Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.172
Filtrer
1.
Infect Dis Poverty ; 13(1): 56, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39090685

RÉSUMÉ

BACKGROUND: Non-pharmaceutical measures and travel restrictions have halted the spread of coronavirus disease 2019 (COVID-19) and influenza. Nonetheless, with COVID-19 restrictions lifted, an unanticipated outbreak of the influenza B/Victoria virus in late 2021 and another influenza H3N2 outbreak in mid-2022 occurred in Guangdong, southern China. The mechanism underlying this phenomenon remains unknown. To better prepare for potential influenza outbreaks during COVID-19 pandemic, we studied the molecular epidemiology and phylogenetics of influenza A(H3N2) and B/Victoria that circulated during the COVID-19 pandemic in this region. METHODS: From January 1, 2018 to December 31, 2022, we collected throat swabs from 173,401 patients in Guangdong who had acute respiratory tract infections. Influenza viruses in the samples were tested using reverse transcription-polymerase chain reaction, followed by subtype identification and sequencing of hemagglutinin (HA) and neuraminidase (NA) genes. Phylogenetic and genetic diversity analyses were performed on both genes from 403 samples. A rigorous molecular clock was aligned with the phylogenetic tree to measure the rate of viral evolution and the root-to-tip distance within strains in different years was assessed using regression curve models to determine the correlation. RESULTS: During the early period of COVID-19 control, various influenza viruses were nearly undetectable in respiratory specimens. When control measures were relaxed in January 2020, the influenza infection rate peaked at 4.94% (39/789) in December 2021, with the influenza B/Victoria accounting for 87.18% (34/39) of the total influenza cases. Six months later, the influenza infection rate again increased and peaked at 11.34% (255/2248) in June 2022; influenza A/H3N2 accounted for 94.51% (241/255) of the total influenza cases in autumn 2022. The diverse geographic distribution of HA genes of B/Victoria and A/H3N2 had drastically reduced, and most strains originated from China. The rate of B/Victoria HA evolution (3.11 × 10-3, P < 0.05) was 1.7 times faster than before the COVID-19 outbreak (1.80 × 10-3, P < 0.05). Likewise, the H3N2 HA gene's evolution rate was 7.96 × 10-3 (P < 0.05), which is 2.1 times faster than the strains' pre-COVID-19 evolution rate (3.81 × 10-3, P < 0.05). CONCLUSIONS: Despite the extraordinarily low detection rate of influenza infection, concealed influenza transmission may occur between individuals during strict COVID-19 control. This ultimately leads to the accumulation of viral mutations and accelerated evolution of H3N2 and B/Victoria viruses. Monitoring the evolution of influenza may provide insights and alerts regarding potential epidemics in the future.


Sujet(s)
COVID-19 , Sous-type H3N2 du virus de la grippe A , Virus influenza B , Grippe humaine , Épidémiologie moléculaire , Phylogenèse , SARS-CoV-2 , Humains , COVID-19/épidémiologie , COVID-19/virologie , COVID-19/transmission , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/isolement et purification , Chine/épidémiologie , Grippe humaine/épidémiologie , Grippe humaine/virologie , Virus influenza B/génétique , Virus influenza B/isolement et purification , Virus influenza B/classification , SARS-CoV-2/génétique , Adulte , Adulte d'âge moyen , Mâle , Femelle , Pandémies , Jeune adulte , Sujet âgé , Glycoprotéine hémagglutinine du virus influenza/génétique , Adolescent , Sialidase/génétique , Enfant , Enfant d'âge préscolaire
2.
mSphere ; 9(8): e0028324, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39087764

RÉSUMÉ

In 2009, a novel swine-origin H1N1 virus emerged, causing a pandemic. The virus, known as H1N1pdm09, quickly displaced the circulating H1 lineage and became the dominant seasonal influenza A virus subtype infecting humans. Human-to-swine spillovers of the H1N1pdm09 have occurred frequently, and each occurrence has led to sustained transmission of the human-origin H1N1pdm09 within swine populations. In the present study, we developed a lipid nanoparticle-based DNA vaccine (LNP-DNA) containing the hemagglutinin gene of a swine-origin H1N1pdm09. In pigs, this LNP-DNA vaccine induced a robust antibody response after a single intramuscular immunization and protected the pigs against challenge infection with the homologous swine-origin H1N1pdm09 virus. In a mouse model, the LNP-DNA vaccine induced antibody and T-cell responses and protected mice against lethal challenge with a mouse-adapted human-origin H1N1pdm09 virus. These findings demonstrate the potential of the LNP-DNA vaccine to protect against both swine- and human-origin H1N1pdm09 viruses. IMPORTANCE: Swine influenza A virus (IAV) is widespread and causes significant economic losses to the swine industry. Moreover, bidirectional transmission of IAV between swine and humans commonly occurs. Once introduced into the swine population, human-origin IAV often reassorts with endemic swine IAV, resulting in reassortant viruses. Thus, it is imperative to develop a vaccine that is not only effective against IAV strains endemic in swine but also capable of preventing the spillover of human-origin IAV. In this study, we developed a lipid nanoparticle-encapsulated DNA plasmid vaccine (LNP-DNA) that demonstrates efficacy against both swine- and human-origin H1N1 viruses. The LNP-DNA vaccines are non-infectious and non-viable, meeting the criteria to serve as a vaccine platform for rapidly updating vaccines. Collectively, this LNP-DNA vaccine approach holds great potential for alleviating the impact of IAV on the swine industry and preventing the emergence of reassortant IAV strains.


Sujet(s)
Anticorps antiviraux , Sous-type H1N1 du virus de la grippe A , Vaccins antigrippaux , Grippe humaine , Nanoparticules , Infections à Orthomyxoviridae , Maladies des porcs , Vaccins à ADN , Animaux , Sous-type H1N1 du virus de la grippe A/immunologie , Sous-type H1N1 du virus de la grippe A/génétique , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Suidae , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/médecine vétérinaire , Nanoparticules/administration et posologie , Humains , Souris , Maladies des porcs/prévention et contrôle , Maladies des porcs/virologie , Maladies des porcs/immunologie , Anticorps antiviraux/sang , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Femelle , Souris de lignée BALB C , Liposomes/administration et posologie
3.
Brief Bioinform ; 25(5)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39129362

RÉSUMÉ

Influenza viruses rapidly evolve to evade previously acquired human immunity. Maintaining vaccine efficacy necessitates continuous monitoring of antigenic differences among strains. Traditional serological methods for assessing these differences are labor-intensive and time-consuming, highlighting the need for efficient computational approaches. This paper proposes MetaFluAD, a meta-learning-based method designed to predict quantitative antigenic distances among strains. This method models antigenic relationships between strains, represented by their hemagglutinin (HA) sequences, as a weighted attributed network. Employing a graph neural network (GNN)-based encoder combined with a robust meta-learning framework, MetaFluAD learns comprehensive strain representations within a unified space encompassing both antigenic and genetic features. Furthermore, the meta-learning framework enables knowledge transfer across different influenza subtypes, allowing MetaFluAD to achieve remarkable performance with limited data. MetaFluAD demonstrates excellent performance and overall robustness across various influenza subtypes, including A/H3N2, A/H1N1, A/H5N1, B/Victoria, and B/Yamagata. MetaFluAD synthesizes the strengths of GNN-based encoding and meta-learning to offer a promising approach for accurate antigenic distance prediction. Additionally, MetaFluAD can effectively identify dominant antigenic clusters within seasonal influenza viruses, aiding in the development of effective vaccines and efficient monitoring of viral evolution.


Sujet(s)
Antigènes viraux , Humains , Antigènes viraux/génétique , Antigènes viraux/immunologie , , Grippe humaine/immunologie , Grippe humaine/virologie , Grippe humaine/prévention et contrôle , Biologie informatique/méthodes , Orthomyxoviridae/immunologie , Orthomyxoviridae/génétique , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Apprentissage machine
4.
PLoS One ; 19(8): e0308547, 2024.
Article de Anglais | MEDLINE | ID: mdl-39150957

RÉSUMÉ

The baculovirus expression vector system (BEVS) is a powerful tool in pharmaceutical biotechnology to infect insect cells and produce the recombinant proteins of interest. It has been well documented that optimizing the culture condition and its supplementation through designed experiments is critical for maximum protein production. In this study, besides physicochemical parameters including incubation temperature, cell count of infection, multiplicity of infection, and feeding percentage, potential supplementary factors such as cholesterol, polyamine, galactose, pluronic-F68, glucose, L-glutamine, and ZnSO4 were screened for Spodoptera frugiperda (Sf9) cell culture and expression of hemagglutinin (HA) protein of Influenza virus via Placket-Burman design and then optimized through Box-Behnken approach. The optimized conditions were then applied for scale-up culture and the expressed r-HA protein was characterized. Optimization of selected parameters via the Box-Behnken approach indicated that feed percentage, cell count, and multiplicity of infection are the main parameters affecting r-HA expression level and potency compared to the previously established culture condition. This study demonstrated the effectiveness of designing experiments to select and optimize important parameters that potentially affect Sf9 cell culture, r-HA expression, and its potency in the BEVS system.


Sujet(s)
Glycoprotéine hémagglutinine du virus influenza , Spodoptera , Animaux , Cellules Sf9 , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Glycoprotéine hémagglutinine du virus influenza/génétique , Techniques de culture cellulaire/méthodes , Milieux de culture , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Protéines recombinantes/biosynthèse , Baculoviridae/génétique , Baculoviridae/métabolisme
5.
Trop Biomed ; 41(2): 183-189, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-39154271

RÉSUMÉ

Low pathogenic avian influenza (LPAI) subtype H9N2 is a causative agent that has raised increasing concern about its impact on poultry and potential public health threats. Even though H9N2 is endemic in Peninsular Malaysia, it was first reported in Sabah in August 2022, after an outbreak associated with high mortality in broiler chickens. In the present study, based on the hemagglutinin (HA) gene, we report the genetic variations and phylogenetic analysis of a H9N2 virus isolated from broiler chickens in Sabah. The sequence analysis of the HA gene revealed a 98% similarity to the H9N2 virus recently isolated from China in 2018. The amino acids in the HA cleavage site displayed a characteristic LPAI motif (PARSSR/ GLF). Notably, at position 226, the isolate had amino acid Leucine (L) demonstrating its ability to bind to the receptor of mammals, resulting in the potential risk of transmission to humans. In addition, the H9N2 isolate harboured seven potential N-glycosylation sites. The phylogenetic analysis revealed that the isolate belonged to clade h9.4.2.5 in the Y280 lineage, similar to previously reported in Malaysia. However, we observed that the isolate in this study falls in a different cluster compared with previous Malaysian isolates, suggesting different source of H9N2 introduction into the country. This prompts us to propose continuous and thorough surveillance of poultry across the country and the necessity of implementing farm biosecurity to minimize economic losses and potential threats to public health.


Sujet(s)
Poulets , Sous-type H9N2 du virus de la grippe A , Grippe chez les oiseaux , Phylogenèse , Maladies de la volaille , Animaux , Sous-type H9N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/isolement et purification , Sous-type H9N2 du virus de la grippe A/classification , Grippe chez les oiseaux/virologie , Malaisie , Maladies de la volaille/virologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Variation génétique
6.
Sci Rep ; 14(1): 18750, 2024 08 13.
Article de Anglais | MEDLINE | ID: mdl-39138310

RÉSUMÉ

In 2004, the equine-origin H3N8 canine influenza virus (CIV) first caused an outbreak with lethal cases in racing greyhounds in Florida, USA, and then spread to domestic dogs nationwide. Although transmission of this canine virus to humans has not been reported, it is important to evaluate its zoonotic potential because of the high contact opportunities between companion dogs and humans. To gain insight into the interspecies transmissibility of H3N8 CIV, we tested its adaptability to human respiratory A549 cells through successive passages. We found that CIV acquired high growth properties in these cells mainly through mutations in surface glycoproteins, such as hemagglutinin (HA) and neuraminidase (NA). Our reverse genetics approach revealed that HA2-K82E, HA2-R163K, and NA-S18L mutations were responsible for the increased growth of CIV in human cells. Molecular analyses revealed that both HA2 mutations altered the optimum pH for HA membrane fusion activity and that the NA mutation changed the HA-NA functional balance. These findings suggest that H3N8 CIV could evolve into a human pathogen with pandemic potential through a small number of mutations, thereby posing a threat to public health in the future.


Sujet(s)
Sous-type H3N8 du virus de la grippe A , Mutation , Sialidase , Humains , Animaux , Chiens , Sous-type H3N8 du virus de la grippe A/génétique , Sous-type H3N8 du virus de la grippe A/physiologie , Sialidase/génétique , Sialidase/métabolisme , Cellules A549 , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/transmission , Adaptation physiologique/génétique , Grippe humaine/virologie , Grippe humaine/transmission
7.
Emerg Microbes Infect ; 13(1): 2389095, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39101691

RÉSUMÉ

Influenza virus infection poses a continual menace to public health. Here, we developed soluble trimeric HA ectodomain vaccines by establishing interprotomer disulfide bonds in the stem region, which effectively preserve the native antigenicity of stem epitopes. The stable trimeric H1 ectodomain proteins exhibited higher thermal stabilities in comparison with unmodified HAs and showed strong binding activities towards a panel of anti-stem cross-reactive antibodies that recognize either interprotomer or intraprotomer epitopes. Negative stain transmission electron microscopy (TEM) analysis revealed the stable trimer architecture of the interprotomer disulfide-stapled WA11#5, NC99#2, and FLD#1 proteins as well as the irregular aggregation of unmodified HA molecules. Immunizations of mice with those trimeric HA ectodomain vaccines formulated with incomplete Freund's adjuvant elicited significantly more potent cross-neutralizing antibody responses and offered broader immuno-protection against lethal infections with heterologous influenza strains compared to unmodified HA proteins. Additionally, the findings of our study indicate that elevated levels of HA stem-specific antibody responses correlate with strengthened cross-protections. Our design strategy has proven effective in trimerizing HA ectodomains derived from both influenza A and B viruses, thereby providing a valuable reference for designing future influenza HA immunogens.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Disulfures , Glycoprotéine hémagglutinine du virus influenza , Vaccins antigrippaux , Souris de lignée BALB C , Infections à Orthomyxoviridae , Animaux , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Anticorps antiviraux/immunologie , Souris , Disulfures/composition chimique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Anticorps neutralisants/immunologie , Femelle , Protection croisée/immunologie , Réactions croisées , Humains , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Épitopes/immunologie , Épitopes/génétique , Épitopes/composition chimique , Multimérisation de protéines , Virus influenza B/immunologie , Virus influenza B/génétique , Virus influenza B/composition chimique
8.
J Med Microbiol ; 73(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39073070

RÉSUMÉ

Introduction. After two seasons of absence and low circulation, influenza activity increased significantly in the winter of 2022-2023. This study aims to characterize virological and epidemiological aspects of influenza infection in Bulgaria during the 2022-2023 season and perform a phylogenetic/molecular analysis of the hemagglutinin (HA) and neuraminidase (NA) sequences of representative influenza strains.Hypothesis/Gap Statement. Influenza A and B viruses generate new genetic groups/clades each season, replacing previously circulating variants. This results in increased antigenic distances from current vaccine strains. Strengthening existing influenza surveillance is essential to meet the challenges posed by the co-circulation of influenza and SARS-CoV-2.Methodology. We tested 2713 clinical samples from patients with acute respiratory illnesses using a multiplex real-time RT-PCR kit (FluSC2) to detect influenza A/B and Severe acute respiratory syndrome coronavirus-2(SARS-CoV-2) simultaneously. Representative Bulgarian influenza strains were sequenced at the WHO Collaborating Centres in London, UK, and Atlanta, USA.Results. Influenza virus was detected in 694 (25.6 %) patients. Of these, 364 (52.4 %), 213 (30.7 %) and 117 (16.9 %) were positive for influenza A(H1N1)pdm09, A(H3N2) and B/Victoria lineage virus, respectively. HA genes of the 47 influenza A(H1N1)pdm09 viruses fell into clades 5a.2. and 5a.2a.1 within the 6B.5A.1A.5a.2 group. Twenty-seven A(H3N2) viruses belonging to subclades 2b, 2a.1, 2a.1b and 2a.3a.1 within the 3C.2a1b.2a.2 group were analysed. All 23 sequenced B/Victoria lineage viruses were classified into the V1A.3a.2 group. We identified amino acid substitutions in HA and NA compared with the vaccine strains, including several substitutions in the HA antigenic sites.Conclusion. The study's findings showed genetic diversity among the influenza A viruses and, to a lesser extent, among B viruses, circulating in the first season after the lifting of anti-COVID-19 measures.


Sujet(s)
Variation génétique , Virus influenza B , Grippe humaine , Sialidase , Phylogenèse , SARS-CoV-2 , Humains , Grippe humaine/virologie , Grippe humaine/épidémiologie , Virus influenza B/génétique , Virus influenza B/classification , Virus influenza B/isolement et purification , SARS-CoV-2/génétique , SARS-CoV-2/classification , Sialidase/génétique , Adulte , Mâle , Adulte d'âge moyen , Femelle , Bulgarie/épidémiologie , Jeune adulte , Sujet âgé , Glycoprotéine hémagglutinine du virus influenza/génétique , Enfant d'âge préscolaire , Enfant , Adolescent , COVID-19/épidémiologie , COVID-19/virologie , Nourrisson , Saisons , Virus de la grippe A/génétique , Virus de la grippe A/classification , Virus de la grippe A/isolement et purification , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/classification , Sous-type H1N1 du virus de la grippe A/isolement et purification , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/classification , Sous-type H3N2 du virus de la grippe A/isolement et purification
10.
Nat Commun ; 15(1): 5800, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987276

RÉSUMÉ

Enhancing influenza vaccine cross-protection is imperative to alleviate the significant public health burden of influenza. Heterologous sequential immunization may synergize diverse vaccine formulations and routes to improve vaccine potency and breadth. Here we investigate the effects of immunization strategies on the generation of cross-protective immune responses in female Balb/c mice, utilizing mRNA lipid nanoparticle (LNP) and protein-based PHC nanoparticle vaccines targeting influenza hemagglutinin. Our findings emphasize the crucial role of priming vaccination in shaping Th bias and immunodominance hierarchies. mRNA LNP prime favors Th1-leaning responses, while PHC prime elicits Th2-skewing responses. We demonstrate that cellular and mucosal immune responses are pivotal correlates of cross-protection against influenza. Notably, intranasal PHC immunization outperforms its intramuscular counterpart in inducing mucosal immunity and conferring cross-protection. Sequential mRNA LNP prime and intranasal PHC boost demonstrate optimal cross-protection against antigenically drifted and shifted influenza strains. Our study offers valuable insights into tailoring immunization strategies to optimize influenza vaccine effectiveness.


Sujet(s)
Administration par voie nasale , Protection croisée , Vaccins antigrippaux , Souris de lignée BALB C , Nanoparticules , Infections à Orthomyxoviridae , Animaux , Femelle , Humains , Souris , Anticorps antiviraux/immunologie , Protection croisée/immunologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Immunité muqueuse/immunologie , Immunisation/méthodes , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Lipides/composition chimique , Liposomes , Nanoparticules/composition chimique , /administration et posologie , /immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , ARN messager/génétique , ARN messager/immunologie , Vaccination/méthodes
11.
Vet Microbiol ; 296: 110188, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39018942

RÉSUMÉ

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.


Sujet(s)
Poulets , Glycoprotéine hémagglutinine du virus influenza , Sous-type H9N2 du virus de la grippe A , Grippe chez les oiseaux , Sous-type H9N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/immunologie , Animaux , Poulets/virologie , Grippe chez les oiseaux/virologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Chine , Substitution d'acide aminé , Maladies de la volaille/virologie , Mutation , Antigènes viraux/immunologie , Antigènes viraux/génétique , Réplication virale , Phylogenèse , Sialidase/génétique , Sialidase/immunologie , Acides aminés/génétique
12.
PLoS One ; 19(7): e0307100, 2024.
Article de Anglais | MEDLINE | ID: mdl-39012858

RÉSUMÉ

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


Sujet(s)
Poulets , Sous-type H5N1 du virus de la grippe A , Vaccins antigrippaux , Grippe chez les oiseaux , Animaux , Poulets/virologie , Poulets/immunologie , Grippe chez les oiseaux/prévention et contrôle , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/immunologie , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/génétique , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Amérique du Nord , Vaccination , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/virologie , Maladies de la volaille/immunologie , Herpèsvirus de type 1 du dindon/immunologie , Herpèsvirus de type 1 du dindon/génétique
13.
Front Cell Infect Microbiol ; 14: 1433661, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979510

RÉSUMÉ

In recent years, the avian influenza virus has emerged as a significant threat to both human and public health. This study focuses on a patient infected with the H10N3 subtype of avian influenza virus, admitted to the Third People's Hospital of Kunming City on March 6, 2024. Metagenomic RNA sequencing and polymerase chain reaction (PCR) analysis were conducted on the patient's sputum, confirming the H10N3 infection. The patient presented severe pneumonia symptoms such as fever, expectoration, chest tightness, shortness of breath, and cough. Phylogenetic analysis of the Haemagglutinin (HA) and neuraminidase (NA) genes of the virus showed that the virus was most closely related to a case of human infection with the H10N3 subtype of avian influenza virus found in Zhejiang Province, China. Analysis of amino acid mutation sites identified four mutations potentially hazardous to human health. Consequently, this underscores the importance of continuous and vigilant monitoring of the dynamics surrounding the H10N3 subtype of avian influenza virus, utilizing advanced genomic surveillance techniques.


Sujet(s)
Glycoprotéine hémagglutinine du virus influenza , Virus de la grippe A , Grippe humaine , Sialidase , Phylogenèse , Humains , Chine/épidémiologie , Grippe humaine/virologie , Sialidase/génétique , Glycoprotéine hémagglutinine du virus influenza/génétique , Virus de la grippe A/génétique , Virus de la grippe A/classification , Virus de la grippe A/isolement et purification , Mutation , Analyse de mutations d'ADN , Animaux , Grippe chez les oiseaux/virologie , Protéines virales/génétique , Expectoration/virologie , Oiseaux/virologie , Mâle , ARN viral/génétique
14.
Cell Host Microbe ; 32(8): 1397-1411.e11, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39032493

RÉSUMÉ

Human influenza virus evolves to escape neutralization by polyclonal antibodies. However, we have a limited understanding of how the antigenic effects of viral mutations vary across the human population and how this heterogeneity affects virus evolution. Here, we use deep mutational scanning to map how mutations to the hemagglutinin (HA) proteins of two H3N2 strains, A/Hong Kong/45/2019 and A/Perth/16/2009, affect neutralization by serum from individuals of a variety of ages. The effects of HA mutations on serum neutralization differ across age groups in ways that can be partially rationalized in terms of exposure histories. Mutations that were fixed in influenza variants after 2020 cause greater escape from sera from younger individuals compared with adults. Overall, these results demonstrate that influenza faces distinct antigenic selection regimes from different age groups and suggest approaches to understand how this heterogeneous selection shapes viral evolution.


Sujet(s)
Anticorps antiviraux , Glycoprotéine hémagglutinine du virus influenza , Sous-type H3N2 du virus de la grippe A , Grippe humaine , Mutation , Humains , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/immunologie , Adulte , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Grippe humaine/virologie , Grippe humaine/immunologie , Facteurs âges , Adulte d'âge moyen , Jeune adulte , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Antigènes viraux/génétique , Antigènes viraux/immunologie , Adolescent , Évolution moléculaire , Sujet âgé , Enfant
15.
Poult Sci ; 103(9): 103988, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38970848

RÉSUMÉ

Inactivated vaccines play an important role in preventing and controlling the epidemic caused by the H5 subtype avian influenza virus. The vaccine strains are updated in response to alterations in surface protein antigens, while an avian-derived vaccine internal backbone with a high replicative capacity in chicken embryonated eggs and MDCK cells is essential for vaccine development. In this study, we constructed recombinant viruses using the clade 2.3.4.4d A/chicken/Jiangsu/GY5/2017(H5N6, CkG) strain as the surface protein donor and the clade 2.3.4.4b A/duck/Jiangsu/84512/2017(H5N6, Dk8) strain with high replicative ability as an internal donor. After optimization, the integration of the M gene from the CkG into the internal genes from Dk8 (8GM) was selected as the high-yield vaccine internal backbone, as the combination improved the hemagglutinin1/nucleoprotein (HA1/NP) ratio in recombinant viruses. The r8GMΔG with attenuated hemagglutinin and neuraminidase from the CkG exhibited high-growth capacity in both chicken embryos and MDCK cell cultures. The inactivated r8GMΔG vaccine candidate also induced a higher hemagglutination inhibition antibody titer and microneutralization titer than the vaccine strain using PR8 as the internal backbone. Further, the inactivated r8GMΔG vaccine candidate provided complete protection against wild-type strain challenge. Therefore, our study provides a high-yield, easy-to-cultivate candidate donor as an internal gene backbone for vaccine development.


Sujet(s)
Poulets , Vaccins antigrippaux , Grippe chez les oiseaux , Animaux , Vaccins antigrippaux/immunologie , Chiens , Cellules rénales canines Madin-Darby , Embryon de poulet , Grippe chez les oiseaux/prévention et contrôle , Virus de la grippe A/immunologie , Vaccins inactivés/immunologie , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/virologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie
16.
J Virol ; 98(8): e0078124, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39078191

RÉSUMÉ

Influenza remains a worldwide public health threat. Although seasonal influenza vaccines are currently the best means of preventing severe disease, the standard-of-care vaccines require frequent updating due to antigenic drift and can have low efficacy, particularly in vulnerable populations. Here, we demonstrate that a single administration of a recombinant adenovirus-associated virus (rAAV) vector expressing a computationally optimized broadly reactive antigen (COBRA)-derived influenza H1 hemagglutinin (HA) induces strongly neutralizing and broadly protective antibodies in naïve mice and ferrets with pre-existing influenza immunity. Following a lethal viral challenge, the rAAV-COBRA vaccine allowed for significantly reduced viral loads in the upper and lower respiratory tracts and complete protection from morbidity and mortality that lasted for at least 5 months post-vaccination. We observed no signs of antibody waning during this study. CpG motif enrichment of the antigen can act as an internal adjuvant to further enhance the immune responses to allow for lower vaccine dosages with the induction of unique interferon-producing CD4+ and CD8+ T cells specific to HA head and stem peptide sequences. Our studies highlight the utility of rAAV as an effective platform to improve seasonal influenza vaccines. IMPORTANCE: Developing an improved seasonal influenza vaccine remains an ambitious goal of researchers and clinicians alike. With influenza routinely causing severe epidemics with the potential to rise to pandemic levels, it is critical to create an effective, broadly protective, and durable vaccine to improve public health worldwide. As a potential solution, we created a rAAV viral vector expressing a COBRA-optimized influenza hemagglutinin antigen with modestly enriched CpG motifs to evoke a robust and long-lasting immune response after a single intramuscular dose without needing boosts or adjuvants. Importantly, the rAAV vaccine boosted antibody breadth to future strains in ferrets with pre-existing influenza immunity. Together, our data support further investigation into the utility of viral vectors as a potential avenue to improve our seasonal influenza vaccines.


Sujet(s)
Immunité acquise , Anticorps antiviraux , Dependovirus , Furets , Glycoprotéine hémagglutinine du virus influenza , Vaccins antigrippaux , Infections à Orthomyxoviridae , Animaux , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Souris , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Dependovirus/génétique , Dependovirus/immunologie , Anticorps neutralisants/immunologie , Humains , Femelle , Vecteurs génétiques , Souris de lignée BALB C , Vaccination , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Lymphocytes T CD8+/immunologie
17.
mBio ; 15(8): e0180424, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39037231

RÉSUMÉ

Influenza A virus (IAV) is well known for its pandemic potential. While current surveillance and vaccination strategies are highly effective, therapeutic approaches are often short-lived due to the high mutation rates of IAV. Recently, monoclonal antibodies (mAbs) have emerged as a promising therapeutic approach, both against current strains and future IAV pandemics. In addition to mAbs, several antibody-like alternatives exist, which aim to improve upon mAbs. Among these, Affimers stand out for their short development time, high expression levels in Escherichia coli, and animal-free production. In this study, we utilized the Affimer platform to isolate and produce specific and potent inhibitors of IAV. Using a monomeric version of the IAV trimeric hemagglutinin (HA) fusion protein, we isolated 12 Affimers that inhibit IAV infection in vitro. Two of these Affimers were characterized in detail and exhibited nanomolar-binding affinities to the target H3 HA protein, specifically binding to the HA1 head domain. Cryo-electron microscopy (cryo-EM), employing a novel spray approach to prepare cryo-grids, allowed us to image HA-Affimer complexes. Combined with functional assays, we determined that these Affimers inhibit IAV by blocking the interaction of HA with the host-cell receptor, sialic acid. Furthermore, these Affimers inhibited IAV strains closely related to the one used for their isolation. Overall, our results support the use of Affimers as a viable alternative to existing targeted therapies for IAV and highlight their potential as diagnostic reagents. IMPORTANCE: Influenza A virus is one of the few viruses that can cause devastating pandemics. Due to the high mutation rates of this virus, annual vaccination is required, and antivirals are short-lived. Monoclonal antibodies present a promising approach to tackle influenza virus infections but are associated with some limitations. To improve on this strategy, we explored the Affimer platform, which are antibody-like proteins made in bacteria. By performing phage-display against a monomeric version of influenza virus fusion protein, an established viral target, we were able to isolate Affimers that inhibit influenza virus infection in vitro. We characterized the mechanism of inhibition of the Affimers by using assays targeting different stages of the viral replication cycle. We additionally characterized HA-Affimer complex structure, using a novel approach to prepare samples for cryo-electron microscopy. Overall, these results show that Affimers are a promising tool against influenza virus infection.


Sujet(s)
Cryomicroscopie électronique , Glycoprotéine hémagglutinine du virus influenza , Virus de la grippe A , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/génétique , Humains , Animaux , Antiviraux/pharmacologie , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/immunologie , Protéines de fusion recombinantes/composition chimique , Chiens , Grippe humaine/virologie , Cellules rénales canines Madin-Darby
18.
Protein Expr Purif ; 223: 106541, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-38971212

RÉSUMÉ

Avian influenza poses a significant global health threat, with the potential for widespread pandemics and devastating consequences. Hemagglutinin (HA), a critical surface glycoprotein of influenza viruses, plays a pivotal role in viral entry and serves as a primary target for subunit vaccine development. In this study, we successfully cloned, expressed, and purified hemagglutinin from the circulating strain of H5N1 influenza virus using a robust molecular biology approach. The cloning process involved insertion of the synthetic HA gene into the pET21b vector, confirmed through double digestion and sequencing. SDS-PAGE analysis confirmed the presence of the expected 60 kDa protein band post-induction. Following expression, the protein was subjected to purification via Ni-NTA affinity chromatography, yielding pure protein fractions. Native PAGE analysis confirmed the protein's oligomeric forms, essential for optimal antigenicity. Western blot analysis further validated protein identity using anti-His and anti-HA antibodies. MALDI-TOF analysis confirmed the protein's sequence integrity, while hemagglutination assay demonstrated its biological activity in binding to N-acetyl neuraminic acid. These findings underscore the potential of recombinant hemagglutinin as a valuable antigen for diagnosis and biochemical assays as well as for vaccine development against avian influenza. In conclusion, this study represents a critical guide for bacterial production of H5N1 HA, which can be a cost-effective and simpler strategy compared to mammalian protein expression. Further research into optimizing vaccine candidates and production methods will be essential in combating the ongoing threat of avian influenza pandemics.


Sujet(s)
Glycoprotéine hémagglutinine du virus influenza , Sous-type H5N1 du virus de la grippe A , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/composition chimique , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Glycoprotéine hémagglutinine du virus influenza/biosynthèse , Protéines recombinantes/génétique , Protéines recombinantes/composition chimique , Protéines recombinantes/biosynthèse , Protéines recombinantes/immunologie , Protéines recombinantes/isolement et purification , Protéines recombinantes/métabolisme , Animaux , Escherichia coli/génétique , Escherichia coli/métabolisme , Hémagglutination , Grippe chez les oiseaux/prévention et contrôle , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/immunologie , Grippe chez les oiseaux/génétique , Clonage moléculaire , Expression des gènes , Multimérisation de protéines , Humains , Oiseaux
19.
Emerg Infect Dis ; 30(8): 1-13, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39043566

RÉSUMÉ

Influenza A/H9 viruses circulate worldwide in wild and domestic avian species, continuing to evolve and posing a zoonotic risk. A substantial increase in human infections with A/H9N2 subtype avian influenza viruses (AIVs) and the emergence of novel reassortants carrying A/H9N2-origin internal genes has occurred in recent years. Different names have been used to describe the circulating and emerging A/H9 lineages. To address this issue, an international group of experts from animal and public health laboratories, endorsed by the WOAH/FAO Network of Expertise on Animal Influenza, has created a practical lineage classification and nomenclature system based on the analysis of 10,638 hemagglutinin sequences from A/H9 AIVs sampled worldwide. This system incorporates phylogenetic relationships and epidemiologic characteristics designed to trace emerging and circulating lineages and clades. To aid in lineage and clade assignment, an online tool has been created. This proposed classification enables rapid comprehension of the global spread and evolution of A/H9 AIVs.


Sujet(s)
Grippe chez les oiseaux , Grippe humaine , Phylogenèse , Terminologie comme sujet , Animaux , Humains , Grippe humaine/épidémiologie , Grippe humaine/virologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/épidémiologie , Oiseaux/virologie , Sous-type H9N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/classification , Glycoprotéine hémagglutinine du virus influenza/génétique
20.
J Gen Virol ; 105(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38980150

RÉSUMÉ

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


Sujet(s)
Poulets , Furets , Sous-type H7N9 du virus de la grippe A , Grippe chez les oiseaux , Dindons , Animaux , Dindons/virologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/transmission , Sous-type H7N9 du virus de la grippe A/génétique , Sous-type H7N9 du virus de la grippe A/pathogénicité , Poulets/virologie , Virulence , Chine/épidémiologie , Maladies de la volaille/virologie , Maladies de la volaille/transmission , Glycoprotéine hémagglutinine du virus influenza/génétique , Humains , Excrétion virale , Réplication virale , Zoonoses/virologie , Grippe humaine/virologie , Grippe humaine/transmission
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE