Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24.168
Filtrer
1.
Front Immunol ; 15: 1399960, 2024.
Article de Anglais | MEDLINE | ID: mdl-38873606

RÉSUMÉ

The VH6-1 class of antibodies includes some of the broadest and most potent antibodies that neutralize influenza A virus. Here, we elicit and isolate anti-idiotype antibodies against germline versions of VH6-1 antibodies, use these to sort human leukocytes, and isolate a new VH6-1-class member, antibody L5A7, which potently neutralized diverse group 1 and group 2 influenza A strains. While its heavy chain derived from the canonical IGHV6-1 heavy chain gene used by the class, L5A7 utilized a light chain gene, IGKV1-9, which had not been previously observed in other VH6-1-class antibodies. The cryo-EM structure of L5A7 in complex with Indonesia 2005 hemagglutinin revealed a nearly identical binding mode to other VH6-1-class members. The structure of L5A7 bound to the isolating anti-idiotype antibody, 28H6E11, revealed a shared surface for binding anti-idiotype and hemagglutinin that included two critical L5A7 regions: an FG motif in the third heavy chain-complementary determining region (CDR H3) and the CDR L1 loop. Surprisingly, the chemistries of L5A7 interactions with hemagglutinin and with anti-idiotype were substantially different. Overall, we demonstrate anti-idiotype-based isolation of a broad and potent influenza A virus-neutralizing antibody, revealing that anti-idiotypic selection of antibodies can involve features other than chemical mimicry of the target antigen.


Sujet(s)
Anticorps anti-idiotypiques , Anticorps neutralisants , Anticorps antiviraux , Glycoprotéine hémagglutinine du virus influenza , Virus de la grippe A , Humains , Virus de la grippe A/immunologie , Anticorps antiviraux/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/isolement et purification , Anticorps anti-idiotypiques/immunologie , Anticorps anti-idiotypiques/isolement et purification , Glycoprotéine hémagglutinine du virus influenza/immunologie , Grippe humaine/immunologie , Grippe humaine/virologie , Animaux , Chaines lourdes des immunoglobulines/immunologie , Chaines lourdes des immunoglobulines/composition chimique
2.
Sci Rep ; 14(1): 13524, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38866913

RÉSUMÉ

Myxovirus resistance (Mx) proteins are products of interferon stimulated genes (ISGs) and Mx proteins of different species have been reported to mediate antiviral activity against a number of viruses, including influenza A viruses (IAV). Ferrets are widely considered to represent the 'gold standard' small animal model for studying pathogenesis and immunity to human IAV infections, however little is known regarding the antiviral activity of ferret Mx proteins. Herein, we report induction of ferret (f)Mx1/2 in a ferret lung cell line and in airway tissues from IAV-infected ferrets, noting that fMx1 was induced to higher levels that fMx2 both in vitro and in vivo. Overexpression confirmed cytoplasmic expression of fMx1 as well as its ability to inhibit infection and replication of IAV, noting that this antiviral effect of fMx1was modest when compared to cells overexpressing either human MxA or mouse Mx1. Together, these studies provide the first insights regarding the role of fMx1 in cell innate antiviral immunity to influenza viruses. Understanding similarities and differences in the antiviral activities of human and ferret ISGs provides critical context for evaluating results when studying human IAV infections in the ferret model.


Sujet(s)
Furets , Virus de la grippe A , Protéines de résistance aux myxovirus , Infections à Orthomyxoviridae , Animaux , Protéines de résistance aux myxovirus/génétique , Protéines de résistance aux myxovirus/métabolisme , Virus de la grippe A/immunologie , Humains , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Réplication virale/effets des médicaments et des substances chimiques , Antiviraux/pharmacologie , Lignée cellulaire , Souris , Immunité innée , Poumon/virologie , Poumon/immunologie
3.
Respir Res ; 25(1): 234, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840154

RÉSUMÉ

BACKGROUND: The concurrent circulation of SARS-CoV-2 with other respiratory viruses is unstoppable and represents a new diagnostic reality for clinicians and clinical microbiology laboratories. Multiplexed molecular testing on automated platforms that focus on the simultaneous detection of multiple respiratory viruses in a single tube is a useful approach for current and future diagnosis of respiratory infections in the clinical setting. METHODS: Two time periods were included in the study: from February to April 2022, an early 2022 period, during the gradual lifting of COVID-19 prevention measures in the country, and from October 2022 to April 2023, the 2022/23 respiratory infections season. We analysed a total of 1,918 samples in the first period and 18,131 respiratory samples in the second period using a multiplex molecular assay for the simultaneous detection of Influenza A (Flu-A), Influenza B (Flu-B), Human Respiratory Syncytial Virus (HRSV) and SARS-CoV-2. RESULTS: The results from early 2022 showed a strong dominance of SARS-CoV-2 infections with 1,267/1,918 (66.1%) cases. Flu-A was detected in 30/1,918 (1.6%) samples, HRSV in 14/1,918 (0.7%) samples, and Flu-B in 2/1,918 (0.1%) samples. Flu-A/SARS-CoV-2 co-detections were observed in 11/1,267 (0.9%) samples, and HRSV/SARS-CoV-2 co-detection in 5/1,267 (0.4%) samples. During the 2022/23 winter respiratory season, SARS-CoV-2 was detected in 1,738/18,131 (9.6%), Flu-A in 628/18,131 (3.5%), Flu-B in 106/18,131 (0.6%), and HRSV in 505/18,131 (2.8%) samples. Interestingly, co-detections were present to a similar extent as in early 2022. CONCLUSION: The results show that the multiplex molecular approach is a valuable tool for the simultaneous laboratory diagnosis of SARS-CoV-2, Flu-A/B, and HRSV in hospitalized and outpatients. Infections with Flu-A/B, and HRSV occurred shortly after the COVID-19 control measures were lifted, so a strong reoccurrence of various respiratory infections and co-detections in the post COVID-19 period was to be expected.


Sujet(s)
COVID-19 , Virus de la grippe A , Virus influenza B , Grippe humaine , Infections à virus respiratoire syncytial , Virus respiratoire syncytial humain , SARS-CoV-2 , Humains , COVID-19/épidémiologie , COVID-19/diagnostic , Virus influenza B/isolement et purification , Virus influenza B/génétique , Grippe humaine/épidémiologie , Grippe humaine/diagnostic , Grippe humaine/virologie , SARS-CoV-2/génétique , SARS-CoV-2/isolement et purification , Infections à virus respiratoire syncytial/épidémiologie , Infections à virus respiratoire syncytial/diagnostic , Virus respiratoire syncytial humain/isolement et purification , Virus respiratoire syncytial humain/génétique , Virus de la grippe A/isolement et purification , Virus de la grippe A/génétique , Mâle , Femelle , Co-infection/épidémiologie , Co-infection/diagnostic , Adulte d'âge moyen , Adulte , Techniques de diagnostic moléculaire/méthodes , Saisons , Sujet âgé
4.
PLoS One ; 19(6): e0303756, 2024.
Article de Anglais | MEDLINE | ID: mdl-38829903

RÉSUMÉ

The rapid spread of highly pathogenic avian influenza (HPAI) A (H5N1) viruses in Southeast Asia in 2004 prompted the New Zealand Ministry for Primary Industries to expand its avian influenza surveillance in wild birds. A total of 18,693 birds were sampled between 2004 and 2020, including migratory shorebirds (in 2004-2009), other coastal species (in 2009-2010), and resident waterfowl (in 2004-2020). No avian influenza viruses (AIVs) were isolated from cloacal or oropharyngeal samples from migratory shorebirds or resident coastal species. Two samples from red knots (Calidris canutus) tested positive by influenza A RT-qPCR, but virus could not be isolated and no further characterization could be undertaken. In contrast, 6179 samples from 15,740 mallards (Anas platyrhynchos) tested positive by influenza A RT-qPCR. Of these, 344 were positive for H5 and 51 for H7. All H5 and H7 viruses detected were of low pathogenicity confirmed by a lack of multiple basic amino acids at the hemagglutinin (HA) cleavage site. Twenty H5 viruses (six different neuraminidase [NA] subtypes) and 10 H7 viruses (two different NA subtypes) were propagated and characterized genetically. From H5- or H7-negative samples that tested positive by influenza A RT-qPCR, 326 AIVs were isolated, representing 41 HA/NA combinations. The most frequently isolated subtypes were H4N6, H3N8, H3N2, and H10N3. Multivariable logistic regression analysis of the relations between the location and year of sampling, and presence of AIV in individual waterfowl showed that the AIV risk at a given location varied from year to year. The H5 and H7 isolates both formed monophyletic HA groups. The H5 viruses were most closely related to North American lineages, whereas the H7 viruses formed a sister cluster relationship with wild bird viruses of the Eurasian and Australian lineages. Bayesian analysis indicates that the H5 and H7 viruses have circulated in resident mallards in New Zealand for some time. Correspondingly, we found limited evidence of influenza viruses in the major migratory bird populations visiting New Zealand. Findings suggest a low probability of introduction of HPAI viruses via long-distance bird migration and a unique epidemiology of AIV in New Zealand.


Sujet(s)
Animaux sauvages , Oiseaux , Grippe chez les oiseaux , Phylogenèse , Animaux , Nouvelle-Zélande/épidémiologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/épidémiologie , Animaux sauvages/virologie , Oiseaux/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 , Génome viral , Canards/virologie
5.
J Transl Med ; 22(1): 570, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879538

RÉSUMÉ

BACKGROUND: Gut microbiota (GM) have been implicated as important regulators of gastrointestinal symptom which is commonly occurred along with respiratory influenza A virus (IAV) infection, suggesting the involvement of the gut-to-lung axis in a host's response to IAV. IAV primarily destroys airway epithelium tight junctions (TJs) and consequently causes acute respiratory disease syndrome. It is known that GM and their metabolism produce an anti-influenza effect, but their role in IAV-induced airway epithelial integrity remains unknown. METHODS: A mouse model of IAV infection was established. GM were analyzed using 16S rRNA gene sequencing, and short-chain fatty acids (SCFAs) levels were measured. GM depletion and fecal microbiota transplantation (FMT) were conducted to validate the role of GM in IAV infection. A pair-feeding experiment was conducted to reveal whether IAV-induced GM dysbiosis is attributed to impaired food intake. Furthermore, human bronchial epithelial (HBE) cells were cocultured with IAV in the presence or absence of acetate. TJs function was analyzed by paracellular permeability and transepithelial electronic resistance (TEER). The mechanism of how acetate affects TJs integrity was evaluated in HBE cells transfected with G protein-coupled receptor 43 (GPR43) short hairpin RNA (shRNA). RESULTS: IAV-infected mice exhibited lower relative abundance of acetate-producing bacteria (Bacteroides, Bifidobacterium, and Akkermansia) and decreased acetate levels in gut and serum. These changes were partly caused by a decrease in food consumption (due to anorexia). GM depletion exacerbated and FMT restored IAV-induced lung inflammatory injury. IAV infection suppressed expressions of TJs (occludin, ZO-1) leading to disrupted airway epithelial barrier function as evidenced by decreased TEER and increased permeability. Acetate pretreatment activated GPR43, partially restored IAV-induced airway epithelial barrier function, and reduced inflammatory cytokines levels (TNF-α, IL-6, and IL-1ß). Such protective effects of acetate were absent in HBE cells transfected with GPR43 shRNA. Acetate and GPR43 improved TJs in an AMP-activated protein kinase (AMPK)-dependent manner. CONCLUSION: Collectively, our results demonstrated that GM protected airway TJs by modulating GPR43-AMPK signaling in IAV-induced lung injury. Therefore, improving GM dysbiosis may be a potential therapeutic target for patients with IAV infection.


Sujet(s)
Acétates , Microbiome gastro-intestinal , Lésion pulmonaire , Infections à Orthomyxoviridae , Jonctions serrées , Animaux , Jonctions serrées/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Acétates/métabolisme , Humains , Infections à Orthomyxoviridae/complications , Souris de lignée C57BL , Virus de la grippe A , Transplantation de microbiote fécal , Récepteurs couplés aux protéines G/métabolisme , Souris , Cellules épithéliales/métabolisme , Dysbiose , Acides gras volatils/métabolisme
6.
Emerg Microbes Infect ; 13(1): 2364732, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38832658

RÉSUMÉ

Recently, an outbreak of highly pathogenic avian influenza A (H5N1), which carries the clade 2.3.4.4b hemagglutinin (HA) gene and has been prevalent among North American bird populations since the winter of 2021, was reported in dairy cows in the United States. As of 24 May 2024, the virus has affected 63 dairy herds across nine states and has resulted in two human infections. The virus causes unusual symptoms in dairy cows, including an unexpected drop in milk production, and thick colostrum-like milk. Notably, The US Food and Drug Administration reported that around 20% of tested retail milk samples contained H5N1 viruses, with a higher percentage of positive results from regions with infected cattle herds. Data are scant regarding how effectively pasteurization inactivates the H5N1 virus in milk. Therefore, in this study, we evaluated the thermal stability of the H5 clade 2.3.4.4b viruses, along with one human H3N2 virus and other influenza subtype viruses, including H1, H3, H7, H9, and H10 subtype viruses. We also assessed the effectiveness of pasteurization in inactivating these viruses. We found that the avian H3 virus exhibits the highest thermal stability, whereas the H5N1 viruses that belong to clade 2.3.4.4b display moderate thermal stability. Importantly, our data provide direct evidence that the standard pasteurization methods used by dairy companies are effective in inactivating all tested subtypes of influenza viruses in raw milk. Our findings indicate that thermally pasteurized milk products do not pose a safety risk to consumers.


Sujet(s)
Lait , Pasteurisation , Animaux , Pasteurisation/méthodes , Lait/virologie , Bovins , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/isolement et purification , Humains , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/transmission , Grippe chez les oiseaux/prévention et contrôle , Grippe chez les oiseaux/épidémiologie , Inactivation virale , États-Unis , Grippe humaine/virologie , Grippe humaine/transmission , Grippe humaine/prévention et contrôle , Virus de la grippe A/génétique , Virus de la grippe A/isolement et purification , Femelle
7.
ACS Nano ; 18(24): 15545-15556, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38838261

RÉSUMÉ

Deterministic formation of membrane scission necks by protein machinery with multiplexed functions is critical in biology. A microbial example is M2 viroporin, a proton pump from the influenza A virus that is multiplexed with membrane remodeling activity to induce budding and scission in the host membrane during viral maturation. In comparison, the dynamin family constitutes a class of eukaryotic proteins implicated in mitochondrial fission, as well as various budding and endocytosis pathways. In the case of Dnm1, the mitochondrial fission protein in yeast, the membrane remodeling activity is multiplexed with mechanoenzyme activity to create fission necks. It is not clear why these functions are combined in these scission processes, which occur in drastically different compositions and solution conditions. In general, direct experimental access to changing neck sizes induced by individual proteins or peptide fragments is challenging due to the nanoscale dimensions and influence of thermal fluctuations. Here, we use a mechanical model to estimate the size of scission necks by leveraging small-angle X-ray scattering structural data of protein-lipid systems under different conditions. The influence of interfacial tension, lipid composition, and membrane budding morphology on the size of the induced scission necks is systematically investigated using our data and molecular dynamic simulations. We find that the M2 budding protein from the influenza A virus has robust pH-dependent membrane activity that induces nanoscopic necks within the range of spontaneous hemifission for a broad range of lipid compositions. In contrast, the sizes of scission necks generated by mitochondrial fission proteins strongly depend on lipid composition, which suggests a role for mechanical constriction.


Sujet(s)
Membrane cellulaire , Membrane cellulaire/métabolisme , Membrane cellulaire/composition chimique , Protéines de la matrice virale/métabolisme , Protéines de la matrice virale/composition chimique , Dynamines/métabolisme , Dynamines/composition chimique , Virus de la grippe A/métabolisme , Diffusion aux petits angles , Protéines viroporines
8.
Biotechnol J ; 19(6): e2400074, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38896409

RÉSUMÉ

The ELISA is the most worldwide method for immunoassay. However, the ELISA is losing ground due to low reproducibility of manual experimental processes in both R&D and IVD areas. An automated platform is a good solution, but there are still limitations owning to extremely high cost and requiring large space to set up especially for a small size laboratory. Here, we present a novel all-in-one platform called "VEUS" settable on the laboratory table that offers comprehensive automation of the entire multiplex immunoassay process by exploiting antibody conjugated magnetic particles, quality control and then immunoanalytical reaction, thereby enhancing detection sensitivity and high reproducibility. As a proof of concept, the system exhibits a sensitive LOD of 0.6 and 3.1 pg mL-1 within 1 h run, comparable precision that of molecular diagnostic systems based on PCR method, enabling rapid multiplex diagnosis of Influenza A, Influenza B, and COVID-19 viruses with similar symptoms. Through automation by the all-in-one system, it can be used by novice users, something innovative for immunoassays, relying heavily on user experience. Furthermore, it can contribute to streamline entire immunoassay processes of diverse biomarkers with high reproducibility and convenience in laboratories.


Sujet(s)
SARS-CoV-2 , Humains , Dosage immunologique/méthodes , SARS-CoV-2/immunologie , SARS-CoV-2/isolement et purification , Anticorps immobilisés/immunologie , Anticorps immobilisés/composition chimique , Reproductibilité des résultats , COVID-19/diagnostic , COVID-19/virologie , Test ELISA/méthodes , Virus de la grippe A/immunologie , Virus influenza B/immunologie , Laboratoire automatique/méthodes , Limite de détection
9.
Hum Vaccin Immunother ; 20(1): 2363076, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38847280

RÉSUMÉ

To optimize seasonal influenza control and prevention programs in regions with potentially complicated seasonal patterns. Descriptive epidemiology was used to analyze the etiology of influenza, and chi-square tests were used to compare the epidemic patterns among different influenza virus types and subtypes/lineages. From January 2010 to December 2019, a total of 63,626 ILI cases were reported in Chongqing and 14,136 (22.22%) were laboratory-confirmed influenza cases. The proportions of specimens positive for influenza A and influenza B were 13.32% (8,478/63,626) and 8.86% (5,639/63,626), respectively. The proportion of positive specimens for influenza A reached the highest in winter (23.33%), while the proportion of positive specimens for influenza B reached the highest in spring (11.88%). Children aged 5-14 years old had the highest proportion of positive specimens for influenza. The influenza virus types/subtypes positive was significantly different by seasons and age groups (P<.001), but not by gender (p = .436). The vaccine strains were matched to the circulating influenza virus strains in all other years except for 2018 (vaccine strain was B/Colorado/06/2017; circulating strain was B/Yamagata). The study showed significant variations in epidemic patterns, including seasonal epidemic period and age distributions, among different influenza types, subtypes/lineages in Chongqing. Influenza vaccines matched to the circulating influenza virus strain in nine of the ten years. To prevent and mitigate the influenza outbreaks in this area, high risk population, especially children aged 5-14 years, are encouraged to get vaccinated against influenza before the epidemic seasons.


Sujet(s)
Virus influenza B , Grippe humaine , Saisons , Humains , Enfant , Grippe humaine/épidémiologie , Grippe humaine/prévention et contrôle , Grippe humaine/virologie , Chine/épidémiologie , Adolescent , Enfant d'âge préscolaire , Mâle , Femelle , Virus influenza B/classification , Virus influenza B/isolement et purification , Nourrisson , Jeune adulte , Adulte d'âge moyen , Adulte , Sujet âgé , Virus de la grippe A/classification , Virus de la grippe A/isolement et purification , Vaccins antigrippaux/administration et posologie , Épidémies , Nouveau-né
10.
Rev Med Virol ; 34(4): e2559, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38886173

RÉSUMÉ

The World Organization for Animal Health defines Avian Influenza Virus as a highly infectious disease caused by diverse subtypes that continue to evolve rapidly, impacting poultry species, pet birds, wild birds, non-human mammals, and occasionally humans. The effects of Avian influenza viruses have been recognised as a precursor for serious health concerns among affected birds, poultry, and human populations in the Middle East. Furthermore, low and high pathogenic avian influenza viruses lead to respiratory illness with varying severity, depending on the virus subtype (e.g., H5, H7, H9, etc.). Possible future outbreaks and endemics of newly emerging subtypes are expected to occur, as many studies have reported the emergence of novel mutations and viral subtypes. However, proper surveillance programs and biosecurity applications should be developed, and countries with incapacitated defences against such outbreaks should be encouraged to undergo complete reinstation and reinforcement in their health and research sectors. Public education regarding biosafety and virus prevention is necessary to ensure minimal spread of avian influenza endemic.


Sujet(s)
Oiseaux , Virus de la grippe A , Grippe chez les oiseaux , Grippe humaine , Animaux , Grippe chez les oiseaux/épidémiologie , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/prévention et contrôle , Grippe chez les oiseaux/transmission , Humains , Grippe humaine/prévention et contrôle , Grippe humaine/épidémiologie , Grippe humaine/virologie , Région méditerranéenne/épidémiologie , Oiseaux/virologie , Virus de la grippe A/génétique , Virus de la grippe A/physiologie , Virus de la grippe A/pathogénicité , Épidémies de maladies/prévention et contrôle , Épidémies de maladies/médecine vétérinaire
11.
Immun Inflamm Dis ; 12(6): e1309, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38860765

RÉSUMÉ

BACKGROUND: Astragaloside IV (AS-IV) is the most active monomer in the traditional Chinese herbal medicine Radix Astragali, which has a wide range of antiviral, anti-inflammatory, and antifibrosis pharmacological effects, and shows protective effects in acute lung injury. METHODS: This study utilized the immunofluorescence, flow cytometry, enzyme-linked immunosorbent assay, quantitative reverse transcription-polymerase chain reaction, western blot, and hematoxylin and eosin staining methods to investigate the mechanism of AS-IV in reducing viral pneumonia caused by influenza A virus in A549 cells and BALB/c mice. RESULTS: The results showed that AS-IV suppressed reactive oxygen species production in influenza virus-infected A549 cells in a dose-dependent manner, and subsequently inhibited the activation of nucleotide-binding oligomerization domain-like receptor thermal protein domain associated protein 3 inflammasome and Caspase-1, decreased interleukin (IL) -1ß and IL-18 secretion. In BALB/c mice infected with Poly (I:C), oral administration of AS-IV can significantly reduce Poly (I:C)-induced acute pneumonia and lung pathological injury. CONCLUSIONS: AS-IV alleviates the inflammatory response induced by influenza virus in vitro and lung flammation and structural damage caused by poly (I:C) in vivo.


Sujet(s)
Caspase-1 , Souris de lignée BALB C , Protéine-3 de la famille des NLR contenant un domaine pyrine , Infections à Orthomyxoviridae , Espèces réactives de l'oxygène , Saponines , Transduction du signal , Triterpènes , Animaux , Saponines/pharmacologie , Triterpènes/pharmacologie , Triterpènes/usage thérapeutique , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Humains , Espèces réactives de l'oxygène/métabolisme , Cellules A549 , Caspase-1/métabolisme , Infections à Orthomyxoviridae/traitement médicamenteux , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammation/traitement médicamenteux , Virus de la grippe A/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique
12.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892096

RÉSUMÉ

Influenza A viruses (IAVs) pose a significant global threat to human health. A tightly controlled host immune response is critical to avoid any detrimental effects of IAV infection. It is critical to investigate the association between the response of Toll-like receptors (TLRs) and influenza virus. Because TLRs may act as a double-edged sword, a balanced TLR response is critical for the overall benefit of the host. Consequently, a thorough understanding of the TLR response is essential for targeting TLRs as a novel therapeutic and prophylactic intervention. To date, a limited number of studies have assessed TLR and IAV interactions. Therefore, further research on TLR interactions in IAV infection should be conducted to determine their role in host-virus interactions in disease causation or clearance of the virus. Although influenza virus vaccines are available, they have limited efficacy, which should be enhanced to improve their efficacy. In this study, we discuss the current status of our understanding of the TLR response in IAV infection and the strategies adopted by IAVs to avoid TLR-mediated immune surveillance, which may help in devising new therapeutic or preventive strategies. Furthermore, recent advances in the use of TLR agonists as vaccine adjuvants to enhance influenza vaccine efficacy are discussed.


Sujet(s)
Virus de la grippe A , Vaccins antigrippaux , Grippe humaine , Récepteurs de type Toll , Humains , Récepteurs de type Toll/métabolisme , Grippe humaine/immunologie , Grippe humaine/virologie , Animaux , Vaccins antigrippaux/immunologie , Virus de la grippe A/immunologie , Interactions hôte-pathogène/immunologie , Immunité innée , Transduction du signal
14.
Viruses ; 16(5)2024 05 10.
Article de Anglais | MEDLINE | ID: mdl-38793634

RÉSUMÉ

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


Sujet(s)
Systèmes CRISPR-Cas , Grippe chez les oiseaux , Sensibilité et spécificité , Animaux , Grippe chez les oiseaux/virologie , Grippe chez les oiseaux/diagnostic , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/isolement et purification , Sous-type H5N1 du virus de la grippe A/classification , Virus de la grippe A/génétique , Virus de la grippe A/isolement et purification , Virus de la grippe A/classification , Volaille/virologie , Maladies de la volaille/virologie , Maladies de la volaille/diagnostic , Poulets/virologie , Oiseaux/virologie
16.
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
17.
Proc Natl Acad Sci U S A ; 121(22): e2310677121, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38753503

RÉSUMÉ

Seasonal and pandemic-associated influenza strains cause highly contagious viral respiratory infections that can lead to severe illness and excess mortality. Here, we report on the optimization of our small-molecule inhibitor F0045(S) targeting the influenza hemagglutinin (HA) stem with our Sulfur-Fluoride Exchange (SuFEx) click chemistry-based high-throughput medicinal chemistry (HTMC) strategy. A combination of SuFEx- and amide-based lead molecule diversification and structure-guided design led to identification and validation of ultrapotent influenza fusion inhibitors with subnanomolar EC50 cellular antiviral activity against several influenza A group 1 strains. X-ray structures of six of these compounds with HA indicate that the appended moieties occupy additional pockets on the HA surface and increase the binding interaction, where the accumulation of several polar interactions also contributes to the improved affinity. The compounds here represent the most potent HA small-molecule inhibitors to date. Our divergent HTMC platform is therefore a powerful, rapid, and cost-effective approach to develop bioactive chemical probes and drug-like candidates against viral targets.


Sujet(s)
Antiviraux , Glycoprotéine hémagglutinine du virus influenza , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Humains , Antiviraux/pharmacologie , Antiviraux/composition chimique , Chimie pharmaceutique/méthodes , Tests de criblage à haut débit/méthodes , Grippe humaine/traitement médicamenteux , Grippe humaine/virologie , Cristallographie aux rayons X/méthodes , Chimie click/méthodes , Animaux , Virus de la grippe A/effets des médicaments et des substances chimiques , Cellules rénales canines Madin-Darby , Inhibiteurs des protéines de fusion virale/pharmacologie , Inhibiteurs des protéines de fusion virale/composition chimique , Chiens
18.
Sci Rep ; 14(1): 12184, 2024 05 28.
Article de Anglais | MEDLINE | ID: mdl-38806597

RÉSUMÉ

Catalytic antibodies possess a dual function that enables both antigen recognition and degradation. However, their time-consuming preparation is a significant drawback. This study developed a new method for quickly converting mice monoclonal antibodies into catalytic antibodies using site-directed mutagenesis. Three mice type monoclonal antibodies targeting hemagglutinin molecule of influenza A virus could be transformed into the catalytic antibodies by deleting Pro95 in CDR-3 of the light chain. No catalytic activity was observed for monoclonal antibodies and light chains. In contrast, the Pro95-deleted light chains exhibited a catalytic activity to cleave the antigenic peptide including the portion of conserved region of hemagglutinin molecule. The affinity of the Pro95-deleted light chains to the antigen increased approximately 100-fold compared to the wild-type light chains. In the mutants, three residues (Asp1, Ser92, and His93) come closer to the appropriate position to create the catalytic site and contributing to the enhancement of both catalytic function and immunoreactivity. Notably, the Pro95-deleted catalytic light chains could suppress influenza virus infection in vitro assay, whereas the parent antibody and the light chain did not. This strategy offers a rapid and efficient way to create catalytic antibodies from existing antibodies, accelerating the development for various applications in diagnostic and therapeutic applications.


Sujet(s)
Anticorps catalytiques , Anticorps monoclonaux , Animaux , Souris , Anticorps monoclonaux/immunologie , Anticorps catalytiques/métabolisme , Anticorps catalytiques/immunologie , Anticorps catalytiques/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Mutagenèse dirigée , Virus de la grippe A/immunologie , Domaine catalytique , Humains , Chaines légères des immunoglobulines/génétique , Chaines légères des immunoglobulines/immunologie , Chaines légères des immunoglobulines/métabolisme , Anticorps antiviraux/immunologie , Souris de lignée BALB C
19.
Curr Microbiol ; 81(7): 198, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38819647

RÉSUMÉ

Although usefulness of masks for protection against respiratory pathogens, accumulation of pathogens on their surface represents a source of infection spread. Here we prepared a plant extract-based disinfecting layer to be used in coating masks thus inhibiting their capacity to transmit airborne pathogens. To reach this, a polypropylene membrane base was coated with a layer of polyvinyledine difluoride polymer containing 500 µg/ml of Camellia sinensis (Black tea) methanolic extract. Direct inhibitory effects of C. sinensis were initially demonstrated against Staphylococcus aureus (respiratory bacteria), influenza A virus (enveloped virus) and adenovirus 1 (non-enveloped virus) which were directly proportional to both extract concentration and incubation time with the pathogen. This was later confirmed by the capacity of the supplemented membrane with the plant extract to block infectivity of the above mentioned pathogens, recorded % inhibition values were 61, 72 and 50 for S. aureus, influenza and adenovirus, respectively. In addition to the disinfecting capacity of the membrane its hydrophobic nature and pore size (154 nm) prevented penetration of dust particles or water droplets carrying respiratory pathogens. In summary, introducing this layer could protect users from infection and decrease infection risk upon handling contaminated masks surfaces.


Sujet(s)
Camellia sinensis , Masques , Extraits de plantes , Staphylococcus aureus , Camellia sinensis/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Staphylococcus aureus/effets des médicaments et des substances chimiques , Masques/virologie , Désinfectants/pharmacologie , Virus de la grippe A/effets des médicaments et des substances chimiques , Humains
20.
Nat Commun ; 15(1): 4500, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802391

RÉSUMÉ

Jamaican fruit bats (Artibeus jamaicensis) naturally harbor a wide range of viruses of human relevance. These infections are typically mild in bats, suggesting unique features of their immune system. To better understand the immune response to viral infections in bats, we infected male Jamaican fruit bats with the bat-derived influenza A virus (IAV) H18N11. Using comparative single-cell RNA sequencing, we generated single-cell atlases of the Jamaican fruit bat intestine and mesentery. Gene expression profiling showed that H18N11 infection resulted in a moderate induction of interferon-stimulated genes and transcriptional activation of immune cells. H18N11 infection was predominant in various leukocytes, including macrophages, B cells, and NK/T cells. Confirming these findings, human leukocytes, particularly macrophages, were also susceptible to H18N11, highlighting the zoonotic potential of this bat-derived IAV. Our study provides insight into a natural virus-host relationship and thus serves as a fundamental resource for future in-depth characterization of bat immunology.


Sujet(s)
Chiroptera , Infections à Orthomyxoviridae , Analyse sur cellule unique , Animaux , Chiroptera/virologie , Chiroptera/immunologie , Chiroptera/génétique , Mâle , Humains , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/médecine vétérinaire , Macrophages/immunologie , Macrophages/virologie , Virus de la grippe A/génétique , Virus de la grippe A/immunologie , Analyse de profil d'expression de gènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...