Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.521
Filtrer
1.
Front Immunol ; 15: 1360698, 2024.
Article de Anglais | MEDLINE | ID: mdl-38979428

RÉSUMÉ

Regulatory T cells (Tregs) play a crucial and complex role in balancing the immune response to viral infection. Primarily, they serve to regulate the immune response by limiting the expression of proinflammatory cytokines, reducing inflammation in infected tissue, and limiting virus-specific T cell responses. But excessive activity of Tregs can also be detrimental and hinder the ability to effectively clear viral infection, leading to prolonged disease and potential worsening of disease severity. Not much is known about the impact of Tregs during severe influenza. In the present study, we show that CD4+/CD25+FoxP3+ Tregs are strongly involved in disease progression during influenza A virus (IAV) infection in mice. By comparing sublethal with lethal dose infection in vivo, we found that not the viral load but an increased number of CD4+/CD25+FoxP3+ Tregs may impair the immune response by suppressing virus specific CD8+ T cells and favors disease progression. Moreover, the transfer of induced Tregs into mice with mild disease symptoms had a negative and prolonged effect on disease outcome, emphasizing their importance for pathogenesis. Furthermore, treatment with MEK-inhibitors resulted in a significant reduction of induced Tregs in vitro and in vivo and positively influenced the progression of the disease. Our results demonstrate that CD4+/CD25+FoxP3+ Tregs are involved in the pathogenesis of severe influenza and indicate the potential of the MEK-inhibitor zapnometinib to modulate CD4+/CD25+FoxP3+ Tregs. Thus, making MEK-inhibitors even more promising for the treatment of severe influenza virus infections.


Sujet(s)
Virus de la grippe A , Infections à Orthomyxoviridae , Lymphocytes T régulateurs , Animaux , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/traitement médicamenteux , Souris , Virus de la grippe A/immunologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Femelle , Souris de lignée C57BL , Facteurs de transcription Forkhead/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Charge virale/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
2.
J Immunotoxicol ; 21(1): 2340495, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38946256

RÉSUMÉ

Per- and polyfluoroalkyl substances (PFAS) are anthropogenic organofluorine compounds that persist indefinitely in the environment and bioaccumulate throughout all trophic levels. Biomonitoring efforts have detected multiple PFAS in the serum of most people. Immune suppression has been among the most consistent effects of exposure to PFAS. PFAS often co-occur as mixtures in the environment, however, few studies have examined immunosuppression of PFAS mixtures or determined whether PFAS exposure affects immune function in the context of infection. In this study, mixtures containing two or four different PFAS and a mouse model of infection with influenza A virus (IAV) were used to assess immunotoxicity of PFAS mixtures. PFAS were administered via the drinking water as either a binary mixture of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) or quaternary mixture of PFOS, PFOA, perfluorohexane sulfonate (PFHxS), and perfluorononanoic acid (PFNA). The results indicated that the binary mixture affected the T-cell response, while the quaternary mixture affected the B-cell response to infection. These findings indicate that the immunomodulatory effects of PFAS mixtures are not simply additive, and that the sensitivity of immune responses to PFAS varies by cell type and mixture. The study also demonstrates the importance of studying adverse health effects of PFAS mixtures.


Sujet(s)
Acides alcanesulfoniques , Caprylates , Fluorocarbones , Virus de la grippe A , Infections à Orthomyxoviridae , Fluorocarbones/effets indésirables , Fluorocarbones/toxicité , Animaux , Souris , Virus de la grippe A/immunologie , Acides alcanesulfoniques/toxicité , Acides alcanesulfoniques/effets indésirables , Infections à Orthomyxoviridae/immunologie , Caprylates/toxicité , Caprylates/effets indésirables , Humains , Femelle , Souris de lignée C57BL , Grippe humaine/immunologie , Modèles animaux de maladie humaine , Lymphocytes T/immunologie , Lymphocytes T/effets des médicaments et des substances chimiques
3.
Front Immunol ; 15: 1376395, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975350

RÉSUMÉ

Influenza A Virus (IAV) and Respiratory Syncytial Virus (RSV) are both responsible for millions of severe respiratory tract infections every year worldwide. Effective vaccines able to prevent transmission and severe disease, are important measures to reduce the burden for the global health system. Despite the strong systemic immune responses induced upon current parental immunizations, this vaccination strategy fails to promote a robust mucosal immune response. Here, we investigated the immunogenicity and efficacy of a mucosal adenoviral vector vaccine to tackle both pathogens simultaneously at their entry site. For this purpose, BALB/c mice were immunized intranasally with adenoviral vectors (Ad) encoding the influenza-derived proteins, hemagglutinin (HA) and nucleoprotein (NP), in combination with an Ad encoding for the RSV fusion (F) protein. The mucosal combinatory vaccine induced neutralizing antibodies as well as local IgA responses against both viruses. Moreover, the vaccine elicited pulmonary CD8+ and CD4+ tissue resident memory T cells (TRM) against the immunodominant epitopes of RSV-F and IAV-NP. Furthermore, the addition of Ad-TGFß or Ad-CCL17 as mucosal adjuvant enhanced the formation of functional CD8+ TRM responses against the conserved IAV-NP. Consequently, the combinatory vaccine not only provided protection against subsequent infections with RSV, but also against heterosubtypic challenges with pH1N1 or H3N2 strains. In conclusion, we present here a potent combinatory vaccine for mucosal applications, which provides protection against two of the most relevant respiratory viruses.


Sujet(s)
Anticorps antiviraux , Immunité muqueuse , Virus de la grippe A , Vaccins antigrippaux , Souris de lignée BALB C , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Animaux , Souris , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Anticorps antiviraux/immunologie , Virus de la grippe A/immunologie , Femelle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Virus respiratoires syncytiaux/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Vaccins combinés/immunologie , Vaccins combinés/administration et posologie , Humains , Adenoviridae/immunologie , Adenoviridae/génétique , Vecteurs génétiques
4.
Arch Virol ; 169(8): 163, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990396

RÉSUMÉ

Antigenically divergent H7N9 viruses pose a potential threat to public health, with the poor immunogenicity of candidate H7N9 vaccines demonstrated in clinical trials underscoring the urgent need for more-effective H7N9 vaccines. In the present study, mice were immunized with various doses of a suspended-MDCK-cell-derived inactivated H7N9 vaccine, which was based on a low-pathogenic H7N9 virus, to assess cross-reactive immunity and cross-protection against antigenically divergent H7N9 viruses. We found that the CRX-527 adjuvant, a synthetic TLR4 agonist, significantly enhanced the humoral immune responses of the suspended-MDCK-cell-derived H7N9 vaccine, with significant antigen-sparing and immune-enhancing effects, including robust virus-specific IgG, hemagglutination-inhibiting (HI), neuraminidase-inhibiting (NI), and virus-neutralizing (VN) antibody responses, which are crucial for protection against influenza virus infection. Moreover, the CRX-527-adjuvanted H7N9 vaccine also elicited cross-protective immunity and cross-protection against a highly pathogenic H7N9 virus with a single vaccination. Notably, NI and VN antibodies might play an important role in cross-protection against lethal influenza virus infections. This study showed that a synthetic TLR4 agonist adjuvant has a potent immunopotentiating effect, which might be considered worth further development as a means of increasing vaccine effectiveness.


Sujet(s)
Anticorps antiviraux , Immunité humorale , Sous-type H7N9 du virus de la grippe A , Vaccins antigrippaux , Souris de lignée BALB C , Infections à Orthomyxoviridae , Récepteur de type Toll-4 , Vaccins inactivés , Animaux , Sous-type H7N9 du virus de la grippe A/immunologie , Récepteur de type Toll-4/agonistes , Récepteur de type Toll-4/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Souris , Anticorps antiviraux/immunologie , Chiens , Cellules rénales canines Madin-Darby , Vaccins inactivés/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Femelle , Anticorps neutralisants/immunologie , Protection croisée/immunologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Adjuvants vaccinaux , Immunoglobuline G/immunologie , Immunoglobuline G/sang
5.
Immunohorizons ; 8(7): 478-491, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39007717

RÉSUMÉ

IκB kinase (IKK)α controls noncanonical NF-κB signaling required for lymphoid organ development. We showed previously that lymph node formation is ablated in IkkαLyve-1 mice constitutively lacking IKKα in lymphatic endothelial cells (LECs). We now reveal that loss of IKKα in LECs leads to the formation of BALT in the lung. Tertiary lymphoid structures appear only in the lungs of IkkαLyve-1 mice and are not present in any other tissues, and these highly organized BALT structures form after birth and in the absence of inflammation. Additionally, we show that IkkαLyve-1 mice challenged with influenza A virus (IAV) exhibit markedly improved survival and reduced weight loss compared with littermate controls. Importantly, we determine that the improved morbidity and mortality of IkkαLyve-1 mice is independent of viral load and rate of clearance because both mice control and clear IAV infection similarly. Instead, we show that IFN-γ levels are decreased, and infiltration of CD8 T cells and monocytes into IkkαLyve-1 lungs is reduced. We conclude that ablating IKKα in LECs promotes BALT formation and reduces the susceptibility of IkkαLyve-1 mice to IAV infection through a decrease in proinflammatory stimuli.


Sujet(s)
Homéostasie , I-kappa B Kinase , Virus de la grippe A , Poumon , Infections à Orthomyxoviridae , Animaux , I-kappa B Kinase/métabolisme , I-kappa B Kinase/génétique , Souris , Poumon/immunologie , Poumon/virologie , Poumon/anatomopathologie , Infections à Orthomyxoviridae/immunologie , Virus de la grippe A/immunologie , Cellules endothéliales/immunologie , Cellules endothéliales/métabolisme , Lymphocytes T CD8+/immunologie , Souris de lignée C57BL , Souris knockout , Transduction du signal/immunologie , Interféron gamma/métabolisme
6.
Nat Commun ; 15(1): 5800, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987276

RÉSUMÉ

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


Sujet(s)
Administration par voie nasale , Protection croisée , Vaccins antigrippaux , Souris de lignée BALB C , Nanoparticules , Infections à Orthomyxoviridae , Animaux , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Nanoparticules/composition chimique , Femelle , Protection croisée/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Souris , Immunité muqueuse/immunologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , ARN messager/génétique , ARN messager/immunologie , Lipides/composition chimique , Anticorps antiviraux/immunologie , Humains , Immunisation/méthodes , Vaccination/méthodes , , Liposomes
7.
PLoS One ; 19(7): e0301664, 2024.
Article de Anglais | MEDLINE | ID: mdl-38985719

RÉSUMÉ

Influenza viruses constitute a major threat to human health globally. The viral surface glycoprotein hemagglutinin (HA) is the immunodominant antigen, contains the site for binding to the cellular receptor (RBS), and it is the major target of neutralizing antibody responses post-infection. We developed llama-derived single chain antibody fragments (VHHs) specific for type A influenza virus. Four VHHs were identified and further characterized. VHH D81 bound residues in the proximity of the C-terminal region of HA1 of H1 and H5 subtypes, and showed weak neutralizing activity, whereas VHH B33 bound residues in the proximity of the N-terminal region of the HA's stem domain (HA2) of H1, H5, and H9 subtypes, and showed no neutralizing activity. Of most relevance, VHHs E13 and G41 recognized highly conserved conformational epitopes on the H1 HA's globular domain (HA1) and showed high virus neutralizing activity (ranging between 0.94 to 0.01µM), when tested against several human H1N1 isolates. Additionally, E13 displayed abrogated virus replication of a panel of H1N1 strains spanning over 80 years of antigenic drift and isolated from human, avian, and swine origin. Interestingly, E13 conferred protection in vivo at a dose as low as 0.05 mg/kg. Mice treated with E13 intranasally resulted in undetectable virus challenge loads in the lungs at day 4 post-challenge. The transfer of sterilizing pan-H1 immunity, by a dose in the range of micrograms given intranasally, is of major significance for a monomeric VHH and supports the further development of E13 as an immunotherapeutic agent for the mitigation of influenza infections.


Sujet(s)
Anticorps neutralisants , Camélidés du Nouveau Monde , Glycoprotéine hémagglutinine du virus influenza , Sous-type H1N1 du virus de la grippe A , Infections à Orthomyxoviridae , Anticorps à domaine unique , Animaux , Glycoprotéine hémagglutinine du virus influenza/immunologie , Humains , Sous-type H1N1 du virus de la grippe A/immunologie , Anticorps à domaine unique/immunologie , Anticorps neutralisants/immunologie , Souris , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/virologie , Camélidés du Nouveau Monde/immunologie , Anticorps antiviraux/immunologie , Femelle , Grippe humaine/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/virologie , Épitopes/immunologie , Chiens , Souris de lignée BALB C
8.
Nat Commun ; 15(1): 5593, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961067

RÉSUMÉ

Human cases of avian influenza virus (AIV) infections are associated with an age-specific disease burden. As the influenza virus N2 neuraminidase (NA) gene was introduced from avian sources during the 1957 pandemic, we investigate the reactivity of N2 antibodies against A(H9N2) AIVs. Serosurvey of healthy individuals reveal the highest rates of AIV N2 antibodies in individuals aged ≥65 years. Exposure to the 1968 pandemic N2, but not recent N2, protected against A(H9N2) AIV challenge in female mice. In some older adults, infection with contemporary A(H3N2) virus could recall cross-reactive AIV NA antibodies, showing discernable human- or avian-NA type reactivity. Individuals born before 1957 have higher anti-AIV N2 titers compared to those born between 1957 and 1968. The anti-AIV N2 antibodies titers correlate with antibody titers to the 1957 N2, suggesting that exposure to the A(H2N2) virus contribute to this reactivity. These findings underscore the critical role of neuraminidase immunity in zoonotic and pandemic influenza risk assessment.


Sujet(s)
Anticorps antiviraux , Réactions croisées , Sous-type H3N2 du virus de la grippe A , Grippe humaine , Sialidase , Pandémies , Sialidase/immunologie , Sialidase/génétique , Animaux , Humains , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Sous-type H3N2 du virus de la grippe A/immunologie , Femelle , Réactions croisées/immunologie , Souris , Grippe humaine/immunologie , Grippe humaine/épidémiologie , Grippe humaine/virologie , Sujet âgé , Sous-type H2N2 du virus de la grippe A/immunologie , Sous-type H2N2 du virus de la grippe A/génétique , Mâle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/épidémiologie , Infections à Orthomyxoviridae/médecine vétérinaire , Oiseaux/virologie , Adulte d'âge moyen , Grippe chez les oiseaux/épidémiologie , Grippe chez les oiseaux/immunologie , Grippe chez les oiseaux/virologie , Sous-type H9N2 du virus de la grippe A/immunologie , Adulte , Protéines virales/immunologie , Protéines virales/génétique
9.
Curr Microbiol ; 81(9): 267, 2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39003673

RÉSUMÉ

In this study, we evaluated the impact of human gut microbiota on the immune pathways in the respiratory tract using a gnotobiotic (Gn) piglet model. We humanized piglets with rural and urban infant fecal microbiota (RIFM and UIFM, respectively) and then infected them with a H1N1 swine influenza virus. We analyzed the microbial diversity and structure of the intestinal and respiratory tracts of the piglets before and after the influenza virus infection and measured the viral load and immune responses. We found that the viral load in the upper respiratory tract of UIFM transplanted piglets was higher than their rural cohorts (RIFM), while virus-specific antibody responses were comparable. The relative cytokine gene expression in the tracheobronchial (respiratory tract) and mesenteric (gastrointestinal) lymph nodes, lungs, blood, and spleen of RIFM and UIFM piglets revealed a trend in reciprocal regulation of proinflammatory, innate, and adaptive immune-associated cytokines as well as the frequency of T-helper/memory cells, cytotoxic T cells, and myeloid immune cell subsets. We also observed different phylum-level shifts of the fecal microbiota in response to influenza virus infection between the two piglet groups, suggesting the potential impact of the gut microbiota on the immune responses to influenza virus infection and lung microbiota. In conclusion, Gn piglets humanized with diverse infant fecal microbiota had differential immune regulation, with UIFM favoring the activation of proinflammatory immune mediators following an influenza virus infection compared to their rural RIFM cohorts. Furthermore, Gn piglets can be a useful model in investigating the impact of diverse human microbiota of the gastrointestinal tract, probably also the respiratory tract, on respiratory health and testing specific probiotic- or prebiotic-based therapeutics.


Sujet(s)
Cytokines , Modèles animaux de maladie humaine , Fèces , Microbiome gastro-intestinal , Axénie , Immunité muqueuse , Sous-type H1N1 du virus de la grippe A , Animaux , Suidae , Fèces/microbiologie , Fèces/virologie , Humains , Sous-type H1N1 du virus de la grippe A/immunologie , Cytokines/métabolisme , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Charge virale , Nourrisson , Grippe humaine/immunologie , Grippe humaine/microbiologie , Grippe humaine/virologie
10.
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
11.
Viruses ; 16(6)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38932148

RÉSUMÉ

The devastating effects of COVID-19 have highlighted the importance of prophylactic and therapeutic strategies to combat respiratory diseases. Stimulator of interferon gene (STING) is an essential component of the host defense mechanisms against respiratory viral infections. Although the role of the cGAS/STING signaling axis in the innate immune response to DNA viruses has been thoroughly characterized, mounting evidence shows that it also plays a key role in the prevention of RNA virus infections. In this study, we investigated the role of STING activation during Influenza virus (IFV) infection. In both mouse bone marrow-derived macrophages and monocytic cell line THP-1 differentiated with PMA, we found that dimeric amidobenzimidazole (diABZI), a STING agonist, had substantial anti-IFV activity against multiple strains of IFV, including A/H1N1, A/H3N2, B/Yamagata, and B/Victoria. On the other hand, a pharmacological antagonist of STING (H-151) or the loss of STING in human macrophages leads to enhanced viral replication but suppressed IFN expression. Furthermore, diABZI was antiviral against IFV in primary air-liquid interface cultures of nasal epithelial cells. Our data suggest that STING agonists may serve as promising therapeutic antiviral agents to combat IFV.


Sujet(s)
Antiviraux , Immunité innée , Macrophages , Protéines membranaires , Animaux , Humains , Immunité innée/effets des médicaments et des substances chimiques , Souris , Antiviraux/pharmacologie , Macrophages/immunologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/virologie , Protéines membranaires/agonistes , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Cellules THP-1 , Réplication virale/effets des médicaments et des substances chimiques , Grippe humaine/immunologie , Grippe humaine/virologie , Grippe humaine/traitement médicamenteux , Chiens , Souris de lignée C57BL , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/traitement médicamenteux , Infections à Orthomyxoviridae/virologie , Orthomyxoviridae/effets des médicaments et des substances chimiques , Orthomyxoviridae/immunologie , Orthomyxoviridae/physiologie , Benzimidazoles/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
12.
Viruses ; 16(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38932155

RÉSUMÉ

COVID-19 is a spectrum of clinical symptoms in humans caused by infection with SARS-CoV-2. The coalescence of SARS-CoV-2 with seasonal respiratory viruses, particularly influenza viruses, is a global health concern. To understand this, transgenic mice expressing the human ACE2 receptor (K18-hACE2) were infected with influenza A virus (IAV) followed by SARS-CoV-2 and the host response and effect on virus biology was compared to K18-hACE2 mice infected with IAV or SARS-CoV-2 alone. The sequentially infected mice showed reduced SARS-CoV-2 RNA synthesis, yet exhibited more rapid weight loss, more severe lung damage and a prolongation of the innate response compared to the singly infected or control mice. Sequential infection also exacerbated the extrapulmonary encephalitic manifestations associated with SARS-CoV-2 infection. Conversely, prior infection with a commercially available, multivalent live-attenuated influenza vaccine (Fluenz Tetra) elicited the same reduction in SARS-CoV-2 RNA synthesis, albeit without the associated increase in disease severity. This suggests that the innate immune response stimulated by IAV inhibits SARS-CoV-2. Interestingly, infection with an attenuated, apathogenic influenza vaccine does not result in an aberrant immune response and enhanced disease severity. Taken together, the data suggest coinfection ('twinfection') is deleterious and mitigation steps should be instituted as part of the comprehensive public health and management strategy of COVID-19.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Modèles animaux de maladie humaine , Virus de la grippe A , Souris transgéniques , Infections à Orthomyxoviridae , SARS-CoV-2 , Animaux , COVID-19/immunologie , COVID-19/virologie , Souris , SARS-CoV-2/immunologie , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/immunologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Humains , Co-infection/virologie , Poumon/virologie , Poumon/anatomopathologie , Encéphalite virale/virologie , Encéphalite virale/immunologie , Vaccins antigrippaux/immunologie , Femelle , Immunité innée
13.
Viruses ; 16(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38932247

RÉSUMÉ

Influenza A virus (IAV) infections in swine are usually subclinical, but they can reach high morbidity rates. The mortality rate is normally low. In this study, six vaccinated, spontaneously deceased sows revealed IAV infection and enhanced neutrophilic bronchopneumonia with unexpectedly large numbers of infiltrating eosinophils. The purpose of this study was to characterize these lung lesions with special emphasis on the phenotypes of inflammatory cells, the presence of eosinophilic peroxidase (EPO), and neutrophil extracellular traps (NETs). The number of Sirius red-stained eosinophils was significantly higher in the lungs of IAV-infected sows compared to healthy pigs, indicating a migration of eosinophils from blood vessels into the lung tissue stimulated by IAV infection. The detection of intra- and extracellular EPO in the lungs suggests its contribution to pulmonary damage. The presence of CD3+ T lymphocytes, CD20+ B lymphocytes, and Iba-1+ macrophages indicates the involvement of cell-mediated immune responses in disease progression. Furthermore, high numbers of myeloperoxidase-positive cells were detected. However, DNA-histone-1 complexes were reduced in IAV-infected sows, leading to the hypothesis that NETs are not formed in the IAV-infected sows. In conclusion, our findings in the lungs of IAV-infected vaccinated sows suggest the presence of so far unreported field cases of vaccine-associated enhanced respiratory disease.


Sujet(s)
Virus de la grippe A , Vaccins antigrippaux , Poumon , Infections à Orthomyxoviridae , Maladies des porcs , Animaux , Suidae , Poumon/anatomopathologie , Poumon/virologie , Poumon/immunologie , Maladies des porcs/virologie , Maladies des porcs/immunologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/médecine vétérinaire , Femelle , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Virus de la grippe A/immunologie , Épidémies de maladies/médecine vétérinaire , Granulocytes éosinophiles/immunologie , Pièges extracellulaires/immunologie , Vaccination/médecine vétérinaire , Eosinophil Peroxidase/métabolisme
14.
Front Immunol ; 15: 1361323, 2024.
Article de Anglais | MEDLINE | ID: mdl-38835763

RÉSUMÉ

Introduction: Swine influenza viruses (SIVs) pose significant economic losses to the pig industry and are a burden on global public health systems. The increasing complexity of the distribution and evolution of different serotypes of influenza strains in swine herds escalates the potential for the emergence of novel pandemic viruses, so it is essential to develop new vaccines based on swine influenza. Methods: Here, we constructed a self-assembling ferritin nanoparticle vaccine based on the hemagglutinin (HA) extracellular domain of swine influenza A (H1N1) virus using insect baculovirus expression vector system (IBEVS), and after two immunizations, the immunogenicities and protective efficacies of the HA-Ferritin nanoparticle vaccine against the swine influenza virus H1N1 strain in mice and piglets were evaluated. Results: Our results demonstrated that HA-Ferritin nanoparticle vaccine induced more efficient immunity than traditional swine influenza vaccines. Vaccination with the HA-Ferritin nanoparticle vaccine elicited robust hemagglutinin inhibition titers and antigen-specific IgG antibodies and increased cytokine levels in serum. MF59 adjuvant can significantly promote the humoral immunity of HA-Ferritin nanoparticle vaccine. Furthermore, challenge tests showed that HA-Ferritin nanoparticle vaccine conferred full protection against lethal challenge with H1N1 virus and significantly decreased the severity of virus-associated lung lesions after challenge in both BALB/c mice and piglets. Conclusion: Taken together, these results indicate that the hemagglutinin extracellular-based ferritin nanoparticle vaccine may be a promising vaccine candidate against SIVs infection.


Sujet(s)
Anticorps antiviraux , Ferritines , Glycoprotéine hémagglutinine du virus influenza , Sous-type H1N1 du virus de la grippe A , Vaccins antigrippaux , Souris de lignée BALB C , Nanoparticules , Infections à Orthomyxoviridae , Animaux , Sous-type H1N1 du virus de la grippe A/immunologie , Ferritines/immunologie , Vaccins antigrippaux/immunologie , Suidae , Souris , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Maladies des porcs/virologie , Femelle ,
15.
Sci Rep ; 14(1): 13800, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877101

RÉSUMÉ

Adjuvants enhance, prolong, and modulate immune responses by vaccine antigens to maximize protective immunity and enable more effective immunization in the young and elderly. Most adjuvants are formulated with injectable vaccines. However, an intranasal route of vaccination may induce mucosal and systemic immune responses for enhancing protective immunity in individuals and be easier to administer compared to injectable vaccines. In this study, a next generation of broadly-reactive influenza hemagglutinin (HA) vaccines were developed using the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology. These HA vaccines were formulated with Mastoparan 7 (M7-NH2) mast cell degranulating peptide adjuvant and administered intranasally to determine vaccine-induced seroconversion of antibodies against a panel of influenza viruses and protection following infection with H1N1 and H3N2 viruses in mice. Mice vaccinated intranasally with M7-NH2-adjuvanted COBRA HA vaccines had high HAIs against a panel of H1N1 and H3N2 influenza viruses and were protected against both morbidity and mortality, with reduced viral lung titers, following challenge with an H1N1 influenza virus. Additionally, M7-NH2 adjuvanted COBRA HA vaccines induced Th2 skewed immune responses with robust IgG and isotype antibodies in the serum and mucosal lung lavages. Overall, this intranasally delivered M7-NH2 -adjuvanted COBRA HA vaccine provides effective protection against drifted H1N1 and H3N2 viruses.


Sujet(s)
Adjuvants immunologiques , Administration par voie nasale , Anticorps antiviraux , Glycoprotéine hémagglutinine du virus influenza , Sous-type H1N1 du virus de la grippe A , Sous-type H3N2 du virus de la grippe A , Vaccins antigrippaux , Infections à Orthomyxoviridae , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Animaux , Souris , Glycoprotéine hémagglutinine du virus influenza/immunologie , Sous-type H1N1 du virus de la grippe A/immunologie , Sous-type H3N2 du virus de la grippe A/immunologie , Adjuvants immunologiques/administration et posologie , Anticorps antiviraux/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Femelle , Souris de lignée BALB C , Protéines et peptides de signalisation intercellulaire/immunologie , Adjuvants vaccinaux/administration et posologie
16.
Hum Vaccin Immunother ; 20(1): 2356269, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38826029

RÉSUMÉ

The influenza viruses cause seasonal respiratory illness that affect millions of people globally every year. Prophylactic vaccines are the recommended method to prevent the breakout of influenza epidemics. One of the current commercial influenza vaccines consists of inactivated viruses that are selected months prior to the start of a new influenza season. In many seasons, the vaccine effectiveness (VE) of these vaccines can be relatively low. Therefore, there is an urgent need to develop an improved, more universal influenza vaccine (UIV) that can provide broad protection against various drifted strains in all age groups. To meet this need, the computationally optimized broadly reactive antigen (COBRA) methodology was developed to design a hemagglutinin (HA) molecule as a new influenza vaccine. In this study, COBRA HA-based inactivated influenza viruses (IIV) expressing the COBRA HA from H1 or H3 influenza viruses were developed and characterized for the elicitation of immediate and long-term protective immunity in both immunologically naïve or influenza pre-immune animal models. These results were compared to animals vaccinated with IIV vaccines expressing wild-type H1 or H3 HA proteins (WT-IIV). The COBRA-IIV elicited long-lasting broadly reactive antibodies that had hemagglutination-inhibition (HAI) activity against drifted influenza variants.


Sujet(s)
Anticorps antiviraux , Glycoprotéine hémagglutinine du virus influenza , Vaccins antigrippaux , Infections à Orthomyxoviridae , Vaccins inactivés , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Animaux , Vaccins inactivés/immunologie , Vaccins inactivés/administration et posologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Glycoprotéine hémagglutinine du virus influenza/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie , Souris , Femelle , Souris de lignée BALB C , Humains , Grippe humaine/prévention et contrôle , Grippe humaine/immunologie , , Tests d'inhibition de l'hémagglutination
17.
Nat Commun ; 15(1): 5025, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38871701

RÉSUMÉ

Influenza A viruses in swine have considerable genetic diversity and continue to pose a pandemic threat to humans due to a potential lack of population level immunity. Here we describe a pipeline to characterize and triage influenza viruses for their pandemic risk and examine the pandemic potential of two widespread swine origin viruses. Our analysis reveals that a panel of human sera collected from healthy adults in 2020 has no cross-reactive neutralizing antibodies against a α-H1 clade strain (α-swH1N2) but do against a γ-H1 clade strain. The α-swH1N2 virus replicates efficiently in human airway cultures and exhibits phenotypic signatures similar to the human H1N1 pandemic strain from 2009 (H1N1pdm09). Furthermore, α-swH1N2 is capable of efficient airborne transmission to both naïve ferrets and ferrets with prior seasonal influenza immunity. Ferrets with H1N1pdm09 pre-existing immunity show reduced α-swH1N2 viral shedding and less severe disease signs. Despite this, H1N1pdm09-immune ferrets that became infected via the air can still onward transmit α-swH1N2 with an efficiency of 50%. These results indicate that this α-swH1N2 strain has a higher pandemic potential, but a moderate level of impact since there is reduced replication fitness and pathology in animals with prior immunity.


Sujet(s)
Furets , Sous-type H1N1 du virus de la grippe A , Sous-type H1N2 du virus de la grippe A , Grippe humaine , Infections à Orthomyxoviridae , Pandémies , Animaux , Furets/virologie , Humains , Suidae , Grippe humaine/virologie , Grippe humaine/épidémiologie , Grippe humaine/immunologie , Grippe humaine/sang , Grippe humaine/transmission , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/épidémiologie , Infections à Orthomyxoviridae/transmission , Infections à Orthomyxoviridae/sang , Sous-type H1N1 du virus de la grippe A/immunologie , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/isolement et purification , Sous-type H1N2 du virus de la grippe A/génétique , Sous-type H1N2 du virus de la grippe A/immunologie , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Maladies des porcs/virologie , Maladies des porcs/épidémiologie , Maladies des porcs/immunologie , Maladies des porcs/transmission , Maladies des porcs/sang , Femelle , Excrétion virale , Mâle , Adulte , Réplication virale
18.
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
19.
Cell Death Differ ; 31(7): 924-937, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38849575

RÉSUMÉ

Mitochondria react to infection with sub-lethal signals in the apoptosis pathway. Mitochondrial signals can be inflammatory but mechanisms are only partially understood. We show that activation of the caspase-activated DNase (CAD) mediates mitochondrial pro-inflammatory functions and substantially contributes to host defense against viral infection. In cells lacking CAD, the pro-inflammatory activity of sub-lethal signals was reduced. Experimental activation of CAD caused transient DNA-damage and a pronounced DNA damage response, involving major kinase signaling pathways, NF-κB and cGAS/STING, driving the production of interferon, cytokines/chemokines and attracting neutrophils. The transcriptional response to CAD-activation was reminiscent of the reaction to microbial infection. CAD-deficient cells had a diminished response to viral infection. Influenza virus infected CAD-deficient mice displayed reduced inflammation in lung tissue, higher viral titers and increased weight loss. Thus, CAD links the mitochondrial apoptosis system and cell death caspases to host defense. CAD-driven DNA damage is a physiological element of the inflammatory response to infection.


Sujet(s)
Altération de l'ADN , Inflammation , Mitochondries , Animaux , Inflammation/anatomopathologie , Inflammation/métabolisme , Souris , Mitochondries/métabolisme , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/anatomopathologie , Infections à Orthomyxoviridae/métabolisme , Souris de lignée C57BL , Apoptose , Humains , Facteur de transcription NF-kappa B/métabolisme , Désoxyribonucléases/métabolisme , Désoxyribonucléases/génétique , Souris knockout , Transduction du signal , Endodeoxyribonucleases/métabolisme , Endodeoxyribonucleases/génétique , Endodeoxyribonucleases/déficit , Nucleotidyltransferases
20.
Toxicol Appl Pharmacol ; 489: 117010, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38901696

RÉSUMÉ

Humoral responses to respiratory viruses, such as influenza viruses, develop over time and are central to protection from repeated infection with the same or similar viruses. Epidemiological and experimental studies have linked exposures to environmental contaminants that bind the aryl hydrocarbon receptor (AHR) with modulated antibody responses to pathogenic microorganisms and common vaccinations. Other studies have prompted investigation into the potential therapeutic applications of compounds that activate AHR. Herein, using two different AHR ligands [2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester (ITE), to modulate the duration of AHR activity, we show that the humoral response to viral infection is dependent upon the duration and timing of AHR signaling, and that different cellular elements of the response have different sensitivities. When AHR activation was initiated prior to infection with influenza A virus, there was suppression of all measured elements of the humoral response (i.e., the frequency of T follicular helper cells, germinal center B cells, plasma cells, and circulating virus-specific antibody). However, when the timing of AHR activation was adjusted to either early (days -1 to +5 relative to infection) or later (days +5 onwards), then AHR activation affected different aspects of the overall humoral response. These findings highlight the importance of considering the timing of AHR activation in relation to triggering an immune response, particularly when targeting the AHR to manipulate disease processes.


Sujet(s)
Immunité humorale , Dibenzodioxines polychlorées , Récepteurs à hydrocarbure aromatique , Récepteurs à hydrocarbure aromatique/métabolisme , Animaux , Immunité humorale/effets des médicaments et des substances chimiques , Dibenzodioxines polychlorées/toxicité , Femelle , Facteurs temps , Souris , Souris de lignée C57BL , Indoles/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Anticorps antiviraux , Infections à Orthomyxoviridae/immunologie , Infections de l'appareil respiratoire/immunologie , Infections de l'appareil respiratoire/virologie , Infections de l'appareil respiratoire/métabolisme , Ligands , Thiazoles
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...