence
9.
J Gen Virol ; 102(3)2021 03.
Article in English | MEDLINE | ID: mdl-33507145

ABSTRACT

Newcastle disease virus (NDV) is endemic in Bangladesh and is a major threat to commercial poultry operations. While complete fusion (F) genes are recommended for molecular characterization and classification of NDV isolates, heretofore, only partial F gene data have been available for Bangladeshi NDVs. To this end, we obtained the full-length F gene coding sequences of 11 representative NDVs isolated in Bangladesh between 2010 and 2017. In addition, one of the viruses (MK934289/chicken/Bangladesh/C161/2010) was used in an experimental infection of chickens to establish the viral pathotype and study gross and microscopic lesions. Phylogenetic analysis provided evidence that all studied Bangladeshi isolates belong to genotype XIII.2 of class II NDVs. Six of the viruses were isolated between 2010 and 2017 and grouped together with isolates from neighbouring India during 2013-2016. Another four Bangladeshi isolates (2010-2016) formed a separate monophyletic branch within XIII.2 and showed high nucleotide distance from the isolates from India and the other six Bangladeshi viruses within the sub-genotype; however, none of these groups fulfils all classification criteria to be named as a separate sub-genotype. The eleventh Bangladeshi virus studied here (C162) was genetically more distant from the remaining isolates. It out-grouped the viruses from sub-genotypes XIII.2.1 and XIII.2.2 and showed more than 9.5 % nucleotide distance from all genotype XIII sub-genotypes. This isolate may represent an NDV variant that is evolving independently from the other viruses in the region. The experimental infection in chickens revealed that the tested isolate (C161) is a velogenic viscerotropic virus. Massive haemorrhages, congestion and necrosis in different visceral organs, and lymphoid depletion in lymphoid tissues, typical for infection with velogenic NDV, were observed. Our findings demonstrate the endemic circulation of sub-genotype XIII.2 in Southcentral Asia and further genetic diversification of these viruses in Bangladesh and neighbouring India. This constant evolution of the viruses may lead to the establishment of new genetic groups in the region. Additional historical and prospective virus and surveillance data from the region and neighbouring countries will allow a more detailed epidemiological inference.


Subject(s)
Genetic Variation , Newcastle Disease/virology , Newcastle disease virus/genetics , Animals , Asia , Bangladesh/epidemiology , Chickens/virology , Evolution, Molecular , Genotype , India , Lung/pathology , Newcastle Disease/epidemiology , Newcastle Disease/pathology , Newcastle disease virus/classification , Newcastle disease virus/isolation & purification , Newcastle disease virus/pathogenicity , Phylogeny , Poultry Diseases/epidemiology , Poultry Diseases/pathology , Poultry Diseases/virology , RNA, Viral/genetics , Virulence
10.
Viruses ; 12(4)2020 03 26.
Article in English | MEDLINE | ID: mdl-32224965

ABSTRACT

Newcastle disease is an important poultry disease that also affects Columbiform birds. The viruses adapted to pigeons and doves are referred to as pigeon paramyxoviruses 1 (PPMV-1). PPMV-1 are frequently isolated from pigeons worldwide and have the potential to cause disease in chickens. The complete genomes of 18 PPMV-1 isolated in China during 2012-2018 were sequenced by next-generation sequencing (NGS). Comprehensive phylogenetic analyses showed that five of the viruses belong to sub-genotype VI1.2.1.1.2.1 and 13 isolates belong to sub-genotype VI.2.1.1.2.2. The results demonstrate that these sub-genotypes have been predominant in China during the last decade. The viruses of these sub-genotypes have been independently maintained and continuously evolved for over 20 years, and differ significantly from those causing outbreaks worldwide during the 1980s to 2010s. The viral reservoir remains unknown and possibilities of the viruses being maintained in both pigeon farms and wild bird populations are viable. In vivo characterization of the isolates' pathogenicity estimated mean death times between 62 and 114 hours and intracerebral pathogenicity indices between 0.00 and 0.63. Cross-reactivity testing showed minor antigenic differences between the studied viruses and the genotype II LaSota vaccine. These data will facilitate PPMV-1 epidemiology studies, vaccine development, and control of Newcastle disease in pigeons and poultry.


Subject(s)
Avulavirus Infections/veterinary , Avulavirus/genetics , Columbidae/virology , Genome, Viral , Genomics , Poultry Diseases/epidemiology , Poultry Diseases/virology , Animals , Avulavirus/immunology , Avulavirus/isolation & purification , China/epidemiology , Cross Reactions , Genomics/methods , Genotype , History, 21st Century , Molecular Epidemiology , Phylogeny , Poultry Diseases/history , Poultry Diseases/immunology , Whole Genome Sequencing
11.
Microbiol Resour Announc ; 9(5)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32001570

ABSTRACT

We report the complete genome sequences of 11 virulent Newcastle disease viruses. The isolates were obtained from vaccinated broiler and layer chickens in three different provinces of Indonesia in 2013 and 2014. Phylogenetic analysis revealed that all isolates belong to subgenotype VII.2 in the class II cluster.

12.
Sci Rep ; 9(1): 16350, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704961

ABSTRACT

U.S. public health agencies have employed next-generation sequencing (NGS) as a tool to quickly identify foodborne pathogens during outbreaks. Although established short-read NGS technologies are known to provide highly accurate data, long-read sequencing is still needed to resolve highly-repetitive genomic regions and genomic arrangement, and to close the sequences of bacterial chromosomes and plasmids. Here, we report the use of long-read nanopore sequencing to simultaneously sequence the entire chromosome and plasmid of Salmonella enterica subsp. enterica serovar Bareilly and Escherichia coli O157:H7. We developed a rapid and random sequencing approach coupled with de novo genome assembly within a customized data analysis workflow that uses publicly-available tools. In sequencing runs as short as four hours, using the MinION instrument, we obtained full-length genomes with an average identity of 99.87% for Salmonella Bareilly and 99.89% for E. coli in comparison to the respective MiSeq references. These nanopore-only assemblies provided readily available information on serotype, virulence factors, and antimicrobial resistance genes. We also demonstrate the potential of nanopore sequencing assemblies for rapid preliminary phylogenetic inference. Nanopore sequencing provides additional advantages as very low capital investment and footprint, and shorter (10 hours library preparation and sequencing) turnaround time compared to other NGS technologies.


Subject(s)
Bacterial Proteins/genetics , Escherichia coli/isolation & purification , Foodborne Diseases/genetics , Genome, Bacterial , Plasmids/genetics , Salmonella/isolation & purification , Whole Genome Sequencing/methods , Animals , Escherichia coli/genetics , Foodborne Diseases/microbiology , High-Throughput Nucleotide Sequencing/methods , Nanopore Sequencing/methods , Salmonella/genetics , Sequence Analysis, DNA/methods , Virulence Factors/genetics
13.
Microbiol Resour Announc ; 8(27)2019 Jul 03.
Article in English | MEDLINE | ID: mdl-31270191

ABSTRACT

A Newcastle disease virus was isolated from a chicken from a live bird market in the Mbeya region of Tanzania. Complete genome characterization of the isolate identified it as a member of subgenotype Vd. This is the first complete genome sequence of this subgenotype.

14.
Vet Microbiol ; 235: 25-34, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31282376

ABSTRACT

Five, class II, virulent Newcastle disease virus (vNDV) isolates of different genotypes from different host species were evaluated for their ability to infect, cause disease, and transmit to naïve chickens. Groups of five birds received a low, medium, or high dose, by the oculonasal route, of one of the following vNDV: three chicken-origin, one cormorant-origin, and one pigeon-origin. Three naïve birds were added to each group at two days post-inoculation (DPI) to evaluate transmission. Virus shedding was quantified from swabs (2/4/7 DPI), and seroconversion was evaluated at 14 DPI. All inoculated and contact birds in the chicken-origin vNDV groups succumbed to infection, displaying clinical signs typical of Newcastle disease and shed virus titers above 6 log10 EID50/ml. Birds receiving a high and medium dose of the cormorant virus showed primarily neurological clinical signs with 80% and 60% mortality, respectively. The chickens showing clinical disease shed virus at titers below 4 log10 EID50/ml, and the remaining bird in the high dose group seroconverted with a high HI titer. For the pigeon-origin virus, no clinical signs were observed in any of the birds, but all 5 chickens in the high challenge dose and one bird in the medium challenge group shed virus at mean titers of 3.1 and 2.2 log10 EID50/ml, respectively. Overall, the chicken-origin viruses infected chickens and efficiently transmitted to naïve birds, while the cormorant- and pigeon-origin viruses infected chickens only at the higher doses and did not transmit to other birds.


Subject(s)
Chickens/virology , Columbidae/virology , Newcastle Disease/transmission , Newcastle disease virus/pathogenicity , Poultry Diseases/transmission , Animals , Animals, Wild/virology , Vaccination , Virulence , Virus Shedding
15.
Infect Genet Evol ; 74: 103917, 2019 10.
Article in English | MEDLINE | ID: mdl-31200111

ABSTRACT

Several Avian paramyxoviruses 1 (synonymous with Newcastle disease virus or NDV, used hereafter) classification systems have been proposed for strain identification and differentiation. These systems pioneered classification efforts; however, they were based on different approaches and lacked objective criteria for the differentiation of isolates. These differences have created discrepancies among systems, rendering discussions and comparisons across studies difficult. Although a system that used objective classification criteria was proposed by Diel and co-workers in 2012, the ample worldwide circulation and constant evolution of NDV, and utilization of only some of the criteria, led to identical naming and/or incorrect assigning of new sub/genotypes. To address these issues, an international consortium of experts was convened to undertake in-depth analyses of NDV genetic diversity. This consortium generated curated, up-to-date, complete fusion gene class I and class II datasets of all known NDV for public use, performed comprehensive phylogenetic neighbor-Joining, maximum-likelihood, Bayesian and nucleotide distance analyses, and compared these inference methods. An updated NDV classification and nomenclature system that incorporates phylogenetic topology, genetic distances, branch support, and epidemiological independence was developed. This new consensus system maintains two NDV classes and existing genotypes, identifies three new class II genotypes, and reduces the number of sub-genotypes. In order to track the ancestry of viruses, a dichotomous naming system for designating sub-genotypes was introduced. In addition, a pilot dataset and sub-trees rooting guidelines for rapid preliminary genotype identification of new isolates are provided. Guidelines for sequence dataset curation and phylogenetic inference, and a detailed comparison between the updated and previous systems are included. To increase the speed of phylogenetic inference and ensure consistency between laboratories, detailed guidelines for the use of a supercomputer are also provided. The proposed unified classification system will facilitate future studies of NDV evolution and epidemiology, and comparison of results obtained across the world.


Subject(s)
Newcastle disease virus/classification , RNA, Viral/genetics , Sequence Analysis, RNA/methods , Bayes Theorem , Consensus , Data Curation , Databases, Genetic , Genotype , Guidelines as Topic , International Cooperation , Likelihood Functions , Newcastle disease virus/genetics , Phylogeny
16.
Avian Dis ; 63(1): 75-83, 2019 03 01.
Article in English | MEDLINE | ID: mdl-31251522

ABSTRACT

Turkey herpesvirus (HVT) is widely used as a vaccine against Marek's disease in chickens and recently as a vector for foreign genes from infectious bursal disease virus, Newcastle disease (ND) virus, infectious laryngotracheitis (ILT) virus, and avian influenza virus. Advantages of HVT-vector vaccines are that the vaccines do not contain live respiratory viruses or live infectious bursal disease virus able to replicate and cause disease or embryo mortality, they can be administered at hatch or in ovo, and they are relatively insensitive to interference from maternally derived antibodies. As producers have tried to combine HVT-vector vaccines to protect against additional diseases, reports have indicated that applying two vectored vaccines using the same HVT vector is reported to reduce the efficacy of one or both vaccines. To confirm this interference, we evaluated commercial vaccines from multiple companies, including products with inserts designed to protect against ND, infectious ILT, and infectious bursal disease (IBD). Using a standard dosage, we found that the ILT product was most severely affected by the addition of other vaccines, as demonstrated by a significant increase in clinical signs, significant decrease in weight gain, and increase in quantity of challenge virus observed from tracheal swabs collected from Days 3-5 postchallenge. The ND and IBD products were also affected by the addition of other vaccines, although in most cases differences compared to vaccination with the vector alone were not statistically significant. This study demonstrates the importance of following manufacturer guidelines and the need for validating alternative strategies to benefit from the high level of protection offered by vector vaccines.


Evaluación de la eficacia de la protección cuando se combinan vacunas recombinantes con base en el virus herpes del pavo como vector. El virus herpes de los pavos (HVT) se usa ampliamente como una vacuna contra la enfermedad de Marek en pollos y recientemente como un vector para genes externos como del virus de la enfermedad infecciosa de la bolsa, del virus de la enfermedad de Newcastle (ND), del virus de la laringotraqueítis infecciosa (ILT) y del virus de la influenza aviar. Las ventajas de las vacunas con vector de HVT consisten en que las vacunas no contienen virus vivos respiratorios o virus de la enfermedad infecciosa de la bolsa, no son capaces de replicarse y causar enfermedad o mortalidad embrionaria, pueden administrarse en el momento de la eclosión o in ovo y son relativamente insensibles a la interferencia de anticuerpos de origen materno. A medida que los productores han intentado combinar las vacunas con el vector HVT para inducir protección contra enfermedades adicionales, los informes han indicado que la aplicación de dos vacunas vectorizadas utilizando el mismo vector HVT reduce la eficacia de una o de ambas vacunas. Para confirmar esta interferencia, se evaluaron las vacunas comerciales de múltiples compañías, incluidos los productos con inserciones diseñadas para proteger contra la enfermedad de Newcastle, la laringotraqueítis infecciosa aviar y contra la enfermedad infecciosa de la bolsa. Utilizando una dosis estándar, se encontró que el producto para la laringotraqueítis infecciosa se vio más afectado por la adición de otras vacunas, como lo demuestra un aumento significativo en los signos clínicos, una disminución significativa en el aumento de peso y un aumento en la cantidad de virus de desafío observados en los hisopos traqueales recolectados de tres a cinco días después del desafío. Los productos para la enfermedad de Newcastle y para la enfermedad de Gumboro también se vieron afectados por la adición de otras vacunas, aunque en la mayoría de los casos las diferencias en comparación con la vacunación únicamente con el vector no fueron estadísticamente significativas. Este estudio demuestra la importancia de seguir las pautas del fabricante y la necesidad de validar estrategias alternativas para beneficiarse del alto nivel de protección ofrecido por las vacunas con vectores.


Subject(s)
Chickens , Herpesvirus 1, Meleagrid/immunology , Marek Disease/prevention & control , Poultry Diseases/prevention & control , Vaccination/veterinary , Viral Vaccines/immunology , Animals , Genetic Vectors , Herpesvirus 1, Gallid/immunology , Infectious bursal disease virus/immunology , Influenza A virus/immunology , Newcastle disease virus/immunology , Vaccines, Combined/immunology
17.
Virus Genes ; 55(4): 502-512, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31089865

ABSTRACT

Highly virulent Newcastle disease virus (NDV) causes Newcastle disease (ND), which is a threat to poultry production worldwide. Effective disease management requires approaches to accurately determine sources of infection, which involves tracking of closely related viruses. Next-generation sequencing (NGS) has emerged as a research tool for thorough genetic characterization of infectious organisms. Previously formalin-fixed paraffin-embedded (FFPE) tissues have been used to conduct retrospective epidemiological studies of related but genetically distinct viruses. However, this study extends the applicability of NGS for complete genome analysis of viruses from FFPE tissues to track the evolution of closely related viruses. Total RNA was obtained from FFPE spleens, lungs, brains, and small intestines of chickens in 11 poultry flocks during disease outbreaks in Pakistan. The RNA was randomly sequenced on an Illumina MiSeq instrument and the raw data were analyzed using a custom data analysis pipeline that includes de novo assembly. Genomes of virulent NDV were detected in 10/11 birds: eight nearly complete (> 95% coverage of concatenated coding sequence) and two partial genomes. Phylogeny of the NDV complete genome coding sequences was compared to current methods of analysis based on the full and partial fusion genes and determined that the approach provided a better phylogenetic resolution. Two distinct lineages of sub-genotype VIIi NDV were identified to be simultaneously circulating in Pakistani poultry. Non-targeted NGS of total RNA from FFPE tissues coupled with de novo assembly provided a reliable, safe, and affordable method to conduct epidemiological and evolutionary studies to facilitate management of ND in Pakistan.


Subject(s)
Chickens , Newcastle Disease/virology , Newcastle disease virus/genetics , Poultry Diseases/virology , Animals , Databases, Nucleic Acid , Disease Outbreaks/veterinary , Genome, Viral , High-Throughput Nucleotide Sequencing , Newcastle Disease/epidemiology , Newcastle disease virus/isolation & purification , Pakistan/epidemiology , Phylogeny , RNA, Viral , Sequence Analysis, RNA , Viral Fusion Proteins/genetics , Viral Structural Proteins/analysis
18.
Arch Virol ; 164(8): 2031-2047, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31123963

ABSTRACT

Newcastle disease virus (NDV) has a wide avian host range and a high degree of genetic variability, and virulent strains cause Newcastle disease (ND), a worldwide concern for poultry health. Although NDV has been studied in Nigeria, genetic information about the viruses involved in the endemicity of the disease and the transmission that likely occurs at the poultry-wildlife interface is still largely incomplete. Next-generation and Sanger sequencing was performed to provide complete (n = 73) and partial genomic sequence data (n = 38) for NDV isolates collected from domestic and wild birds in Nigeria during 2002-2015, including the first complete genome sequences of genotype IV and subgenotype VIh from the African continent. Phylogenetic analysis revealed that viruses of seven different genotypes circulated in that period, demonstrating high genetic diversity of NDV for a single country. In addition, a high degree of similarity between NDV isolates from domestic and wild birds was observed, suggesting that spillovers had occurred, including to three species that had not previously been shown to be susceptible to NDV infection. Furthermore, the first spillover of a mesogenic Komarov vaccine virus is documented, suggesting a previous spillover and evolution of this virus. The similarities between viruses from poultry and multiple bird species and the lack of evidence for host adaptation in codon usage suggest that transmission of NDV between poultry and non-poultry birds occurred recently. This is especially significant when considering that some viruses were isolated from species of conservation concern. The high diversity of NDV observed in both domestic and wild birds in Nigeria emphasizes the need for active surveillance and epidemiology of NDV in all bird species.


Subject(s)
Animals, Wild/virology , Birds/virology , Newcastle Disease/virology , Newcastle disease virus/genetics , Animals , Genetic Variation/genetics , Genomics/methods , Genotype , Nigeria , Phylogeny , Poultry/virology , Whole Genome Sequencing/methods
19.
BMC Evol Biol ; 19(1): 108, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31126244

ABSTRACT

BACKGROUND: Avian avulavirus (commonly known as avian paramyxovirus-1 or APMV-1) can cause disease of varying severity in both domestic and wild birds. Understanding how viruses move among hosts and geography would be useful for informing prevention and control efforts. A Bayesian statistical framework was employed to estimate the evolutionary history of 1602 complete fusion gene APMV-1 sequences collected from 1970 to 2016 in order to infer viral transmission between avian host orders and diffusion among geographic regions. Ancestral states were estimated with a non-reversible continuous-time Markov chain model, allowing transition rates between discrete states to be calculated. The evolutionary analyses were stratified by APMV-1 classes I (n = 198) and II (n = 1404), and only those sequences collected between 2006 and 2016 were allowed to contribute host and location information to the viral migration networks. RESULTS: While the current data was unable to assess impact of host domestication status on APMV-1 diffusion, these analyses supported the sharing of APMV-1 among divergent host taxa. The highest supported transition rate for both classes existed from domestic chickens to Anseriformes (class I:6.18 transitions/year, 95% highest posterior density (HPD) 0.31-20.02, Bayes factor (BF) = 367.2; class II:2.88 transitions/year, 95%HPD 1.9-4.06, BF = 34,582.9). Further, among class II viruses, domestic chickens also acted as a source for Columbiformes (BF = 34,582.9), other Galliformes (BF = 34,582.9), and Psittaciformes (BF = 34,582.9). Columbiformes was also a highly supported source to Anseriformes (BF = 322.0) and domestic chickens (BF = 402.6). Additionally, our results provide support for the diffusion of viruses among continents and regions, but no interhemispheric viral exchange between 2006 and 2016. Among class II viruses, the highest transition rates were estimated from South Asia to the Middle East (1.21 transitions/year; 95%HPD 0.36-2.45; BF = 67,107.8), from Europe to East Asia (1.17 transitions/year; 95%HPD 0.12-2.61; BF = 436.2) and from Europe to Africa (1.06 transitions/year, 95%HPD 0.07-2.51; BF = 169.3). CONCLUSIONS: While migration appears to occur infrequently, geographic movement may be important in determining viral diversification and population structure. In contrast, inter-order transmission of APMV-1 may occur readily, but most events are transient with few lineages persisting in novel hosts.


Subject(s)
Host-Pathogen Interactions , Internationality , Newcastle Disease/transmission , Newcastle Disease/virology , Newcastle disease virus/classification , Phylogeny , Africa , Animals , Asia , Bias , Chickens/virology , Europe , Genotype , Geography , Newcastle disease virus/genetics , United States
20.
Virology ; 531: 203-218, 2019 05.
Article in English | MEDLINE | ID: mdl-30928699

ABSTRACT

In May of 2018, virulent Newcastle disease virus was detected in sick, backyard, exhibition chickens in southern California. Since, the virus has affected 401 backyard and four commercial flocks, and one live bird market in California, and one backyard flock in Utah. The pathogenesis and transmission potential of this virus, along with two genetically related and widely studied viruses, chicken/California/2002 and chicken/Belize/2008, were evaluated in both 3-week- and 62-week-old chickens given a low, medium, or high challenge dose. All three viruses were highly virulent causing clinical signs, killing all the chickens in the medium and high dose groups, and efficiently transmitting to contacts. The three viruses also replicated in the reproductive tract of the adult hens. Virus shedding for all viruses was detected 24 hours after challenge, peaking with high titers at day 4 post challenge. Although not genetically identical, the studied isolates were shown to be phenotypically very similar, which allows the utilization of the available literature in the control of the current outbreak.


Subject(s)
Newcastle Disease/transmission , Newcastle disease virus/physiology , Newcastle disease virus/pathogenicity , Poultry Diseases/transmission , Age Factors , Animals , California/epidemiology , Chickens , Disease Outbreaks , Female , Male , Newcastle Disease/epidemiology , Newcastle Disease/virology , Newcastle disease virus/genetics , Newcastle disease virus/isolation & purification , Poultry Diseases/epidemiology , Poultry Diseases/virology , Virulence , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL
...