Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.281
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.
J Ethnopharmacol ; 336: 118654, 2025 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-39098621

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Chaihu Guizhi Decoction (CGD) has a long history of use in China for the treatment of influenza, which involves the use of a variety of aromatic herbs. Our previous studies have found that the contents of aromatic constituents in CGD affected the efficacy of treatment of influenza-infected mice, suggesting a clue that essential oil from CGD may play a relatively important role in ameliorating influenza induced pneumonia. AIM OF THE STUDY: To evaluate the anti-influenza potential of essential oil derived from Chaihu Guizhi Decoction (CGD-EO), to characterize and predict the key active components in CGD-EO, and to explore the mechanism of action of CGD-EO. MATERIALS AND METHODS: CGD-EO was obtained by steam distillation, and the components of the essential oil were characterized by gas chromatography-mass spectrometry (GC-MS) in conjunction with the retention index. The constituents absorbed into the blood of mice treated with CGD-EO were analyzed by headspace solid phase microextraction gas chromatography/mass spectrometry (HS-SPME-GC/MS). The potential anti-influenza active constituents and their possible action pathway were predicted by simulation using a network pharmacology approach. The protective effect of CGD-EO and its major components on H1N1/PR8-infected cells was determined using the CCK8 assay kit. Mice infected with influenza A virus H1N1/PR8 were administered different doses of CGD-EO orally and the body weights and lung weights were recorded. Mice with varying degrees of H1N1/PR8 infection were administered CGD-EO orally, and their daily weight, water consumption, and clinical indicators were recorded. Necropsies were conducted on days 3 and 5, during which lung weights were measured and lung tissues were preserved. Furthermore, the mRNA expression of the H1N1/PR8 virus and inflammatory factors in lung tissue was analyzed using RT-qPCR. RESULTS: (E)-cinnamaldehyde was the most abundant compound in the CGD-EO. The results of serum medicinal chemistry combined with network pharmacological analysis indicated that (E)-cinnamaldehyde and 3-phenyl-2-propenal may be potential active components of the CGD-EO anti-influenza, and may be involved in the NF-κB signalling pathway. In vitro studies have demonstrated that both CGD-EO and cinnamaldehyde exert a protective effect on MDCK cells infected with H1N1/PR8. In a 0.5 TCID50 H1N1/PR8-induced influenza model, mice treated with CGD-EO at a dose of 63.50 µg/kg exhibited a reduction in lung index, pathological lung lesions, and H1N1/PR8 viral gene levels. In addition, CGD-EO treatment was found to regulate the levels of inflammatory cytokines, including IL-6, TNF-α, and IFN-γ. Moreover, following three days of administration, an upregulation of NF-κB mRNA levels in mouse lung tissue was observed in response to CGD-EO treatment. CONCLUSIONS: The findings of our study indicate CGD-EO exerts a protective effect against H1N1-induced cytopathic lesions in vitro and is capable of alleviating H1N1-induced pneumonitis in mice. Moreover, it appears to be more efficacious in the treatment of mild symptoms of H1N1 infection. Studies have demonstrated that CGD-EO has antiviral potential to attenuate influenza-induced lung injury by modulating inflammatory cytokines and NF-κB signalling pathways during the early stages of influenza infection. It is possible that (E)-cinnamaldehyde is a potential active ingredient in the anti-influenza efficacy of CGD-EO.


Sujet(s)
Antiviraux , Médicaments issus de plantes chinoises , Huile essentielle , Infections à Orthomyxoviridae , Animaux , Huile essentielle/pharmacologie , Médicaments issus de plantes chinoises/pharmacologie , Souris , Infections à Orthomyxoviridae/traitement médicamenteux , Antiviraux/pharmacologie , Souris de lignée BALB C , Pneumopathie virale/traitement médicamenteux , Mâle , Cellules rénales canines Madin-Darby , Chiens , Sous-type H1N1 du virus de la grippe A/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/virologie , Poumon/métabolisme , Humains , Femelle , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/virologie , Pneumopathie infectieuse/métabolisme
3.
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
4.
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
5.
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.
Appl Microbiol Biotechnol ; 108(1): 469, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39298023

RÉSUMÉ

Prior research has indicated that the gut-lung-axis can be influenced by the intestinal microbiota, thereby impacting lung immunity. Rifaximin is a broad-spectrum antibacterial drug that can maintain the homeostasis of intestinal microflora. In this study, we established an influenza A virus (IAV)-infected mice model with or without rifaximin supplementation to investigate whether rifaximin could ameliorate lung injury induced by IAV and explore the molecular mechanism involved. Our results showed that IAV caused significant weight loss and disrupted the structure of the lung and intestine. The analysis results of 16S rRNA and metabolomics indicated a notable reduction in the levels of probiotics Lachnoclostridium, Ruminococcaceae_UCG-013, and tryptophan metabolites in the fecal samples of mice infected with IAV. In contrast, supplementation with 50 mg/kg rifaximin reversed these changes, including promoting the repair of the lung barrier and increasing the abundance of Muribaculum, Papillibacter and tryptophan-related metabolites content in the feces. Additionally, rifaximin treatment increased ILC3 cell numbers, IL-22 level, and the expression of RORγ and STAT-3 protein in the lung. Furthermore, our findings demonstrated that the administration of rifaximin can mitigate damage to the intestinal barrier while enhancing the expression of AHR, IDO-1, and tight junction proteins in the small intestine. Overall, our results provided that rifaximin alleviated the imbalance in gut microbiota homeostasis induced by IAV infection and promoted the production of tryptophan-related metabolites. Tryptophan functions as a signal to facilitate the activation and movement of ILC3 cells from the intestine to the lung through the AHR/STAT3/IL-22 pathway, thereby aiding in the restoration of the barrier. KEY POINTS: • Rifaximin ameliorated IAV infection-caused lung barrier injury and induced ILC3 cell activation. • Rifaximin alleviated IAV-induced gut dysbiosis and recovered tryptophan metabolism. • Tryptophan mediates rifaximin-induced ILC3 cell activation via the AHR/STAT3/IL-22 pathway.


Sujet(s)
Microbiome gastro-intestinal , Virus de la grippe A , Poumon , Infections à Orthomyxoviridae , Rifaximine , Animaux , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Rifaximine/usage thérapeutique , Souris , Poumon/microbiologie , Poumon/effets des médicaments et des substances chimiques , Infections à Orthomyxoviridae/traitement médicamenteux , Virus de la grippe A/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , ARN ribosomique 16S/génétique , Interleukines/métabolisme , Interleukines/génétique , , Souris de lignée C57BL , Antibactériens/pharmacologie , Facteur de transcription STAT-3/métabolisme , Fèces/microbiologie , Tryptophane/métabolisme , Lésion pulmonaire/traitement médicamenteux , Probiotiques/administration et posologie , Probiotiques/pharmacologie
7.
PLoS Pathog ; 20(9): e1012508, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39303003

RÉSUMÉ

Influenza and coronavirus disease 2019 (COVID-19) represent two respiratory diseases that have significantly impacted global health, resulting in substantial disease burden and mortality. An optimal solution would be a combined vaccine capable of addressing both diseases, thereby obviating the need for multiple vaccinations. Previously, we conceived a chimeric protein subunit vaccine targeting both influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), utilizing the receptor binding domain of spike protein (S-RBD) and the stalk region of hemagglutinin protein (HA-stalk) components. By integrating the S-RBD from the SARS-CoV-2 Delta variant with the headless hemagglutinin (HA) from H1N1 influenza virus, we constructed stable trimeric structures that remain accessible to neutralizing antibodies. This vaccine has demonstrated its potential by conferring protection against a spectrum of strains in mouse models. In this study, we designed an mRNA vaccine candidate encoding the chimeric antigen. The resultant humoral and cellular immune responses were meticulously evaluated in mouse models. Furthermore, the protective efficacy of the vaccine was rigorously examined through challenges with either homologous or heterologous influenza viruses or SARS-CoV-2 strains. Our findings reveal that the mRNA vaccine exhibited robust immunogenicity, engendering high and sustained levels of neutralizing antibodies accompanied by robust and persistent cellular immunity. Notably, this vaccine effectively afforded complete protection to mice against H1N1 or heterosubtypic H5N8 subtypes, as well as the SARS-CoV-2 Delta and Omicron BA.2 variants. Additionally, our mRNA vaccine design can be easily adapted from Delta RBD to Omicron RBD antigens, providing protection against emerging variants. The development of two-in-one vaccine targeting both influenza and COVID-19, incorporating the mRNA platform, may provide a versatile approach to combating future pandemics.


Sujet(s)
Vaccins contre la COVID-19 , COVID-19 , Glycoprotéine hémagglutinine du virus influenza , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Vaccins à ARNm , Animaux , Souris , SARS-CoV-2/immunologie , COVID-19/prévention et contrôle , COVID-19/immunologie , Vaccins à ARNm/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Glycoprotéine de spicule des coronavirus/génétique , Humains , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Vaccins contre la COVID-19/immunologie , Vaccins antigrippaux/immunologie , Anticorps antiviraux/immunologie , Souris de lignée BALB C , Femelle , Sous-type H1N1 du virus de la grippe A/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Vaccins synthétiques/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Anticorps neutralisants/immunologie
8.
Vaccine ; 42(24): 126269, 2024 Oct 24.
Article de Anglais | MEDLINE | ID: mdl-39241354

RÉSUMÉ

Recombinant influenza virus neuraminidase (NA) is a promising broadly protective influenza vaccine candidate. However, the recombinant protein alone is not sufficient to induce durable and protective immune responses and requires the coadministration of immunostimulatory molecules. Here, we evaluated the immunogenicity and cross-protective potential of a recombinant influenza virus N2 neuraminidase vaccine construct, adjuvanted with a toll-like receptor 9 (TLR9) agonist (CpG 1018® adjuvant), and alum. The combination of CpG 1018 adjuvant and alum induced a balanced and robust humoral and T-cellular immune response against the NA, which provided protection and reduced morbidity against homologous and heterologous viral challenges in mouse and hamster models. This study supports Syrian hamsters as a useful complementary animal model to mice for pre-clinical evaluation of influenza virus vaccines.


Sujet(s)
Adjuvants immunologiques , Anticorps antiviraux , Vaccins antigrippaux , Sialidase , Infections à Orthomyxoviridae , Animaux , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Sialidase/immunologie , Sialidase/génétique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Souris , Adjuvants immunologiques/administration et posologie , Femelle , Cricetinae , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Adjuvants vaccinaux , Souris de lignée BALB C , Protection croisée/immunologie , Mesocricetus , Oligodésoxyribonucléotides/administration et posologie , Oligodésoxyribonucléotides/immunologie , Alun/administration et posologie , Modèles animaux de maladie humaine , Immunité cellulaire
9.
Hum Vaccin Immunother ; 20(1): 2403175, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39291424

RÉSUMÉ

Influenza neuraminidase (NA) is a promising target for a broadly protective vaccine. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to develop N2 NA vaccine candidates. The unique wild type (WT) N2 sequences of human and swine influenza strains isolated between 1957 and 2019 were used to design the COBRA N2-A NA vaccine, while the unique WT N2 sequences of human influenza strains isolated between 2000 and 2019 were used to design the COBRA N2-B NA vaccine. Sera collected from COBRA N2 NA vaccinated mice showed more broadly reactive antibody responses against a broad panel of H×N2 influenza virus strains than sera collected from mice vaccinated with WT N2 NA vaccines. Antibodies elicited by COBRA or WT N2 NA antigens cross react with recent human H3N2 influenza viruses from different clades, while the antibodies elicited by A/Switzerland/9715293/2013 hemagglutinin (HA) reacted with viruses from the same clade. Furthermore, mice vaccinated with COBRA N2-B NA vaccine had lower viral lung titers compared to mock vaccinated mice when challenged with human H3N2 influenza viruses. Thus, the COBRA N2 NA vaccines elicit broadly protective murine anti-NA antibodies against multiple strains across subtypes and the viral loads were significantly decreased in the lungs of the mice in the COBRA N2 NA vaccine groups, compared to the mice in the mock vaccinated group, indicating that the COBRA-based N2 subtype NA vaccines have a potential to be a component in a universal influenza vaccine.


Sujet(s)
Anticorps antiviraux , Sous-type H3N2 du virus de la grippe A , Vaccins antigrippaux , Sialidase , Infections à Orthomyxoviridae , Animaux , Femelle , Humains , Souris , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Réactions croisées/immunologie , Sous-type H3N2 du virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , Poumon/virologie , Poumon/immunologie , Souris de lignée BALB C , Sialidase/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Charge virale , Protéines virales/immunologie
11.
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
12.
PLoS Pathog ; 20(9): e1011639, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39283916

RÉSUMÉ

Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection or immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed.


Sujet(s)
Lymphocytes T CD4+ , Centre germinatif , Vaccins antigrippaux , Infections à Orthomyxoviridae , Animaux , Centre germinatif/immunologie , Souris , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Lymphocytes T CD4+/immunologie , Anticorps antiviraux/immunologie , Souris de lignée C57BL , Lymphocytes B/immunologie , Mémoire immunologique , Cellules T mémoire/immunologie , Immunisation/méthodes , Femelle , Antigènes viraux/immunologie
13.
Physiol Rep ; 12(17): e70047, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39267201

RÉSUMÉ

Increased circulating tissue inhibitor of metalloproteinases-1 (TIMP-1) levels have been observed in patients with acute lung injury (ALI). However, the sex-specific regulation of TIMP-1 and the underlying molecular mechanisms have not been well elucidated. In this study, we found that plasma TIMP-1 levels were significantly higher in COVID-19 and H1N1 patients compared with those in healthy subjects (n = 25). TIMP-1 concentrations were significantly different between males and females in each disease group. Among female but not male patients, TIMP-1 levels significantly correlated with the PaO2/FiO2 ratio and hospital length of stay. Using the mouse model of ALI induced by the H1N1 virus, we found that TIMP-1 is strikingly induced in PDGFRα-positive cells in the murine lungs. Moreover, female mice showed a higher Timp-1 expression in the lungs on day 3 postinfection. Mechanistically, we observed that estrogen can upregulate TIMP-1 expression in lung fibroblasts, not epithelial cells. In addition, overexpression of estrogen receptor α (ERα) increased the TIMP-1 promoter activity. In summary, TIMP-1 is an estrogen-responsive gene, and its promoter activity is regulated by ERα. Circulating TIMP-1 may serve as a sex-specific marker, reflecting the severity and worst outcomes in female patients with SARS-CoV2- and IAV-related ALI.


Sujet(s)
Lésion pulmonaire aigüe , Marqueurs biologiques , COVID-19 , Récepteur alpha des oestrogènes , Inhibiteur tissulaire de métalloprotéinase-1 , Lésion pulmonaire aigüe/génétique , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/sang , Animaux , Inhibiteur tissulaire de métalloprotéinase-1/génétique , Inhibiteur tissulaire de métalloprotéinase-1/sang , Inhibiteur tissulaire de métalloprotéinase-1/métabolisme , Femelle , Mâle , Humains , Souris , COVID-19/métabolisme , COVID-19/génétique , COVID-19/sang , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Marqueurs biologiques/sang , Marqueurs biologiques/métabolisme , Oestrogènes/sang , Adulte d'âge moyen , Sous-type H1N1 du virus de la grippe A , Poumon/métabolisme , SARS-CoV-2 , Adulte , Régulation de l'expression des gènes , Souris de lignée C57BL , Facteurs sexuels , Caractères sexuels , Infections à Orthomyxoviridae/métabolisme , Infections à Orthomyxoviridae/sang , Infections à Orthomyxoviridae/génétique
14.
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
16.
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
17.
J Extracell Vesicles ; 13(9): e12495, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39254228

RÉSUMÉ

Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have shown anti-inflammatory potential in multiple inflammatory diseases. In the March 2022 issue of the Journal of Extracellular Vesicles, it was shown that EVs from human MSCs can suppress severe acute respiratory distress syndrome, coronavirus 2 (SARS-CoV-2) replication and can mitigate the production and release of infectious virions. We therefore hypothesized that MSC-EVs have an anti-viral effect in SARS-CoV-2 infection in vivo. We extended this question to ask whether also other respiratory viral infections could be treated by MSC-EVs. Adipose stem cell-derived EVs (ASC-EVs) were isolated using tangential flow filtration from conditioned media obtained from a multi-flask cell culture system. The effects of the ASC-EVs were tested  in Vero E6 cells in vitro. ASC-EVs were also given i.v. to SARS-CoV-2 infected Syrian Hamsters, and H1N1 influenza virus infected mice. The ASC-EVs attenuated SARS-CoV-2 virus replication in Vero E6 cells and reduced body weight and signs of lung injury in infected Syrian hamsters. Furthermore, ASC-EVs increased the survival rate of influenza A-infected mice and attenuated signs of lung injury. In summary, this study suggests that ASC-EVs can have beneficial therapeutic effects in models of virus-infection-associated acute lung injury and may potentially be developed to treat lung injury in humans.


Sujet(s)
Lésion pulmonaire aigüe , COVID-19 , Vésicules extracellulaires , Sous-type H1N1 du virus de la grippe A , Cellules souches mésenchymateuses , SARS-CoV-2 , Animaux , Vésicules extracellulaires/métabolisme , Cellules souches mésenchymateuses/métabolisme , SARS-CoV-2/physiologie , COVID-19/thérapie , Lésion pulmonaire aigüe/thérapie , Lésion pulmonaire aigüe/virologie , Souris , Cellules Vero , Humains , Chlorocebus aethiops , Infections à Orthomyxoviridae/thérapie , Réplication virale , Mesocricetus , Modèles animaux de maladie humaine , Mâle , Grippe humaine/thérapie , Femelle
18.
J Immunotoxicol ; 21(1): 2400624, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39319829

RÉSUMÉ

Infections caused by the influenza virus lead to both epidemic and pandemic outbreaks in humans and animals. Owing to their rapid production, safety, and stability, DNA vaccines represent a promising avenue for eliciting immunity and thwarting viral infections. While DNA vaccines have demonstrated substantial efficacy in murine models, their effectiveness in larger animals remains subdued. This limitation may be addressed by augmenting the immunogenicity of DNA-based vaccines. In the investigation here, protein expression was enhanced via codon optimization and then mouse cytotoxic T-lymphocyte antigen 4 (CTLA-4) was harnessed as a modulatory adjunct to bind directly to antigen-presenting cells. Further, the study evaluated the immunogenicity of two variants of the hemagglutinin (HA) antigen, i.e. the full-length and the C-terminal deletion versions. The study findings revealed that the codon-optimized HA gene (pcHA) led to increased protein synthesis, as evidenced by elevated mRNA levels. Codon optimization also significantly bolstered both cellular and humoral immune responses. In cytokine assays, all plasmid constructs, particularly pCTLA4-cHA, induced robust interferon (IFN)-γ production, while interleukin (IL)-4 levels remained uniformly non-significant. Mice immunized with pcHA displayed an augmented presence of IFNγ+ T-cells, underscoring the enhanced potency of the codon-optimized HA vaccine. Contrarily, CTLA-4-fused DNA vaccines did not significantly amplify the immune response.


Sujet(s)
Antigène CTLA-4 , Codon , Glycoprotéine hémagglutinine du virus influenza , Vaccins antigrippaux , Infections à Orthomyxoviridae , Vaccins à ADN , Animaux , Vaccins à ADN/immunologie , Vaccins à ADN/génétique , Souris , Antigène CTLA-4/génétique , Antigène CTLA-4/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Codon/génétique , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Humains , Femelle , Souris de lignée BALB C , Modèles animaux de maladie humaine , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Grippe humaine/immunologie , Grippe humaine/prévention et contrôle , Sous-type H1N1 du virus de la grippe A/immunologie
19.
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
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE