Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.303
Filtrer
1.
J Ethnopharmacol ; 336: 118720, 2025 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-39197802

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Jinye Baidu granules (JYBD) have been used to treat acute respiratory tract infections and demonstrated clinical efficacy for the treatment of emerging or epidemic respiratory viruses such as SARS-CoV-2 and influenza virus. AIM OF THE STUDY: This study is to investigate the antiviral effect of JYBD against influenza A viruses (IAV) in vitro and in vivo and elucidate its underlying mechanism. MATERIALS AND METHODS: Ultra-high-performance liquid chromatography connected with Orbitrap mass spectrometer (UHPLC-Orbitrap MS) was employed to describe the chemical profile of JYBD. The potential pathways and targets involved in JYBD against IAV infection were predicted by network pharmacology. The efficacy and mechanism of JYBD were validated through both in vivo and in vitro experiments. Moreover, combination therapy with JYBD and the classic anti-influenza drugs was also investigated. RESULTS: A total of 126 compounds were identified by UHPLC-Orbitrap MS, of which 9 compounds were unambiguously confirmed with reference standards. JYBD could significantly inhibit the replication of multiple strains of IAV, especially oseltamivir-resistant strains. The results of qRT-PCR and WB demonstrated that JYBD could inhibit the excessive induction of pro-inflammatory cytokines induced by IAV infection and regulate inflammatory response through inhibiting JAK/STAT, NF-κB and MAPK pathways. Moreover, both JYBD monotherapy or in combination with oseltamivir could alleviate IAV-induced severe lung injury in mice. CONCLUSIONS: JYBD could inhibit IAV replication and mitigate virus-induced excessive inflammatory response. Combinations of JYBD and neuraminidase inhibitors conferred synergistic suppression of IAV both in vitro and in vivo. It might provide a scientific basis for clinical applications of JYBD against influenza virus infected diseases.


Sujet(s)
Antiviraux , Médicaments issus de plantes chinoises , Virus de la grippe A , Pharmacologie des réseaux , Infections à Orthomyxoviridae , Antiviraux/pharmacologie , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Virus de la grippe A/effets des médicaments et des substances chimiques , Chiens , Souris , Humains , Infections à Orthomyxoviridae/traitement médicamenteux , Infections à Orthomyxoviridae/virologie , Cellules rénales canines Madin-Darby , Réplication virale/effets des médicaments et des substances chimiques , Cellules A549 , Souris de lignée BALB C , Mâle , Femelle , Chromatographie en phase liquide à haute performance
2.
Biomaterials ; 312: 122721, 2025 Jan.
Article de Anglais | MEDLINE | ID: mdl-39106817

RÉSUMÉ

Silver nanoparticles (AgNPs) are a potential antiviral agent due to their ability to disrupt the viral particle or alter the virus metabolism inside the host cell. In vitro, AgNPs exhibit antiviral activity against the most common human respiratory viruses. However, their capacity to modulate immune responses during respiratory viral infections has yet to be explored. This study demonstrates that administering AgNPs directly into the lungs prior to infection can reduce viral loads and therefore virus-induced cytokines in mice infected with influenza virus or murine pneumonia virus. The prophylactic effect was diminished in mice with depleted lymphoid cells. We showed that AgNPs-treatment resulted in the recruitment and activation of lymphocytes in the lungs, particularly natural killer (NK) cells. Mechanistically, AgNPs enhanced the ability of alveolar macrophages to promote both NK cell migration and IFN-γ production. By contrast, following infection, in mice treated with AgNPs, NK cells exhibited decreased activation, indicating that these nanoparticles can regulate the potentially deleterious activation of these cells. Overall, the data suggest that AgNPs may possess prophylactic antiviral properties by recruiting and controlling the activation of lymphoid cells through interaction with alveolar macrophages.


Sujet(s)
Cellules tueuses naturelles , Poumon , Nanoparticules métalliques , Infections à Orthomyxoviridae , Argent , Animaux , Argent/composition chimique , Argent/pharmacologie , Nanoparticules métalliques/composition chimique , Poumon/virologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/traitement médicamenteux , Infections à Orthomyxoviridae/virologie , Souris , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Macrophages alvéolaires/effets des médicaments et des substances chimiques , Macrophages alvéolaires/métabolisme , Macrophages alvéolaires/virologie , Souris de lignée C57BL , Lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes/métabolisme , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Femelle , Activation des lymphocytes/effets des médicaments et des substances chimiques
3.
Biomaterials ; 312: 122736, 2025 Jan.
Article de Anglais | MEDLINE | ID: mdl-39121728

RÉSUMÉ

The resurgence of influenza viruses as a significant global threat emphasizes the urgent need for innovative antiviral strategies beyond existing treatments. Here, we present the development and evaluation of a novel super-multivalent sialyllactosylated filamentous phage, termed t-6SLPhage, as a potent entry blocker for influenza A viruses. Structural variations in sialyllactosyl ligands, including linkage type, valency, net charge, and spacer length, were systematically explored to identify optimal binding characteristics against target hemagglutinins and influenza viruses. The selected SLPhage equipped with optimal ligands, exhibited exceptional inhibitory potency in in vitro infection inhibition assays. Furthermore, in vivo studies demonstrated its efficacy as both a preventive and therapeutic intervention, even when administered post-exposure at 2 days post-infection, under 4 lethal dose 50% conditions. Remarkably, co-administration with oseltamivir revealed a synergistic effect, suggesting potential combination therapies to enhance efficacy and mitigate resistance. Our findings highlight the efficacy and safety of sialylated filamentous bacteriophages as promising influenza inhibitors. Moreover, the versatility of M13 phages for surface modifications offers avenues for further engineering to enhance therapeutic and preventive performance.


Sujet(s)
Antiviraux , Animaux , Antiviraux/pharmacologie , Antiviraux/composition chimique , Humains , Chiens , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/traitement médicamenteux , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/physiologie , Cellules rénales canines Madin-Darby , Inovirus/effets des médicaments et des substances chimiques , Oséltamivir/pharmacologie , Oséltamivir/composition chimique , Souris , Grippe humaine/virologie , Grippe humaine/traitement médicamenteux , Souris de lignée BALB C , Acide N-acétyl-neuraminique/composition chimique , Acide N-acétyl-neuraminique/métabolisme , Femelle
4.
PLoS One ; 19(9): e0308648, 2024.
Article de Anglais | MEDLINE | ID: mdl-39312544

RÉSUMÉ

BACKGROUND: The alveolar epithelium is protected by a heparan sulfate-rich, glycosaminoglycan layer called the epithelial glycocalyx. It is cleaved in patients with acute respiratory distress syndrome (ARDS) and in murine models of influenza A (IAV) infection, shedding fragments into the airspace from the cell surface. Glycocalyx shedding results in increased permeability of the alveolar-capillary barrier, amplifying acute lung injury. The mechanisms underlying alveolar epithelial glycocalyx shedding in IAV infection are unknown. We hypothesized that induction of host sheddases such as matrix metalloproteinases (MMPs) during IAV infection results in glycocalyx shedding and increased lung injury. MATERIALS AND METHODS: We measured glycocalyx shedding and lung injury during IAV infection with and without treatment with the pan-MMP inhibitor Ilomastat (ILO) and in an MMP-7 knock out (MMP-7KO) mouse. C57BL/6 or MMP-7KO male and female mice were given IAV A/PR/8/34 (H1N1) at 30,000 PFU/mouse or PBS intratracheally. For some experiments, C56BL/6 mice were infected in the presence of ILO (100mg/kg) or vehicle given daily by IP injection. Bronchoalveolar lavage (BAL) and lung tissue were collected on day 1, 3, and 7 for analysis of glycocalyx shedding (BAL Syndecan-1) and lung injury (histology, BAL protein, BAL cytokines, BAL immune cell infiltrates, BAL RAGE). Expression and localization of the sheddase MMP-7 and its inhibitor TIMP-1 was examined by RNAScope. For in vitro experiments, MLE-12 mouse lung epithelial cells were cultured and treated with active or heat-inactivated heparinase (2.5 U/mL) prior to infection with IAV (MOI 1) and viral load and MMP-7 and TIMP-1 expression analyzed. RESULTS: IAV infection caused shedding of the epithelial glycocalyx into the BAL. Inhibition of MMPs with ILO reduced glycocalyx shedding by 36% (p = 0.0051) and reduced lung epithelial injury by 40% (p = 0.0404). ILO also reduced viral load by 68% (p = 0.027), despite having no significant effect on lung cytokine production. Both MMP-7 and its inhibitor TIMP-1 were upregulated in IAV infected mice: MMP-7 colocalized with IAV, while TIMP-1 was limited to cells adjacent to infection. However, MMP-7KO mice had similar glycocalyx shedding, epithelial injury, and viral load compared to WT littermates, suggesting redundancy in MMP sheddase function in the lung. In vitro, heparinase treatment before infection led to a 52% increase in viral load (p = 0.0038) without altering MMP-7 or TIMP-1 protein levels. CONCLUSIONS: Glycocalyx shedding and MMPs play key roles in IAV-induced epithelial injury, with significant impact on IAV viral load. Further studies are needed to understand which specific MMPs regulate lung epithelial glycocalyx shedding.


Sujet(s)
Glycocalyx , Matrix metalloproteinase 7 , Souris de lignée C57BL , Infections à Orthomyxoviridae , Animaux , Glycocalyx/métabolisme , Souris , Femelle , Mâle , Matrix metalloproteinase 7/métabolisme , Matrix metalloproteinase 7/génétique , Infections à Orthomyxoviridae/métabolisme , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/anatomopathologie , Sous-type H1N1 du virus de la grippe A/physiologie , Souris knockout , Alvéoles pulmonaires/virologie , Alvéoles pulmonaires/métabolisme , Alvéoles pulmonaires/anatomopathologie , Acides hydroxamiques/pharmacologie , Inhibiteurs de métalloprotéinases matricielles/pharmacologie , Inhibiteur tissulaire de métalloprotéinase-1/métabolisme , Matrix metalloproteinases/métabolisme , Indoles
6.
Sci Rep ; 14(1): 21324, 2024 09 12.
Article de Anglais | MEDLINE | ID: mdl-39266641

RÉSUMÉ

The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases twofold at 7 days post-influenza infection (dpi) and threefold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Cellules endothéliales , Poumon , Lymphangiogenèse , Infections à Orthomyxoviridae , Protéines de signalisation YAP , Animaux , Protéines de signalisation YAP/métabolisme , Cellules endothéliales/métabolisme , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Poumon/métabolisme , Poumon/anatomopathologie , Infections à Orthomyxoviridae/métabolisme , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/anatomopathologie , Virus de la grippe A/physiologie , Vaisseaux lymphatiques/métabolisme , Vaisseaux lymphatiques/anatomopathologie , Souris knockout , Transduction du signal , Prolifération cellulaire , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Souris de lignée C57BL , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique
7.
Emerg Infect Dis ; 30(10): 2033-2041, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39240548

RÉSUMÉ

The prevalence of highly pathogenic avian influenza (HPAI) A(H5N1) viruses has increased in wild birds and poultry worldwide, and concomitant outbreaks in mammals have occurred. During 2023, outbreaks of HPAI H5N1 virus infections were reported in cats in South Korea. The H5N1 clade 2.3.4.4b viruses isolated from 2 cats harbored mutations in the polymerase basic protein 2 gene encoding single amino acid substitutions E627K or D701N, which are associated with virus adaptation in mammals. Hence, we analyzed the pathogenicity and transmission of the cat-derived H5N1 viruses in other mammals. Both isolates caused fatal infections in mice and ferrets. We observed contact infections between ferrets, confirming the viruses had high pathogenicity and transmission in mammals. Most HPAI H5N1 virus infections in humans have occurred through direct contact with poultry or a contaminated environment. Therefore, One Health surveillance of mammals, wild birds, and poultry is needed to prevent potential zoonotic threats.


Sujet(s)
Furets , Sous-type H5N1 du virus de la grippe A , Infections à Orthomyxoviridae , Animaux , Furets/virologie , République de Corée/épidémiologie , Souris , Chats , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/médecine vétérinaire , Infections à Orthomyxoviridae/épidémiologie , Sous-type H5N1 du virus de la grippe A/pathogénicité , Sous-type H5N1 du virus de la grippe A/génétique , Phylogenèse , Maladies des chats/virologie , Maladies des chats/épidémiologie , Virulence , Épidémies de maladies , Humains , Femelle
9.
Open Vet J ; 14(8): 1896-1904, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39308731

RÉSUMÉ

Background: Dogs in close contact with humans can serve as a source of potentially dangerous reassortant influenza viruses (IVs) with zoonotic potential. The dog's body can serve as a vessel for the emergence of new IVs. These new viruses can become a source of infection for other animals and humans. The potential for zoonotic transmission of IVs from dogs to humans poses a public health risk. Aim: Study of the circulation of IVs in the dog population in Almaty, Kazakhstan. Methods: Biosamples (oropharyngeal swabs and blood serum) from dogs were collected from veterinary clinics in Almaty in 2023-2024. Samples were screened using RT-PCR, HI assay, and ELISA. Results: RT-PCR analysis of 355 nasopharyngeal swabs showed the presence of influenza A virus (IAV) in 32 samples (9.01% of the total number of samples analyzed). When subtyping IAV H1N1 RNA was detected in 19 swabs (5.35%). IAV subtype could not be determined in 13 PCR-positive samples (3.66%). The genetic material of IAV H3N2, H5, H7, and H9, as well as coronavirus, bocavirus, and adenovirus has not been identified. In a serological analysis of 180 blood sera using ELISA, antibodies to IAV were detected in 5.56% (n = 10). The results of the HI assay showed the presence of antihemagglutinins to A/H1N1pdm in 6.11% (11 samples), to A/H3N2 in 9.44% (17 samples), and no antibodies to IAV H5, H7, and type B were detected. Conclusion: There is no information about human infection with any canine influenza virus. However, many cases of infection in dogs with human IAVs H1N1, H1N1pdm09, and H3N2 have been described. When dogs are co-infected with different IAVs, new recombinant IAVs may emerge that can infect humans and other animals. Therefore, ongoing global surveillance of animal populations is necessary to monitor the evolution and circulation of viruses dangerous to public health. This is also important for timely preparation for the emergence of a new zoonotic influenza virus that has pandemic potential for humans.


Sujet(s)
Maladies des chiens , Infections à Orthomyxoviridae , Animaux , Chiens , Maladies des chiens/épidémiologie , Maladies des chiens/virologie , Infections à Orthomyxoviridae/médecine vétérinaire , Infections à Orthomyxoviridae/épidémiologie , Infections à Orthomyxoviridae/virologie , Kazakhstan/épidémiologie , Virus de la grippe A/isolement et purification , Humains , Test ELISA/médecine vétérinaire
10.
J Med Virol ; 96(9): e29906, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39262090

RÉSUMÉ

Influenza virus-induced viral pneumonia is a major threat to human health, and specific therapeutic agents for viral pneumonia are still lacking. MoringaA (MA) is an anti-influenza virus active compound isolated from Moringa seeds, which can inhibit influenza virus by activating the TFEB-autophagic lysosomal pathway in host cells. In this study, we obtained exosomes from M2-type macrophages and encapsulated and delivered MA (MA-Exos), and we investigated the efficacy of MA-Exos in antiviral and viral pneumonia in vivo and in vitro, respectively. In addition, we provided insights into the mechanism by which MA-Exos regulates TFEB-lysosomal autophagy by RNA sequencing. The MA-Exos showed broad-spectrum inhibition of IAV, and significant promotion of the autophagic lysosomal pathway. Meanwhile, we found that GCN5 gene and protein were significantly down-regulated in IAV-infected cells after MA-Exos intervention, indicating its blocking the acetylation of TFEB by GCN5. In addition, MA-Exos also significantly promoted autophagy in lung tissue cells of mice with viral pneumonia. MA-Exos can inhibit and clear influenza virus by mediating the TFEB-autophagy lysosomal pathway by a mechanism related to the down-regulation of histone acetyltransferase GCN5. Our study provides a strategy for targeting MA-Exos for the treatment of viral pneumonia from both antiviral and virus-induced inflammation inhibition pathways.


Sujet(s)
Antiviraux , Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Exosomes , Virus de la grippe A , Lysosomes , Animaux , Souris , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/virologie , Exosomes/métabolisme , Antiviraux/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Humains , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/physiologie , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/traitement médicamenteux , Macrophages/virologie , Macrophages/effets des médicaments et des substances chimiques , Poumon/virologie
11.
Emerg Microbes Infect ; 13(1): 2400546, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39221898

RÉSUMÉ

The vast majority of data obtained from sequence analysis of influenza A viruses (IAVs) have revealed that nonstructural 1 (NS1) proteins from H1N1 swine, H3N8 equine, H3N2 avian and the correspondent subtypes from dogs have a conserved four C-terminal amino acid motif when independent cross-species transmission occurs between these species. To test the influence of the C-terminal amino acid motifs of NS1 protein on the replication and virulence of IAVs, we systematically generated 7 recombinants, which carried naturally truncated NS1 proteins, and their last four C-terminal residues were replaced with PEQK and SEQK (for H1N1), EPEV and KPEI (for H3N8) and ESEV and ESEI (for H3N2) IAVs. Another recombinant was generated by removing the C-terminal residues by reverse genetics. Remarkably, the ESEI and KPEI motifs circulating in canines largely contributed efficient replication in cultured cells and these had enhanced virulence. In contrast, the avian ESEV motif was only responsible for high pathogenicity in mice. We examined the effects of these motifs upon interferon (IFN) induction. The 7 mutant viruses replicated in vitro in an IFN-independent manner, and the canine SEQK motif was able to induced higher levels of IFN-ß in human cell lines. These findings shed further new light on the role of the four C-terminal residues in replication and virulence of IAVs and suggest that these motifs can modulate viral replication in a species-specific manner.


Sujet(s)
Motifs d'acides aminés , Sous-type H1N1 du virus de la grippe A , Infections à Orthomyxoviridae , Protéines virales non structurales , Réplication virale , Protéines virales non structurales/génétique , Protéines virales non structurales/métabolisme , Protéines virales non structurales/composition chimique , Animaux , Chiens , Virulence , Souris , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/pathogénicité , Sous-type H1N1 du virus de la grippe A/physiologie , Infections à Orthomyxoviridae/virologie , Humains , Cellules rénales canines Madin-Darby , Souris de lignée BALB C , Maladies des chiens/virologie , Sous-type H3N2 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/pathogénicité , Sous-type H3N2 du virus de la grippe A/physiologie , Sous-type H3N8 du virus de la grippe A/génétique , Sous-type H3N8 du virus de la grippe A/pathogénicité , Femelle
12.
Front Immunol ; 15: 1432743, 2024.
Article de Anglais | MEDLINE | ID: mdl-39247193

RÉSUMÉ

Introduction: Influenza A virus (IAV) infection is a global respiratory disease, which annually leads to 3-5 million cases of severe illness, resulting in 290,000-650,000 deaths. Additionally, during the past century, four global IAV pandemics have claimed millions of human lives. The epithelial lining of the trachea plays a vital role during IAV infection, both as point of viral entry and replication as well as in the antiviral immune response. Tracheal tissue is generally inaccessible from human patients, which makes animal models crucial for the study of the tracheal host immune response. Method: In this study, pigs were inoculated with swine- or human-adapted H1N1 IAV to gain insight into how host adaptation of IAV shapes the innate immune response during infection. In-depth multi-omics analysis (global proteomics and RNA sequencing) of the host response in upper and lower tracheal tissue was conducted, and results were validated by microfluidic qPCR. Additionally, a subset of samples was selected for histopathological examination. Results: A classical innate antiviral immune response was induced in both upper and lower trachea after infection with either swine- or human-adapted IAV with upregulation of genes and higher abundance of proteins associated with viral infection and recognition, accompanied by a significant induction of interferon stimulated genes with corresponding higher proteins concentrations. Infection with the swine-adapted virus induced a much stronger immune response compared to infection with a human-adapted IAV strain in the lower trachea, which could be a consequence of a higher viral load and a higher degree of inflammation. Discussion: Central components of the JAK-STAT pathway, apoptosis, pyrimidine metabolism, and the cytoskeleton were significantly altered depending on infection with swine- or human-adapted virus and might be relevant mechanisms in relation to antiviral immunity against putative zoonotic IAV. Based on our findings, we hypothesize that during host adaptation, IAV evolve to modulate important host cell elements to favor viral infectivity and replication.


Sujet(s)
Infections à Orthomyxoviridae , Protéomique , Trachée , Animaux , Trachée/immunologie , Trachée/virologie , Suidae , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Protéomique/méthodes , Humains , Adaptation à l'hôte/immunologie , Immunité innée , Sous-type H1N1 du virus de la grippe A/immunologie , Interactions hôte-pathogène/immunologie , Multi-omique
13.
Emerg Microbes Infect ; 13(1): 2387450, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39129565

RÉSUMÉ

Throughout history, the influenza A virus has caused numerous devastating global pandemics. Macrophages, as pivotal innate immune cells, exhibit a wide range of immune functions characterized by distinct polarization states, reflecting their intricate heterogeneity. In this study, we employed the time-resolved single-cell sequencing technique coupled with metabolic RNA labelling to elucidate the dynamic transcriptional changes in distinct polarized states of bone marrow-derived macrophages (BMDMs) upon infection with the influenza A virus. Our approach not only captures the temporal dimension of transcriptional activity, which is lacking in conventional scRNA-seq methods, but also reveals that M2-polarized Arg1_macrophage cluster is the sole state supporting successful replication of influenza A virus. Furthermore, we identified distinct antigen presentation capabilities to CD4+ T and CD8+ T cells across diverse polarized states of macrophages. Notably, the M1 phenotype, exhibited by (BMDMs) and murine alveolar macrophages (AMs), demonstrated superior conventional and cross-presentation abilities for exogenous antigens, with a particular emphasis on cross-presentation capacity. Additionally, as CD8+ T cell differentiation progressed, M1 polarization exhibited an enhanced capacity for cross-presentation. All three phenotypes of BMDMs, including M1, demonstrated robust presentation to CD4+ regulatory T cells, while displaying limited ability to present to naive CD4+ T cells. These findings offer novel insights into the immunological regulatory mechanisms governing distinct polarized states of macrophages, particularly their roles in restricting the replication of influenza A virus and modulating antigen-specific T cell responses through innate immunity.


Sujet(s)
Présentation d'antigène , Lymphocytes T CD8+ , Virus de la grippe A , Macrophages , Infections à Orthomyxoviridae , Animaux , Virus de la grippe A/immunologie , Virus de la grippe A/génétique , Souris , Macrophages/immunologie , Macrophages/virologie , Lymphocytes T CD8+/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Lymphocytes T CD4+/immunologie , Souris de lignée C57BL , Analyse sur cellule unique/méthodes , RNA-Seq/méthodes , Analyse de l'expression du gène de la cellule unique
14.
Microb Pathog ; 195: 106895, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39208965

RÉSUMÉ

Deadly outbreaks among poultry, wild birds, and carnivorous mammals by the highly pathogenic H5N1 virus of the clade 2.3.4.4b have been reported in South America. The increasing virus incidence in various mammal species poses a severe zoonotic and pandemic threat. In Uruguay, the clade 2.3.4.4b viruses were first detected in February 2023, affecting wild birds and backyard poultry. Three months after the first reported case in Uruguay, the disease affected a population of 23 coatis (Nasua) in an ecological park. Most animals became infected, likely directly or indirectly from wild birds in the park, and experienced sudden death. Five animals from the colony survived, and four of them developed antibodies. The genomes of the H5N1 strains infecting coatis belonged to the B3.2 genotype of the clade 2.3.4.4b. Genomes from coatis were closely associated with those infecting backyard poultry, but transmission likely occurred through wild birds. Notable, two genomes have a 627K substitution in the RNA polymerase PB2 subunit, a hallmark amino acid linked to mammalian adaptation. Our findings support the ability of the avian influenza virus of the 2.3.4.4b clade to infect and transmit among terrestrial mammals with high pathogenicity and undergo rapid adaptive changes. It also highlights the coatis' ability to develop immunity and naturally clear the infection.


Sujet(s)
Animaux sauvages , Génome viral , Sous-type H5N1 du virus de la grippe A , Grippe chez les oiseaux , Mutation , Phylogenèse , Procyonidae , Animaux , Procyonidae/virologie , Grippe chez les oiseaux/virologie , Sous-type H5N1 du virus de la grippe A/génétique , Sous-type H5N1 du virus de la grippe A/pathogénicité , Sous-type H5N1 du virus de la grippe A/isolement et purification , Génome viral/génétique , Uruguay , Animaux sauvages/virologie , Oiseaux/virologie , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/médecine vétérinaire , Volaille/virologie , Génotype , Mammifères/virologie , Amérique du Sud , Épidémies de maladies/médecine vétérinaire
15.
mBio ; 15(9): e0066824, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39105586

RÉSUMÉ

The COVID-19 pandemic caused by SARS-CoV-2 has had a persistent and significant impact on global public health for 4 years. Recently, there has been a resurgence of seasonal influenza transmission worldwide. The co-circulation of SARS-CoV-2 and seasonal influenza viruses results in a dual burden on communities. Additionally, the pandemic potential of zoonotic influenza viruses, such as avian Influenza A/H5N1 and A/H7N9, remains a concern. Therefore, a combined vaccine against all these respiratory diseases is in urgent need. mRNA vaccines, with their superior efficacy, speed in development, flexibility, and cost-effectiveness, offer a promising solution for such infectious diseases and potential future pandemics. In this study, we present FLUCOV-10, a novel 10-valent mRNA vaccine created from our proven platform. This vaccine encodes hemagglutinin (HA) proteins from four seasonal influenza viruses and two avian influenza viruses with pandemic potential, as well as spike proteins from four SARS-CoV-2 variants. A two-dose immunization with the FLUCOV-10 elicited robust immune responses in mice, producing IgG antibodies, neutralizing antibodies, and antigen-specific cellular immune responses against all the vaccine-matched viruses of influenza and SARS-CoV-2. Remarkably, the FLUCOV-10 immunization provided complete protection in mouse models against both homologous and heterologous strains of influenza and SARS-CoV-2. These results highlight the potential of FLUCOV-10 as an effective vaccine candidate for the prevention of influenza and COVID-19.IMPORTANCEAmidst the ongoing and emerging respiratory viral threats, particularly the concurrent and sequential spread of SARS-CoV-2 and influenza, our research introduces FLUCOV-10. This novel mRNA-based combination vaccine, designed to counteract both influenza and COVID-19, by incorporating genes for surface glycoproteins from various influenza viruses and SARS-CoV-2 variants. This combination vaccine was highly effective in preclinical trials, generating strong immune responses and ensuring protection against both matching and heterologous strains of influenza viruses and SARS-CoV-2. FLUCOV-10 represents a significant step forward in our ability to address respiratory viral threats, showcasing potential as a singular, adaptable vaccine solution for global health challenges.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Vaccins antigrippaux , SARS-CoV-2 , Vaccins à ARNm , Animaux , COVID-19/prévention et contrôle , COVID-19/immunologie , Souris , SARS-CoV-2/immunologie , SARS-CoV-2/génétique , Vaccins contre la COVID-19/immunologie , Vaccins contre la COVID-19/administration et posologie , Humains , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Vaccins synthétiques/immunologie , Vaccins synthétiques/génétique , Vaccins synthétiques/administration et posologie , Souris de lignée BALB C , Femelle , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Sous-type H5N1 du virus de la grippe A/immunologie , Sous-type H5N1 du virus de la grippe A/génétique , Virus de la grippe A/immunologie , Virus de la grippe A/génétique
16.
Antiviral Res ; 230: 105978, 2024 10.
Article de Anglais | MEDLINE | ID: mdl-39117282

RÉSUMÉ

Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.


Sujet(s)
Vaccins antigrippaux , Sialidase , Infections à Orthomyxoviridae , Sialidase/immunologie , Sialidase/génétique , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/génétique , Animaux , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Souris , Lymphocytes T/immunologie , Souris de lignée BALB C , Femelle , Humains , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Grippe humaine/virologie , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Lymphocytes auxiliaires Th1/immunologie , Protéines virales/génétique , Protéines virales/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang
17.
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
18.
J R Soc Interface ; 21(217): 20240168, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39109454

RÉSUMÉ

Viruses that infect animals regularly spill over into the human population, but individual events may lead to anything from a single case to a novel pandemic. Rapidly gaining an understanding of a spillover event is critical to calibrating a public health response. We here propose a novel method, using likelihood-free rejection sampling, to evaluate the properties of an outbreak of swine-origin influenza A(H1N2)v in the United Kingdom, detected in November 2023. From the limited data available, we generate historical estimates of the probability that the outbreak had died out in the days following the detection of the first case. Our method suggests that the outbreak could have been said to be over with 95% certainty between 19 and 29 days after the first case was detected, depending upon the probability of a case being detected. We further estimate the number of undetected cases conditional upon the outbreak still being live, the epidemiological parameter R 0, and the date on which the spillover event itself occurred. Our method requires minimal data to be effective. While our calculations were performed after the event, the real-time application of our method has potential value for public health responses to cases of emerging viral infection.


Sujet(s)
Grippe humaine , Royaume-Uni/épidémiologie , Humains , Grippe humaine/épidémiologie , Sous-type H1N2 du virus de la grippe A , Suidae , Animaux , Épidémies de maladies , Infections à Orthomyxoviridae/épidémiologie , Infections à Orthomyxoviridae/virologie , Maladies des porcs/épidémiologie , Maladies des porcs/virologie
19.
Sci Adv ; 10(33): eado4313, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39141734

RÉSUMÉ

αß T cell receptors (TCRs) principally recognize aberrant peptides bound to major histocompatibility complex molecules (pMHCs) on unhealthy cells, amplifying specificity and sensitivity through physical load placed on the TCR-pMHC bond during immunosurveillance. To understand this mechanobiology, TCRs stimulated by abundantly and sparsely arrayed epitopes (NP366-374/Db and PA224-233/Db, respectively) following in vivo influenza A virus infection were studied with optical tweezers. While certain NP repertoire CD8 T lymphocytes require many ligands for activation, others are digital, needing just few. Conversely, all PA TCRs perform digitally, exhibiting pronounced bond lifetime increases through sustained, energizing volleys of structural transitioning. Optimal digital performance is superior in vivo, correlating with ERK phosphorylation, CD3 loss, and activation marker up-regulation in vitro. Given neoantigen array paucity, digital TCRs are likely critical for immunotherapies.


Sujet(s)
Lymphocytes T CD8+ , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris , Récepteurs aux antigènes des cellules T/métabolisme , Récepteurs aux antigènes des cellules T/immunologie , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme , Récepteur lymphocytaire T antigène, alpha-bêta/composition chimique , Virus de la grippe A/immunologie , Humains , Activation des lymphocytes/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Pinces optiques
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE