Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.312
1.
Viruses ; 16(5)2024 05 10.
Article En | MEDLINE | ID: mdl-38793634

Avian influenza viruses (AIVs) of the H5 subtype rank among the most serious pathogens, leading to significant economic losses in the global poultry industry and posing risks to human health. Therefore, rapid and accurate virus detection is crucial for the prevention and control of H5 AIVs. In this study, we established a novel detection method for H5 viruses by utilizing the precision of CRISPR/Cas12a and the efficiency of RT-RPA technologies. This assay facilitates the direct visualization of detection results through blue light and lateral flow strips, accurately identifying H5 viruses with high specificity and without cross-reactivity against other AIV subtypes, NDV, IBV, and IBDV. With detection thresholds of 1.9 copies/µL (blue light) and 1.9 × 103 copies/µL (lateral flow strips), our method not only competes with but also slightly surpasses RT-qPCR, demonstrating an 80.70% positive detection rate across 81 clinical samples. The RT-RPA/CRISPR-based detection method is characterized by high sensitivity, specificity, and independence from specialized equipment. The immediate field applicability of the RT-RPA/CRISPR approach underscores its importance as an effective tool for the early detection and management of outbreaks caused by the H5 subtype of AIVs.


CRISPR-Cas Systems , Influenza in Birds , Sensitivity and Specificity , Animals , Influenza in Birds/virology , Influenza in Birds/diagnosis , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/classification , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza A virus/classification , Poultry/virology , Poultry Diseases/virology , Poultry Diseases/diagnosis , Chickens/virology , Birds/virology
2.
Viruses ; 16(5)2024 05 15.
Article En | MEDLINE | ID: mdl-38793663

Marek's disease (MD), caused by gallid alphaherpesvirus 2 (GaAHV2) or Marek's disease herpesvirus (MDV), is a devastating disease in chickens characterized by the development of lymphomas throughout the body. Vaccine strains used against MD include gallid alphaherpesvirus 3 (GaAHV3), a non-oncogenic chicken alphaherpesvirus homologous to MDV, and homologous meleagrid alphaherpesvirus 1 (MeAHV1) or turkey herpesvirus (HVT). Previous work has shown most of the MDV gC produced during in vitro passage is secreted into the media of infected cells although the predicted protein contains a transmembrane domain. We formerly identified two alternatively spliced gC mRNAs that are secreted during MDV replication in vitro, termed gC104 and gC145 based on the size of the intron removed for each UL44 (gC) transcript. Since gC is conserved within the Alphaherpesvirinae subfamily, we hypothesized GaAHV3 (strain 301B/1) and HVT also secrete gC due to mRNA splicing. To address this, we collected media from 301B/1- and HVT-infected cell cultures and used Western blot analyses and determined that both 301B/1 and HVT produced secreted gC. Next, we extracted RNAs from 301B/1- and HVT-infected cell cultures and chicken feather follicle epithelial (FFE) skin cells. RT-PCR analyses confirmed one splicing variant for 301B/1 gC (gC104) and two variants for HVT gC (gC104 and gC145). Interestingly, the splicing between all three viruses was remarkably conserved. Further analysis of predicted and validated mRNA splicing donor, branch point (BP), and acceptor sites suggested single nucleotide polymorphisms (SNPs) within the 301B/1 UL44 transcript sequence resulted in no gC145 being produced. However, modification of the 301B/1 gC145 donor, BP, and acceptor sites to the MDV UL44 sequences did not result in gC145 mRNA splice variant, suggesting mRNA splicing is more complex than originally hypothesized. In all, our results show that mRNA splicing of avian herpesviruses is conserved and this information may be important in developing the next generation of MD vaccines or therapies to block transmission.


Chickens , RNA Splicing , Viral Envelope Proteins , Animals , Chickens/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Marek Disease/virology , Mardivirus/genetics , Mardivirus/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , Herpesvirus 2, Gallid/genetics , Alternative Splicing , Antigens, Viral
3.
Viruses ; 16(5)2024 05 16.
Article En | MEDLINE | ID: mdl-38793675

The emergence of new virulent genotypes and the continued genetic drift of Newcastle disease virus (NDV) implies that distinct genotypes of NDV are simultaneously evolving in different geographic locations across the globe, including throughout Africa, where NDV is an important veterinary pathogen. Expanding the genomic diversity of NDV increases the possibility of diagnostic and vaccine failures. In this review, we systematically analyzed the genetic diversity of NDV genotypes in Africa using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Information published between 1999 and 2022 were used to obtain the genetic background of different genotypes of NDV and their geographic distributions in Africa. The following genotypes were reported in Africa: I, II, III, IV, V, VI, VII, VIII, XI, XIII, XIV, XVII, XVIII, XX, and XXI. A new putative genotype has been detected in the Democratic Republic of the Congo. However, of 54 African countries, only 26 countries regularly report information on NDV outbreaks, suggesting that this number may be vastly underestimated. With eight different genotypes, Nigeria is the country with the greatest genotypic diversity of NDV among African countries. Genotype VII is the most prevalent group of NDV in Africa, which was reported in 15 countries. A phylogeographic analysis of NDV sequences revealed transboundary transmission of the virus in Eastern Africa, Western and Central Africa, and in Southern Africa. A regional and continental collaboration is recommended for improved NDV risk management in Africa.


Genetic Variation , Genotype , Newcastle Disease , Newcastle disease virus , Phylogeny , Newcastle disease virus/genetics , Newcastle disease virus/classification , Newcastle disease virus/isolation & purification , Newcastle Disease/virology , Newcastle Disease/epidemiology , Africa/epidemiology , Animals , Genome, Viral , Vaccination/veterinary , Chickens/virology , Viral Vaccines/genetics , Viral Vaccines/immunology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Phylogeography
4.
Adv Exp Med Biol ; 1451: 55-74, 2024.
Article En | MEDLINE | ID: mdl-38801571

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.


Fowlpox virus , Fowlpox , Fowlpox virus/genetics , Animals , Fowlpox/virology , Chickens/virology , Genome, Viral
5.
Virology ; 595: 110094, 2024 Jul.
Article En | MEDLINE | ID: mdl-38692133

Stress-induced immunosuppression (SIIS) is one of common problems in the intensive poultry industry, affecting the effect of vaccine immunization and leading to high incidences of diseases. In this study, the expression characteristics and regulatory mechanisms of miR-214 in the processes of SIIS and its influence on the immune response to avian influenza virus (AIV) vaccine in chicken were explored. The qRT-PCR results showed that serum circulating miR-214 was significantly differentially expressed (especially on 2, 5, and 28 days post immunization (dpi)) in the processes, so had the potential as a molecular marker. MiR-214 expressions from multiple tissues were closely associated with the changes in circulating miR-214 expression levels. MiR-214-PTEN regulatory network was a potential key regulatory mechanism for the heart, bursa of Fabricius, and glandular stomach to participate in the process of SIIS affecting AIV immune response. This study can provide references for further understanding of stress affecting immune response.


Chickens , Influenza Vaccines , Influenza in Birds , MicroRNAs , PTEN Phosphohydrolase , Stress, Physiological , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Chickens/virology , Influenza Vaccines/immunology , Influenza in Birds/virology , Influenza in Birds/immunology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Poultry Diseases/virology , Poultry Diseases/immunology , Immune Tolerance , Signal Transduction , Influenza A virus/immunology
6.
J Med Microbiol ; 73(5)2024 May.
Article En | MEDLINE | ID: mdl-38771617

Infectious bronchitis virus (IBV) is a highly contagious avian Gammacoronavirus that affects mainly chickens (Gallus gallus) but can circulate in other avian species. IBV constitutes a significant threat to the poultry industry, causing reduced egg yield, growth and mortality levels that can vary in impact. The virus can be transmitted horizontally by inhalation or direct/indirect contact with infected birds or contaminated fomites, vehicles, farm personnel and litter (Figure 1). The error-prone viral polymerase and recombination mechanisms mean diverse viral population results, with multiple genotypes, serotypes, pathotypes and protectotypes. This significantly complicates control and mitigation strategies based on vigilance in biosecurity and the deployment of vaccination.


Chickens , Coronavirus Infections , Infectious bronchitis virus , Poultry Diseases , Infectious bronchitis virus/genetics , Infectious bronchitis virus/classification , Infectious bronchitis virus/physiology , Animals , Chickens/virology , Poultry Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/veterinary
7.
Nat Commun ; 15(1): 3494, 2024 May 01.
Article En | MEDLINE | ID: mdl-38693163

H9N2 avian influenza viruses (AIVs) are a major concern for the poultry sector and human health in countries where this subtype is endemic. By fitting a model simulating H9N2 AIV transmission to data from a field experiment, we characterise the epidemiology of the virus in a live bird market in Bangladesh. Many supplied birds arrive already exposed to H9N2 AIVs, resulting in many broiler chickens entering the market as infected, and many indigenous backyard chickens entering with pre-existing immunity. Most susceptible chickens become infected within one day spent at the market, owing to high levels of viral transmission within market and short latent periods, as brief as 5.3 hours. Although H9N2 AIV transmission can be substantially reduced under moderate levels of cleaning and disinfection, effective risk mitigation also requires a range of additional interventions targeting markets and other nodes along the poultry production and distribution network.


Chickens , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Animals , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/transmission , Influenza in Birds/epidemiology , Influenza in Birds/virology , Chickens/virology , Bangladesh/epidemiology , Poultry Diseases/transmission , Poultry Diseases/virology , Poultry Diseases/epidemiology , Models, Biological
8.
PLoS One ; 19(5): e0303371, 2024.
Article En | MEDLINE | ID: mdl-38728352

Marek's disease (MD) is an important neoplastic disease caused by serotype 1 Marek's disease virus (MDV-1), which results in severe economic losses worldwide. Despite vaccination practices that have controlled the MD epidemic, current increasing MD-suspected cases indicate the persistent viral infections circulating among vaccinated chicken farms in many countries. However, the lack of available information about phylogeny and molecular characterization of circulating MDV-1 field strains in Taiwan reveals a potential risk in MD outbreaks. This study investigated the genetic characteristics of 18 MDV-1 strains obtained from 17 vaccinated chicken flocks in Taiwan between 2018 and 2020. Based on the sequences of the meq oncogene, the phylogenetic analysis demonstrated that the circulating Taiwanese MDV-1 field strains were predominantly in a single cluster that showed high similarity with strains from countries of the East Asian region. Because the strains were obtained from CVI988/Rispens vaccinated chicken flocks and the molecular characteristics of the Meq oncoprotein showed features like vvMDV and vv+MDV strains, the circulating Taiwanese MDV-1 field strains may have higher virulence compared with vvMDV pathotype. In conclusion, the data presented demonstrates the circulation of hypervirulent MDV-1 strains in Taiwan and highlights the importance of routine surveillance and precaution strategies in response to the emergence of enhanced virulent MDV-1.


Chickens , Herpesvirus 2, Gallid , Marek Disease , Oncogene Proteins, Viral , Phylogeny , Animals , Chickens/virology , Taiwan/epidemiology , Marek Disease/virology , Marek Disease/prevention & control , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/pathogenicity , Virulence/genetics , Oncogene Proteins, Viral/genetics , Poultry Diseases/virology , Poultry Diseases/epidemiology , Poultry Diseases/prevention & control , Marek Disease Vaccines/genetics , Marek Disease Vaccines/immunology , Vaccination/veterinary
9.
J Virol Methods ; 327: 114942, 2024 Jun.
Article En | MEDLINE | ID: mdl-38670532

H5, H7 and H9 are the major subtypes of avian influenza virus (AIV) that cause economic losses in the poultry industry and sporadic zoonotic infection. Early detection of AIV is essential for preventing disease spread. Therefore, molecular diagnosis and subtyping of AIV via real-time RT-PCR (rRT-PCR) is preferred over other classical diagnostic methods, such as egg inoculation, RT-PCR and HI test, due to its high sensitivity, specificity and convenience. The singleplex rRT-PCRs for the Matrix, H5 and H7 gene used for the national surveillance program in Korea have been developed in 2017; however, these methods were not designed for multiplexing, and does not reflect the sequences of currently circulating strains completely. In this study, the multiplex H5/7/9 rRT-PCR assay was developed with sets of primers and probe updated or newly designed to simultaneously detect the H5, H7 and H9 genes. Multiplex H5/7/9 rRT-PCR showed 100% specificity without cross-reactivity with other subtypes of AIVs and avian disease-causing viruses or bacteria, and the limit of detection was 1-10 EID50/0.1 ml (50% egg infectious dose). Artificial mixed infections with the three different subtypes could be detected accurately with high analytical sensitivity even under highly biased relative molecular ratios by balancing the reactivities of each subtype by modifying the concentration of the primers and probes. The multiplex H5/7/9 rRT-PCR assay developed in this study could be a useful tool for large-scale surveillance programs for viral detection as well as subtyping due to its high specificity, sensitivity and robustness in discriminating viruses in mixed infections, and this approach would greatly decrease the time, cost, effort and chance of cross-contamination compared to the conventional method of testing three subtypes by different singleplex rRT-PCR methods in parallel or in series.


Chickens , Influenza A virus , Influenza in Birds , Multiplex Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Influenza in Birds/virology , Influenza in Birds/diagnosis , Animals , Multiplex Polymerase Chain Reaction/methods , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Real-Time Polymerase Chain Reaction/methods , Chickens/virology , Republic of Korea , Poultry Diseases/virology , Poultry Diseases/diagnosis , DNA Primers/genetics , Poultry/virology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Birds/virology
10.
Vet Microbiol ; 293: 110094, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636175

Infectious bursa disease (IBD) is an acute, highly contactable, lethal, immunosuppressive infectious disease caused by the Infectious bursa disease virus (IBDV). Currently, the emerged novel variant IBDV (nVarIBDV) and the sustainedly prevalent very virulent IBDV (vvIBDV) are the two most prevalent strains of IBDV in China. The antigenic properties of the two prevalent strains differed significantly, which led to the escape of nVarIBDV from the immune protection provided by the existing vvIBDV vaccine. However, the molecular basis of the nVarIBDV immune escape remains unclear. In this study, we demonstrated, for the first time, that residues 252, 254, and 256 in the PDE of VP2 are involved in the immune escape of the emerging nVarIBDV. Firstly, the IFA-mediated antigen-antibody affinity assay showed that PBC and PDE of VP2 could affect the affinity of vvIBDV antiserum to VP2, of which PDE was more significant. The key amino acids of PDE influencing the antigen-antibody affinity were also identified, with G254N being the most significant, followed by V252I and I256V. Then the mutated virus with point or combined mutations was rescued by reverse genetics. it was further demonstrated that mutations of V252I, G254N, and I256V in PDE could individually or collaboratively reduce antigen-antibody affinity and interfere with antiserum neutralization, with G254N being the most significant. This study revealed the reasons for the widespread prevalence of nVarIBDV in immunized chicken flocks and provided innovative ideas for designing novel vaccines that match the antigen of the epidemic strain.


Birnaviridae Infections , Capsid Proteins , Chickens , Immune Evasion , Infectious bursal disease virus , Poultry Diseases , Infectious bursal disease virus/genetics , Infectious bursal disease virus/immunology , Animals , Chickens/virology , Capsid Proteins/genetics , Capsid Proteins/immunology , Poultry Diseases/virology , Poultry Diseases/immunology , Birnaviridae Infections/veterinary , Birnaviridae Infections/virology , Birnaviridae Infections/immunology , China , Antibodies, Viral/immunology , Mutation , Viral Vaccines/immunology , Viral Structural Proteins
11.
Viruses ; 16(4)2024 03 26.
Article En | MEDLINE | ID: mdl-38675851

Avian metapneumovirus (aMPV), classified within the Pneumoviridae family, wreaks havoc on poultry health. It typically causes upper respiratory tract and reproductive tract infections, mainly in turkeys, chickens, and ducks. Four subtypes of AMPV (A, B, C, D) and two unclassified subtypes have been identified, of which subtypes A and B are widely distributed across the world. In January 2024, an outbreak of severe respiratory disease occurred on turkey and chicken farms across different states in the US. Metagenomics sequencing of selected tissue and swab samples confirmed the presence of aMPV subtype B. Subsequently, all samples were screened using an aMPV subtype A and B multiplex real-time RT-PCR kit. Of the 221 farms, 124 (56%) were found to be positive for aMPV-B. All samples were negative for subtype A. Six whole genomes were assembled, five from turkeys and one from chickens; all six assembled genomes showed 99.29 to 99.98% nucleotide identity, indicating a clonal expansion event for aMPV-B within the country. In addition, all six sequences showed 97.74 to 98.58% nucleotide identity with previously reported subtype B sequences, e.g., VCO3/60616, Hungary/657/4, and BR/1890/E1/19. In comparison to these two reference strains, the study sequences showed unique 49-62 amino acid changes across the genome, with maximum changes in glycoprotein (G). One unique AA change from T (Threonine) to I (Isoleucine) at position 153 in G protein was reported only in the chicken aMPV sequence, which differentiated it from turkey sequences. The twelve unique AA changes along with change in polarity of the G protein may indicate that these unique changes played a role in the adaptation of this virus in the US poultry. This is the first documented report of aMPV subtype B in US poultry, highlighting the need for further investigations into its genotypic characterization, pathogenesis, and evolutionary dynamics.


Genome, Viral , Metapneumovirus , Paramyxoviridae Infections , Phylogeny , Poultry Diseases , Turkeys , Animals , Metapneumovirus/genetics , Metapneumovirus/classification , Metapneumovirus/isolation & purification , Paramyxoviridae Infections/veterinary , Paramyxoviridae Infections/virology , Paramyxoviridae Infections/epidemiology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Turkeys/virology , United States/epidemiology , Chickens/virology , Poultry/virology , Metagenomics , Disease Outbreaks/veterinary
12.
Viruses ; 16(4)2024 03 27.
Article En | MEDLINE | ID: mdl-38675858

Infectious bronchitis virus (IBV) causes a highly contagious respiratory disease in chickens, leading to significant economic losses in the poultry industry worldwide. IBV exhibits a high mutation rate, resulting in the continuous emergence of new variants and strains. A complete genome analysis of IBV is crucial for understanding its characteristics. However, it is challenging to obtain whole-genome sequences from IBV-infected clinical samples due to the low abundance of IBV relative to the host genome. Here, we present a novel approach employing next-generation sequencing (NGS) to directly sequence the complete genome of IBV. Through in silico analysis, six primer pairs were designed to match various genotypes, including the GI-19 lineage of IBV. The primer sets successfully amplified six overlapping fragments by long-range PCR and the size of the amplicons ranged from 3.7 to 6.4 kb, resulting in full coverage of the IBV genome. Furthermore, utilizing Illumina sequencing, we obtained the complete genome sequences of two strains belonging to the GI-19 lineage (QX genotype) from clinical samples, with 100% coverage rates, over 1000 × mean depth coverage, and a high percentage of mapped reads to the reference genomes (96.63% and 97.66%). The reported method significantly improves the whole-genome sequencing of IBVs from clinical samples; thus, it can improve understanding of the epidemiology and evolution of IBVs.


Chickens , Coronavirus Infections , Genome, Viral , Genotype , High-Throughput Nucleotide Sequencing , Infectious bronchitis virus , Phylogeny , Poultry Diseases , Whole Genome Sequencing , Infectious bronchitis virus/genetics , Infectious bronchitis virus/isolation & purification , Infectious bronchitis virus/classification , Animals , Whole Genome Sequencing/methods , Chickens/virology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Coronavirus Infections/epidemiology , RNA, Viral/genetics
13.
Viruses ; 16(4)2024 04 04.
Article En | MEDLINE | ID: mdl-38675905

Highly pathogenic avian influenza (HPAI) H5-viruses are circulating in wild birds and are repeatedly introduced to poultry causing outbreaks in the Netherlands since 2014. The largest epizootic ever recorded in Europe was caused by HPAI H5N1 clade 2.3.4.4b viruses in the period 2021-2022. The recent H5-clade 2.3.4.4 viruses were found to differ in their virulence for chickens and ducks. Viruses causing only mild disease may remain undetected, increasing the risk of virus spread to other farms, wild birds and mammals. We developed in ovo models to determine the virulence of HPAI viruses for chickens and ducks, which are fast and have low costs. The virulence of five contemporary H5-viruses was compared studying replication rate, average time to death and virus spread in the embryo. Remarkable differences in virulence were observed between H5-viruses and between poultry species. The H5N1-2021 virus was found to have a fast replication rate in both the chicken and duck in ovo models, but a slower systemic virus dissemination compared to three other H5-clade 2.3.4.4b viruses. The results show the potential of in ovo models to quickly determine the virulence of novel HPAI viruses, and study potential virulence factors which can help to better guide the surveillance in poultry.


Chickens , Ducks , Influenza in Birds , Virus Replication , Animals , Ducks/virology , Influenza in Birds/virology , Chickens/virology , Virulence , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/genetics , Chick Embryo , Poultry Diseases/virology
14.
Viruses ; 16(4)2024 04 07.
Article En | MEDLINE | ID: mdl-38675911

Zika virus (ZIKV) remains a public health concern, with epidemics in endemic regions and sporadic outbreaks in new areas posing significant threats. Several mosquito-borne flaviviruses that can cause human illness, including West Nile, Usutu, and St. Louis encephalitis, have associations with birds. However, the susceptibility of chickens to ZIKV and their role in viral epidemiology is not currently known. We investigated the susceptibility of chickens to experimental ZIKV infection using chickens ranging from 1-day-old chicks to 6-week-old birds. ZIKV caused no clinical signs in chickens of all age groups tested. Viral RNA was detected in the blood and tissues during the first 5 days post-inoculation in 1-day and 4-day-old chicks inoculated with a high viral dose, but ZIKV was undetectable in 6-week-old birds at all timepoints. Minimal antibody responses were observed in 6-week-old birds, and while present in younger chicks, they waned by 28 days post-infection. Innate immune responses varied significantly between age groups. Robust type I interferon and inflammasome responses were measured in older chickens, while limited innate immune activation was observed in younger chicks. Signal transducer and activator of transcription 2 (STAT2) is a major driver of host restriction to ZIKV, and chicken STAT2 is distinct from human STAT2, potentially contributing to the observed resistance to ZIKV infection. The rapid clearance of the virus in older chickens coincided with an effective innate immune response, highlighting age-dependent susceptibility. Our study indicates that chickens are not susceptible to productive ZIKV infection and are unlikely to play a role in the ZIKV epidemiology.


Chickens , Immunity, Innate , Poultry Diseases , Zika Virus Infection , Zika Virus , Animals , Chickens/virology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Disease Susceptibility , Poultry Diseases/virology , Poultry Diseases/immunology , Age Factors , Antibodies, Viral/blood , RNA, Viral/genetics
15.
Viruses ; 16(4)2024 04 14.
Article En | MEDLINE | ID: mdl-38675946

Infectious bronchitis virus (IBV) is a highly contagious Gammacoronavirus causing moderate to severe respiratory infection in chickens. Understanding the initial antiviral response in the respiratory mucosa is crucial for controlling viral spread. We aimed to characterize the impact of IBV Delmarva (DMV)/1639 and IBV Massachusetts (Mass) 41 at the primary site of infection, namely, in chicken tracheal epithelial cells (cTECs) in vitro and the trachea in vivo. We hypothesized that some elements of the induced antiviral responses are distinct in both infection models. We inoculated cTECs and infected young specific pathogen-free (SPF) chickens with IBV DMV/1639 or IBV Mass41, along with mock-inoculated controls, and studied the transcriptome using RNA-sequencing (RNA-seq) at 3 and 18 h post-infection (hpi) for cTECs and at 4 and 11 days post-infection (dpi) in the trachea. We showed that IBV DMV/1639 and IBV Mass41 replicate in cTECs in vitro and the trachea in vivo, inducing host mRNA expression profiles that are strain- and time-dependent. We demonstrated the different gene expression patterns between in vitro and in vivo tracheal IBV infection. Ultimately, characterizing host-pathogen interactions with various IBV strains reveals potential mechanisms for inducing and modulating the immune response during IBV infection in the chicken trachea.


Chickens , Coronavirus Infections , Gene Expression Profiling , Infectious bronchitis virus , Poultry Diseases , Trachea , Animals , Trachea/virology , Trachea/immunology , Chickens/virology , Infectious bronchitis virus/physiology , Infectious bronchitis virus/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/immunology , Coronavirus Infections/virology , Poultry Diseases/virology , Poultry Diseases/immunology , Poultry Diseases/genetics , Epithelial Cells/virology , Epithelial Cells/immunology , Transcriptome , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Virus Replication , Specific Pathogen-Free Organisms
16.
Microb Pathog ; 190: 106638, 2024 May.
Article En | MEDLINE | ID: mdl-38574829

Autophagy plays an important role in the lifecycle of viruses. However, there is currently a lack of systematic research on the relationship between Infectious Bronchitis Virus (IBV) and autophagy. This study aims to investigate the impact of IBV on autophagy and the role of autophagy in viral replication. We observed that IBV infection increased the expression of microtubule-associated protein 1 light chain 3, a marker of autophagy, decreased the expression of sequestosome 1, and led to elevated intracellular LC3 puncta levels. These findings suggest that IBV infection activates the autophagic process in cells. To investigate the impact of autophagy on the replication of IBV, we utilized rapamycin as an autophagy activator and 3-methyladenine as an autophagy inhibitor. Our results indicate that IBV promotes viral replication by inducing autophagy. Further investigation revealed that IBV induces autophagosome formation by inhibiting the mTOR-ULK1 pathway and activating the activity of vacuolar protein sorting 34 (VPS34), autophagy-related gene 14, and the Beclin-1 complex. VPS34 plays a crucial role in this process, as inhibiting VPS34 protein activity enhances cell proliferation after IBV infection. Additionally, inhibiting VPS34 significantly improves the survival rate of IBV-infected chicks, suppresses IBV replication in the kidney, and alleviates tracheal, lung, and kidney damage caused by IBV infection. In summary, IBV infection can induce autophagy by modulating the mTOR/ULK1 signaling pathway and activating the VPS34 complex, while autophagy serves to promote virus replication.


Autophagy , Chickens , Class III Phosphatidylinositol 3-Kinases , Infectious bronchitis virus , Virus Replication , Infectious bronchitis virus/physiology , Animals , Class III Phosphatidylinositol 3-Kinases/metabolism , Chickens/virology , Coronavirus Infections/virology , Coronavirus Infections/metabolism , Sirolimus/pharmacology , Beclin-1/metabolism , Beclin-1/genetics , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Cell Line , Poultry Diseases/virology , Autophagosomes/metabolism , Autophagosomes/virology , Chlorocebus aethiops , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
17.
Zhonghua Liu Xing Bing Xue Za Zhi ; 45(4): 574-578, 2024 Apr 10.
Article Zh | MEDLINE | ID: mdl-38678355

Objective: To identify a novel reassortant H3N2 avian influenza virus using nanopore sequencing technology and analyze its genetic characteristics. Methods: The positive samples of the H3N2 avian influenza virus, collected from the external environment in the farmers' market of Guangzhou, were cultured in chicken embryos. The whole genome was sequenced by targeted amplification and nanopore sequencing technology. The genetic characteristics were analyzed using bioinformatics software. Results: The phylogenetic trees showed that each gene fragment of the strain belonged to the Eurasian evolutionary branch, and the host source was of avian origin. The HA gene was closely related to the origin of the H3N6 virus. The NA gene was closely related to the H3N2 avian influenza virus from 2017 to 2020. The PB1 gene was closely related to the H5N6 avian influenza virus in Guangxi Zhuang Autonomous Region and Fujian Province from 2016 to 2022 and was not related to the PB1 gene of the H5N6 avian influenza epidemic strain in Guangzhou. The other internal gene fragments had complex sources with significant genetic diversity. Molecular characteristics indicated that the strain exhibited the molecular characteristics of a typical low pathogenic avian influenza virus and tended to bind to the receptors of avian origin. On important protein sites related to biological characteristics, this strain had mutations of PB2-L89V, PB1-L473V, NP-A184K, M1-N30D/T215A, and NS1-P42S/N205S. Conclusions: This study identified a novel reassortant H3N2 avian influenza virus by nanopore sequencing, with the PB1 gene derived from the H5N6 avian influenza virus. The virus had a low ability to spread across species, but further exploration was needed to determine whether its pathogenicity to the host was affected.


Influenza A Virus, H3N2 Subtype , Influenza in Birds , Nanopore Sequencing , Phylogeny , Reassortant Viruses , Animals , Reassortant Viruses/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza in Birds/virology , Influenza in Birds/epidemiology , Genome, Viral , Chick Embryo , Chickens/virology , Viral Proteins/genetics , Genetic Variation
18.
Emerg Microbes Infect ; 13(1): 2343912, 2024 Dec.
Article En | MEDLINE | ID: mdl-38629574

Human infections with the H7N9 influenza virus have been eliminated in China through vaccination of poultry; however, the H7N9 virus has not yet been eradicated from poultry. Carefully analysis of H7N9 viruses in poultry that have sub-optimal immunity may provide a unique opportunity to witness the evolution of highly pathogenic avian influenza virus in the context of vaccination. Between January 2020 and June 2023, we isolated 16 H7N9 viruses from samples we collected during surveillance and samples that were sent to us for disease diagnosis. Genetic analysis indicated that these viruses belonged to a single genotype previously detected in poultry. Antigenic analysis indicated that 12 of the 16 viruses were antigenically close to the H7-Re4 vaccine virus that has been used since January 2022, and the other four viruses showed reduced reactivity with the vaccine. Animal studies indicated that all 16 viruses were nonlethal in mice, and four of six viruses showed reduced virulence in chickens upon intranasally inoculation. Importantly, the H7N9 viruses detected in this study exclusively bound to the avian-type receptors, having lost the capacity to bind to human-type receptors. Our study shows that vaccination slows the evolution of H7N9 virus by preventing its reassortment with other viruses and eliminates a harmful characteristic of H7N9 virus, namely its ability to bind to human-type receptors.


Chickens , Influenza A Virus, H7N9 Subtype , Influenza Vaccines , Influenza in Birds , Vaccination , Animals , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H7N9 Subtype/pathogenicity , Chickens/virology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza in Birds/virology , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Mice , Humans , China , Evolution, Molecular , Influenza, Human/prevention & control , Influenza, Human/virology , Influenza, Human/immunology , Mice, Inbred BALB C , Virulence , Phylogeny , Female , Poultry Diseases/virology , Poultry Diseases/prevention & control , Poultry/virology
19.
Emerg Microbes Infect ; 13(1): 2341142, 2024 Dec.
Article En | MEDLINE | ID: mdl-38581279

H6N6 avian influenza viruses (AIVs) have been widely detected in wild birds, poultry, and even mammals. Recently, H6N6 viruses were reported to be involved in the generation of H5 and H7 subtype viruses. To investigate the emergence, evolutionary pattern, and potential for an epidemic of H6N6 viruses, the complete genomes of 198 H6N6 viruses were analyzed, including 168 H6N6 viruses deposited in the NCBI and GISAID databases from inception to January 2019 and 30 isolates collected from China between November 2014 and January 2019. Using phylogenetic analysis, the 198 strains of H6N6 viruses were identified as 98 genotypes. Molecular clock analysis indicated that the evolution of H6N6 viruses in China was constant and not interrupted by selective pressure. Notably, the laboratory isolates reassorted with six subtype viruses: H6N2, H5N6, H7N9, H5N2, H4N2, and H6N8, resulting in nine novel H6N6 reassortment events. These results suggested that H6N6 viruses can act as an intermediary in the evolution of H5N6, H6N6, and H7N9 viruses. Animal experiments demonstrated that the 10 representative H6N6 viruses showed low pathogenicity in chickens and were capable of infecting mice without prior adaptation. Our findings suggest that H6N6 viruses play an important role in the evolution of AIVs, and it is necessary to continuously monitor and evaluate the potential epidemic of the H6N6 subtype viruses.


Chickens , Evolution, Molecular , Genome, Viral , Influenza A virus , Influenza in Birds , Phylogeny , Reassortant Viruses , Animals , China/epidemiology , Reassortant Viruses/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Influenza in Birds/virology , Influenza in Birds/epidemiology , Mice , Chickens/virology , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Genotype , Humans
20.
J Virol ; 98(4): e0024824, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38466094

The H9N2 avian influenza virus (AIV) represents a significant risk to both the poultry industry and public health. Our surveillance efforts in China have revealed a growing trend of recent H9N2 AIV strains exhibiting a loss of hemagglutination activity at 37°C, posing challenges to detection and monitoring protocols. This study identified a single K141N substitution in the hemagglutinin (HA) glycoprotein as the culprit behind this diminished hemagglutination activity. The study evaluated the evolutionary dynamics of residue HA141 and studied the impact of the N141K substitution on aspects such as virus growth, thermostability, receptor-binding properties, and antigenic properties. Our findings indicate a polymorphism at residue 141, with the N variant becoming increasingly prevalent in recent Chinese H9N2 isolates. Although both wild-type and N141K mutant strains exclusively target α,2-6 sialic acid receptors, the N141K mutation notably impedes the virus's ability to bind to these receptors. Despite the mutation exerting minimal influence on viral titers, antigenicity, and pathogenicity in chicken embryos, it significantly enhances viral thermostability and reduces plaque size on Madin-Darby canine kidney (MDCK) cells. Additionally, the N141K mutation leads to decreased expression levels of HA protein in both MDCK cells and eggs. These findings highlight the critical role of the K141N substitution in altering the hemagglutination characteristics of recent H9N2 AIV strains under elevated temperatures. This emphasizes the need for ongoing surveillance and genetic analysis of circulating H9N2 AIV strains to develop effective control and prevention measures.IMPORTANCEThe H9N2 subtype of avian influenza virus (AIV) is currently the most prevalent low-pathogenicity AIV circulating in domestic poultry globally. Recently, there has been an emerging trend of H9N2 AIV strains acquiring increased affinity for human-type receptors and even losing their ability to bind to avian-type receptors, which raises concerns about their pandemic potential. In China, there has been a growing number of H9N2 AIV strains that have lost their ability to agglutinate chicken red blood cells, leading to false-negative results during surveillance efforts. In this study, we identified a K141N mutation in the HA protein of H9N2 AIV to be responsible for the loss of hemagglutination activity. This finding provides insight into the development of effective surveillance, prevention, and control strategies to mitigate the threat posed by H9N2 AIV to both animal and human health.


Amino Acid Substitution , Hemagglutination , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Mutation , Animals , Chick Embryo , Dogs , Humans , Chickens/virology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/growth & development , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/metabolism , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza in Birds/virology , Poultry , Female , Mice , Cell Line , Evolution, Molecular , Temperature , Receptors, Virus/metabolism
...