Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.641
Filtrer
1.
PeerJ ; 12: e17523, 2024.
Article de Anglais | MEDLINE | ID: mdl-38846750

RÉSUMÉ

Background: Influenza A(H3N2) virus evolves continuously. Its hemagglutinin (HA) and neuraminidase (NA) genes have high genetic variation due to the antigenic drift. This study aimed to investigate the characteristics and evolution of HA and NA genes of the influenza A(H3N2) virus in Thailand. Methods: Influenza A positive respiratory samples from 2015 to 2018 were subtyped by multiplex real-time RT-PCR. Full-length HA and NA genes from the positive samples of influenza A(H3N2) were amplified and sequenced. Phylogenetic analysis with the maximum likelihood method was used to investigate the evolution of the virus compared with the WHO-recommended influenza vaccine strain. Homology modeling and N-glycosylation site prediction were also performed. Results: Out of 443 samples, 147 (33.18%) were A(H1N1)pdm09 and 296 (66.82%) were A(H3N2). The A(H3N2) viruses circulating in 2015 were clade 3C.2a whereas sub-clade 3C.2a1 and 3C.2a2 dominated in 2016-2017 and 2018, respectively. Amino acid substitutions were found in all antigenic sites A, B, C, D, and E of HA but the majority of the substitutions were located at antigenic sites A and B. The S245N and N329S substitutions in the NA gene affect the N-glycosylation. None of the mutations associated with resistance to NA inhibitors were observed. Mean evolutionary rates of the HA and NA genes were 3.47 × 10 -3 and 2.98 × 10-3 substitutions per site per year. Conclusion: The influenza A(H3N2) virus is very genetically diverse and is always evolving to evade host defenses. The HA and NA gene features including the evolutionary rate of the influenza A(H3N2) viruses that were circulating in Thailand between 2015 and 2018 are described. This information is useful for monitoring the genetic characteristics and evolution in HA and NA genes of influenza A(H3N2) virus in Thailand which is crucial for predicting the influenza vaccine strains resulting in high vaccine effectiveness.


Sujet(s)
Évolution moléculaire , Glycoprotéine hémagglutinine du virus influenza , Sous-type H3N2 du virus de la grippe A , Grippe humaine , Sialidase , Phylogenèse , Thaïlande/épidémiologie , Sialidase/génétique , Sous-type H3N2 du virus de la grippe A/génétique , Humains , Grippe humaine/virologie , Grippe humaine/épidémiologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Substitution d'acide aminé
3.
Front Immunol ; 15: 1425842, 2024.
Article de Anglais | MEDLINE | ID: mdl-38915410

RÉSUMÉ

Vaccination against influenza virus can reduce the risk of influenza by 40% to 60%, they rely on the production of neutralizing antibodies specific to influenza hemagglutinin (HA) ignoring the neuraminidase (NA) as an important surface target. Vaccination with standardized NA concentration may offer broader and longer-lasting protection against influenza infection. In this regard, we aimed to compare the potency of a NA displayed on the surface of a VLP with a soluble NA. The baculovirus expression system (BEVS) and the novel virus-free Tnms42 insect cell line were used to express N2 NA on gag-based VLPs. To produce VLP immunogens with high levels of purity and concentration, a two-step chromatography purification process combined with ultracentrifugation was used. In a prime/boost vaccination scheme, mice vaccinated with 1 µg of the N2-VLPs were protected from mortality, while mice receiving the same dose of unadjuvanted NA in soluble form succumbed to the lethal infection. Moreover, NA inhibition assays and NA-ELISAs of pre-boost and pre-challenge sera confirm that the VLP preparation induced higher levels of NA-specific antibodies outperforming the soluble unadjuvanted NA.


Sujet(s)
Anticorps antiviraux , Vaccins antigrippaux , Sialidase , Infections à Orthomyxoviridae , Vaccins à pseudo-particules virales , Animaux , Sialidase/immunologie , Sialidase/génétique , Vaccins antigrippaux/immunologie , Vaccins à pseudo-particules virales/immunologie , Vaccins à pseudo-particules virales/génétique , Vaccins à pseudo-particules virales/administration et posologie , Souris , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Femelle , Souris de lignée BALB C , Protéines recombinantes/immunologie , Protéines recombinantes/génétique , , Humains , Vaccination/méthodes
4.
Elife ; 122024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38805550

RÉSUMÉ

Human H3N2 influenza viruses are subject to rapid antigenic evolution which translates into frequent updates of the composition of seasonal influenza vaccines. Despite these updates, the effectiveness of influenza vaccines against H3N2-associated disease is suboptimal. Seasonal influenza vaccines primarily induce hemagglutinin-specific antibody responses. However, antibodies directed against influenza neuraminidase (NA) also contribute to protection. Here, we analysed the antigenic diversity of a panel of N2 NAs derived from human H3N2 viruses that circulated between 2009 and 2017. The antigenic breadth of these NAs was determined based on the NA inhibition (NAI) of a broad panel of ferret and mouse immune sera that were raised by infection and recombinant N2 NA immunisation. This assessment allowed us to distinguish at least four antigenic groups in the N2 NAs derived from human H3N2 viruses that circulated between 2009 and 2017. Computational analysis further revealed that the amino acid residues in N2 NA that have a major impact on susceptibility to NAI by immune sera are in proximity of the catalytic site. Finally, a machine learning method was developed that allowed to accurately predict the impact of mutations that are present in our N2 NA panel on NAI. These findings have important implications for the renewed interest to develop improved influenza vaccines based on the inclusion of a protective NA antigen formulation.


Two proteins, the hemagglutinin and the neuraminidase, protrude from the surface of the influenza virus. Their detection by the immune system allows the host organism to mount defences against the viral threat. The virus evolves in response to this pressure, which manifests as changes in the appearance of its hemagglutinin and neuraminidase. This process, known as antigenic drift, leads to the proteins evading detection. It is also why flu vaccines require frequent updates, as they rely on 'training' the immune system to recognise the most important strains in circulation ­ primarily by exposing it to appropriate versions of hemagglutinin. While the antigenic drift of hemagglutinin has been extensively studied, much less is known about how the neuraminidase accumulates mutations, and how these affect the immune response. To investigate this question, Catani et al. selected 43 genetically distant neuraminidases from human viral samples isolated between 2009 and 2017. Statistical analyses were applied to define their relatedness, revealing that a group of closely related neuraminidases predominated from 2009 to 2015, before they were being taken over by a second group. A third group, which was identified in viruses isolated in 2013, was remarkably close to the neuraminidase of strains that circulated in the late 1990s. The fourth and final group of neuraminidases was derived from influenza viruses that normally circulate in pigs but can also occasionally infect humans. Next, Catani et al. examined the immune response that these 43 neuraminidases could elicit in mice, as well as in ferrets ­ the animal most traditionally used in influenza research. This allowed them to pinpoint which changes in the neuraminidase sequences were important to escape recognition by the host. Data obtained from the two model species were comparable, suggesting that these experiments could be conducted on mice going forward, which are easier to work with than ferrets. Finally, Catani et al. used machine learning to build a computational model that could predict how strongly the immune system would respond to a specific neuraminidase variant. These findings could help guide the development of new vaccines that include neuraminidases tailored to best prime and train the immune system against a larger variety of strains. This may aid the development of 'supra-seasonal' vaccines that protect against a broad range of influenza viruses, reducing the need for yearly updates.


Sujet(s)
Antigènes viraux , Furets , Sous-type H3N2 du virus de la grippe A , Grippe humaine , Sialidase , Sialidase/immunologie , Sialidase/génétique , Sous-type H3N2 du virus de la grippe A/immunologie , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/enzymologie , Humains , Animaux , Antigènes viraux/immunologie , Antigènes viraux/génétique , Souris , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Anticorps antiviraux/immunologie , Vaccins antigrippaux/immunologie , Variation des antigènes , Protéines virales/immunologie , Protéines virales/génétique , Protéines virales/composition chimique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie
5.
Arch Biochem Biophys ; 757: 110041, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38750923

RÉSUMÉ

The influenza virus neuraminidase (NA) protein is responsible for actively cleaving the sialic acid (SA) bound to the viral hemagglutinin. In the present study, we identified a combination of five novel amino acid substitutions in the NA, conferring increased substrate binding and altered surface characteristics to a low pathogenic avian influenza (LPAI) H9N2 virus strain. The H9N2 strain reported from India, A/Environmental/India/1726265/2017 (H9N2-1726265) showed the combination of amino acid substitutions T149I, R249W, G346A, W403R and G435R, which were in the vicinity of the enzyme active site cavity. The strain A/chicken/India/99321/2009 (H9N2-99321) did not show these substitutions and was used for comparison. Virus elution was studied using turkey red blood cells (tRBCs). NA enzyme kinetics assays were carried out using the MUNANA substrate, which is an SA analogue. Homology modelling and molecular docking were performed to determine alterations in the surface characteristics and substrate binding. H9N2-1726265 showed enhanced elution from tRBCs. Enzyme kinetics revealed a lower KM of H9N2-1726265 (111.5 µM) as compared to H9N2-99321 (135.2 µM), indicating higher substrate binding affinity of H9N2-1726265, due to which the NA enzyme cleaved the SA more efficiently, leading to faster elution. Molecular docking revealed a greater number of binding interactions of H9N2-1726265 to SA as compared to H9N2-99321 corroborating the greater substrate binding affinity. Changes in the surface charge, hydrophobicity, and contour, were observed in H9N2-1726265 NA due to the five substitutions. Thus, the novel combination of five amino acids near the sialic acid binding site of NA, resulted in altered surface characteristics, higher substrate binding affinity, and virus elution.


Sujet(s)
Sous-type H9N2 du virus de la grippe A , Simulation de docking moléculaire , Mutation , Sialidase , Sialidase/génétique , Sialidase/composition chimique , Sialidase/métabolisme , Sous-type H9N2 du virus de la grippe A/génétique , Sous-type H9N2 du virus de la grippe A/enzymologie , Sous-type H9N2 du virus de la grippe A/composition chimique , Animaux , Substitution d'acide aminé , Protéines virales/composition chimique , Protéines virales/génétique , Protéines virales/métabolisme , Grippe chez les oiseaux/virologie , Dindons , Cinétique , Domaine catalytique
6.
Emerg Infect Dis ; 30(6): 1285-1288, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38703022

RÉSUMÉ

We isolated novel reassortant avian influenza A(H5N6) viruses containing genes from clade 2.3.4.4b H5N1 virus and low pathogenicity avian influenza viruses in carcasses of whooper swans and bean geese in South Korea during December 2023. Neuraminidase gene was from a clade 2.3.4.4b H5N6 virus infecting poultry and humans in China.


Sujet(s)
Animaux sauvages , Oiseaux , Virus de la grippe A , Grippe chez les oiseaux , Phylogenèse , Animaux , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/épidémiologie , République de Corée/épidémiologie , Animaux sauvages/virologie , Virus de la grippe A/génétique , Virus de la grippe A/classification , Oiseaux/virologie , Virus recombinants/génétique , Histoire du 21ème siècle , Humains , Sialidase/génétique
7.
Viruses ; 16(5)2024 04 27.
Article de Anglais | MEDLINE | ID: mdl-38793574

RÉSUMÉ

Influenza viruses are constantly evolving and are therefore monitored worldwide in the hope to reduce the burden of disease by annual updates to vaccine recommendations. We conducted genomic sequencing of 110 influenza A and 30 influenza B viruses from specimens collected between October 2023 and February 2024 in Arizona, USA. We identified mutations in the hemagglutinin (HA) antigenic sites as well as the neuraminidase (NA) gene in our samples. We also found no unique HA and NA mutations in vaccinated yet influenza-infected individuals. Real-time genomic sequencing surveillance is important to ensure influenza vaccine effectiveness.


Sujet(s)
Génome viral , Virus de la grippe A , Virus influenza B , Grippe humaine , Mutation , Sialidase , Arizona/épidémiologie , Humains , Grippe humaine/épidémiologie , Grippe humaine/virologie , Sialidase/génétique , Virus influenza B/génétique , Virus de la grippe A/génétique , Virus de la grippe A/classification , Glycoprotéine hémagglutinine du virus influenza/génétique , Génomique/méthodes , Phylogenèse , Adulte , Surveillance épidémiologique , Enfant , Adolescent , Adulte d'âge moyen , Mâle , Femelle , Enfant d'âge préscolaire , Sujet âgé , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/génétique , Jeune adulte , Séquençage du génome entier
8.
Virus Genes ; 60(3): 320-324, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38722491

RÉSUMÉ

H6 avian influenza virus is widely prevalent in wild birds and poultry and has caused human infection in 2013 in Taiwan, China. During our active influenza surveillance program in wild waterfowl at Poyang Lake, Jiangxi Province, an H6N2 AIV was isolated and named A/bean goose/JiangXi/452-4/2013(H6N2). The isolate was characterized as a typical low pathogenic avian influenza virus (LPAIV) due to the presence of the amino acid sequence PQIETR↓GLFGAI at the cleavage site of the hemagglutinin (HA) protein. The genetic evolution analysis revealed that the NA gene of the isolate originated from North America and exhibited the highest nucleotide identity (99.29%) with a virus recovered from wild bird samples in North America, specifically A/bufflehead/California/4935/2012(H11N2). Additionally, while the HA and PB1 genes belonged to the Eurasian lineage, they displayed frequent genetic interactions with the North American lineage. The remaining genes showed close genetic relationships with Eurasian viruses. The H6N2 isolate possessed a complex genome, indicating it is a multi-gene recombinant virus with genetic material from both Eurasian and North American lineages.


Sujet(s)
Animaux sauvages , Virus de la grippe A , Grippe chez les oiseaux , Phylogenèse , Virus recombinants , Animaux , Chine , Virus recombinants/génétique , Virus recombinants/isolement et purification , Virus recombinants/classification , Grippe chez les oiseaux/virologie , Animaux sauvages/virologie , Virus de la grippe A/génétique , Virus de la grippe A/isolement et purification , Virus de la grippe A/classification , Glycoprotéine hémagglutinine du virus influenza/génétique , Oiseaux/virologie , Évolution moléculaire , Génome viral/génétique , Sialidase/génétique , Protéines virales/génétique
9.
Antiviral Res ; 227: 105918, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38795911

RÉSUMÉ

The most widely used class of antivirals available for Influenza treatment are the neuraminidase inhibitors (NAI) Oseltamivir and Zanamivir. However, amino acid (AA) substitutions in the neuraminidase may cause reduced inhibition or high antiviral resistance. In Mexico, the current state of knowledge about NAI susceptibility is scarce, in this study we report the results of 14 years of Influenza surveillance by phenotypic and genotypic methods. A total of 255 isolates were assessed with the NAI assay, including Influenza A(H1N1)pdm09, A(H3N2) and Influenza B (IBV). Furthermore, 827 sequences contained in the GISAID platform were analyzed in search of relevant mutations.Overall, five isolates showed highly reduced inhibition or reduced inhibition to Oseltamivir, and two showed reduced inhibition to Zanamivir in the NAI assays. Additionally, five A(H1N1)pdm09 sequences from the GISAID possessed AA substitutions associated to reduced inhibition to Oseltamivir and none to Zanamivir. Oseltamivir resistant A(H1N1)pdm09 harbored the H275Y mutation. No genetic mutations were identified in Influenza A(H3N2) and IBV. Overall, these results show that in Mexico the rate of NAI resistance is low (0.6%), but it is essential to continue the Influenza surveillance in order to understand the drug susceptibility of circulating strains.


Sujet(s)
Antiviraux , Résistance virale aux médicaments , Virus influenza B , Grippe humaine , Sialidase , Oséltamivir , Zanamivir , Résistance virale aux médicaments/génétique , Antiviraux/pharmacologie , Mexique/épidémiologie , Humains , Virus influenza B/effets des médicaments et des substances chimiques , Virus influenza B/génétique , Grippe humaine/virologie , Grippe humaine/traitement médicamenteux , Grippe humaine/épidémiologie , Oséltamivir/pharmacologie , Zanamivir/pharmacologie , Sialidase/génétique , Sialidase/antagonistes et inhibiteurs , Sous-type H1N1 du virus de la grippe A/effets des médicaments et des substances chimiques , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/isolement et purification , Mutation , Sous-type H3N2 du virus de la grippe A/effets des médicaments et des substances chimiques , Sous-type H3N2 du virus de la grippe A/génétique , Adulte , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/génétique , Adolescent , Enfant , Substitution d'acide aminé , Jeune adulte , Adulte d'âge moyen , Femelle , Enfant d'âge préscolaire , Génotype , Mâle , Sujet âgé , Tests de sensibilité microbienne , Protéines virales/génétique
10.
FEMS Microbiol Rev ; 48(3)2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38734891

RÉSUMÉ

Avian influenza viruses evolve antigenically to evade host immunity. Two influenza A virus surface glycoproteins, the haemagglutinin and neuraminidase, are the major targets of host immunity and undergo antigenic drift in response to host pre-existing humoral and cellular immune responses. Specific sites have been identified as important epitopes in prominent subtypes such as H5 and H7, which are of animal and public health significance due to their panzootic and pandemic potential. The haemagglutinin is the immunodominant immunogen, it has been extensively studied, and the antigenic reactivity is closely monitored to ensure candidate vaccine viruses are protective. More recently, the neuraminidase has received increasing attention for its role as a protective immunogen. The neuraminidase is expressed at a lower abundance than the haemagglutinin on the virus surface but does elicit a robust antibody response. This review aims to compile the current information on haemagglutinin and neuraminidase epitopes and immune escape mutants of H5 and H7 highly pathogenic avian influenza viruses. Understanding the evolution of immune escape mutants and the location of epitopes is critical for identification of vaccine strains and development of broadly reactive vaccines that can be utilized in humans and animals.


Sujet(s)
Oiseaux , Épitopes , Glycoprotéine hémagglutinine du virus influenza , Grippe chez les oiseaux , Sialidase , Sialidase/immunologie , Sialidase/génétique , Animaux , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Épitopes/immunologie , Épitopes/génétique , Oiseaux/virologie , Grippe chez les oiseaux/immunologie , Grippe chez les oiseaux/virologie , Dérive et cassure antigéniques/immunologie , Humains , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/génétique , Grippe humaine/immunologie , Grippe humaine/virologie , Grippe humaine/prévention et contrôle , Protéines virales/immunologie , Protéines virales/génétique , Protéines virales/composition chimique , Virus de la grippe A/immunologie , Virus de la grippe A/génétique
11.
Int Immunopharmacol ; 132: 112051, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38599098

RÉSUMÉ

BACKGROUND: IgA Nephropathy (IgAN), the primary form of glomerulonephritis, presents significant clinical challenges due to its obscure pathogenesis and lack of targeted treatments. We conducted a proteome-wide Mendelian randomization (MR) study to identify therapeutic targets for IgAN. METHODS: Utilizing a plasma proteome dataset comprising 4907 blood plasma proteins as the exposure variable, and renal biopsy-confirmed IgAN cases as the outcome, this study employed MR to pinpoint proteins potentially pathogenic to IgAN. The robustness of our findings was affirmed through external dataset validation, reverse causation testing, and Bayesian colocalization analysis. Additionally, we conducted phenotypic scanning and analyzed downstream metabolites to investigate candidate proteins's biological function. RESULTS: In our study, a significant association was identified between an increase in neuraminidase 1 (NEU1) expression and the risk of IgAN. Specifically, a one standard deviation increase in NEU1 expression was associated with an odds ratio of 11.80 for the development of IgAN (95% confidence interval: 4.03-34.54). This association was substantiated across various statistical models and external validations. Colocalization analysis indicated a shared causal variant between NEU1 expression and IgAN. Furthermore, an increased influenza risk associated with NEU1 was observed, supporting the therapeutic potential of NEU1 inhibitors for IgAN. However, our study found no significant role for neuraminic acid-related metabolites in IgAN's development, suggesting an independent pathway for NEU1's influence. CONCLUSION: This study identifies NEU1 as a promising therapeutic target for IgAN, backed by robust genetic evidence. Future research should explore NEU1's therapeutic potential in diverse populations and clinical scenarios, further establishing its role in IgAN.


Sujet(s)
Glomérulonéphrite à dépôts d'IgA , Analyse de randomisation mendélienne , Sialidase , Glomérulonéphrite à dépôts d'IgA/génétique , Glomérulonéphrite à dépôts d'IgA/métabolisme , Humains , Sialidase/génétique , Sialidase/métabolisme , Grippe humaine/génétique , Génomique , Protéome , Thérapie moléculaire ciblée
12.
Virology ; 595: 110097, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38685171

RÉSUMÉ

Current influenza vaccine is not effective in providing cross-protection against variants. We evaluated the immunogenicity and efficacy of multi-subtype neuraminidase (NA) and M2 ectodomain virus-like particle (m-cNA-M2e VLP) and chimeric M2e-H3 stalk protein vaccines (M2e-H3 stalk) in ferrets. Our results showed that ferrets with recombinant m-cNA-M2e VLP or M2e-H3 stalk vaccination induced multi-vaccine antigen specific IgG antibodies (M2e, H3 stalk, NA), NA inhibition, antibody-secreting cells, and IFN-γ secreting cell responses. Ferrets immunized with either m-cNA-M2e VLP or M2e-H3 stalk vaccine were protected from H1N1 and H3N2 influenza viruses by lowering viral titers in nasal washes, trachea, and lungs after challenge. Vaccinated ferret antisera conferred broad humoral immunity in naïve mice. Our findings provide evidence that immunity to M2e and HA-stalk or M2e plus multi-subtype NA proteins induces cross-protection in ferrets.


Sujet(s)
Anticorps antiviraux , Protection croisée , Furets , Sous-type H1N1 du virus de la grippe A , Sous-type H3N2 du virus de la grippe A , Vaccins antigrippaux , Sialidase , Infections à Orthomyxoviridae , Vaccins à pseudo-particules virales , Animaux , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Protection croisée/immunologie , Anticorps antiviraux/immunologie , Sialidase/immunologie , Sialidase/génétique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Sous-type H3N2 du virus de la grippe A/immunologie , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/immunologie , Vaccins à pseudo-particules virales/immunologie , Vaccins à pseudo-particules virales/administration et posologie , Souris , Protéines de la matrice virale/immunologie , Protéines de la matrice virale/génétique , Femelle , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Protéines viroporines , Protéines virales
13.
Arch Virol ; 169(5): 111, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664271

RÉSUMÉ

India has reported highly pathogenic avian influenza (HPAI) H5N1 virus outbreaks since 2006, with the first human case reported in 2021. These included viruses belonging to the clades 2.2, 2.2.2, 2.2.2.1, 2.3.2.1a, and 2.3.2.1c. There are currently no data on the gene pool of HPAI H5N1 viruses in India. Molecular clock and phylogeography analysis of the HA and NA genes; and phylogenetic analysis of the internal genes of H5N1 viruses from India were carried out. Sequences reported from 2006 to 2015; and sequences from 2021 that were available in online databases were used in the analysis. Five separate introductions of H5N1 viruses into India were observed, via Indonesia or Korea (2002), Bangladesh (2009), Bhutan (2010), and China (2013, 2018) (clades 2.2, 2.2.2, 2.2.2.1, 2.3.2.1a, 2.3.2.1c, and 2.3.4.4b). Phylogenetic analysis revealed eight reassortant genotypes. The H5N1 virus isolated from the human case showed a unique reassortant genotype. Amino acid markers associated with adaptation to mammals were also present. This is the first report of the spatio-temporal origins and gene pool analysis of H5N1 viruses from India, highlighting the need for increased molecular surveillance.


Sujet(s)
Sous-type H5N1 du virus de la grippe A , Grippe chez les oiseaux , Grippe humaine , Phylogenèse , Phylogéographie , Inde/épidémiologie , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/classification , Sous-type H5N1 du virus de la grippe A/isolement et purification , Animaux , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/épidémiologie , Humains , Grippe humaine/virologie , Grippe humaine/épidémiologie , Génotype , Virus recombinants/génétique , Virus recombinants/classification , Virus recombinants/isolement et purification , Sialidase/génétique , Glycoprotéine hémagglutinine du virus influenza/génétique , Oiseaux/virologie , Épidémies de maladies
14.
J Virol ; 98(5): e0195923, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38634598

RÉSUMÉ

The role of Culex mosquitoes in the transmission of Japanese encephalitis virus (JEV) is crucial, yet the mechanisms of JEV infection in these vectors remain unclear. Previous research has indicated that various host factors participate in JEV infection. Herein, we present evidence that mosquito sialic acids enhance JEV infection both in vivo and in vitro. By treating mosquitoes and C6/36 cells with neuraminidase or lectin, the function of sialic acids is effectively blocked, resulting in significant inhibition of JEV infection. Furthermore, knockdown of the sialic acid biosynthesis genes in Culex mosquitoes also leads to a reduction in JEV infection. Moreover, our research revealed that sialic acids play a role in the attachment of JEV to mosquito cells, but not in its internalization. To further explore the mechanisms underlying the promotion of JEV attachment by sialic acids, we conducted immunoprecipitation experiments to confirm the direct binding of sialic acids to the last α-helix in JEV envelope protein domain III. Overall, our study contributes to a molecular comprehension of the interaction between mosquitoes and JEV and offers potential strategies for preventing the dissemination of flavivirus in natural environments.IMPORTANCEIn this study, we aimed to investigate the impact of glycoconjugate sialic acids on mosquito infection with Japanese encephalitis virus (JEV). Our findings demonstrate that sialic acids play a crucial role in enhancing JEV infection by facilitating the attachment of the virus to the cell membrane. Furthermore, our investigation revealed that sialic acids directly bind to the final α-helix in the JEV envelope protein domain III, thereby accelerating virus adsorption. Collectively, our results highlight the significance of mosquito sialic acids in JEV infection within vectors, contributing to a better understanding of the interaction between mosquitoes and JEV.


Sujet(s)
Culex , Virus de l'encéphalite japonaise (espèce) , Encéphalite japonaise , Acides sialiques , Attachement viral , Animaux , Souris , Lignée cellulaire , Culex/virologie , Culex/métabolisme , Virus de l'encéphalite japonaise (espèce)/physiologie , Virus de l'encéphalite japonaise (espèce)/métabolisme , Encéphalite japonaise/virologie , Encéphalite japonaise/métabolisme , Vecteurs moustiques/virologie , Sialidase/métabolisme , Sialidase/génétique , Acides sialiques/métabolisme , Protéines de l'enveloppe virale/métabolisme , Protéines de l'enveloppe virale/génétique , Pénétration virale
15.
Antiviral Res ; 224: 105853, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38430970

RÉSUMÉ

While clinical trials have illuminated both the virological and clinical efficacy of baloxavir for influenza and post-treatment viral resistance, these aspects warrant further study in real-world settings. In response, we executed a prospective, observational study of the Japanese 2022-2023 influenza season. A cohort of 73 A(H3N2)-diagnosed outpatients-36 treated with baloxavir, 20 with oseltamivir, and 17 with other neuraminidase inhibitors (NAIs)-were analyzed. Viral samples were collected before and after administering an antiviral on days 1, 5, and 10, respectively. Cultured viruses were amplified using RT-PCR and sequenced to detect mutations. Fever and other symptoms were tracked via self-reporting diaries. In the baloxavir cohort, viral detection was 11.1% (4/36) and 0% (0/36) on day 5 and day 10, respectively. Two isolates from day 5 (5.6%, 2/36) manifested I38T/M-substitutions in the polymerase acidic protein (PA). For oseltamivir and other NAIs, viral detection rates were 60.0% (12/20) and 52.9% (9/17) on day 5, and 16.7% (3/18) and 6.3% (1/16) on day 10, respectively. No oseltamivir-resistant neuraminidase mutations were identified after treatment. Median fever durations for the baloxavir, oseltamivir, and other NAI cohorts were 27.0, 38.0, and 36.0 h, respectively, with no significant difference. Two patients harboring PA I38T/M-substitutions did not exhibit prolonged fever or other symptoms. These findings affirm baloxavir's virological and clinical effectiveness against A(H3N2) in the 2022-2023 season and suggest limited clinical influence of post-treatment resistance emergence.


Sujet(s)
Dibenzothiépines , Grippe humaine , Morpholines , Triazines , Humains , Grippe humaine/traitement médicamenteux , Oséltamivir/usage thérapeutique , Oséltamivir/pharmacologie , Sialidase/génétique , Sialidase/usage thérapeutique , Sous-type H3N2 du virus de la grippe A/génétique , Patients en consultation externe , Saisons , Études prospectives , Antiviraux/usage thérapeutique , Antiviraux/pharmacologie , Pyridones/usage thérapeutique , Antienzymes/pharmacologie , Guanidines/pharmacologie , Fièvre/traitement médicamenteux
16.
Hum Vaccin Immunother ; 20(1): 2304393, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38497413

RÉSUMÉ

Current influenza vaccines could be augmented by including recombinant neuraminidase (rNA) protein antigen to broaden protective immunity and improve efficacy. Toward this goal, we investigated formulation conditions to optimize rNA physicochemical stability. When rNA in sodium phosphate saline buffer (NaPBS) was frozen and thawed (F/T), the tetrameric structure transitioned from a "closed" to an "open" conformation, negatively impacting functional activity. Hydrogen deuterium exchange experiments identified differences in anchorage binding sites at the base of the open tetramer, offering a structural mechanistic explanation for the change in conformation and decreased functional activity. Change to the open configuration was triggered by the combined stresses of acidic pH and F/T. The desired closed conformation was preserved in a potassium phosphate buffer (KP), minimizing pH drop upon freezing and including 10% sucrose to control F/T stress. Stability was further evaluated in thermal stress studies where changes in conformation were readily detected by ELISA and size exclusion chromatography (SEC). Both tests were suitable indicators of stability and antigenicity and considered potential critical quality attributes (pCQAs). To understand longer-term stability, the pCQA profiles from thermally stressed rNA at 6 months were modeled to predict stability of at least 24-months at 5°C storage. In summary, a desired rNA closed tetramer was maintained by formulation selection and monitoring of pCQAs to produce a stable rNA vaccine candidate. The study highlights the importance of understanding and controlling vaccine protein structural and functional integrity.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Humains , Grippe humaine/prévention et contrôle , Sialidase/génétique , Vaccins synthétiques/génétique , ARN
17.
Microb Pathog ; 190: 106628, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38508422

RÉSUMÉ

Rotavirus infections in suckling and weaning piglets cause severe dehydration and death, resulting in significant economic losses in the pig breeding industry. With the continuous emergence of porcine rotavirus (PoRV) variants and poor vaccine cross-protection among various genotypes, there is an urgent need to develop alternative strategies such as seeking effective antiviral products from nature, microbial metabolites and virus-host protein interaction. Sialidases play a crucial role in various physiopathological processes and offer a promising target for developing antivirus drugs. However, the effect of bacterial-derived sialidases on the infection of PoRVs remains largely unknown. Herein, we investigated the impact of bacterial-derived sialidases (sialidase Cp and Vc) on PoRV strain OSU(Group A) infection, using differentiated epithelial monkey kidney cells (MA104) as a model. Our results indicated that the pretreatment of MA104 with exogenous sialidases effectively suppressed PoRV OSU in a concentration-dependent manner. Notably, even at a concentration of 0.01 µU/mL, sialidases significantly inhibited the virus (MOI = 0.01). Meanwhile, we found that sialidase Vc pretreatment sharply reduced the binding rate of PoRV OSU. Last, we demonstrated that PoRV OSU might recognize α-2,3-linked sialic acid as the primary attachment factor in MA104. Our findings provide new insights into the underlying mechanism of PoRV OSU infections, shedding lights on the development of alternative antivirus approaches based on bacteria-virus interaction.


Sujet(s)
Sialidase , Infections à rotavirus , Rotavirus , Réplication virale , Animaux , Sialidase/métabolisme , Sialidase/génétique , Rotavirus/effets des médicaments et des substances chimiques , Rotavirus/physiologie , Suidae , Réplication virale/effets des médicaments et des substances chimiques , Lignée cellulaire , Cellules épithéliales/virologie , Cellules épithéliales/microbiologie , Attachement viral/effets des médicaments et des substances chimiques , Acide N-acétyl-neuraminique/métabolisme , Acide N-acétyl-neuraminique/pharmacologie , Antiviraux/pharmacologie , Haplorhini , Maladies des porcs/virologie , Maladies des porcs/microbiologie
18.
Respir Res ; 25(1): 134, 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38500102

RÉSUMÉ

Neu1 is a sialidase enzyme that plays a crucial role in the regulation of glycosylation in a variety of cellular processes, including cellular signaling and inflammation. In recent years, numerous evidence has suggested that human NEU1 is also involved in the pathogenesis of various respiratory diseases, including lung infection, chronic obstructive pulmonary disease (COPD), asthma, and pulmonary fibrosis. This review paper aims to provide an overview of the current research on human NEU1 and respiratory diseases.


Sujet(s)
Asthme , Troubles respiratoires , Humains , Sialidase/génétique , Inflammation
19.
Crit Rev Eukaryot Gene Expr ; 34(4): 45-54, 2024.
Article de Anglais | MEDLINE | ID: mdl-38505872

RÉSUMÉ

HDAC1 functions as an oncogene in multi-type cancers. This study aimed to investigate the roles of histone deacetylase 1 (HDAC1) in cervical cancer (CC). mRNA expression was determined using reverse transcription quantitative polymerase chain reaction. The protein-protein complexes was analyzed using co-immunoprecipitation assay. The binding sites between NRF2 and NEU1 were confirmed by chromatin immunoprecipitation assay. Cell viability was detected by CCK-8. Cell proliferation was measured using CCK-8 and colony formation assays. Cell migrative and invasive ability were determined using transwell assay. We found that HDAC1 was upregulated in CC patients and cells. Trichostatin A (TSA) treatment decreased the number of colonies and migrated and invaded cells. Moreover, HDAC1 interacted with NRF2 to downregulate NEU1 expression. NEU1 knockdown attenuated the effects of TSA and enhanced the aggressiveness of CC cells. In conclusion, HDAC1 functions as an oncogene in CC. Targeting HDAC1 may be an alternative strategy for CC.


Sujet(s)
Tumeurs du col de l'utérus , Femelle , Humains , Régulation négative , Tumeurs du col de l'utérus/génétique , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme , Sincalide/génétique , Sincalide/métabolisme , Sialidase/génétique , Sialidase/métabolisme
20.
Viruses ; 16(3)2024 03 01.
Article de Anglais | MEDLINE | ID: mdl-38543754

RÉSUMÉ

The H274Y substitution (N2 numbering) in neuraminidase (NA) N1 confers oseltamivir resistance to A(H1N1) influenza viruses. This resistance has been associated with reduced N1 expression using transfected cells, but the effect of this substitution on the enzymatic properties and on the expression of other group-1-NA subtypes is unknown. The aim of the present study was to evaluate the antiviral resistance, enzymatic properties, and expression of wild-type (WT) and H274Y-substituted NA for each group-1-NA. To this end, viruses with WT or H274Y-substituted NA (N1pdm09 or avian N4, N5 or N8) were generated by reverse genetics, and for each reverse-genetic virus, antiviral susceptibility, NA affinity (Km), and maximum velocity (Vm) were measured. The enzymatic properties were coupled with NA quantification on concentrated reverse genetic viruses using mass spectrometry. The H274Y-NA substitution resulted in highly reduced inhibition by oseltamivir and normal inhibition by zanamivir and laninamivir. This resistance was associated with a reduced affinity for MUNANA substrate and a conserved Vm in all viruses. NA quantification was not significantly different between viruses carrying WT or H274Y-N1, N4 or N8, but was lower for viruses carrying H274Y-N5 compared to those carrying a WT-N5. In conclusion, the H274Y-NA substitution of different group-1-NAs systematically reduced their affinity for MUNANA substrate without a significant impact on NA Vm. The impact of the H274Y-NA substitution on viral NA expression was different according to the studied NA.


Sujet(s)
Sous-type H1N1 du virus de la grippe A , Virus de la grippe A , Grippe humaine , Humains , Oséltamivir/pharmacologie , Antiviraux/pharmacologie , Virus de la grippe A/génétique , Sialidase/génétique , Sialidase/métabolisme , Sous-type H1N1 du virus de la grippe A/génétique , Génétique inverse , Résistance virale aux médicaments/génétique , Substitution d'acide aminé , Antienzymes/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...