Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 363
Filter
1.
Microb Pathog ; 195: 106871, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39163919

ABSTRACT

The H9N2 avian influenza virus (AIV) is spreading worldwide. Presence of H9N2 virus tends to increase the chances of infection with other pathogens which can lead to more serious economic losses. In a previous study, a regulated delayed lysis Salmonella vector was used to deliver a DNA vaccine named pYL233 encoding M1 protein, mosaic HA protein and chicken GM-CSF adjuvant. To further increase its efficiency, chitosan as a natural adjuvant was applied in this study. The purified plasmid pYL233 was coated with chitosan to form a DNA containing nanoparticles (named CS233) by ionic gel method and immunized by intranasal boost immunization in birds primed by oral administration with Salmonella strain. The CS233 DNA nanoparticle has a particle size of about 150 nm, with an encapsulation efficiency of 93.2 ± 0.12 % which protected the DNA plasmid from DNase I digestion and could be stable for a period of time at 37°. After intranasal boost immunization, the CS233 immunized chickens elicited higher antibody response, elevated CD4+ T cells and CD8+ T cells activation and increased T-lymphocyte proliferation, as well as increased productions of IL-4 and IFN-γ. After challenge, chickens immunized with CS233 resulted in the lowest levels of pulmonary virus titer and viral shedding as compared to the other challenge groups. The results showed that the combination of intranasal immunization with chitosan-coated DNA vaccine and oral immunization with regulatory delayed lytic Salmonella strain could enhance the immune response and able to provide protection against H9N2 challenge.


Subject(s)
Administration, Intranasal , Antibodies, Viral , Chickens , Chitosan , Immunity, Cellular , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Plasmids , Vaccines, DNA , Virus Shedding , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Chickens/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Viral/blood , Plasmids/genetics , Nanoparticles , Immunization, Secondary , CD8-Positive T-Lymphocytes/immunology , Adjuvants, Immunologic/administration & dosage , Interferon-gamma , Interleukin-4 , Adjuvants, Vaccine , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/virology , CD4-Positive T-Lymphocytes/immunology , Salmonella/immunology , Salmonella/genetics
2.
Vet Microbiol ; 296: 110188, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018942

ABSTRACT

H9N2 avian influenza virus (AIV), one of the predominant subtypes circulating in the poultry industry, inflicts substantial economic damage. Mutations in the hemagglutinin (HA) and neuraminidase (NA) proteins of H9N2 frequently alter viral antigenicity and replication. In this paper, we analyzed the HA genetic sequences and antigenic properties of 26 H9N2 isolates obtained from chickens in China between 2012 and 2019. The results showed that these H9N2 viruses all belonged to h9.4.2.5, and were divided into two clades. We assessed the impact of amino acid substitutions at HA sites 145, 149, 153, 164, 167, 168, and 200 on antigenicity, and found that a mutation at site 164 significantly modified antigenic characteristics. Amino acid variations at sites 145, 153, 164 and 200 affected virus's hemagglutination and the growth kinetics in mammalian cells. These results underscore the critical need for ongoing surveillance of the H9N2 virus and provide valuable insights for vaccine development.


Subject(s)
Chickens , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Animals , Chickens/virology , Influenza in Birds/virology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , China , Amino Acid Substitution , Poultry Diseases/virology , Mutation , Antigens, Viral/immunology , Antigens, Viral/genetics , Virus Replication , Phylogeny , Neuraminidase/genetics , Neuraminidase/immunology , Amino Acids/genetics
3.
Nat Commun ; 15(1): 5593, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961067

ABSTRACT

Human cases of avian influenza virus (AIV) infections are associated with an age-specific disease burden. As the influenza virus N2 neuraminidase (NA) gene was introduced from avian sources during the 1957 pandemic, we investigate the reactivity of N2 antibodies against A(H9N2) AIVs. Serosurvey of healthy individuals reveal the highest rates of AIV N2 antibodies in individuals aged ≥65 years. Exposure to the 1968 pandemic N2, but not recent N2, protected against A(H9N2) AIV challenge in female mice. In some older adults, infection with contemporary A(H3N2) virus could recall cross-reactive AIV NA antibodies, showing discernable human- or avian-NA type reactivity. Individuals born before 1957 have higher anti-AIV N2 titers compared to those born between 1957 and 1968. The anti-AIV N2 antibodies titers correlate with antibody titers to the 1957 N2, suggesting that exposure to the A(H2N2) virus contribute to this reactivity. These findings underscore the critical role of neuraminidase immunity in zoonotic and pandemic influenza risk assessment.


Subject(s)
Antibodies, Viral , Cross Reactions , Influenza A Virus, H3N2 Subtype , Influenza, Human , Neuraminidase , Pandemics , Neuraminidase/immunology , Neuraminidase/genetics , Animals , Humans , Antibodies, Viral/immunology , Antibodies, Viral/blood , Influenza A Virus, H3N2 Subtype/immunology , Female , Cross Reactions/immunology , Mice , Influenza, Human/immunology , Influenza, Human/epidemiology , Influenza, Human/virology , Aged , Influenza A Virus, H2N2 Subtype/immunology , Influenza A Virus, H2N2 Subtype/genetics , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/veterinary , Birds/virology , Middle Aged , Influenza in Birds/epidemiology , Influenza in Birds/immunology , Influenza in Birds/virology , Influenza A Virus, H9N2 Subtype/immunology , Adult , Viral Proteins/immunology , Viral Proteins/genetics
4.
Emerg Microbes Infect ; 13(1): 2364736, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38847071

ABSTRACT

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.


Subject(s)
Chickens , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Glycosylation , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/metabolism , Animals , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza in Birds/virology , Chickens/virology , Mutation , Polysaccharides/metabolism , Virus Replication , Madin Darby Canine Kidney Cells , Poultry Diseases/virology
5.
J Infect ; 89(2): 106199, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38901571

ABSTRACT

The sustained circulation of H9N2 avian influenza viruses (AIVs) poses a significant threat for contributing to a new pandemic. Given the temporal and spatial uncertainty in the antigenicity of H9N2 AIVs, the immune protection efficiency of vaccines remains challenging. By developing an antigenicity prediction method for H9N2 AIVs, named PREDAC-H9, the global antigenic landscape of H9N2 AIVs was mapped. PREDAC-H9 utilizes the XGBoost model with 14 well-designed features. The XGBoost model was built and evaluated to predict the antigenic relationship between any two viruses with high values of 81.1 %, 81.4 %, 81.3 %, 81.1 %, and 89.4 % in accuracy, precision, recall, F1 value, and area under curve (AUC), respectively. Then the antigenic correlation network (ACnet) was constructed based on the predicted antigenic relationship for H9N2 AIVs from 1966 to 2022, and ten major antigenic clusters were identified. Of these, four novel clusters were generated in China in the past decade, demonstrating the unique complex situation there. To help tackle this situation, we applied PREDAC-H9 to calculate the cluster-transition determining sites and screen out virus strains with the high cross-protective spectrum, thus providing an in silico reference for vaccine recommendation. The proposed model will reduce the clinical monitoring workload and provide a useful tool for surveillance and control of H9N2 AIVs.


Subject(s)
Antigens, Viral , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Animals , Antigens, Viral/immunology , China , Birds
6.
Int J Pharm ; 660: 124318, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38852750

ABSTRACT

Avian influenza virus subtype H9N2 has the ability to infect birds and humans, further causing significant losses to the poultry industry and even posing a great threat to human health. Oral vaccine received particular interest for preventing majority infection due to its ability to elicit both mucosal and systemic immune responses, but their development is limited by the bad gastrointestinal (GI) environment, compact epithelium and mucus barrier, and the lack of effective mucosal adjuvants. Herein, we developed the dendritic fibrous nano-silica (DFNS) grafted with Cistanche deserticola polysaccharide (CDP) nanoparticles (CDP-DFNS) as an adjuvant for H9N2 vaccine. Encouragingly, CDP-DFNS facilitated the proliferation of T and B cells, and further induced the activation of T lymphocytes in vitro. Moreover, CDP-DFNS/H9N2 significantly promoted the antigen-specific antibodies levels in serum and intestinal mucosal of chickens, indicating the good ability to elicit both systemic and mucosal immunity. Additional, CDP-DFNS facilitate the activation of CD4 + and CD8 + T cells both in spleen and intestinal mucosal, and the indexes of immune organs. This study suggested that CDP-DFNS may be a new avenue for development of oral vaccine against pathogens that are transmitted via mucosal route.


Subject(s)
Adjuvants, Immunologic , Chickens , Immunity, Mucosal , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Nanoparticles , Polysaccharides , Silicon Dioxide , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/drug effects , Polysaccharides/administration & dosage , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/immunology , Silicon Dioxide/administration & dosage , Silicon Dioxide/chemistry , Nanoparticles/administration & dosage , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Immunity, Mucosal/drug effects , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Oral , Intestinal Mucosa/immunology , Intestinal Mucosa/drug effects , Antibodies, Viral/blood , Antibodies, Viral/immunology
7.
Int J Biol Macromol ; 275(Pt 1): 133453, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942402

ABSTRACT

Influenza A virus (IAV) causes annual epidemics and occasional pandemics, resulting in significant economic losses and numerous fatalities. Current vaccines, typically administered through injection, provide limited protection due to the frequent antigenic shift and drift of IAV strains. Therefore, the development of alternative broad-spectrum vaccine strategies is imperative. Lactic acid bacteria (LAB) represent promising candidates for vaccine engineering due to their low cost, high safety profile, and suitability for oral administration. In this study, we identified a strain of Lactobacillus plantarum (Lp) that is resistant to acid and bile salts and capable of colonizing the intestines of mice. Subsequently, we employed the RecE/T gene editing system to integrate headless hemagglutinins (mini-HA) into the genome of Lp, generating Lp-mini-HA-SP. Remarkably, immunization with Lp-mini-HA-SP elicited serum IgG antibody responses and conferred immune protection against H9N2 and H1N1 influenza virus challenges. Collectively, our findings offer a novel approach for the development of orally administered IAV vaccines and hold significant potential for future drug development endeavors.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Lactobacillus plantarum , Orthomyxoviridae Infections , Vaccines, Synthetic , Lactobacillus plantarum/genetics , Lactobacillus plantarum/immunology , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Mice , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Vaccines, Synthetic/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Mice, Inbred BALB C
8.
J Biol Chem ; 300(6): 107395, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38768812

ABSTRACT

B2 haplotype major histocompatibility complex (MHC) has been extensively reported to confer resistance to various avian diseases. But its peptide-binding motif is unknown, and the presenting peptide is rarely identified. Here, we identified its peptide-binding motif (X-A/V/I/L/P/S/G-X-X-X-X-X-X-V/I/L) in vitro using Random Peptide Library-based MHC I LC-MS/MS analysis. To further clarify the structure basis of motif, we determined the crystal structure of the BF2∗02:01-PB2552-560 complex at 1.9 Å resolution. We found that BF2∗02:01 had a relatively wide antigen-binding groove, and the structural characterization of pockets was consistent with the characterization of peptide-binding motif. The wider features of the peptide-binding motif and increased number of peptides bound by BF2∗02:01 than BF2∗04:01 might resolve the puzzles for the presence of potential H9N2 resistance in B2 chickens. Afterward, we explored the H9N2 avian influenza virus (AIV)-induced cellular immune response in B2 haplotype chickens in vivo. We found that ratio of CD8+ T cell and kinetic expression of cytotoxicity genes including Granzyme K, interferon-γ, NK lysin, and poly-(ADP-ribose) polymerase in peripheral blood mononuclear cells were significantly increased in defending against H9N2 AIV infection. Especially, we selected 425 epitopes as candidate epitopes based on the peptide-binding motif and further identified four CD8+ T-cell epitopes on H9N2 AIV including NS198-106, PB2552-560, NP182-190, and NP455-463 via ELI-spot interferon-γ detections after stimulating memory lymphocytes with peptides. More importantly, these epitopes were found to be conserved in H7N9 AIV and H9N2 AIV. These findings provide direction for developing effective T cell epitope vaccines using well-conserved internal viral antigens in chickens.


Subject(s)
Chickens , Epitopes, T-Lymphocyte , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Influenza A Virus, H9N2 Subtype/immunology , Animals , Epitopes, T-Lymphocyte/immunology , Influenza in Birds/immunology , Influenza in Birds/virology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism
9.
Front Immunol ; 15: 1352022, 2024.
Article in English | MEDLINE | ID: mdl-38698856

ABSTRACT

The complement system is an innate immune mechanism against microbial infections. It involves a cascade of effector molecules that is activated via classical, lectin and alternative pathways. Consequently, many pathogens bind to or incorporate in their structures host negative regulators of the complement pathways as an evasion mechanism. Factor H (FH) is a negative regulator of the complement alternative pathway that protects "self" cells of the host from non-specific complement attack. FH has been shown to bind viruses including human influenza A viruses (IAVs). In addition to its involvement in the regulation of complement activation, FH has also been shown to perform a range of functions on its own including its direct interaction with pathogens. Here, we show that human FH can bind directly to IAVs of both human and avian origin, and the interaction is mediated via the IAV surface glycoprotein haemagglutinin (HA). HA bound to common pathogen binding footprints on the FH structure, complement control protein modules, CCP 5-7 and CCP 15-20. The FH binding to H1 and H3 showed that the interaction overlapped with the receptor binding site of both HAs, but the footprint was more extensive for the H3 HA than the H1 HA. The HA - FH interaction impeded the initial entry of H1N1 and H3N2 IAV strains but its impact on viral multicycle replication in human lung cells was strain-specific. The H3N2 virus binding to cells was significantly inhibited by preincubation with FH, whereas there was no alteration in replicative rate and progeny virus release for human H1N1, or avian H9N2 and H5N3 IAV strains. We have mapped the interaction between FH and IAV, the in vivo significance of which for the virus or host is yet to be elucidated.


Subject(s)
Complement Factor H , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A virus , Influenza, Human , Protein Binding , Humans , Complement Factor H/metabolism , Complement Factor H/immunology , Animals , Influenza, Human/immunology , Influenza, Human/virology , Influenza, Human/metabolism , Influenza A virus/immunology , Influenza A virus/physiology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Binding Sites , Influenza in Birds/virology , Influenza in Birds/immunology , Influenza in Birds/metabolism , Birds/virology , Host-Pathogen Interactions/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology
10.
Nat Commun ; 15(1): 3494, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693163

ABSTRACT

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.


Subject(s)
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
11.
Hum Vaccin Immunother ; 20(1): 2347019, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38807261

ABSTRACT

Influenza A viruses pose a significant threat to global health, impacting both humans and animals. Zoonotic transmission, particularly from swine and avian species, is the primary source of human influenza outbreaks. Notably, avian influenza viruses of the H5N1, H7N9, and H9N2 subtypes are of pandemic concern through their global spread and sporadic human infections. Preventing and controlling these viruses is critical due to their high threat level. Vaccination remains the most effective strategy for influenza prevention and control in humans, despite varying vaccine efficacy across strains. This review focuses specifically on pandemic preparedness for avian influenza viruses. We delve into vaccines tested in animal models and summarize clinical trials conducted on H5N1, H7N9, and H9N2 vaccines in humans.


Subject(s)
Birds , Influenza Vaccines , Influenza in Birds , Influenza, Human , Pandemics , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Humans , Influenza, Human/prevention & control , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza in Birds/prevention & control , Influenza in Birds/epidemiology , Pandemics/prevention & control , Vaccine Development , Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Clinical Trials as Topic , Disease Models, Animal , Vaccination , Pandemic Preparedness
12.
Vaccine ; 42(15): 3410-3419, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38641498

ABSTRACT

The application of recombinant herpesvirus of turkey, expressing the H9 hemagglutinin gene from low pathogenic avian influenza virus (LPAIV) H9N2 and the avian orthoavulavirus-1 (AOAV-1) (commonly known as Newcastle Disease virus (NDV)) fusion protein (F) as an rHVT-H9-F vaccine, is an alternative to currently used classical vaccines. This study investigated H9- and ND-specific humoral and mucosal responses, H9-specific cell-mediated immunity, and protection conferred by the rHVT-H9-F vaccine in specific pathogen-free (SPF) chickens. Vaccination elicited systemic NDV F- and AIV H9-specific antibody response but also local antibodies in eye wash fluid and oropharyngeal swabs. The ex vivo H9-specific stimulation of splenic and pulmonary T cells in the vaccinated group demonstrated the ability of vaccination to induce systemic and local cellular responses. The clinical protection against a challenge using a LPAIV H9N2 strain of the G1 lineage isolated in Morocco in 2016 was associated with a shorter duration of shedding along with reduced viral genome load in the upper respiratory tract and reduced cloacal shedding compared to unvaccinated controls.


Subject(s)
Antibodies, Viral , Chickens , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Virus Shedding , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Chickens/immunology , Influenza in Birds/prevention & control , Influenza in Birds/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , Virus Shedding/immunology , Specific Pathogen-Free Organisms , Newcastle disease virus/immunology , Newcastle disease virus/genetics , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Poultry Diseases/virology , Immunity, Cellular , Herpesvirus 1, Meleagrid/immunology , Herpesvirus 1, Meleagrid/genetics , Vaccination/methods , Immunity, Humoral , Genetic Vectors/immunology , Immunogenicity, Vaccine , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics
13.
Avian Pathol ; 53(5): 390-399, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38563198

ABSTRACT

Avian influenza (AI), caused by H9N2 subtype avian influenza virus (AIV), poses a serious threat to poultry farming and public health due to its transmissibility and pathogenicity. The PB2 protein is a major component of the viral RNA polymerase complex. It is of great importance to identify the antigenic determinants of the PB2 protein to explore the function of the PB2 protein. In this study, the PB2 sequence of H9N2 subtype AIV, from 1090 to 1689 bp, was cloned and expressed. The recombinant PB2 protein with cutting gel was used to immunize BALB/c mice. After cell fusion, the hybridoma cell lines secreting monoclonal antibodies (mAbs) targeting the PB2 protein were screened by indirect ELISA and western blotting, and the antigenic epitopes of mAbs were identified by constructing truncated overlapping fragments in the PB2 protein of H9N2 subtype AIV. The results showed that three hybridoma cell lines (4B7, 4D10, and 5H1) that stably secreted mAbs specific to the PB2 protein were screened; the heavy chain of 4B7 was IgG2α, those of 4D10 and 5H1 were IgG1, and all three mAbs had kappa light chain. Also, the minimum B-cell epitope recognized was 475LRGVRVSK482 and 528TITYSSPMMW537. Homology analysis showed that these two epitopes were conserved among the different subtypes of AIV strains and located on the surface of the PB2 protein. The above findings provide an experimental foundation for further investigation of the function of the PB2 protein and developing monoclonal antibody-based diagnostic kits.


Subject(s)
Antibodies, Monoclonal , Epitopes, B-Lymphocyte , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Mice, Inbred BALB C , Viral Proteins , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Animals , Antibodies, Monoclonal/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Mice , Influenza in Birds/virology , Influenza in Birds/immunology , Epitopes, B-Lymphocyte/immunology , Hybridomas , RNA-Dependent RNA Polymerase/genetics , Antibodies, Viral/immunology , Chickens/virology , Female
14.
Avian Dis ; 68(1): 43-51, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38687107

ABSTRACT

The aim of the current study was to map the genetic diversity in the haemagglutinin (HA) glycoprotein of influenza A viruses (IAVs) of the H9N2 subtype. Twenty-five H9N2 IAVs were isolated from broiler chickens from March to July 2019. The HA gene was amplified, and phylogenetic analysis was performed to determine the evolutionary relationship. Important antigenic amino acid residues of HA attributed to immune escape and zoonotic potential were compared among H9N2 IAVs. Phylogenetic analysis revealed that sublineage B2 under the G1 lineage in Pakistan was found to be diversified, and newly sequenced H9N2 isolates were nested into two clades (A and B). Mutations linked to the antigenic variation and potential immune escape were observed as G72E (1/25, 4%), A180T (3/25, 12%), and A180V (1/25, 4%). A twofold significant reduction (P < 0.01) in log2 hemagglutination inhibition titers was observed with H9N2 IAV naturally harboring amino acid V180 instead of A180 in HA protein. Moreover, in the last 20 years, complete substitution at residues (T127D, D135N, and L150N) and partial substitution at residues (72, 74, 131, 148, 180, 183, 188, 216, 217, and 249, mature H9 HA numbering) associated with changes in antigenicity were observed. The presence of L216 in all H9N2 IAV isolates and T/V180 in four isolates in the receptor-binding site reveals the potential of these viruses to cross the species barrier to infect human or mammals. The current study observed the circulation of antigenically diverse H9N2 IAV variants that possess potential mutations that can escape the host immune system.


Nota de investigación- Mapeo de marcadores genéticos asociados con la antigenicidad y el rango de huéspedes en los virus de la influenza tipo A subtipo H9N2 que infectan a la avicultura en Pakistán. El objetivo del presente estudio fue mapear la diversidad genética en la glicoproteína hemaglutinina (HA) de los virus de la influenza A (IAV) del subtipo H9N2. Se aislaron veinticinco virus de influenza H9N2 de pollos de engorde de marzo a julio del 2019. Se amplificó el gene HA y se realizó un análisis filogenético para determinar la relación evolutiva. Se compararon importantes residuos de aminoácidos antigénicos de la hemaglutinina atribuidos al escape inmunológico y al potencial zoonótico entre los virus de la influenza aviar H9N2. El análisis filogenético reveló que el sublinaje B2 bajo el linaje G1 en Pakistán estaba diversificado, y los aislados de H9N2 recién secuenciados se agruparon en dos clados (A y B). Se observaron mutaciones relacionadas con la variación antigénica y el posible escape inmunológico como los residuos de aminoácidos G72E (1/25, 4%), A180T (3/25, 12%) y A180V (1/25, 4%). Se observó una reducción significativa al doble (P < 0.01) en los títulos de inhibición de la hemaglutinación log2 cuando el virus de la influenza aviar H9N2 albergaba naturalmente el aminoácido V180 en lugar del A180 en la proteína HA. Además, en los últimos 20 años, sustitución completa en los residuos (T127D, D135N y L150N) y sustitución parcial en los residuos (72, 74, 131, 148, 180, 183, 188, 216, 217 y 249, de acuerdo con la numeración de la HA subtipo madura) asociados con cambios en la antigenicidad. La presencia del residuo L216 en todos los aislados de influenza aviar H9N2 y T/V180 en cuatro aislados en el sitio de unión al receptor revela el potencial de estos virus para cruzar la barrera de las especies para infectar a humanos o mamíferos. El estudio actual observó la circulación de variantes antigénicamente diversas del virus de influenza aviar H9N2 que poseen mutaciones potenciales que pueden escapar del sistema inmunológico del huésped.


Subject(s)
Chickens , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Phylogeny , Poultry Diseases , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Animals , Pakistan , Influenza in Birds/virology , Influenza in Birds/immunology , Poultry Diseases/virology , Host Specificity , Genetic Markers , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Antigenic Variation , Genetic Variation
15.
J Virol ; 98(4): e0024824, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38466094

ABSTRACT

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.


Subject(s)
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
16.
Arch Razi Inst ; 78(6): 1746-1752, 2023 12.
Article in English | MEDLINE | ID: mdl-38828164

ABSTRACT

Influenza viruses can multiply in quails and be transmitted to other animal species. As vaccination reduces virus shedding in chickens, the effect of the killed H9N2 avian influenza virus (AIV) on tissue distribution and virus shedding was evaluated in quails. One hundred 20-day-old quails were divided into six equal groups, kept in separate pens, and fed ad libitum. Before vaccination, blood samples were randomly collected from the wing veins. Four groups were vaccinated with the inactivated H9N2 Razi Institute vaccine at 21 days subcutaneously at the back of neck. Three weeks later, two groups were re-vaccinated. Two weeks later, at the age of 56 days, three groups were challenged with 100 µL of allantoic fluid containing 105 EID50 H9N2 through the oculonasal route. Blood samples were collected from quails at 42, 56, 63, and 70 days from each group to determine AIV antibodies by the hemagglutination inhibition test. Three quails were randomly selected and euthanized from each group on days 1, 3, and 6 post-inoculation (PI). Tissue samples were collected, and the RT-PCR test was performed. No clinical signs or gross lesions existed in any of the groups during the experiment. However, the virus was detected in different tissues on the first, third, and sixth days after the challenge in unvaccinated challenged birds. Virus detection was significantly more frequent in the quails vaccinated once and challenged than in the twice-vaccinated challenged group (P≤0.05). On the third day of PI, the virus was detected in some organs of the challenged groups. On the sixth day of PI, the virus was detected only in the lungs of two unvaccinated and once-vaccinated challenged birds. It was concluded that the vaccination of quails against AIV H9 is necessary to protect them from clinical signs, as well as respiratory tract and intestine replication. Two-time vaccination significantly protects the respiratory and intestine tracts, compared to one-time vaccination (P≤0.05).


Subject(s)
Coturnix , Influenza A Virus, H9N2 Subtype , Influenza Vaccines , Influenza in Birds , Vaccination , Virus Shedding , Animals , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/physiology , Influenza in Birds/prevention & control , Influenza in Birds/virology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Vaccination/veterinary , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Poultry Diseases/virology , Poultry Diseases/prevention & control , Antibodies, Viral/blood
17.
Sci Rep ; 12(1): 2311, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35145121

ABSTRACT

Many studies have been conducted on measuring avian influenza viruses and their hemagglutinin (HA) antigens via electrochemical principles; most of these studies have used gold electrodes on ceramic, glass, or silicon substrates, and/or labeling for signal enhancement. Herein, we present a paper-based immunosensor for label-free measurement of multiple avian influenza virus (H5N1, H7N9, and H9N2) antigens using flexible screen-printed carbon nanotube-polydimethylsiloxane electrodes. These flexible electrodes on a paper substrate can complement the physical weakness of the paper-based sensors when wetted, without affecting flexibility. The relative standard deviation of the peak currents was 1.88% when the electrodes were repeatedly bent and unfolded twenty times with deionized water provided each cycle, showing the stability of the electrodes. For the detection of HA antigens, approximately 10-µl samples (concentration: 100 pg/ml-100 ng/ml) were needed to form the antigen-antibody complexes during 20-30 min incubation, and the immune responses were measured via differential pulse voltammetry. The limits of detections were 55.7 pg/ml (0.95 pM) for H5N1 HA, 99.6 pg/ml (1.69 pM) for H7N9 HA, and 54.0 pg/ml (0.72 pM) for H9N2 HA antigens in phosphate buffered saline, and the sensors showed good selectivity and reproducibility. Such paper-based sensors are economical, flexible, robust, and easy-to-manufacture, with the ability to detect several avian influenza viruses.


Subject(s)
Antigens, Viral/analysis , Biosensing Techniques/methods , Dimethylpolysiloxanes , Electrochemical Techniques/methods , Electrodes , Immunoassay/methods , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Nanotubes, Carbon , Paper , Virology/methods , Animals , Birds , Humans , Influenza in Birds/diagnosis , Influenza in Birds/virology , Influenza, Human/diagnosis , Influenza, Human/virology , Limit of Detection , Reproducibility of Results
18.
Microbiol Spectr ; 10(1): e0082221, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35019707

ABSTRACT

The H9N2 subtype avian influenza virus (AIV) has become endemic in poultry globally; however due to its low pathogenicity, it is not under primary surveillance and control in many countries. Recent reports of human infection caused by H9N2 AIV has increased public concern. This study investigated the genetic and antigenic characteristics of H9N2 AIV isolated from local markets in nine provinces in Southern China from 2013 to 2018. We detected an increasing annual isolation rate of H9N2 AIV. Phylogenetic analyses of hemagglutinin (HA) genes suggests that isolated strains were rooted in BJ94 lineage but have evolved into new subgroups (II and III), which derived from subgroup I. The estimated substitution rate of the subgroup III strains was 6.23 × 10-3 substitutions/site/year, which was 1.5-fold faster than that of the average H9N2 HA rate (3.95 × 10-3 substitutions/site/year). Based on the antigenic distances, subgroup II and III strains resulted in two clear antigenic clusters 2 and 3, separated from the vaccine strain F98, cluster 1. New antigenic properties of subgroup III viruses were associated with 11 amino acid changes in the HA protein, suggesting antigenic drift in H9N2 viruses. Our phylogenetic and antigenic analyses of the H9N2 strains circulating in local markets in Southern China provide new insights on the antigenic diversification of H9N2 viruses. IMPORTANCE The H9N2 low pathogenicity avian influenza (LPAI) virus has become endemic in poultry globally. In several Asian countries, vaccination against H9N2 avian influenza virus (AIV) was approved to reduce economic losses in the poultry industry. However, surveillance programs initiated after the introduction of vaccination identified the persistence of H9N2 AIV in poultry (especially in chicken in South Korea and China). Recent reports of human infection caused by H9N2 AIV has increased public concern. Surveillance of H9N2 circulating in poultry in the fields or markets was essential to update the vaccination strategies. This study investigated the genetic and antigenic characteristics of H9N2 AIVs isolated from local markets in nine provinces in Southern China from 2013 to 2018. The discovery of mutations in the hemagglutinin (HA) gene that result in antigenic changes provides a baseline reference for evolutionary studies of H9N2 viruses and vaccination strategies in poultry.


Subject(s)
Evolution, Molecular , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/virology , Poultry Diseases/virology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antigenic Drift and Shift , Antigenic Variation , Chickens , China/epidemiology , Hemagglutinins, Viral/chemistry , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza in Birds/epidemiology , Phylogeny , Poultry Diseases/epidemiology
19.
Sci Rep ; 11(1): 20462, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650121

ABSTRACT

Influenza viruses cause severe respiratory infections in humans and birds, triggering global health concerns and economic burden. Influenza infection is a dynamic process involving complex biological host responses. The objective of this study was to illustrate global biological processes in ileum and cecal tonsils at early time points after chickens were infected with low pathogenic avian influenza virus (LPAIV) H9N2 through transcriptome analysis. Total RNA isolated from ileum and cecal tonsils of non-infected and infected layers at 12-, 24- and 72-h post-infection (hpi) was used for mRNA sequencing analyses to characterize differentially expressed genes and overrepresented pathways. Statistical analysis highlighted transcriptomic signatures significantly occurring 24 and 72 hpi, but not earlier at 12 hpi. Interferon (IFN)-inducible and IFN-stimulated gene (ISG) expression was increased, followed by continued expression of various heat-shock proteins (HSP), including HSP60, HSP70, HSP90 and HSP110. Some upregulated genes involved in innate antiviral responses included DDX60, MX1, RSAD2 and CMPK2. The ISG15 antiviral mechanism pathway was highly enriched in ileum and cecal tonsils at 24 hpi. Overall, most affected pathways were related to interferon production and the heat-shock response. Research on these candidate genes and pathways is warranted to decipher underlying mechanisms of immunity against LPAIV in chickens.


Subject(s)
Cecum/immunology , Ileum/immunology , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/immunology , Animals , Cecum/metabolism , Chickens , Female , Gene Expression Profiling , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Ileum/metabolism , Immunity, Innate , Influenza in Birds/genetics , Influenza in Birds/metabolism , Interferons/genetics , Interferons/metabolism , RNA, Messenger
20.
Cells ; 10(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34571913

ABSTRACT

Immunological memory is a cardinal feature of the immune system. The intestinal mucosa is the primary exposure and entry site of infectious organisms. For an effective and long-lasting safeguard, a robust immune memory system is required, especially by the mucosal immunity. It is well known that tissue-resident memory T cells (Trms) provide a first response against infections reencountered at mucosal tissues surfaces, where they accelerate pathogen clearance. However, their function in intestinal immunization remains to be investigated. Here, we report enhanced local mucosal and systemic immune responses through oral administration of H9N2 influenza whole inactivated virus (H9N2 WIV) plus Bacillus subtilis spores. Subsequently, H9N2 WIV plus spores led to the generation of CD103+ CD69+ Trms, which were independent of circulating T cells during the immune period. Meanwhile, we also found that Bacillus subtilis spores could stimulate Acrp30 expression in 3T3-L1 adipocytes. Moreover, spore-stimulated adipocyte supernatant also upregulated the expression of intercellular adhesion molecule-1 (ICAM1) in dendritic cells (DCs). Furthermore, the proportion of HA-tetramer+ cells was severely curtailed upon suppressed ICAM1 expression, which also depended on HA-loaded DCs. Taken together, our data demonstrated that spore-promoted H9N2 WIV induced an immune response by enhancing Trms populations, which were associated with the activation of ICAM1 in DCs.


Subject(s)
Bacillus subtilis/immunology , Dendritic Cells/immunology , Immunologic Memory/immunology , Intercellular Adhesion Molecule-1/metabolism , Intestinal Mucosa/immunology , Spores, Bacterial/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic , Animals , Antibodies, Viral/immunology , Immunization , Influenza A Virus, H9N2 Subtype/immunology , Intercellular Adhesion Molecule-1/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL