Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Emerg Microbes Infect ; 12(2): 2272644, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37847060

ABSTRACT

Immature feathers are known replication sites for high pathogenicity avian influenza viruses (HPAIVs) in poultry. However, it is unclear whether feathers play an active role in viral transmission. This study aims to investigate the contribution of the feather epithelium to the dissemination of clade 2.3.4.4b goose/Guangdong/1996 lineage H5 HPAIVs in the environment, based on natural and experimental infections of domestic mule and Muscovy ducks. During the 2016-2022 outbreaks, H5 HPAIVs exhibited persistent and marked feather epitheliotropism in naturally infected commercial ducks. Infection of the feather epithelium resulted in epithelial necrosis and disruption, as well as the production and environmental shedding of infectious virions. Viral and feather antigens colocalized in dust samples obtained from poultry barns housing naturally infected birds. In summary, the feather epithelium contributes to viral replication, and it is a likely source of environmental infectious material. This underestimated excretion route could greatly impact the ecology of HPAIVs, facilitating airborne and preening-related infections within a flock, and promoting prolonged viral infectivity and long-distance viral transmission between poultry farms.


Subject(s)
Influenza A virus , Influenza in Birds , Poultry Diseases , Animals , Ducks , Feathers , Virulence , Poultry , Epithelium
2.
Emerg Microbes Infect ; 12(1): 2218945, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37309051

ABSTRACT

Clade 2.3.4.4 Eurasian lineage H5Nx highly pathogenic avian influenza virus (HPAIV) has become the globally dominant clade and caused global outbreaks since 2014. The clade 2.3.4.4 viruses have evolved into eight hemagglutinin subgroups (2.3.4.4a-h). In this study, we evaluated the infectivity, pathobiology, and transmissibility of seven clade 2.3.4.4 viruses (two 2.3.4.4a, two 2.3.4.4b, one 2.3.4.4c and two 2.3.4.4e) in chickens. The two clade 2.3.4.4e viruses caused 100% mortality and transmissibility in chickens. However, clade 2.3.4.4a and c viruses showed 80-90% mortality and 67% transmissibility. Clade 2.3.4.4b viruses showed 100% mortality, but no transmission to co-housed chickens was observed based on lack of seroconversion. All the infected chickens died showing systemic infection, irrespective of subgroup. The results highlight that all the clade 2.3.4.4 HPAIVs used in this study caused high mortality in infected chickens, but the transmissibility of the viruses in chickens was variable in contrast to that of previous Eurasian-lineage H5N1 HPAIVs. Changes in the pathogenicity and transmissibility of clade 2.3.4.4 HPAIVs warrant careful monitoring of the viruses to establish effective control strategies.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Sepsis , Animals , Chickens , Disease Outbreaks
3.
Vaccine ; 41(18): 2893-2904, 2023 05 02.
Article in English | MEDLINE | ID: mdl-37012117

ABSTRACT

Vaccines are an essential tool for the control of viral infections in domestic animals. We generated recombinant vector herpesvirus of turkeys (vHVT) vaccines expressing computationally optimized broadly reactive antigen (COBRA) H5 of avian influenza virus (AIV) alone (vHVT-AI) or in combination with virus protein 2 (VP2) of infectious bursal disease virus (IBDV) (vHVT-IBD-AI) or fusion (F) protein of Newcastle disease virus (NDV) (vHVT-ND-AI). In vaccinated chickens, all three vHVT vaccines provided 90-100% clinical protection against three divergent clades of high pathogenicity avian influenza viruses (HPAIVs), and significantly decreased number of birds and oral viral shedding titers at 2 days post-challenge compared to shams. Four weeks after vaccination, most vaccinated birds had H5 hemagglutination inhibition antibody titers, which significantly increased post-challenge. The vHVT-IBD-AI and vHVT-ND-AI vaccines provided 100% clinical protection against IBDVs and NDV, respectively. Our findings demonstrate that multivalent HVT vector vaccines were efficacious for simultaneous control of HPAIV and other viral infections.


Subject(s)
Birnaviridae Infections , Herpesviridae , Infectious bursal disease virus , Influenza A virus , Influenza in Birds , Newcastle Disease , Poultry Diseases , Viral Vaccines , Animals , Newcastle disease virus/genetics , Newcastle Disease/prevention & control , Chickens , Turkeys , Virulence , Vaccines, Synthetic/genetics , Birnaviridae Infections/prevention & control , Birnaviridae Infections/veterinary , Herpesvirus 1, Meleagrid/genetics , Vaccines, Combined , Poultry Diseases/prevention & control
4.
Sci Total Environ ; 863: 160902, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36526195

ABSTRACT

Avian influenza viruses (AIVs) can affect wildlife, poultry, and humans, so a One Health perspective is needed to optimize mitigation strategies. Migratory waterfowl globally spread AIVs over long distances. Therefore, the study of AIV persistence in waterfowl staging and breeding areas is key to understanding their transmission dynamics and optimizing management strategies. Here, we used artificial streams mimicking natural conditions of waterfowl habitats in the Mediterranean climate (day/night cycles of photosynthetic active radiation and temperature, low water velocity, and similar microbiome to lowland rivers and stagnant water bodies) and then manipulated temperature and sediment presence (i.e., 10-13 °C vs. 16-18 °C, and presence vs. absence of sediments). An H1N1 low pathogenic AIV (LPAIV) strain was spiked in the streams, and water and sediment samples were collected at different time points until 14 days post-spike to quantify viral RNA and detect infectious particles. Viral RNA was detected until the end of the experiment in both water and sediment samples. In water samples, we observed a significant combined effect of temperature and sediments in viral decay, with higher viral genome loads in colder streams without sediments. In sediment samples, we didn't observe any significant effect of temperature. In contrast to prior laboratory-controlled studies that detect longer persistence times, infectious H1N1 LPAIV was isolated in water samples till 2 days post-spike, and none beyond. Infectious H1N1 LPAIV wasn't isolated from any sediment sample. Our results suggest that slow flowing freshwater surface waters may provide conditions facilitating bird-to-bird transmission for a short period when water temperature are between 10 and 18 °C, though persistence for extended periods (e.g., weeks or months) may be less likely. We hypothesize that experiments simulating real environments, like the one described here, provide a more realistic approach for assessing environmental persistence of AIVs.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A virus , Influenza in Birds , Animals , Humans , Rivers , Influenza A Virus, H1N1 Subtype/genetics , Ecosystem , Water , RNA, Viral
6.
Viruses ; 14(6)2022 05 30.
Article in English | MEDLINE | ID: mdl-35746661

ABSTRACT

The papers published in this Special Issue represent only a glimpse of the vast diversity of viral infectious diseases, and the complexity of their interactions with the host, that have an impact on human and animal health [...].


Subject(s)
Zoonoses , Animals , Virus Shedding
7.
Viruses ; 13(11)2021 10 28.
Article in English | MEDLINE | ID: mdl-34834976

ABSTRACT

A surge in fowl adenovirus (FAdV) causing inclusion body hepatitis (IBH) outbreaks has occurred in several countries in the last two decades. In Spain, a sharp increase in case numbers in broilers and broiler breeder pullets arose since 2011, which prompted the vaccination of breeders in some regions. Our retrospective study of IBH cases in Spain from 2011 to 2021 revealed that most cases were reported in broilers (92.21%) and were caused by serotypes FAdV-8b and -11, while cases in broiler breeder pullets were caused by serotypes FAdV-2, -11, and -8b. Vertical transmission was the main route of infection, although horizontal transmission likely happened in some broiler cases. Despite the inconsistent and heterogeneous use of vaccines among regions and over time, the number of cases mirrored the use of vaccines in the country. While IBH outbreaks were recorded year-long, significantly more cases occurred during the cooler and rainier months. The geographic distribution suggested a widespread incidence of IBH and revealed the importance of a highly integrated system. Our findings contribute to a better understanding of FAdV infection dynamics under field conditions and reiterate the importance of surveillance, serological monitoring of breeders, and vaccination of breeders against circulating serotypes to protect progenies.


Subject(s)
Chickens/virology , Hepatitis, Viral, Animal/epidemiology , Inclusion Bodies/virology , Poultry Diseases/epidemiology , Poultry Diseases/virology , Adenoviridae Infections/veterinary , Animals , Aviadenovirus/immunology , Disease Outbreaks , Hepatitis, Viral, Animal/classification , Hepatitis, Viral, Animal/diagnosis , Phylogeny , Poultry/virology , Poultry Diseases/diagnosis , Retrospective Studies , Serogroup , Spain/epidemiology
8.
Vaccine ; 39(14): 1933-1942, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33715903

ABSTRACT

The genetic and antigenic drift associated with the high pathogenicity avian influenza (HPAI) viruses of Goose/Guangdong (Gs/GD) lineage and the emergence of vaccine-resistant field viruses underscores the need for a broadly protective H5 influenza A vaccine. Here, we tested experimental vector herpesvirus of turkey (vHVT)-H5 vaccines containing either wild-type clade 2.3.4.4A-derived H5 inserts or computationally optimized broadly reactive antigen (COBRA) inserts with challenge by homologous and genetically divergent H5 HPAI Gs/GD lineage viruses in chickens. Direct assessment of protection was confirmed for all the tested constructs, which provided clinical protection against the homologous and heterologous H5 HPAI Gs/GD challenge viruses and significantly decreased oropharyngeal shedding titers compared to the sham vaccine. The cross reactivity was assessed by hemagglutinin inhibition (HI) and focus reduction assay against a panel of phylogenetically and antigenically diverse H5 strains. The COBRA-derived H5 inserts elicited antibody responses against antigenically diverse strains, while the wild-type-derived H5 vaccines elicited protection mostly against close antigenically related clades 2.3.4.4A and 2.3.4.4D viruses. In conclusion, the HVT vector, a widely used replicating vaccine platform in poultry, with H5 insert provides clinical protection and significant reduction of viral shedding against homologous and heterologous challenge. In addition, the COBRA-derived inserts have the potential to be used against antigenically distinct co-circulating viruses and future drift variants.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Influenza in Birds , Marek Disease , Animals , Antibodies, Viral , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinins , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/prevention & control , Marek Disease/prevention & control , Vaccines, Synthetic/genetics , Virulence
9.
Front Immunol ; 12: 800188, 2021.
Article in English | MEDLINE | ID: mdl-35003125

ABSTRACT

Highly pathogenic avian influenza viruses (HPAIVs) cause severe systemic disease and high mortality rates in chickens, leading to a huge economic impact in the poultry sector. However, some chickens are resistant to the disease. This study aimed at evaluating the mechanisms behind HPAIV disease resistance. Chickens of different breeds were challenged with H7N1 HPAIV or clade 2.3.4.4b H5N8 HPAIV, euthanized at 3 days post-inoculation (dpi), and classified as resistant or susceptible depending on the following criteria: chickens that presented i) clinical signs, ii) histopathological lesions, and iii) presence of HPAIV antigen in tissues were classified as susceptible, while chickens lacking all these criteria were classified as resistant. Once classified, we performed RNA-Seq from lung and spleen samples in order to compare the transcriptomic signatures between resistant and susceptible chickens. We identified minor transcriptomic changes in resistant chickens in contrast with huge alterations observed in susceptible chickens. Interestingly, six differentially expressed genes were downregulated in resistant birds and upregulated in susceptible birds. Some of these genes belong to the NF-kappa B and/or mitogen-activated protein kinase signaling pathways. Among these six genes, the serine protease-encoding gene PLAU was of particular interest, being the most significantly downregulated gene in resistant chickens. Expression levels of this protease were further validated by RT-qPCR in a larger number of experimentally infected chickens. Furthermore, HPAIV quasi-species populations were constructed using 3 dpi oral swabs. No substantial changes were found in the viral segments that interact with the innate immune response and with the host cell receptors, reinforcing the role of the immune system of the host in the clinical outcome. Altogether, our results suggest that an early inactivation of important host genes could prevent an exaggerated immune response and/or viral replication, conferring resistance to HPAIV in chickens.


Subject(s)
Chickens/genetics , Chickens/virology , Disease Resistance/genetics , Influenza A Virus, H7N1 Subtype , Influenza in Birds/genetics , Animals , Influenza in Birds/virology , RNA-Seq
10.
Poult Sci ; 99(12): 6503-6512, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33248565

ABSTRACT

Identifying which diseases represent a priority is crucial to optimize resources for diagnostics, control, and prevention. Here, the impact of 111 poultry pathogens belonging to Viruses (n = 31), Bacteria (n = 33), and Other (n = 47) was assessed using the H-index. The overall mean H-indexes suggested that poultry Viruses have statistically greater impact than Bacteria, which in turn are statistically more relevant than Others. Among the 20 highest H-indexes, 45% were zoonotic, and almost a third was Office International des Epizooties-listed. Avian influenza virus (H-index 127), Salmonella enteritidis and Salmonella typhimurium (H-index 72), and Eimeria spp (H-index 70) ranked the highest in Virus, Bacteria, and Other, respectively. Pathogens that produce overt clinical diseases and economic damage, cause immunosuppression, and/or are zoonotic had the highest H-index scores. The evolution of citations of particular pathogens reflected severe poultry outbreaks and/or zoonotic outbreaks in relatively wide geographic areas. Also, the evolution of citations based on taxonomic groups mirrored major changes in poultry production practices and management throughout history. Thus, Others were the most cited pathogens until the 1970s and, following 3 decades of unpopularity because of widespread use of intensive production practices, regained importance in the 2000s thanks to welfare regulation changes. Citations for Bacteria increased especially from the 1990s onward, probably because of the ban of growth promoters in western countries and the need to find new control methods for bacterial and protozoal infections. In general, countries with the greatest poultry production and research budgets had higher research production, that is the United States of America (USA) and China. Interestingly, the United Kingdom was among the top research producers despite falling behind other countries in poultry production and research budget. Moreover, the USA exhibited the strongest poultry research production based on number and diversity of publications (Dcos-index). In conclusion, the H-index could be a valid, simple tool to prioritize funding or interest in poultry diseases, especially when used as a preliminary selection approach in combination with other metrics.


Subject(s)
Periodicals as Topic , Poultry Diseases , Research , Animals , Chickens , China , Periodicals as Topic/statistics & numerical data , Poultry , Research/economics , Research/trends , United States
11.
Vaccine ; 38(6): 1526-1534, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31862196

ABSTRACT

Despite decades of vaccination, surveillance, and biosecurity measures, H5N2 low pathogenicity avian influenza (LPAI) virus infections continue in Mexico and neighboring countries. One explanation for tenacity of H5N2 LPAI in Mexico is the antigenic divergence of circulating field viruses compared to licensed vaccines due to antigenic drift. Our phylogenetic analysis indicates that the H5N2 LPAI viruses circulating in Mexico and neighboring countries since 1994 have undergone antigenic drift away from vaccine seed strains. Here we evaluated the efficacy of a new recombinant fowlpox virus vector containing an updated H5 insert (rFPV-H5/2016), more relevant to the current strains circulating in Mexico. We tested the vaccine efficacy against a closely related subcluster 4 Mexican H5N2 LPAI (2010 H5/LP) virus and the historic H5N2 HPAI (1995 H5/HP) virus in White Leghorn chickens. The rFPV-H5/2016 vaccine provided hemagglutinin inhibition (HI) titers pre-challenge against viral antigens from both challenge viruses in almost 100% of the immunized birds, with no differences in number of birds seroconverting or HI titers among all tested doses (1.5, 2.0, and 3.1 log10 mean tissue culture infectious doses/bird). The vaccine conferred 100% clinical protection and a significant decrease in oral and cloacal virus shedding from 1995 H5/HP virus challenged birds when compared to the sham controls at all tested doses. Virus shedding titers from vaccinated 2010 H5/LP virus challenged birds significantly decreased compared to sham birds especially at earlier time points. Our results confirm the efficacy of the new rFPV-H5/2016 against antigenic drift of LPAI virus in Mexico and suggest that this vaccine would be a good candidate, likely as a primer in a prime-boost vaccination program.


Subject(s)
Fowlpox/prevention & control , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Animals , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza Vaccines/genetics , Mexico , Phylogeny , Vaccines, Synthetic/genetics
12.
Vet Res ; 50(1): 89, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31675983

ABSTRACT

In the 2014-2015 Eurasian lineage clade 2.3.4.4A H5 highly pathogenic avian influenza (HPAI) outbreak in the U.S., backyard flocks with minor gallinaceous poultry and large commercial poultry (chickens and turkeys) operations were affected. The pathogenesis of the first H5N8 and reassortant H5N2 clade 2.3.4.4A HPAI U.S. isolates was investigated in six gallinaceous species: chickens, Japanese quail, Bobwhite quail, Pearl guinea fowl, Chukar partridges, and Ring-necked pheasants. Both viruses caused 80-100% mortality in all species, except for H5N2 virus that caused 60% mortality in chickens. The surviving challenged birds remained uninfected based on lack of clinical disease and lack of seroconversion. Among the infected birds, chickens and Japanese quail in early clinical stages (asymptomatic and listless) lacked histopathologic findings. In contrast, birds of all species in later clinical stages (moribund and dead) had histopathologic lesions and systemic virus replication consistent with HPAI virus infection in gallinaceous poultry. These birds had widespread multifocal areas of necrosis, sometimes with heterophilic or lymphoplasmacytic inflammatory infiltrate, and viral antigen in parenchymal cells of most tissues. In general, lesions and antigen distribution were similar regardless of virus and species. However, endotheliotropism was the most striking difference among species, with only Pearl guinea fowl showing widespread replication of both viruses in endothelial cells of most tissues. The expression of IFN-γ and IL-10 in Japanese quail, and IL-6 in chickens, were up-regulated in later clinical stages compared to asymptomatic birds.


Subject(s)
Galliformes , Immunity, Innate , Influenza A virus/physiology , Influenza in Birds/immunology , Influenza in Birds/virology , Poultry Diseases/immunology , Poultry Diseases/virology , Animals , United States
14.
Vaccine ; 37(17): 2369-2376, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30905528

ABSTRACT

Since the first identification of the H5N1 Goose/Guangdong lineage in 1996, this highly pathogenic avian influenza virus has spread worldwide, becoming endemic in domestic poultry. Sporadic transmission to humans has raised concerns of a potential pandemic and underscores the need for a broad cross-protective influenza vaccine. Here, we tested our previously described methodology, termed Computationally Optimized Broadly Reactive Antigen (COBRA), to generate a novel hemagglutinin (HA) gene, termed COBRA-2, that was based on H5 HA sequences from 2005 to 2006. The COBRA-2 HA virus-like particle (VLP) vaccines were used to vaccinate chickens and the immune responses were compared to responses elicited by VLP's expressing HA from A/whooper swan/Mongolia/244/2005 (WS/05), a representative 2005 vaccine virus from clade 2.2. To support this evaluation a hemagglutination inhibition (HAI) breadth panel was developed consisting of phylogenetically and antigenically diverse H5 strains in circulation from 2005 to 2006, as well as recent drift variants (2008 - 2014). We found that the COBRA-2 VLP vaccines elicited robust HAI titers against this entire breadth panel, whereas the VLP vaccine based upon the recommended WS/05 HA only elicited HAI responses against a subset of strains. Furthermore, while all vaccines protected chickens against challenge with the WS/05 virus, only the human COBRA-2 VLP vaccinated birds were protected (80%) against a recent drifted clade 2.3.2.1B, A/duck/Vietnam/NCVD-672/2011 (VN/11) virus. This is the first report to demonstrate seroprotective antibody responses against genetically diverse clades and sub-clades of H5 viruses and protective efficacy against a recent drifted variant using a globular head based design strategy.


Subject(s)
Antigenic Variation/immunology , Antigens, Viral/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Vaccinology , Animals , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/prevention & control , Influenza, Human/prevention & control , Phylogeny , Vaccines, Virus-Like Particle/immunology
15.
Vaccine ; 37(16): 2232-2243, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30885512

ABSTRACT

Since 2012, H7N3 highly pathogenic avian influenza (HPAI) has produced negative economic and animal welfare impacts on poultry in central Mexico. In the present study, chickens were vaccinated with two different recombinant fowlpox virus vaccines (rFPV-H7/3002 with 2015 H7 hemagglutinin [HA] gene insert, and rFPV-H7/2155 with 2002 H7 HA gene insert), and were then challenged three weeks later with H7N3 HPAI virus (A/chicken/Jalisco/CPA-37905/2015). The rFPV-H7/3002 vaccine conferred 100% protection against mortality and morbidity, and significantly reduced virus shed titers from the respiratory and gastrointestinal tracts. In contrast, 100% of sham and rFPV-H7/2155 vaccinated birds shed virus at higher titers and died within 4 days. Pre- (15/20) and post- (20/20) challenge serum of birds vaccinated with rFPV-H7/3002 had antibodies detectable by hemagglutination inhibition (HI) assay using challenge virus antigen. However, only a few birds (3/20) in the rFPV-H7/2155 vaccinated group had antibodies that reacted against the challenge strain but all birds had antibodies that reacted against the homologous vaccine antigen (A/turkey/Virginia/SEP-66/2002) (20/20). One possible explanation for differences in vaccines efficacy is the antigenic drift between circulating viruses and vaccines. Molecular analysis demonstrated that the Mexican H7N3 strains have continued to rapidly evolve since 2012. In addition, we identified in silico three potential new N-glycosylation sites on the globular head of the H7 HA of A/chicken/Jalisco/CPA-37905/2015 challenge virus, which were absent in 2012 H7N3 outbreak virus. Our results suggested that mutations in the HA antigenic sites including increased glycosylation sites, accumulated in the new circulating Mexican H7 HPAIV strains, altered the recognition of neutralizing antibodies from the older vaccine strain rFPV-H7/2155. Therefore, the protective efficacy of novel rFPV-H7/3002 against recent outbreak Mexican H7N3 HPAIV confirms the importance of frequent updating of vaccines seed strains for long-term effective control of H7 HPAI virus.


Subject(s)
Fowlpox/prevention & control , Influenza A Virus, H7N3 Subtype/immunology , Influenza Vaccines/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/immunology , Fowlpox/immunology , Fowlpox/mortality , Fowlpox/virology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H7N3 Subtype/classification , Influenza A Virus, H7N3 Subtype/genetics , Influenza Vaccines/administration & dosage , Mexico , Phylogeny , Vaccines, DNA/administration & dosage , Virus Shedding
16.
Vaccine ; 36(43): 6361-6372, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30241684

ABSTRACT

Maternally-derived antibodies (MDA) provide early protection from disease, but may interfere with active immunity in young chicks. In highly pathogenic avian influenza virus (HPAIV)-enzootic countries, broiler chickens typically have MDA to Newcastle disease virus (NDV) and H5 HPAIV, and their impact on active immunity from recombinant vectored vaccines is unclear. We assessed the effectiveness of a spray-applied recombinant NDV vaccine with H5 AIV insert (rNDV-H5) and a recombinant turkey herpesvirus (HVT) vaccine with H5 AIV insert (rHVT-H5) in commercial broilers with MDA to NDV alone (MDA:AIV-NDV+) or to NDV plus AIV (MDA:AIV+NDV+) to provide protection against homologous HPAIV challenge. In Experiment 1, chicks were spray-vaccinated with rNDV-H5 at 3 weeks (3w) and challenged at 5 weeks (5w). All sham-vaccinated progeny lacked AIV antibodies and died following challenge. In rNDV-H5 vaccine groups, AIV and NDV MDA had completely declined to non-detectable levels by vaccination, enabling rNDV-H5 spray vaccine to elicit a protective AIV antibody response by 5w, with 70-78% survival and significant reduction of virus shedding compared to shams. In Experiment 2, progeny were vaccinated with rHVT-H5 and rNDV-H5 at 1 day (1d) or 3w and challenged at 5w. All sham-vaccinated progeny lacked AIV antibodies and died following challenge. In rHVT-H5(1d) vaccine groups, irrespective of rNDV-H5(3w) boost, AIV antibodies reached protective levels pre-challenge, as all progeny survived and virus shedding significantly decreased compared to shams. In contrast, rNDV-H5-vaccinated progeny had AIV and/or NDV MDA at the time of vaccination (1d and/or 3w) and failed to develop a protective immune response by 5w, resulting in 100% mortality after challenge. Our results demonstrate that MDA to AIV had minimal impact on the effectiveness of rHVT-H5, but MDA to AIV and/or NDV at the time of vaccination can prevent development of protective immunity from a primary or booster rNDV-H5 vaccine.


Subject(s)
Immunity, Maternally-Acquired , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Newcastle disease virus/genetics , Poultry Diseases/prevention & control , Animals , Antibodies, Viral/blood , Chickens/immunology , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization, Secondary , Influenza A Virus, H5N1 Subtype , Influenza in Birds/immunology , Poultry Diseases/virology , Vaccines, Synthetic/immunology , Virus Shedding
17.
Vet Res ; 49(1): 82, 2018 Aug 29.
Article in English | MEDLINE | ID: mdl-30157963

ABSTRACT

In March 2017, H7N9 highly pathogenic avian influenza (HPAI) virus was detected in 2 broiler breeder farms in the state of Tennessee, USA. Subsequent surveillance detected the low pathogenicity avian influenza (LPAI) virus precursor in multiple broiler breeder farms and backyard poultry in Tennessee and neighboring states. The pathogenesis of the H7N9 LPAI virus was investigated in commercial broiler breeders, the bird type mostly affected in this outbreak. Infectivity, transmissibility, and pathogenesis of the H7N9 HPAI and LPAI viruses were also studied in 4-week-old specific pathogen free (SPF) leghorn chickens. The mean bird infectious doses (BID50) for the LPAI isolate was 5.6 log10 mean egg infectious dose (EID50) for broiler breeders and 4.3 log10 EID50 for SPF layer chickens, and no transmission to contact-exposed birds was observed. In both bird types, virus shedding was almost exclusively from the oropharyngeal route. These findings suggest sub-optimal adaptation for sustained transmission with the H7N9 LPAI isolate, indicating that factors other than the birds genetic background may explain the epidemiology of the outbreak. The BID50 for the HPAI isolate in SPF layer chickens was more than 2 logs lower (<2 log10 EID50) than the LPAI isolate. Also, the HPAI virus was shed by both the oropharyngeal and cloacal routes and transmitted to contacts. Greater susceptibility and easier transmission of the H7N9 HPAI virus are features of the HP phenotype that could favor the spread of HPAI over LPAI viruses during outbreaks.


Subject(s)
Chickens , Influenza A Virus, H7N9 Subtype/physiology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza in Birds/transmission , Influenza in Birds/virology , Poultry Diseases/transmission , Poultry Diseases/virology , Animals , Specific Pathogen-Free Organisms , Tennessee , Virulence
18.
Int J Hyg Environ Health ; 221(6): 893-900, 2018 07.
Article in English | MEDLINE | ID: mdl-29891217

ABSTRACT

Airborne transmission of H5N1 highly pathogenic avian influenza (HPAI) viruses has occurred among poultry and from poultry to humans during home or live-poultry market slaughter of infected poultry, and such transmission has been experimentally reproduced. In this study, we investigated simple, practical changes in the processing of H5N1 virus-infected chickens to reduce infectious airborne particles and their transmission. Our findings suggest that containing the birds during the killing and bleeding first step by using a disposable plastic bag, a commonly available cooking pot widely used in Egypt (halla), or a bucket significantly reduces generation of infectious airborne particles and transmission to ferrets. Similarly, lack of infectious airborne particles was observed when processing vaccinated chickens that had been challenged with HPAI virus. Moreover, the use of a mechanical defeatherer significantly increased total number of particles in the air compared to manual defeathering. This study confirms that simple changes in poultry processing can efficiently mitigate generation of infectious airborne particles and their transmission to humans.


Subject(s)
Food-Processing Industry/methods , Influenza in Birds/transmission , Poultry Diseases/transmission , Animals , Chickens/virology , Female , Ferrets , Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Vaccination/veterinary
19.
Vaccine ; 36(1): 84-90, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29180030

ABSTRACT

The outbreak of highly pathogenic avian influenza virus in North American poultry during 2014 and 2015 demonstrated the devastating effects of the disease and highlighted the need for effective emergency vaccine prevention and control strategies targeted at currently circulating strains. This study evaluated the efficacy of experimental recombinant turkey herpesvirus vector vaccines with three different inserts targeting the hemagglutinin gene of an isolate from the recent North American influenza outbreak. White leghorn chickens were vaccinated at one day of age and challenged with A/Turkey/Minnesota/12582/2015 H5N2 at 4 weeks of age. Birds were analyzed for survival, viral shedding at two and four days after infection, and specific antibody prior to challenge and from surviving birds. The three experimental vaccines demonstrated 100%, 45% and 15% survival with the most effective vaccine significantly reducing oral and cloacal viral shedding compared to all other groups and generated specific antibody prior to challenge with highly pathogenic avian influenza virus. More studies are needed using diverse H5Nx highly pathogenic virus isolates to fully determine the breadth of coverage against possible exposure strains, as well as possible impact of maternally derived antibody on protection and vaccine efficacy.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Herpesvirus 1, Meleagrid/immunology , Herpesvirus Vaccines/genetics , Influenza A Virus, H5N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Animals , Antibodies, Viral/blood , Chickens , Disease Outbreaks/prevention & control , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Herpesvirus 1, Meleagrid/genetics , Herpesvirus Vaccines/administration & dosage , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H5N2 Subtype/isolation & purification , Influenza A Virus, H5N2 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza in Birds/epidemiology , Influenza in Birds/immunology , Poultry Diseases/prevention & control , Poultry Diseases/virology , United States/epidemiology , Vaccination , Vaccine Potency , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virus Shedding
20.
Emerg Infect Dis ; 23(11): 1806-1814, 2017 11.
Article in English | MEDLINE | ID: mdl-29047426

ABSTRACT

Exposure to infected poultry is a suspected cause of avian influenza (H5N1) virus infections in humans. We detected infectious droplets and aerosols during laboratory-simulated processing of asymptomatic chickens infected with human- (clades 1 and 2.2.1) and avian- (clades 1.1, 2.2, and 2.1) origin H5N1 viruses. We detected fewer airborne infectious particles in simulated processing of infected ducks. Influenza virus-naive chickens and ferrets exposed to the air space in which virus-infected chickens were processed became infected and died, suggesting that the slaughter of infected chickens is an efficient source of airborne virus that can infect birds and mammals. We did not detect consistent infections in ducks and ferrets exposed to the air space in which virus-infected ducks were processed. Our results support the hypothesis that airborne transmission of HPAI viruses can occur among poultry and from poultry to humans during home or live-poultry market slaughter of infected poultry.


Subject(s)
Air Microbiology , Chickens , Ducks , Influenza in Birds/transmission , Poultry Diseases/transmission , Animal Husbandry , Animals , Ferrets , Influenza A Virus, H5N1 Subtype , Inhalation Exposure , Poultry Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...