Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.343
Filtrer
1.
Sci Rep ; 14(1): 16021, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992055

RÉSUMÉ

Environmental conditions profoundly impact the health, welfare, and productivity of laying hens in commercial poultry farming. We investigated the association between microclimate variations, production indices, and histopathological responses to accidental Newcastle disease virus (NDV) infection within a controlled closed-house system. The study was conducted over seven months in a laying hen facility in Cairo, Egypt. Microclimate measurements included temperature, relative humidity (RH%), air velocity (AV), and the temperature humidity index (THI) that were obtained from specific locations on the front and back sides of the facility. Productivity indices, including the egg production percentage (EPP), egg weight (EW), average daily feed intake, and feed conversion ratio, were assessed monthly. During an NDV outbreak, humoral immune responses, gross pathology, and histopathological changes were evaluated. The results demonstrated significant (p < 0.05) variations in EPP and EW between the front and back sides except in April and May. AV had a significant (p = 0.006) positive effect (Beta = 0.346) on EW on the front side. On the back side, AV had a significant (p = 0.001) positive effect (Beta = 0.474) on EW, while it negatively influenced (p = 0.027) EPP (Beta = - 0.281). However, temperature, RH%, and THI had no impact and could not serve as predictors for EPP or EW on either farm side. The humoral immune response to NDV was consistent across microclimates, highlighting the resilience of hens. Histopathological examination revealed characteristic NDV-associated lesions, with no significant differences between the microclimates. This study underscores the significance of optimizing microclimate conditions to enhance laying performance by providing tailored environmental management strategies based on seasonal variations, ensuring consistent airflow, particularly near cooling pads and exhaust fans, and reinforcing the importance of biosecurity measures under field challenges with continuous monitoring and adjustment.


Sujet(s)
Poulets , Maladie de Newcastle , Virus de la maladie de Newcastle , Maladies de la volaille , Animaux , Maladie de Newcastle/virologie , Poulets/virologie , Femelle , Virus de la maladie de Newcastle/physiologie , Maladies de la volaille/virologie , Égypte , Microclimat , Température
2.
Vet Microbiol ; 295: 110163, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38959807

RÉSUMÉ

Avian influenza virus (AIV) infection and vaccination against live attenuated infectious bronchitis virus (aIBV) are frequent in poultry worldwide. Here, we evaluated the clinical effect of H9N2 subtype AIV and QX genotype aIBV co-infection in specific-pathogen-free (SPF) white leghorn chickens and explored the potential mechanisms underlying the observed effects using by 4D-FastDIA-based proteomics. The results showed that co-infection of H9N2 AIV and QX aIBV increased mortality and suppressed the growth of SPF chickens. In particular, severe lesions in the kidneys and slight respiratory signs similar to the symptoms of virulent QX IBV infection were observed in some co-infected chickens, with no such clinical signs observed in single-infected chickens. The replication of H9N2 AIV was significantly enhanced in both the trachea and kidneys, whereas there was only a slight effect on the replication of the QX aIBV. Proteomics analysis showed that the IL-17 signaling pathway was one of the unique pathways enriched in co-infected chickens compared to single infected-chickens. A series of metabolism and immune response-related pathways linked with co-infection were also significantly enriched. Moreover, co-infection of the two pathogens resulted in the enrichment of the negative regulation of telomerase activity. Collectively, our study supports the synergistic effect of the two pathogens, and pointed out that aIBV vaccines might increased IBV-associated lesions due to pathogenic co-infections. Exacerbation of the pathogenicity and mortality in H9N2 AIV and QX aIBV co-infected chickens possibly occurred because of an increase in H9N2 AIV replication, the regulation of telomerase activity, and the disturbance of cell metabolism and the immune system.


Sujet(s)
Poulets , Co-infection , Infections à coronavirus , Virus de la bronchite infectieuse , Sous-type H9N2 du virus de la grippe A , Grippe chez les oiseaux , Maladies de la volaille , Animaux , Poulets/virologie , Sous-type H9N2 du virus de la grippe A/pathogénicité , Sous-type H9N2 du virus de la grippe A/génétique , Virus de la bronchite infectieuse/pathogénicité , Virus de la bronchite infectieuse/génétique , Co-infection/virologie , Co-infection/médecine vétérinaire , Grippe chez les oiseaux/virologie , Maladies de la volaille/virologie , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/virologie , Organismes exempts d'organismes pathogènes spécifiques , Réplication virale , Vaccins atténués/immunologie , Génotype , Virulence , Protéomique , Rein/virologie , Rein/anatomopathologie
3.
Appl Microbiol Biotechnol ; 108(1): 414, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38985204

RÉSUMÉ

Airborne animal viral pathogens can rapidly spread and become a global threat, resulting in substantial socioeconomic and health consequences. To prevent and control potential epidemic outbreaks, accurate, fast, and affordable point-of-care (POC) tests are essential. As a proof-of-concept, we have developed a molecular system based on the loop-mediated isothermal amplification (LAMP) technique for avian metapneumovirus (aMPV) detection, an airborne communicable agent mainly infecting turkeys and chickens. For this purpose, a colorimetric system was obtained by coupling the LAMP technique with specific DNA-functionalized AuNPs (gold nanoparticles). The system was validated using 50 different samples (pharyngeal swabs and tracheal tissue) collected from aMPV-infected and non-infected chickens and turkeys. Viral detection can be achieved in about 60 min with the naked eye, with 100% specificity and 87.88% sensitivity for aMPV. In summary, this novel molecular detection system allows suitable virus testing in the field, with accuracy and limit of detection (LOD) values highly close to qRT-PCR-based diagnosis. Furthermore, this system can be easily scalable to a platform for the detection of other viruses, addressing the current gap in the availability of POC tests for viral detection in poultry farming. KEY POINTS: •aMPV diagnosis using RT-LAMP is achieved with high sensitivity and specificity. •Fifty field samples have been visualized using DNA-nanoprobe validation. •The developed system is a reliable, fast, and cost-effective option for POCT.


Sujet(s)
Poulets , Or , Metapneumovirus , Techniques de diagnostic moléculaire , Techniques d'amplification d'acides nucléiques , Infections à Paramyxoviridae , Maladies de la volaille , Sensibilité et spécificité , Metapneumovirus/génétique , Metapneumovirus/isolement et purification , Animaux , Techniques d'amplification d'acides nucléiques/méthodes , Techniques d'amplification d'acides nucléiques/économie , Poulets/virologie , Techniques de diagnostic moléculaire/méthodes , Techniques de diagnostic moléculaire/économie , Infections à Paramyxoviridae/diagnostic , Infections à Paramyxoviridae/médecine vétérinaire , Infections à Paramyxoviridae/virologie , Maladies de la volaille/virologie , Maladies de la volaille/diagnostic , Or/composition chimique , Dindons , Nanoparticules métalliques/composition chimique , Limite de détection , Colorimétrie/méthodes , ADN viral/génétique
4.
J Gen Virol ; 105(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38980150

RÉSUMÉ

Between 2013 and 2017, the A/Anhui/1/13-lineage (H7N9) low-pathogenicity avian influenza virus (LPAIV) was epizootic in chickens in China, causing mild disease, with 616 fatal human cases. Despite poultry vaccination, H7N9 has not been eradicated. Previously, we demonstrated increased pathogenesis in turkeys infected with H7N9, correlating with the emergence of the L217Q (L226Q H3 numbering) polymorphism in the haemagglutinin (HA) protein. A Q217-containing virus also arose and is now dominant in China following vaccination. We compared infection and transmission of this Q217-containing 'turkey-adapted' (ty-ad) isolate alongside the H7N9 (L217) wild-type (wt) virus in different poultry species and investigated the zoonotic potential in the ferret model. Both wt and ty-ad viruses demonstrated similar shedding and transmission in turkeys and chickens. However, the ty-ad virus was significantly more pathogenic than the wt virus in turkeys but not in chickens, causing 100 and 33% mortality in turkeys respectively. Expanded tissue tropism was seen for the ty-ad virus in turkeys but not in chickens, yet the viral cell receptor distribution was broadly similar in the visceral organs of both species. The ty-ad virus required exogenous trypsin for in vitro replication yet had increased replication in primary avian cells. Replication was comparable in mammalian cells, and the ty-ad virus replicated successfully in ferrets. The L217Q polymorphism also affected antigenicity. Therefore, H7N9 infection in turkeys can generate novel variants with increased risk through altered pathogenicity and potential HA antigenic escape. These findings emphasize the requirement for enhanced surveillance and understanding of A/Anhui/1/13-lineage viruses and their risk to different species.


Sujet(s)
Poulets , Furets , Sous-type H7N9 du virus de la grippe A , Grippe chez les oiseaux , Dindons , Animaux , Dindons/virologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/transmission , Sous-type H7N9 du virus de la grippe A/génétique , Sous-type H7N9 du virus de la grippe A/pathogénicité , Poulets/virologie , Virulence , Chine/épidémiologie , Maladies de la volaille/virologie , Maladies de la volaille/transmission , Glycoprotéine hémagglutinine du virus influenza/génétique , Humains , Excrétion virale , Réplication virale , Zoonoses/virologie , Grippe humaine/virologie , Grippe humaine/transmission
5.
Arch Virol ; 169(7): 155, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951272

RÉSUMÉ

Given the high prevalence of avian leukosis virus subgroup K (ALV-K) in chickens in China, the positive rate of ALV-K in local chickens in Henan province was investigated, and the genetic region encoding the glycoprotein gp85 of isolates from positive chickens was analyzed. The positive rate of ALV-K in local chickens in Henan was found to be 87.2% (41/47). Phylogenetic analysis of gp85 sequences revealed six clusters that differed in their host range regions (hr1 and hr2) and variable regions (vr1, vr2, and vr3). Evidence of recombination of hr1, hr2, vr1, vr2, and vr3 was observed between the different clusters. The isolate HN23LS02 appears to have obtained its hr1 and hr2 regions from separate lineages via recombination but without having a significant affect on the replication capacity of the virus.


Sujet(s)
Virus de la leucose aviaire , Leucose aviaire , Poulets , Spécificité d'hôte , Phylogenèse , Maladies de la volaille , Recombinaison génétique , Protéines de l'enveloppe virale , Animaux , Virus de la leucose aviaire/génétique , Virus de la leucose aviaire/classification , Virus de la leucose aviaire/isolement et purification , Poulets/virologie , Leucose aviaire/virologie , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/métabolisme , Maladies de la volaille/virologie , Chine
6.
Virulence ; 15(1): 2379371, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39014540

RÉSUMÉ

The economic losses caused by high pathogenicity (HP) avian influenza viruses (AIV) in the poultry industry worldwide are enormous. Although chickens and turkeys are closely related Galliformes, turkeys are thought to be a bridging host for the adaptation of AIV from wild birds to poultry because of their high susceptibility to AIV infections. HPAIV evolve from low pathogenicity (LP) AIV after circulation in poultry through mutations in different viral proteins, including the non-structural protein (NS1), a major interferon (IFN) antagonist of AIV. At present, it is largely unknown whether the virulence determinants of HPAIV are the same in turkeys and chickens. Previously, we showed that mutations in the NS1 of HPAIV H7N1 significantly reduced viral replication in chickens in vitro and in vivo. Here, we investigated the effect of NS1 on the replication and virulence of HPAIV H7N1 in turkeys after inoculation with recombinant H7N1 carrying a naturally truncated wild-type NS1 (with 224 amino-acid "aa" in length) or an extended NS1 with 230-aa similar to the LP H7N1 ancestor. There were no significant differences in multiple-cycle viral replication or in the efficiency of NS1 in blocking IFN induction in the cell culture. Similarly, all viruses were highly virulent in turkeys and replicated at similar levels in various organs and swabs collected from the inoculated turkeys. These results suggest that NS1 does not play a role in the virulence or replication of HPAIV H7N1 in turkeys and further indicate that the genetic determinants of HPAIV differ in these two closely related galliform species.


Sujet(s)
Poulets , Sous-type H7N1 du virus de la grippe A , Grippe chez les oiseaux , Dindons , Protéines virales non structurales , Tropisme viral , Réplication virale , Animaux , Dindons/virologie , Protéines virales non structurales/génétique , Protéines virales non structurales/métabolisme , Grippe chez les oiseaux/virologie , Sous-type H7N1 du virus de la grippe A/génétique , Sous-type H7N1 du virus de la grippe A/pathogénicité , Poulets/virologie , Virulence , Maladies de la volaille/virologie
7.
PLoS One ; 19(7): e0307100, 2024.
Article de Anglais | MEDLINE | ID: mdl-39012858

RÉSUMÉ

The outbreak of clade 2.3.4.4b H5 highly pathogenic avian influenza (HPAI) in North America that started in 2021 has increased interest in applying vaccination as a strategy to help control and prevent the disease in poultry. Two commercially available vaccines based on the recombinant herpes virus of turkeys (rHVT) vector were tested against a recent North American clade 2.3.4.4b H5 HPAI virus isolate: A/turkey/Indiana/22-003707-003/2022 H5N1 in specific pathogen free white leghorn (WL) chickens and commercial broiler chickens. One rHVT-H5 vaccine encodes a hemagglutinin (HA) gene designed by the computationally optimized broadly reactive antigen method (COBRA-HVT vaccine). The other encodes an HA gene of a clade 2.2 virus (2.2-HVT vaccine). There was 100% survival of both chicken types COBRA-HVT vaccinated groups and in the 2.2-HVT vaccinated groups there was 94.8% and 90% survival of the WL and broilers respectively. Compared to the 2.2-HVT vaccinated groups, WL in the COBRA-HVT vaccinated group shed significantly lower mean viral titers by the cloacal route and broilers shed significantly lower titers by the oropharyngeal route than broilers. Virus titers detected in oral and cloacal swabs were otherwise similar among both vaccine groups and chicken types. To assess antibody-based tests to identify birds that have been infected after vaccination (DIVA-VI), sera collected after the challenge were tested with enzyme-linked lectin assay-neuraminidase inhibition (ELLA-NI) for N1 neuraminidase antibody detection and by commercial ELISA for detection of antibodies to the NP protein. As early as 7 days post challenge (DPC) 100% of the chickens were positive by ELLA-NI. ELISA was less sensitive with a maximum of 75% positive at 10DPC in broilers vaccinated with 2.2-HVT. Both vaccines provided protection from challenge to both types of chickens and ELLA-NI was sensitive at identifying antibodies to the challenge virus therefore should be evaluated further for DIVA-VI.


Sujet(s)
Poulets , Sous-type H5N1 du virus de la grippe A , Vaccins antigrippaux , Grippe chez les oiseaux , Animaux , Poulets/virologie , Poulets/immunologie , Grippe chez les oiseaux/prévention et contrôle , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/immunologie , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/génétique , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Amérique du Nord , Vaccination , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/virologie , Maladies de la volaille/immunologie , Herpèsvirus de type 1 du dindon/immunologie , Herpèsvirus de type 1 du dindon/génétique
8.
BMC Genomics ; 25(1): 629, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38914944

RÉSUMÉ

BACKGROUND: Virome studies on birds, including chickens are relatively scarce, particularly from the African continent. Despite the continuous evolution of RNA viruses and severe losses recorded in poultry from seasonal viral outbreaks, the information on RNA virome composition is even scantier as a result of their highly unstable nature, genetic diversity, and difficulties associated with characterization. Also, information on factors that may modulate the occurrence of some viruses in birds is limited, particularly for domesticated birds. Viral metagenomics through advancements in sequencing technologies, has enabled the characterization of the entire virome of diverse host species using various samples. METHODS: The complex RNA viral constituents present in 27 faecal samples of asymptomatic chickens from a South African farm collected at 3-time points from two independent seasons were determined, and the impact of the chicken's age and collection season on viral abundance and diversity was further investigated. The study utilized the non-invasive faecal sampling method, mRNA viral targeted enrichment steps, a whole transcriptome amplification strategy, Illumina sequencing, and bioinformatics tools. RESULTS: The results obtained revealed a total of 48 viral species spanning across 11 orders, 15 families and 21 genera. Viral RNA families such as Coronaviridae, Picornaviridae, Reoviridae, Astroviridae, Caliciviridae, Picorbirnaviridae and Retroviridae were abundant, among which picornaviruses, demonstrated a 100% prevalence across the three age groups (2, 4 and 7 weeks) and two seasons (summer and winter) of the 27 faecal samples investigated. A further probe into the extent of variation between the different chicken groups investigated indicated that viral diversity and abundance were significantly influenced by age (P = 0.01099) and season (P = 0.00099) between chicken groups, while there was no effect on viral shedding within samples in a group (alpha diversity) for age (P = 0.146) and season (P = 0.242). CONCLUSION: The presence of an exceedingly varied chicken RNA virome, encompassing avian, mammalian, fungal, and dietary-associated viruses, underscores the complexities inherent in comprehending the causation, dynamics, and interspecies transmission of RNA viruses within the investigated chicken population. Hence, chickens, even in the absence of discernible symptoms, can harbour viruses that may exhibit opportunistic, commensal, or pathogenic characteristics.


Sujet(s)
Poulets , Fèces , Métagénomique , ARN viral , Virome , Animaux , Poulets/virologie , République d'Afrique du Sud/épidémiologie , Fèces/virologie , Virome/génétique , Métagénomique/méthodes , ARN viral/génétique , Virus à ARN/génétique , Virus à ARN/classification , Virus à ARN/isolement et purification , Fermes , Métagénome , Saisons
9.
Emerg Microbes Infect ; 13(1): 2364736, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38847071

RÉSUMÉ

Since 2007, h9.4.2.5 has emerged as the most predominant branch of H9N2 avian influenza viruses (AIVs) that affects the majority of the global poultry population. The spread of this viral branch in vaccinated chicken flocks has not been considerably curbed despite numerous efforts. The evolutionary fitness of h9.4.2.5-branched AIVs must consequently be taken into consideration. The glycosylation modifications of hemagglutinin (HA) play a pivotal role in regulating the balance between receptor affinity and immune evasion for influenza viruses. Sequence alignment showed that five major HA glycosylation patterns have evolved over time in h9.4.2.5-branched AIVs. Here, we compared the adaptive phenotypes of five virus mutants with different HA glycosylation patterns. According to the results, the mutant with 6 N-linked glycans displayed the best acid and thermal stability and a better capacity for multiplication, although having a relatively lower receptor affinity than 7 glycans. The antigenic profile between the five mutants revealed a distinct antigenic distance, indicating that variations in glycosylation level have an impact on antigenic drift. These findings suggest that changes in the number of glycans on HA can not only modulate the receptor affinity and antigenicity of H9N2 AIVs, but also affect their stability and multiplication. These adaptive phenotypes may underlie the biological basis for the dominant strain switchover of h9.4.2.5-branched AIVs. Overall, our study provides a systematic insight into how changes in HA glycosylation patterns regulate the evolutionary fitness and epidemiological dominance drift of h9.4.2.5-branched H9N2 AIVs, which will be of great benefit for the glycosylation-dependent vaccine design.


Sujet(s)
Poulets , Glycoprotéine hémagglutinine du virus influenza , Sous-type H9N2 du virus de la grippe A , Grippe chez les oiseaux , Glycosylation , Sous-type H9N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/immunologie , Sous-type H9N2 du virus de la grippe A/métabolisme , Animaux , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Grippe chez les oiseaux/virologie , Poulets/virologie , Mutation , Polyosides/métabolisme , Réplication virale , Cellules rénales canines Madin-Darby , Maladies de la volaille/virologie
10.
Viruses ; 16(6)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38932187

RÉSUMÉ

In 2023, South Africa continued to experience sporadic cases of clade 2.3.4.4b H5N1 high-pathogenicity avian influenza (HPAI) in coastal seabirds and poultry. Active environmental surveillance determined that H5Nx, H7Nx, H9Nx, H11Nx, H6N2, and H12N2, amongst other unidentified subtypes, circulated in wild birds and ostriches in 2023, but that H5Nx was predominant. Genome sequencing and phylogenetic analysis of confirmed H5N1 HPAI cases determined that only two of the fifteen sub-genotypes that circulated in South Africa in 2021-2022 still persisted in 2023. Sub-genotype SA13 remained restricted to coastal seabirds, with accelerated mutations observed in the neuraminidase protein. SA15 caused the chicken outbreaks, but outbreaks in the Paardeberg and George areas, in the Western Cape province, and the Camperdown region of the KwaZulu-Natal province were unrelated to each other, implicating wild birds as the source. All SA15 viruses contained a truncation in the PB1-F2 gene, but in the Western Cape SA15 chicken viruses, PA-X was putatively expressed as a novel isoform with eight additional amino acids. South African clade 2.3.4.4b H5N1 viruses had comparatively fewer markers of virulence and pathogenicity compared to European strains, a possible reason why no spillover to mammals has occurred here yet.


Sujet(s)
Oiseaux , Épidémies de maladies , Génotype , Sous-type H5N1 du virus de la grippe A , Grippe chez les oiseaux , Phylogenèse , République d'Afrique du Sud/épidémiologie , Animaux , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/épidémiologie , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/pathogénicité , Sous-type H5N1 du virus de la grippe A/classification , Sous-type H5N1 du virus de la grippe A/isolement et purification , Oiseaux/virologie , Poulets/virologie , Volaille/virologie , Génome viral , Virulence , Animaux sauvages/virologie , Sialidase/génétique , Protéines virales/génétique
11.
Front Cell Infect Microbiol ; 14: 1370414, 2024.
Article de Anglais | MEDLINE | ID: mdl-38915924

RÉSUMÉ

Fowl adenovirus serotype 4 (FAdV-4) is highly pathogenic to broilers aged 3 to 5 weeks and has caused considerable economic loss in the poultry industry worldwide. FAdV-4 is the causative agent of hydropericardium-hepatitis syndrome (HHS) or hydropericardium syndrome (HPS). The virus targets mainly the liver, and HPS symptoms are observed in infected chickens. This disease was first reported in Pakistan but has now spread worldwide, and over time, various deletions in the FAdV genome and mutations in its major structural proteins have been detected. This review provides detailed information about FAdV-4 genome organization, physiological features, epidemiology, coinfection with other viruses, and host immune suppression. Moreover, we investigated the role and functions of important structural proteins in FAdV-4 pathogenesis. Finally, the potential regulatory effects of FAdV-4 infection on ncRNAs are also discussed.


Sujet(s)
Infections à Adenoviridae , Aviadenovirus , Poulets , Génome viral , Maladies de la volaille , Sérogroupe , Animaux , Poulets/virologie , Maladies de la volaille/virologie , Aviadenovirus/génétique , Aviadenovirus/classification , Aviadenovirus/pathogénicité , Infections à Adenoviridae/médecine vétérinaire , Infections à Adenoviridae/virologie , Co-infection/virologie , Co-infection/médecine vétérinaire
12.
Int J Biol Macromol ; 273(Pt 2): 132901, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38848854

RÉSUMÉ

H5-subtype avian influenza virus (AIV) is globally prevalent and undergoes frequent antigenic drift, necessitating regular updates to vaccines. One of the many influencing elements that cause incompatibility between vaccinations and epidemic strains is the dynamic alteration of glycosylation sites. However, the biological significance of N-glycosylation in the viral evolution and antigenic changes is unclear. Here, we performed a systematic analysis of glycosylation sites on the HA1 subunit of H5N1, providing insights into the changes of primary glycosylation sites, including 140 N, 156 N, and 170 N within the antigenic epitopes of HA1 protein. Multiple recombinant viruses were then generated based on HA genes of historical vaccine strains and deactivated for immunizing SPF chickens. Inactivated recombinant strains showed relatively closer antigenicity compared to which has identical N-glycosylation patterns. The N-glycosylation modification discrepancy highlights the inter-branch antigenic diversity of H5-subtype viruses in avian influenza and serves as a vital foundation for improving vaccination tactics.


Sujet(s)
Variation des antigènes , Poulets , Glycoprotéine hémagglutinine du virus influenza , Sous-type H5N1 du virus de la grippe A , Grippe chez les oiseaux , Glycosylation , Animaux , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Poulets/virologie , Grippe chez les oiseaux/immunologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/prévention et contrôle , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/génétique , Vaccins antigrippaux/immunologie , Épitopes/immunologie , Épitopes/composition chimique , Antigènes viraux/immunologie , Antigènes viraux/génétique
13.
Vet Microbiol ; 295: 110136, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38875877

RÉSUMÉ

This study aimed to analyze the species and abundance of viruses carried by avian species in live poultry markets. In 2022, we collected 196 bird samples from two representative live poultry markets in Guangdong, China, of which 147 were randomly selected for metatranscriptome sequencing to construct a metatranscriptome library. This analysis yielded 17 viral families. Statistical analysis of the virus abundance of the six libraries showed that Picornaviridae, Retroviridae, Coronaviridae, and Othomyxoviridae were more abundant in the J1, J2, and J3 libraries, and Coronaviridae, Retroviridae, and Faviviridae were more abundant in the Y1, Y2, and E1 libraries. Finally, samples were screened using nested PCR and three viruses were identified. The positive results combined with high-throughput sequencing abundance data showed a positive correlation between virus abundance and the number of positive samples. This study provides scientific data to support the diagnosis and prevention of avian viral diseases.


Sujet(s)
Séquençage nucléotidique à haut débit , Maladies de la volaille , Volaille , Virus , Animaux , Séquençage nucléotidique à haut débit/médecine vétérinaire , Chine/épidémiologie , Volaille/virologie , Maladies de la volaille/virologie , Maladies de la volaille/épidémiologie , Virus/génétique , Virus/isolement et purification , Virus/classification , Poulets/virologie
14.
Virology ; 597: 110154, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38917693

RÉSUMÉ

To determine the pathogenicity of two different genotypes of avian hepatitis E strains in two species of birds, a total of thirty healthy 12-week-old birds were used. After inoculation, fecal virus shedding, viremia, seroconversion, serum alanine aminotransferase (ALT) increases and liver lesions were evaluated. The results revealed that CHN-GS-aHEV and CaHEV could both infect Hy-Line hens and silkie fowls, respectively. Compared to the original avian HEV strain, the cross-infected virus exhibited a delay of 2 weeks and 1 week in emerged seroconversion, viremia, fecal virus shedding, and increased ALT level, and also showed mild liver lesions. These findings suggested that CHN-GS-aHEV may have circulated in chickens. Overall, these two different genotypes of avian HEV showed some variant pathogenicity in different bird species. This study provides valuable data for further analysis of the epidemic conditions of two avian HEVs in Hy-Line hens and silkie fowls.


Sujet(s)
Poulets , Génotype , Hépatite virale animale , Hepevirus , Maladies de la volaille , Excrétion virale , Animaux , Poulets/virologie , Maladies de la volaille/virologie , Hepevirus/génétique , Hepevirus/pathogénicité , Hepevirus/isolement et purification , Hepevirus/classification , Hépatite virale animale/virologie , Hépatite virale animale/anatomopathologie , Femelle , Fèces/virologie , Foie/virologie , Foie/anatomopathologie , Virémie/médecine vétérinaire , Virémie/virologie , Infections à virus à ARN/médecine vétérinaire , Infections à virus à ARN/virologie , Virulence , Alanine transaminase/sang
15.
Virology ; 597: 110121, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38917688

RÉSUMÉ

The H7 subtype avian influenza viruses are circulating widely worldwide, causing significant economic losses to the poultry industry and posing a serious threat to human health. In 2019, H7N2 and H7N9 co-circulated in Chinese poultry, yet the risk of H7N2 remained unclear. We isolated and sequenced four H7N2 viruses from chickens, revealing them as novel reassortants with H7N9-derived HA, M, NS genes and H9N2-derived PB2, PB1, PA,NP, NA genes. To further explore the key segment of pathogenicity, H7N2-H7N9NA and H7N2-H9N2HA single-substitution were constructed. Pathogenicity study showed H7N2 isolates to be highly pathogenic in chickens, with H7N2-H7N9NA slightly weaker than H7N2-Wild type. Transcriptomic analysis suggested that H7N9-derived HA genes primarily drove the high pathogenicity of H7N2 isolates, eliciting a strong inflammatory response. These findings underscored the increased threat posed by reassorted H7N2 viruses to chickens, emphasizing the necessity of long-term monitoring of H7 subtype avian influenza viruses.


Sujet(s)
Poulets , Sous-type H7N2 du virus de la grippe A , Sous-type H7N9 du virus de la grippe A , Grippe chez les oiseaux , Virus recombinants , Animaux , Poulets/virologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/transmission , Sous-type H7N9 du virus de la grippe A/génétique , Sous-type H7N9 du virus de la grippe A/pathogénicité , Sous-type H7N9 du virus de la grippe A/isolement et purification , Virus recombinants/pathogénicité , Virus recombinants/génétique , Sous-type H7N2 du virus de la grippe A/pathogénicité , Sous-type H7N2 du virus de la grippe A/génétique , Maladies de la volaille/virologie , Maladies de la volaille/transmission , Virulence , Phylogenèse , Sous-type H9N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/pathogénicité , Sous-type H9N2 du virus de la grippe A/physiologie , Chine
16.
Parasit Vectors ; 17(1): 262, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886805

RÉSUMÉ

BACKGROUND: In recent years the Asian bush mosquito Aedes japonicus has invaded Europe, including the Netherlands. This species is a known vector for a range of arboviruses, possibly including West Nile virus (WNV). As WNV emerged in the Netherlands in 2020, it is important to investigate the vectorial capacity of mosquito species present in the Netherlands to estimate the risk of future outbreaks and further spread of the virus. Therefore, this study evaluates the potential role of Ae. japonicus in WNV transmission and spillover from birds to dead-end hosts in the Netherlands. METHODS: We conducted human landing collections in allotment gardens (Lelystad, the Netherlands) in June, August and September 2021 to study the diurnal and seasonal host-seeking behaviour of Ae. japonicus. Furthermore, their host preference in relation to birds using live chicken-baited traps was investigated. Vector competence of field-collected Ae. japonicus mosquitoes for two isolates of WNV at two different temperatures was determined. Based on the data generated from these studies, we developed a Susceptible-Exposed-Infectious-Recovered (SEIR) model to calculate the risk of WNV spillover from birds to humans via Ae. japonicus, under the condition that the virus is introduced and circulates in an enzootic cycle in a given area. RESULTS: Our results show that Ae. japonicus mosquitoes are actively host seeking throughout the day, with peaks in activity in the morning and evening. Their abundance in August was higher than in June and September. For the host-preference experiment, we documented a small number of mosquitoes feeding on birds: only six blood-fed females were caught over 4 full days of sampling. Finally, our vector competence experiments with Ae. japonicus compared to its natural vector Culex pipiens showed a higher infection and transmission rate when infected with a local, Dutch, WNV isolate compared to a Greek isolate of the virus. Interestingly, we also found a small number of infected Cx. pipiens males with virus-positive leg and saliva samples. CONCLUSIONS: Combining the field and laboratory derived data, our model predicts that Ae. japonicus could act as a spillover vector for WNV and could be responsible for a high initial invasion risk of WNV when present in large numbers.


Sujet(s)
Aedes , Vecteurs moustiques , Fièvre à virus West Nile , Virus du Nil occidental , Animaux , Aedes/virologie , Aedes/physiologie , Pays-Bas/épidémiologie , Vecteurs moustiques/virologie , Vecteurs moustiques/physiologie , Virus du Nil occidental/physiologie , Fièvre à virus West Nile/transmission , Fièvre à virus West Nile/virologie , Humains , Femelle , Oiseaux/virologie , Poulets/virologie , Comportement de recherche d'hôte , Saisons
17.
Vet Immunol Immunopathol ; 273: 110791, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38824909

RÉSUMÉ

Infectious bronchitis virus (IBV) strains of the Delmarva (DMV)/1639 genotype have been causing false layer syndrome (FLS) in the Eastern Canadian layer operations since the end of 2015. FLS is characterized by the development of cystic oviducts in layer pullets infected at an early age. Currently, there are no homologous vaccines for the control of this IBV genotype. Our previous research showed that a heterologous vaccination regimen incorporating Massachusetts (Mass) and Connecticut (Conn) IBV types protects layers against DMV/1639 genotype IBV. The aim of this study was to investigate the role of maternal antibodies conferred by breeders received the same vaccination regimen in the protection against the development of DMV/1639-induced FLS in pullets. Maternal antibody-positive (MA+) and maternal antibody-negative (MA-) female progeny chicks were challenged at 1 day of age and kept under observation for 16 weeks. Oviductal cystic formations were observed in 3 of 14 birds (21.4 %) in the MA- pullets, while the lesions were notably absent in the MA+ pullets. Milder histopathological lesions were observed in the examined tissues of the MA+ pullets. However, the maternal derived immunity failed to demonstrate protection against the damage to the tracheal ciliary activity, viral shedding, and viral tissue distribution. Overall, this study underscores the limitations of maternal derived immunity in preventing certain aspects of viral pathogenesis, emphasizing the need for comprehensive strategies to address different aspects of IBV infection.


Sujet(s)
Anticorps antiviraux , Poulets , Infections à coronavirus , Virus de la bronchite infectieuse , Maladies de la volaille , Vaccins antiviraux , Animaux , Virus de la bronchite infectieuse/immunologie , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/immunologie , Maladies de la volaille/virologie , Poulets/immunologie , Poulets/virologie , Femelle , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Infections à coronavirus/prévention et contrôle , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/immunologie , Infections à coronavirus/virologie , Immunité acquise d'origine maternelle , Trachée/immunologie , Trachée/virologie , Oviductes/immunologie , Oviductes/anatomopathologie , Oviductes/virologie
18.
PLoS Pathog ; 20(5): e1012261, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38805555

RÉSUMÉ

Marek's disease virus (MDV) vaccines were the first vaccines that protected against cancer. The avirulent turkey herpesvirus (HVT) was widely employed and protected billions of chickens from a deadly MDV infection. It is also among the most common vaccine vectors providing protection against a plethora of pathogens. HVT establishes latency in T-cells, allowing the vaccine virus to persist in the host for life. Intriguingly, the HVT genome contains telomeric repeat arrays (TMRs) at both ends; however, their role in the HVT life cycle remains elusive. We have previously shown that similar TMRs in the MDV genome facilitate its integration into host telomeres, which ensures efficient maintenance of the virus genome during latency and tumorigenesis. In this study, we investigated the role of the TMRs in HVT genome integration, latency, and reactivation in vitro and in vivo. Additionally, we examined HVT infection of feather follicles. We generated an HVT mutant lacking both TMRs (vΔTMR) that efficiently replicated in cell culture. We could demonstrate that wild type HVT integrates at the ends of chromosomes containing the telomeres in T-cells, while integration was severely impaired in the absence of the TMRs. To assess the role of TMRs in vivo, we infected one-day-old chickens with HVT or vΔTMR. vΔTMR loads were significantly reduced in the blood and hardly any virus was transported to the feather follicle epithelium where the virus is commonly shed. Strikingly, latency in the spleen and reactivation of the virus were severely impaired in the absence of the TMRs, indicating that the TMRs are crucial for the establishment of latency and reactivation of HVT. Our findings revealed that the TMRs facilitate integration of the HVT genome into host chromosomes, which ensures efficient persistence in the host, reactivation, and transport of the virus to the skin.


Sujet(s)
Poulets , Maladie de Marek , Télomère , Intégration virale , Latence virale , Animaux , Poulets/virologie , Télomère/génétique , Télomère/virologie , Maladie de Marek/virologie , Maladie de Marek/immunologie , Maladie de Marek/prévention et contrôle , Vecteurs génétiques , Herpèsvirus de type 1 du dindon/génétique , Herpèsvirus de type 1 du dindon/immunologie , Vaccins contre la maladie de Marek/immunologie , Vaccins contre la maladie de Marek/génétique , Génome viral , Herpèsvirus aviaire de type 2/génétique , Herpèsvirus aviaire de type 2/immunologie , Séquences répétées d'acides nucléiques , Maladies de la volaille/virologie , Maladies de la volaille/immunologie , Maladies de la volaille/prévention et contrôle
19.
J Wildl Dis ; 60(3): 774-778, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38717893

RÉSUMÉ

Ornithologic study skins are specimens of avian skins that have been preserved by drying after removing the viscera and muscle. Because of the high value of study skins for scientific studies, specimens are shared among researchers. There is concern that study skins might be contaminated with high-consequence diseases such as highly pathogenic avian influenza virus (HPAIV) or Newcastle disease virus (NDV). To mitigate risk, thermal or chemical treatment of study skins may be required before transfer; however, such treatments might damage the specimens. Therefore, a study was conducted to evaluate the duration of infectivity of HPAIV and NDV in study skins prepared from infected chickens (Gallus gallus). Study skins were prepared from 10 chickens infected with each virus. Skin and feather pulp samples were taken at the time of study skin preparation to establish starting titers. Mean starting titers in the skin was 4.2 log10 and 5.1 log10 50% egg infectious doses (EID50) for HPAIV and NDV groups respectively, and were 6.7 log10 EID50 for HPAIV, and 6.4 log10 EID50 for NDV in feather pulp. Samples were collected at 2 and 4 wk of drying to quantify viable virus. At 2 wk, fewer samples had detectable virus and mean titers were 1.8 log10 (skin) and 2.1 log10 (feathers) EID50 for HPAIV, and 1.7 log10 (skin) and 3.5 log10 (feathers) EID50 for NDV. At 4 wk viable virus could not be detected in either tissue type.


Sujet(s)
Poulets , Virus de la grippe A , Grippe chez les oiseaux , Maladie de Newcastle , Virus de la maladie de Newcastle , Peau , Animaux , Virus de la maladie de Newcastle/pathogénicité , Grippe chez les oiseaux/virologie , Maladie de Newcastle/virologie , Poulets/virologie , Peau/virologie , Virus de la grippe A/pathogénicité , Manipulation d'échantillons/médecine vétérinaire , Facteurs temps
20.
Adv Exp Med Biol ; 1451: 55-74, 2024.
Article de Anglais | MEDLINE | ID: mdl-38801571

RÉSUMÉ

The complex cytoplasmic DNA virus known as the fowlpox virus (FWPV) is a member of the avipoxvirus genus, Subfamily Chordopoxvirinae, and Family Poxviridae. The large genome size of FWPV makes it a potential vector for the creation of vaccines against a range of serious veterinary and human ailments. It also allows for multiple gene insertion and the generation of abortive infection in mammalian cells. The virus, which causes fowlpox in chickens and turkeys, is mainly transmitted to poultry through aerosols or biting insects. Fowlpox is a highly contagious disease that affects both domestic and wild birds, causing cutaneous and/or diphtheritic illnesses. To control the illness, strict hygiene practices and immunization with FWPV attenuated strains or antigenically similar pigeon pox virus vaccines are employed. Recent years have seen an increase in fowlpox outbreaks in chicken flocks, primarily due to the introduction of novel forms of FWPV. It is believed that the pathogenic characteristics of these strains are enhanced by the integration of reticuloendotheliosis virus sequences of variable lengths into the FWPV genome. The standard laboratory diagnosis of FPV involves histopathological analysis, electron microscopy, virus isolation on chorioallantoic membrane (CAM) of embryonated chicken eggs or cell cultures, and serologic techniques. For quick and consistent diagnosis, polymerase chain reaction (PCR) has proven to be the most sensitive method. PCR is used in concert with restriction endonuclease enzyme analysis (REA) to identify, differentiate, and characterize the molecular makeup of isolates of the fowlpox virus. Sequencing of the amplified fragments is then done.


Sujet(s)
Virus de la variole de la volaille , Variole aviaire , Virus de la variole de la volaille/génétique , Animaux , Variole aviaire/virologie , Poulets/virologie , Génome viral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE