Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 329
Filter
1.
PLoS One ; 19(5): e0302865, 2024.
Article in English | MEDLINE | ID: mdl-38723016

ABSTRACT

Influenza A viruses (IAVs) continue to pose a huge threat to public health, and their prevention and treatment remain major international issues. Neuraminidase (NA) is the second most abundant surface glycoprotein on influenza viruses, and antibodies to NA have been shown to be effective against influenza infection. In this study, we generated a monoclonal antibody (mAb), named FNA1, directed toward N1 NAs. FNA1 reacted with H1N1 and H5N1 NA, but failed to react with the NA proteins of H3N2 and H7N9. In vitro, FNA1 displayed potent antiviral activity that mediated both NA inhibition (NI) and blocking of pseudovirus release. Moreover, residues 219, 254, 358, and 388 in the NA protein were critical for FNA1 binding to H1N1 NA. However, further validation is necessary to confirm whether FNA1 mAb is indeed a good inhibitor against NA for application against H1N1 and H5N1 viruses.


Subject(s)
Antibodies, Monoclonal , Influenza A Virus, H1N1 Subtype , Neuraminidase , Neuraminidase/immunology , Neuraminidase/metabolism , Neuraminidase/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Influenza A Virus, H1N1 Subtype/immunology , Humans , Animals , Antibodies, Viral/immunology , Mice , Influenza A Virus, H5N1 Subtype/immunology , Mice, Inbred BALB C , Antiviral Agents/pharmacology , Viral Proteins/immunology , Viral Proteins/metabolism , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology
2.
Int J Biol Macromol ; 267(Pt 2): 131458, 2024 May.
Article in English | MEDLINE | ID: mdl-38593899

ABSTRACT

Avian influenza virus (AIV) H7N9 diseases have been recently reported, raising concerns about a potential pandemic. Thus, there is an urgent need for effective therapeutics for AIV H7N9 infections. Herein, camelid immunization and yeast two-hybrid techniques were used to identify potent neutralizing nanobodies (Nbs) targeting the H7 subtype hemagglutinin. First, we evaluated the binding specificity and hemagglutination inhibition activity of the screened Nbs against the H7 subtype hemagglutinin. Nb-Z77, with high hemagglutination inhibition activity was selected from the screened Nbs to optimize the yeast expression conditions and construct oligomeric forms of Nb-Z77 using various ligation methods. The oligomers Nb-Z77-DiGS, Nb-Z77-TriGS, Nb-Z77-Fc and Nb-Z77-Foldon were successfully constructed and expressed. Nb-Z77-DiGS and Nb-Z77-Foldon exhibited considerably greater activity than did Nb-Z77 against H7 subtype hemagglutinin, with median effective concentrations of 384.7 and 27.33 pM and binding affinity values of 213 and 5.21 pM, respectively. Nb-Z77-DiGS and Nb-Z77-Foldon completely inhibited the hemagglutination activity of the inactivated virus H7-Re1 at the lowest concentration of 0.938 µg/mL. This study screened a strain of Nb with high hemagglutination inhibition activity and enhanced its antiviral activity through oligomerization, which may have great potential for developing effective agents for the prevention, diagnosis, and treatment of AIV H7 subtype infection.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus , Single-Domain Antibodies , Single-Domain Antibodies/immunology , Single-Domain Antibodies/chemistry , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H7N9 Subtype/immunology , Humans , Hemagglutination Inhibition Tests , Influenza in Birds/immunology , Influenza in Birds/virology , Influenza in Birds/prevention & control , Antibodies, Viral/immunology , Antibodies, Neutralizing/immunology
3.
Emerg Microbes Infect ; 13(1): 2343912, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38629574

ABSTRACT

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.


Subject(s)
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
4.
5.
J Virol ; 97(1): e0143122, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36541801

ABSTRACT

Since 2013, H7N9 avian influenza viruses (AIVs) have caused more than 1,500 human infections and the culling of millions of poultry. Despite large-scale poultry vaccination, H7N9 AIVs continue to circulate among poultry in China and pose a threat to human health. Previously, we isolated and generated four monoclonal antibodies (mAbs) derived from humans naturally infected with H7N9 AIV. Here, we investigated the hemagglutinin (HA) epitopes of H7N9 AIV targeted by these mAbs (L3A-44, K9B-122, L4A-14, and L4B-18) using immune escape studies. Our results revealed four key antigenic epitopes at HA amino acid positions 125, 133, 149, and 217. The mutant H7N9 viruses representing escape mutations containing an alanine-to-threonine substitution at residue 125 (A125T), a glycine-to-glutamic acid substitution at residue 133 (G133E), an asparagine-to-aspartic acid substitution at residue 149 (N149D), or a leucine-to-glutamine substitution at residue 217 (L217Q) showed reduced or completely abolished cross-reactivity with the mAbs, as measured by a hemagglutination inhibition (HI) assay. We further assessed the potential risk of these mutants to humans should they emerge following mAb treatment by measuring the impact of these HA mutations on virus fitness and evasion of host adaptive immunity. Here, we showed that the L4A-14 mAb had broad neutralizing capabilities, and its escape mutant N149D had reduced viral stability and human receptor binding and could be neutralized by both postinfection and antigen-induced sera. Therefore, the L4A-14 mAb could be a therapeutic candidate for H7N9 AIV infection in humans and warrants further investigation for therapeutic applications. IMPORTANCE Avian influenza virus (AIV) H7N9 continues to circulate and evolve in birds, posing a credible threat to humans. Antiviral drugs have proven useful for the treatment of severe influenza infections in humans; however, concerns have been raised as antiviral-resistant mutants have emerged. Monoclonal antibodies (mAbs) have been studied for both prophylactic and therapeutic applications in infectious disease control and have demonstrated great potential. For example, mAb treatment has significantly reduced the risk of people developing severe disease with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition to the protection efficiency, we should also consider the potential risk of the escape mutants generated by mAb treatment to public health by assessing their viral fitness and potential to compromise host adaptive immunity. Considering these parameters, we assessed four human mAbs derived from humans naturally infected with H7N9 AIV and showed that the mAb L4A-14 displayed potential as a therapeutic candidate.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza, Human , Animals , Humans , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Epitopes , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/therapy , Immune Evasion/genetics , Mutation
6.
Cells ; 11(3)2022 01 30.
Article in English | MEDLINE | ID: mdl-35159296

ABSTRACT

The global outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing, as is research on the molecular mechanisms underlying cellular infection by coronaviruses, with the hope of developing therapeutic agents against this pandemic. Other important respiratory viruses such as 2009 pandemic H1N1 and H7N9 avian influenza virus (AIV), influenza A viruses, are also responsible for a possible outbreak due to their respiratory susceptibility. However, the interaction of these viruses with host cells and the regulation of post-transcriptional genes remains unclear. In this study, we detected and analyzed the comparative transcriptome profiling of SARS-CoV-2, panH1N1 (A/California/07/2009), and H7N9 (A/Shanghai/1/2013) infected cells. The results showed that the commonly upregulated genes among the three groups were mainly involved in autophagy, pertussis, and tuberculosis, which indicated that autophagy plays an important role in viral pathogenicity. There are three groups of commonly downregulated genes involved in metabolic pathways. Notably, unlike panH1N1 and H7N9, SARS-CoV-2 infection can inhibit the m-TOR pathway and activate the p53 signaling pathway, which may be responsible for unique autophagy induction and cell apoptosis. Particularly, upregulated expression of IRF1 was found in SARS-CoV-2, panH1N1, and H7N9 infection. Further analysis showed SARS-CoV-2, panH1N1, and H7N9 infection-induced upregulation of lncRNA-34087.27 could serve as a competitive endogenous RNA to stabilize IRF1 mRNA by competitively binding with miR-302b-3p. This study provides new insights into the molecular mechanisms of influenza A virus and SARS-CoV-2 infection.


Subject(s)
COVID-19/immunology , Immunity/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/immunology , RNA/immunology , Transcriptome/immunology , A549 Cells , Animals , COVID-19/genetics , COVID-19/virology , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H7N9 Subtype/physiology , Influenza, Human/genetics , Influenza, Human/virology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-1/metabolism , MicroRNAs/genetics , MicroRNAs/immunology , MicroRNAs/metabolism , Pandemics/prevention & control , RNA/genetics , RNA/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/immunology , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Messenger/metabolism , RNA-Seq/methods , SARS-CoV-2/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptome/genetics
7.
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
8.
Protein Expr Purif ; 192: 106046, 2022 04.
Article in English | MEDLINE | ID: mdl-35007721

ABSTRACT

Production of broadly-reactive antibodies is critical for universal immunodiagnosis of rapidly-evolving influenza viruses. Most monoclonal antibodies (mAbs) are generated in mice using the hybridoma technology which involves labor- and time-consuming screening and low yield issues. In this study, a recombinant antibody based on a broadly-reactive mAb against the hemagglutinin (HA) stalk of H7N9 avian influenza virus was expressed in CHO cells and its biological characteristics, cross-reactivity and epitope recognition were identified. The variable genes of the parental antibody were amplified and cloned into the antibody-expressing plasmids containing the constant genes of murine IgG1. The recombinant antibody was expressed in high yield and purity in CHO cells and showed similar features to the parental antibody, including negative hemagglutination inhibition activity against H7N9 virus and high binding activity with the H7N9 HA protein. Notably, the recombinant antibody exhibited a broad reactivity with different influenza subtypes belonging to group 1 and group 2, which was associated with its recognition of a highly-conserved epitope in the stalk, as observed for the parental antibody. Our results suggest that cell-based antibody expression system can be utilized as an important alternative to the hybridoma technology for antibody production for influenza virus diagnostics.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Orthomyxoviridae/drug effects , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/genetics , Antibodies, Viral/isolation & purification , CHO Cells , Cricetinae , Cricetulus , Cross Reactions , Gene Expression , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/virology , Mice , Orthomyxoviridae/classification , Orthomyxoviridae/immunology
9.
J Virol ; 96(4): e0157821, 2022 02 23.
Article in English | MEDLINE | ID: mdl-34908443

ABSTRACT

The ongoing SARS-CoV-2 pandemic poses a severe global threat to public health, as do influenza viruses and other coronaviruses. Here, we present chimpanzee adenovirus 68 (AdC68)-based vaccines designed to universally target coronaviruses and influenza. Our design is centered on an immunogen generated by fusing the SARS-CoV-2 receptor-binding domain (RBD) to the conserved stalk of H7N9 hemagglutinin (HA). Remarkably, the constructed vaccine effectively induced both SARS-CoV-2-targeting antibodies and anti-influenza antibodies in mice, consequently affording protection from lethal SARS-CoV-2 and H7N9 challenges as well as effective H3N2 control. We propose our AdC68-vectored coronavirus-influenza vaccine as a universal approach toward curbing respiratory virus-causing pandemics. IMPORTANCE The COVID-19 pandemic exemplifies the severe public health threats of respiratory virus infection and influenza A viruses. The currently envisioned strategy for the prevention of respiratory virus-causing diseases requires the comprehensive administration of vaccines tailored for individual viruses. Here, we present an alternative strategy by designing chimpanzee adenovirus 68-based vaccines which target both the SARS-CoV-2 receptor-binding-domain and the conserved stalk of influenza hemagglutinin. When tested in mice, this strategy attained potent neutralizing antibodies against wild-type SARS-CoV-2 and its emerging variants, enabling an effective protection against lethal SARS-CoV-2 challenge. Notably, it also provided complete protection from lethal H7N9 challenge and efficient control of H3N2-induced morbidity. Our study opens a new avenue to universally curb respiratory virus infection by vaccination.


Subject(s)
COVID-19/prevention & control , ChAdOx1 nCoV-19 , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines , Orthomyxoviridae Infections/prevention & control , SARS-CoV-2/immunology , Animals , COVID-19/epidemiology , COVID-19/genetics , COVID-19/immunology , ChAdOx1 nCoV-19/genetics , ChAdOx1 nCoV-19/immunology , ChAdOx1 nCoV-19/pharmacology , Female , HEK293 Cells , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Transgenic , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Pandemics , SARS-CoV-2/genetics
10.
Vet Microbiol ; 264: 109306, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34923247

ABSTRACT

Currently, highly pathogenic avian influenza (HPAI) H7N9 viruses still pose a potential pandemic threat. Influenza virus-like particle (VLP) is one of the most promising vaccine strategies to complement traditional egg-dependent vaccines. Here, we generated a H7N9 VLP vaccine candidate by baculovirus expression system and evaluated its efficacy in chickens and mice. The H7N9 VLP was produced through co-infection of Sf9 insect cells with three recombinant baculoviruses expressing individual HA, NA and M1 gene of the HPAI H7N9 virus A/chicken/Guangdong/GD15/2016. Intramuscular immunization of the H7N9 VLP elicited robust antibody immune responses and conferred complete clinical protection against lethal H7N9 virus challenge both in chickens and mice. Meanwhile, H7N9 VLP significantly restrained virus shedding and dramatically alleviated pulmonary lesions caused by H7N9 virus infection in birds and mice. Interestingly, chicken antibodies induced by the H7N9 VLP also had a good cross-reactivity with H7N9 field strains isolated in different years. In addition, vaccination with the H7N9 VLP elicited high T cell immunity in mouse lung, evidenced by significantly upregulated expression of IL-2, IL-4 and IFN-γ. Furthermore, the H7N9 VLP significantly decreased the expression of some key inflammatory cytokines, such as IL6, RANTES and TNF-α in mouse lung, which may partially account for its contribution to alleviate lung pathology. Therefore, our study describes the good efficacy of the HA + NA + M1-containing H7N9 VLP both in chicken and mice models, highlighting the potential of VLP-based vaccine as a critical alternative of traditional egg-based vaccine for control of H7N9 influenza virus in both humans and poultry.


Subject(s)
Baculoviridae , Influenza A Virus, H7N9 Subtype , Influenza Vaccines , Influenza in Birds , Orthomyxoviridae Infections , Vaccines, Virus-Like Particle , Animals , Antibodies, Viral/blood , Baculoviridae/immunology , Chickens , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Mice , Orthomyxoviridae Infections/prevention & control , Vaccines, Virus-Like Particle/immunology
11.
Sci Rep ; 11(1): 24485, 2021 12 29.
Article in English | MEDLINE | ID: mdl-34966175

ABSTRACT

A new strain of Influenza A Virus (IAV), so-called "H7N9 Avian Influenza", is the first strain of this virus in which a human is infected by transmitting the N9 of influenza virus. Although continuous human-to-human transmission has not been reported, the occurrence of various H7N9-associated epidemics and the lack of production of strong antibodies against H7N9 in humans warn of the potential for H7N9 to become a new pandemic. Therefore, the need for effective vaccination against H7N9 as a life-threatening viral pathogen has become a major concern. The current study reports the design of a multi-epitope vaccine against Hemagglutinin (HA) and Neuraminidase (NA) proteins of H7N9 Influenza A virus by prediction of Cytotoxic T lymphocyte (CTL), Helper T lymphocyte (HTL), IFN-γ and B-cell epitopes. Human ß-defensin-3 (HßD-3) and pan HLA DR-binding epitope (PADRE) sequence were considered as adjuvant. EAAAK, AAY, GPGPG, HEYGAEALERAG, KK and RVRR linkers were used as a connector for epitopes. The final construct contained 777 amino acids that are expected to be a recombinant protein of about ~ 86.38 kDa with antigenic and non-allergenic properties after expression. Modeled protein analysis based on the tertiary structure validation, docking studies, and molecular dynamics simulations results like Root-mean-square deviation (RMSD), Gyration, Root-mean-square fluctuation (RMSF) and Molecular Mechanics Poisson-Boltzmann Surface Area (MM/PBSA) showed that this protein has a stable construct and capable of being in interaction with Toll-like receptor 7 (TLR7), TLR8 and m826 antibody. Analysis of the obtained data the demonstrates that suggested vaccine has the potential to induce the immune response by stimulating T and Bcells, and may be utilizable for prevention purposes against Avian Influenza A (H7N9).


Subject(s)
Epitopes/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Animals , Birds , Computational Biology , Computer Simulation , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunity , Influenza in Birds/immunology , Models, Immunological , Models, Molecular , Neuraminidase/immunology , Viral Proteins/immunology
12.
Microbiol Spectr ; 9(2): e0068721, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34585985

ABSTRACT

H7N9 avian influenza virus (AIV) is an emerging zoonotic pathogen, and it is necessary to develop a differentiating infected from vaccinated animals (DIVA) vaccine for the purpose of eradication. H7N9 subtype AIV hemagglutinin subunit 2 glycoprotein (HA2) peptide chips and antisera of different AIV subtypes were used to screen H7N9 AIV-specific epitopes. A selected specific epitope in the HA2 protein of H7N9 AIV strain A/Chicken/Huadong/JD/17 (JD/17) was replaced with an epitope from an H3N2 subtype AIV strain by reverse genetics. The protection and serological DIVA characteristics of the recombinant H7N9 AIV strain were evaluated. The results showed that a specific epitope on the HA2 protein of H7N9 AIV, named the H7-12 peptide, was successfully screened. The recombinant H7N9 AIV with a modified epitope in the HA2 protein was rescued and named A/Chicken/Huadong/JD-cHA/17 (JD-cHA/17). The HA titer of JD-cHA/17 was 10 log2, and the 50% egg infective dose (EID50) titer was 9.67 log10 EID50/ml. Inactivated JD-cHA/17 induced a hemagglutination inhibition (HI) antibody titer similar that of the parent strain and provided 100% protection against high-pathogenicity or low-pathogenicity H7N9 AIV challenge. A peptide chip coated with H7-12 peptide was successfully applied to detect the seroconversion of chickens infected or vaccinated with JD/17, while there was no reactivity with antisera of chickens vaccinated with JD-cHA/17. Therefore, the marked vaccine candidate JD-cHA/17 can be used as a DIVA vaccine against H7N9 avian influenza when combined with an H7-12 peptide chip, making it a useful tool for stamping out the H7N9 AIV. IMPORTANCE DIVA vaccine is a useful tool for eradicating avian influenza, especially for highly pathogenic avian influenza. Several different DIVA strategies have been proposed for avian influenza inactivated whole-virus vaccine, involving the neuraminidase (NA), nonstructural protein 1 (NS1), matrix protein 2 ectodomain (M2e), or HA2 gene. However, virus reassortment, residual protein in a vaccine component, or reduced vaccine protection may limit the application of these DIVA strategies. Here, we constructed a novel chimeric H7N9 AIV, JD-cHA/17, that expressed the entire HA protein with substitution of an H3 AIV epitope in HA2. The chimeric H7N9 recombinant vaccine provides full clinical protection against high-pathogenicity or low-pathogenicity H7N9 AIV challenge. Combined with a short-peptide-based microarray chip containing the H7N9 AIV epitope in HA2, our finding is expected to be useful as a marker vaccine designed for avian influenza.


Subject(s)
Epitopes/immunology , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Vaccines, Inactivated/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza in Birds/virology , Vaccines, Synthetic/immunology
13.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34437301

ABSTRACT

Avian H7N9 influenza viruses cause sporadic outbreaks of human infections and threaten to cause a major pandemic. The breadth of B cell responses to natural infection and the dominant antigenic sites recognized during first exposure to H7 HA following infection are incompletely understood. Here, we studied the B cell response to H7 HA of 2 individuals who had recovered from natural H7N9 virus infection. We used competition binding, hydrogen-deuterium mass spectrometry, and single-particle negative stain electron microscopy to identify the patterns of molecular recognition of the antibody responses to H7 HA. We found that circulating H7-reactive B cells recognized a diverse antigenic landscape on the HA molecule, including HA head domain epitopes in antigenic sites A and B and in the trimer interface-II region and epitopes in the stem region. Most H7 antibodies exhibited little heterosubtypic breadth, but many recognized a wide diversity of unrelated H7 strains. We tested the antibodies for functional activity and identified clones with diverse patterns of inhibition, including neutralizing, hemagglutination- or egress-inhibiting, or HA trimer-disrupting activities. Thus, the human B cell response to primary H7 natural infection is diverse, highly functional, and broad for recognition of diverse H7 strains.


Subject(s)
Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/immunology , Humans
14.
Infect Genet Evol ; 93: 104993, 2021 09.
Article in English | MEDLINE | ID: mdl-34242774

ABSTRACT

Avian influenza virus (AIV) H7N9 that emerged in 2013 in eastern China is a novel zoonotic agent mainly circulating in poultry without clinical signs but causing severe disease with high fatality in humans in more than 5 waves. Since the emergence of highly pathogenic (HP) H7N9 variants in 2016, it has induced heavy losses in the poultry industry leading to the implementation of an intensive nationwide vaccination program at the end of wave 5 (September 2017). To characterize the ongoing evolution of H7N9 AIV, we conducted analyses of H7N9 glycoprotein genes obtained from 2013 to 2019. Bayesian analyses revealed a decreasing population size of HP H7N9 variants post wave 5. Phylogenetic topologies revealed that two novel small subclades were formed and carried several fixed amino acid mutations that were along HA and NA phylogenetic trees since wave 5. Some of the mutations were located at antigenic sites or receptor binding sites. The antigenic analysis may reveal a significant antigenic drift evaluated by hemagglutinin inhibition (HI) assay and the antigenicity of H7N9 AIV might evolute in large leaps in wave 7. Molecular simulations found that the mutations (V135T, S145P, and L226Q) around the HA receptor pocket increased the affinity to α2,3-linked sialic acid (SIA) while decreased to α2,6-linked SIA. Altered affinity may suggest that HP H7N9 variations aggravate the pathogenicity to poultry but lessen the threat to public health. Selection analyses showed that the HP H7N9 AIV experienced an increasing selection pressure since wave 5, and the national implementation of vaccination might intensify the role of natural selection during the evolution waves 6 and 7. In summary, our data provide important insights about the genetic and antigenic diversity of circulating HP H7N9 viruses from 2017 to 2019. Enhanced surveillance is urgently warranted to understand the current situation of HP H7N9 AIV.


Subject(s)
Antigenic Variation/immunology , Birds , Genetic Variation , Influenza A Virus, H7N9 Subtype/genetics , Influenza in Birds/virology , Animals , China , Influenza A Virus, H7N9 Subtype/immunology , Phylogeny
15.
Emerg Microbes Infect ; 10(1): 472-480, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33657971

ABSTRACT

Pandemic influenza, typically caused by the reassortment of human and avian influenza viruses, can result in severe or fatal infections in humans. Timely identification of potential pandemic viruses must be a priority in influenza virus surveillance. However, the range of host species responsible for the generation of novel pandemic influenza viruses remains unclear. In this study, we conducted serological surveys for avian and human influenza virus infections in farmed mink and determined the susceptibility of mink to prevailing avian and human virus subtypes. The results showed that farmed mink were commonly infected with human (H3N2 and H1N1/pdm) and avian (H7N9, H5N6, and H9N2) influenza A viruses. Correlational analysis indicated that transmission of human influenza viruses occurred from humans to mink, and that feed source was a probable route of avian influenza virus transmission to farmed mink. Animal experiments showed that mink were susceptible and permissive to circulating avian and human influenza viruses, and that human influenza viruses (H3N2 and H1N1/pdm), but not avian viruses, were capable of aerosol transmission among mink. These results indicate that farmed mink could be highly permissive "mixing vessels" for the reassortment of circulating human and avian influenza viruses. Therefore, to reduce the risk of emergence of novel pandemic viruses, feeding mink with raw poultry by-products should not be permitted, and epidemiological surveillance of influenza viruses in mink farms should be urgently implemented.


Subject(s)
Influenza A virus/pathogenicity , Mink/virology , Orthomyxoviridae Infections/transmission , Animals , Disease Models, Animal , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/immunology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A virus/immunology , Mink/immunology , Neutralization Tests , Orthomyxoviridae Infections/immunology , Reassortant Viruses/immunology , Reassortant Viruses/pathogenicity
16.
PLoS One ; 16(3): e0246613, 2021.
Article in English | MEDLINE | ID: mdl-33735274

ABSTRACT

BACKGROUND: A systemic evaluation of the antigenic differences of the H7 influenza hemagglutinin (HA) proteins, especially for the viruses isolated after 2016, are limited. The purpose of this study was to investigate the antigenic differences of major H7 strains with an ultimate aim to discover H7 HA proteins that can elicit protective receptor-binding antibodies against co-circulating H7 influenza strains. METHOD: A panel of eight H7 influenza strains were selected from 3,633 H7 HA amino acid sequences identified over the past two decades (2000-2018). The sequences were expressed on the surface of virus like particles (VLPs) and used to vaccinate C57BL/6 mice. Serum samples were collected and tested for hemagglutination-inhibition (HAI) activity. The vaccinated mice were challenged with lethal dose of H7N9 virus, A/Anhui/1/2013. RESULTS: VLPs expressing the H7 HA antigens elicited broadly reactive antibodies each of the selected H7 HAs, except the A/Turkey/Italy/589/2000 (Italy/00) H7 HA. A putative glycosylation due to an A169T substitution in antigenic site B was identified as a unique antigenic profile of Italy/00. Introduction of the putative glycosylation site (H7 HA-A169T) significantly altered the antigenic profile of HA of the A/Anhui/1/2013 (H7N9) strain. CONCLUSION: This study identified key amino acid mutations that result in severe vaccine mismatches for future H7 epidemics. Future universal influenza vaccine candidates will need to focus on viral variants with these key mutations.


Subject(s)
Antibodies, Viral/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/administration & dosage , Vaccines, Virus-Like Particle/administration & dosage , Animals , Antibodies, Neutralizing/metabolism , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/genetics , Mice , Mice, Inbred C57BL , Mutation , Phylogeny , Vaccines, Virus-Like Particle/genetics
17.
Sci Rep ; 11(1): 3126, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542302

ABSTRACT

Influenza H7N9 virus continues to cause infections in humans and represents a significant pandemic risk. During the most recent 5th epidemic wave in 2016/17 two distinct lineages with increased human infections and wider geographical spread emerged. In preparation for any future adaptations, broadly reactive antibodies against H7N9 are required for surveillance, therapy and prophylaxis. In this study we have isolated a panel of nanobodies (Nbs) with broad reactivity across H7 influenza strains, including H7N9 strains between 2013 and 2017. We also describe Nbs capable of distinguishing between the most recent high and low pathogenicity Yangtze River Delta lineage H7N9 strains. Nanobodies were classified into 5 distinct groups based on their epitope footprint determined using yeast display and mutational scanning. The epitope footprint of Nbs capable of distinguishing high pathogenic (HP) A/Guangdong/17SF003/2016 from low pathogenic (LP) A/Hong Kong/125/2017 (H7N9) were correlated to natural sequence divergence in the head domain at lysine 164. Several Nbs binding to the head domain were capable of viral neutralisation. The potency of one nanobody NB7-14 could be increased over 1000-fold to 113 pM by linking two Nbs together. Nbs specific for distinct epitopes on H7N9 may be useful for surveillance or therapy in human or veterinary settings.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H7N9 Subtype/drug effects , Peptide Library , Single-Domain Antibodies/biosynthesis , Amino Acid Sequence , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Binding Sites , Birds/virology , Epitopes/chemistry , Epitopes/genetics , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/immunology , Influenza in Birds/immunology , Influenza in Birds/prevention & control , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/transmission , Influenza, Human/virology , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Single-Domain Antibodies/isolation & purification
18.
Biochem Biophys Res Commun ; 545: 145-149, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33550095

ABSTRACT

In March 2013 it was reported by the World Health Organization (WHO) the first cases of human infections with avian influenza virus A (H7N9). From 2013 to December 2019, 1568 cases have been reported with 616 deaths. H7N9 infection has been associated with high morbidity and mortality rates, and vaccination is currently the most effective way to prevent infections and consequently flu-related severe illness. Developing and producing vaccines against pandemic influenza viruses is the main strategy for a response to a possible pandemic. This study aims to present the production of three industrial lots under current Good Manufacturing Practices (cGMP) of the active antigen used to produce the pandemic influenza vaccine candidate against A(H7N9). These batches were characterized and evaluated for quality standards and tested for immunogenicity in mice. The average yield was 173.50 ± 7.88 µg/mL of hemagglutinin and all the preparations met all the required specifications. The formulated H7N9 vaccine is poorly immunogenic and needs to be adjuvanted with an oil in water emulsion adjuvant (IB160) to achieve a best immune response, in a prime and in a boost scheme. These data are important for initial production planning and preparedness in the case of a H7N9 pandemic.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/biosynthesis , Influenza, Human/prevention & control , Pandemics/prevention & control , Animals , Antigens, Viral/biosynthesis , Antigens, Viral/immunology , Drug Compounding/methods , Drug Compounding/statistics & numerical data , Drug Industry/standards , Female , Humans , Influenza Vaccines/immunology , Influenza Vaccines/isolation & purification , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Vaccines, Inactivated/biosynthesis , Vaccines, Inactivated/immunology , Vaccines, Inactivated/isolation & purification
19.
Virus Genes ; 57(2): 164-171, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33606171

ABSTRACT

Influenza vaccines represent the most effective preventive strategy to control influenza virus infections; however, adaptive mutations frequently occur in the hemagglutinin (HA) glycoprotein during the preparation of candidate vaccine virus and production of vaccine in embryonated eggs. In our previous study, we constructed candidate vaccine virus (HA-R) to match the highly pathogenic avian influenza H7N9 viruses A/Guangdong/17SF003/2016 as part of a pandemic preparedness program. However, mixed amino acids (R, G, and I) were presented at position 220 (H3 numbering) in HA during passage in embryonated eggs. The residue at position 220 is located close to the receptor-binding site and the biological characteristics of this site remain to be elucidated. Therefore, in this study, using reverse genetics, we constructed two viruses carrying the single substitution in position 220 of HA (HA-G and HA-I) and evaluated the biological effects of substitution (R with G/I) on receptor binding, neuraminidase (NA) activity, growth characteristics, genetic stability, and antigenicity. The results revealed both mutant viruses exhibited lower HA binding affinities to two receptor types (sialic acid in alpha2,3- and alpha2,6-linkage to galactose, P < 0.001) and significant better growth characteristics compared to HA-R in two cells. Moreover, under similar NA enzymatic activity, the two mutant viruses eluted more easily from agglutinated erythrocytes than HA-R. Collectively, these results implied the balance of HA and NA in mutant viruses was a stronger determinant of viral growth than the individual amino acid in the HA position 220 in HA-R without strong binding between HA and sialylated receptors. Importantly, both the substitutions conferred altered antigenicity to the mutant viruses. In conclusion, amino acid substitutions at position 220 can substantially influence viral biological properties.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H7N9 Subtype/genetics , Influenza Vaccines/genetics , Amino Acid Substitution , Animals , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Madin Darby Canine Kidney Cells , Mutagenesis , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/immunology , Reverse Genetics
20.
Article in English | MEDLINE | ID: mdl-32205415

ABSTRACT

In early 2013, human infections caused by a novel H7N9 avian influenza virus (AIV) were first reported in China; these infections caused severe disease and death. The virus was initially low pathogenic to poultry, enabling it to spread widely in different provinces, especially in live poultry markets. Importantly, the H7N9 low pathogenic AIVs (LPAIVs) evolved into highly pathogenic AIVs (HPAIVs) in the beginning of 2017, causing a greater threat to human health and devastating losses to the poultry industry. Fortunately, nationwide vaccination of chickens with an H5/H7 bivalent inactivated avian influenza vaccine since September 2017 has successfully controlled H7N9 avian influenza infections in poultry and, importantly, has also prevented human infections. In this review, we summarize the biological properties of the H7N9 viruses, specifically their genetic evolution, adaptation, pathogenesis, receptor binding, transmission, drug resistance, and antigenic variation, as well as the prevention and control measures. The information obtained from investigating and managing the H7N9 viruses could improve our ability to understand other novel AIVs and formulate effective measures to control their threat to humans and animals.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/epidemiology , China/epidemiology , Communicable Disease Control , Drug Resistance , Humans , Immunogenicity, Vaccine , Influenza A Virus, H7N9 Subtype/genetics , Influenza, Human/prevention & control , Influenza, Human/transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...