Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.984
Filter
1.
Nat Commun ; 15(1): 8712, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379405

ABSTRACT

Influenza viruses cause substantial morbidity and mortality every year despite seasonal vaccination. mRNA-based vaccines have the potential to elicit more protective immune responses, but for maximal breadth and durability, it is desirable to deliver both the viral hemagglutinin and neuraminidase glycoproteins. Delivering multiple antigens individually, however, complicates manufacturing and increases cost, thus it would be beneficial to express both proteins from a single mRNA. Here, we develop an mRNA genetic configuration that allows the simultaneous expression of unmodified, full-length NA and HA proteins from a single open reading frame. We apply this approach to glycoproteins from contemporary influenza A and B viruses and, after vaccination, observe high levels of functional antibodies and protection from disease in female mouse and male ferret challenge models. This approach may further efforts to utilize mRNA technology to improve seasonal vaccine efficacy by efficiently delivering multiple viral antigens simultaneously and in their native state.


Subject(s)
Antibodies, Viral , Ferrets , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , RNA, Messenger , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Female , Mice , Male , Neuraminidase/immunology , Neuraminidase/genetics , Antibodies, Viral/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Mice, Inbred BALB C , Influenza B virus/immunology , Influenza B virus/genetics , Influenza A virus/immunology , Influenza A virus/genetics , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Glycoproteins/immunology , Glycoproteins/genetics , Viral Proteins/immunology , Viral Proteins/genetics , Antigens, Viral/immunology , Antigens, Viral/genetics , Vaccination/methods
2.
J Med Virol ; 96(10): e29858, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39370830

ABSTRACT

The COVID-19 pandemic had a significant impact on the global influenza vaccination and the epidemics of seasonal influenza. To further explore the molecular epidemiology of influenza viruses and assess vaccine effectiveness, we collected influenza cases in Wuhan during the 2022-2023 influenza season. Among 1312 clinical samples, 312 samples tested positive for influenza viruses using reverse transcription polymerase chain reaction. These positive samples included 146A/H1N1 subtypes (46.8%), 164A/H3N2 subtypes (52.6%) and 2 influenza B virus types (0.6%). Based on the whole genome sequence information of hemagglutinin (HA) and neuraminidase (NA) from 27A/H1N1 influenza virus strains and 26A/H3N2 influenza virus strains obtained in this study, a phylogenetic analysis was conducted. The analysis revealed that all A/H1N1 strains belonged to the evolutionary branch 6B.1A.5a.2a, and they exhibited specific substitutions at positions K71Q, Q206E, E241A, and R276K. Similarly, all A/H3N2 strains were classified into the 3C.2a1b.2a.1a subclade and displayed amino acid substitutions at positions S172H, N175Y, I176T, K187N, and S214P. Notably, the A/H3N2 strains also acquired a new potential glycosylation site at position N174. Using an epitope model, the predicted vaccine effectiveness was assessed for the A/H1N1 and A/H3N2 strains. The predicted vaccine effectiveness against the Wuhan influenza epidemic strain was over 85% for the A/H1N1 vaccine strain. However, the effectiveness against the A/H3N2 vaccine strain was only 48.7%. To further verify the protection of influenza vaccine against circulating influenza viruses in the region, we conducted in vivo and in vitro animal studies. The results of in vitro neutralization experiment showed that rabbit serum antibodies inoculated with quadrivalent isolated influenza vaccine had neutralization ability against all 24 isolated influenza viruses. In vivo experiments showed that vaccinated mice had fewer lung lesions when infected with the influenza strain circulating in Wuhan, suggesting that vaccination can effectively reduce the occurrence of severe lung damage. These findings emphasize the importance of accurately predicting seasonal influenza strains for effective influenza prevention and control, especially during the co-circulation of SARS-CoV-2 and influenza viruses. This study provides valuable information on the seasonal influenza virus in Wuhan during the COVID-19 pandemic and serves as a basis for vaccine prediction and updates.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Influenza, Human , Molecular Epidemiology , Phylogeny , China/epidemiology , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Influenza, Human/virology , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/virology , COVID-19/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Animals , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/classification , Mice , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/classification , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Neuraminidase/genetics , Neuraminidase/immunology , Antibodies, Viral/blood , Mice, Inbred BALB C , Seasons , Vaccine Efficacy , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/classification , COVID-19 Vaccines/immunology
3.
PLoS Biol ; 22(9): e3002800, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39316608

ABSTRACT

Bats are reservoirs of many zoonotic viruses that are fatal in humans but do not cause disease in bats. Moreover, bats generate low neutralizing antibody titers in response to experimental viral infection, although more robust antibody responses have been observed in wild-caught bats during times of food stress. Here, we compared the antibody titers and B cell receptor (BCR) diversity of Jamaican fruit bats (Artibeus jamaicensis; JFBs) and BALB/c mice generated in response to T-dependent and T-independent antigens. We then manipulated the diet of JFBs and challenged them with H18N11 influenza A-like virus or a replication incompetent Nipah virus VSV (Nipah-riVSV). Under standard housing conditions, JFBs generated a lower avidity antibody response and possessed more BCR mRNA diversity compared to BALB/c mice. However, withholding protein from JFBs improved serum neutralization in response to Nipah-riVSV and improved serum antibody titers specific to H18 but reduced BCR mRNA diversity.


Subject(s)
Antibodies, Viral , Chiroptera , Mice, Inbred BALB C , Animals , Chiroptera/immunology , Chiroptera/virology , Mice , Antibodies, Viral/immunology , Nipah Virus/immunology , Antibody Formation/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Diet, Protein-Restricted , Antibodies, Neutralizing/immunology , Antibody Affinity , Influenza A virus/immunology , Female , Antibody Diversity
4.
Immun Inflamm Dis ; 12(9): e70021, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39291404

ABSTRACT

BACKGROUND: Mycoplasma pneumoniae (MP) is a common respiratory pathogen affecting the longevity of the elderly and the health of children. However, the human vaccine against MP has not been successfully developed till now due to the poor immunogenicity and side effects of MP inactivated or attenuated vaccine. Therefore, it is necessary to develop a MP genetic engineering vaccine with influenza virus strain as vector. METHODS: In this study, the major antigen genes P1a of MP adhesion factor P1(3862-4554 bases) and P30a of P30(49-822 bases) were inserted into the nonstructural protein (NS) gene of Influenza A virus strain A/Puerto Rio/8/34(H1N1), PR8 for short, to construct the recombinant vectors NS-P1a or NS-P30a. The recombinant pHW2000 plasmids containing NS-P1a or NS-P30a were cotransfected with the rest 7 fragments of PR8 into HEK293T cells. After inoculating chicken embryos, the recombinant influenza viruses rFLU-P1a and rFLU-P30a were rescued. RT-PCR and sequencing were used to identify the recombinant viruses. The hemagglutination titers of rFLU-P1a and rFLU-P30a were determined after five successive generations in chicken embryos so as to indicate the genetic stability of the recombinant viruses. The morphology of recombinant influenza viruses was observed under electron microscopy. RESULTS: P1a or P30a was designed to be inserted into the modified NS gene sequence separately and synthesized successfully. RT-PCR identification of the recombinant viruses rFLU-P1a and rFLU-P30a showed that P1a (693 bp), P30a (774 bp), NS-P1a (1992bp) and NS-P30a (2073 bp) bands were found, and the sequencing results were correct. After five successive generations, each virus generation has a certain hemagglutination titer (from 1:32 to 1:64), and the band of P1a or P30a can be seen in the corresponding positions. The virus particles under the electron microscope appeared as spheres or long strips connected by several particles, revealing a complete viral membrane structure composed of virus lipid bilayer, hemagglutinin, neuraminidase, and matrix proteins. CONCLUSION: The recombinant viruses rFLU-P1a and rFLU-P30a which carried the advantaged immune regions of the P1 and P30 genes in MP were successfully constructed and identified. And the genetic stability of rFLU-P1a or rFLU-P30a was relatively high. The typical and complete morphology of influenza virus was observed under the electron microscope. Our research provided a foundation for the further development of MP vaccines for human.


Subject(s)
Genetic Vectors , Mycoplasma pneumoniae , Humans , Mycoplasma pneumoniae/genetics , Mycoplasma pneumoniae/immunology , Animals , HEK293 Cells , Genetic Vectors/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Influenza A virus/genetics , Influenza A virus/immunology , Chick Embryo , Pneumonia, Mycoplasma/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/genetics
5.
Viruses ; 16(9)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39339851

ABSTRACT

The emergence and spread of highly pathogenic avian influenza virus A subtype H5N1 (HP H5N1-IAV), particularly clade H5N1 2.3.4.4b, pose a severe global health threat, affecting various species, including mammals. Historically, cattle have been considered less susceptible to IAV, but recent outbreaks of H5N1-IAV 2.3.4.4b in dairy farms suggest a shift in host tropism, underscoring the urgency of expanded surveillance and the need for adaptable diagnostic tools in outbreak management. This study investigated the presence of anti-nucleoprotein (NP) antibodies in serum and milk and viral RNA in milk on dairy farms affected by outbreaks in Texas, Kansas, and Michigan using a multi-species IAV ELISA and RT-qPCR. The analysis of ELISA results from a Michigan dairy farm outbreak demonstrated a positive correlation between paired serum and milk sample results, confirming the reliability of both specimen types. Our findings also revealed high diagnostic performance during the convalescent phase (up to 96%), further improving sensitivity through serial sampling. Additionally, the evaluation of diagnostic specificity using serum and milk samples from IAV-free farms showed an excellent performance (99.6%). This study underscores the efficacy of the IAV NP-blocking ELISA for detecting and monitoring H5N1-IAV 2.3.4.4b circulation in dairy farms, whose recent emergence raises significant animal welfare and zoonotic concerns, necessitating expanded surveillance efforts.


Subject(s)
Cattle Diseases , Disease Outbreaks , Milk , Orthomyxoviridae Infections , Animals , Cattle , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/diagnosis , Disease Outbreaks/veterinary , Milk/virology , Cattle Diseases/epidemiology , Cattle Diseases/virology , Cattle Diseases/diagnosis , Antibodies, Viral/blood , Influenza A virus/isolation & purification , Influenza A virus/genetics , Influenza A virus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/veterinary , United States/epidemiology , RNA, Viral/genetics , Dairying , Female
6.
Sci Adv ; 10(39): eado7087, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331716

ABSTRACT

Pregnancy is associated with profound changes in immunity. However, pregnancy-related respiratory immune adaptations in response to influenza infection and their impact on disease severity remain unclear. Here, we describe, in a preclinical model of mid-gestation pregnancy, a mechanism of enhanced host defense against influenza A virus (IAV) localized to the nasal cavity that limits viral replication and reduces the magnitude of intrapulmonary immune responses. Consequently, the pregnant mice show reduced pulmonary pathology and preserved airway function after IAV infection. The early restriction of viral replication is independent of type I interferon (IFN) but dependent on increased antimicrobial peptides (AMPs) driven by interleukin-17+ (IL-17+) γδ+ T cells within the nasal passages. This pathway of host defense against IAV infection in the upper airways during pregnancy restricts early viral infection and prevents virus dissemination into the lung supporting maternal fitness.


Subject(s)
Influenza A virus , Interferon Type I , Interleukin-17 , Nasal Mucosa , Orthomyxoviridae Infections , Animals , Female , Pregnancy , Interleukin-17/metabolism , Interleukin-17/immunology , Mice , Interferon Type I/metabolism , Interferon Type I/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Nasal Mucosa/immunology , Nasal Mucosa/virology , Nasal Mucosa/metabolism , Influenza A virus/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Virus Replication , Lung/immunology , Lung/virology
7.
Nanotechnology ; 35(50)2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39321818

ABSTRACT

A simple and rapid system based on Raman nanosphere (R-Sphere) immunochromatography was developed in this study for the simultaneous detection of Influenza A, B virus antigens on a single test line (T-line). Two types of R-Sphere with different characteristic Raman spectrum were used as the signal source, which were labeled with monoclonal antibodies against FluA, FluB (tracer antibodies), respectively. A mixture of antibodies containing anti-FluA monoclonal antibody and anti-FluB monoclonal antibody (capture antibody) was sprayed on a single T-line and goat anti-chicken IgY antibody was coated as a C-line, and the antigen solution with known concentration was detected by the strip of lateral flow immunochromatography based on surface-enhanced Raman spectroscopy (SERS). The T-line was scanned with a Raman spectrometer and SERS signals were collected. Simultaneous specific recognition and detection of FluA and FluB were achieved on a single T-line by analyzing the SERS signals. The findings indicated that the test system could identify FluA and FluB in a qualitative manner in just 15 minutes, with a minimum detection threshold of 0.25 ng ml-1, excellent consistency, and specificity. There was no interference with the other four respiratory pathogens, and it exhibited 8 times greater sensitivity compared to the colloidal gold test strip method. The assay system is rapid, sensitive, and does not require repetitive sample pretreatment steps and two viruses can be detected simultaneously on a single T-line by titrating one sample, which improves detection efficiency, and provide a reference for developing multiplexed detection techniques for other respiratory viruses.


Subject(s)
Antigens, Viral , Influenza A virus , Influenza B virus , Nanospheres , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Influenza A virus/isolation & purification , Influenza A virus/immunology , Antigens, Viral/analysis , Antigens, Viral/immunology , Nanospheres/chemistry , Influenza B virus/immunology , Influenza B virus/isolation & purification , Chromatography, Affinity/methods , Chromatography, Affinity/instrumentation , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Animals , Humans
8.
Front Immunol ; 15: 1374838, 2024.
Article in English | MEDLINE | ID: mdl-39281683

ABSTRACT

Dendritic cells (DCs) are professional antigen-presenting cells, which are key components of the immune system and involved in early immune responses. DCs are specialized in capturing, processing, and presenting antigens to facilitate immune interactions. Chickens infected with avian influenza virus (AIV) demonstrate a wide range of clinical symptoms, based on pathogenicity of the virus. Low pathogenic avian influenza (LPAI) viruses typically induce mild clinical signs, whereas high pathogenic avian influenza (HPAI) induce more severe disease, which can lead to death. For this study, chicken bone marrow-derived DC (ckBM-DC)s were produced and infected with high and low pathogenic avian influenza viruses of H5N2 or H7N3 subtypes to characterize innate immune responses, study effect on cell morphologies, and evaluate virus replication. A strong proinflammatory response was observed at 8 hours post infection, via upregulation of chicken interleukin-1ß and stimulation of the interferon response pathway. Microscopically, the DCs underwent morphological changes from classic elongated dendrites to a more general rounded shape that eventually led to cell death with the presence of scattered cellular debris. Differences in onset of morphologic changes were observed between H5 and H7 subtypes. Increases in viral titers demonstrated that both HPAI and LPAI are capable of infecting and replicating in DCs. The increase in activation of infected DCs may be indicative of a dysregulated immune response typically seen with HPAI infections.


Subject(s)
Chickens , Cytokines , Dendritic Cells , Influenza in Birds , Animals , Dendritic Cells/immunology , Dendritic Cells/virology , Chickens/virology , Influenza in Birds/immunology , Influenza in Birds/virology , Influenza in Birds/pathology , Cytokines/metabolism , Cytokines/immunology , Influenza A virus/immunology , Virus Replication , Bone Marrow Cells/immunology , Bone Marrow Cells/virology
9.
EBioMedicine ; 106: 105269, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39111250

ABSTRACT

BACKGROUND: Influenza viruses pose a persistent threat to global public health, necessitating the development of innovative and broadly effective vaccines. METHODS: This study focuses on a multiepitope vaccine (MEV) designed to provide broad-spectrum protection against different influenza viruses. The MEV, containing 19 B-cell linear epitopes, 7 CD4+ T cells, and 11 CD8+ T cells epitopes identified through enzyme-linked immunospot assay (ELISPOT) in influenza viruses infected mice, was administered through a regimen of two doses of DNA vaccine followed by one dose of a protein vaccine in C57BL/6 female mice. FINDINGS: Upon lethal challenge with both seasonal circulating strains (H1N1, H3N2, BV, and BY) and historical strains (H1N1-PR8 and H3N2-X31), MEV demonstrated substantial protection against different influenza seasonal strains, with partial efficacy against historical strains. Notably, the increased germinal centre B cells and antibody-secreting cells, along with robust T cell immune responses, highlighted the comprehensive immune defence elicited by MEV. Elevated hemagglutinin inhibition antibody was also observed against seasonal circulating and historical strains. Additionally, mice vaccinated with MEV exhibited significantly lower counts of inflammatory cells in the lungs compared to negative control groups. INTERPRETATION: Our results demonstrated the efficacy of a broad-spectrum MEV against influenza viruses in mice. Conducting long-term studies to evaluate the durability of MEV-induced immune responses and explore its potential application in diverse populations will offer valuable insights for the continued advancement of this promising vaccine. FUNDING: Funding bodies are described in the Acknowledgments section.


Subject(s)
Epitopes, B-Lymphocyte , Influenza B virus , Influenza Vaccines , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Mice , Influenza B virus/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Female , Epitopes, B-Lymphocyte/immunology , Influenza A virus/immunology , Antibodies, Viral/immunology , Epitopes, T-Lymphocyte/immunology , Disease Models, Animal , Mice, Inbred C57BL , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Seasons , Influenza A Virus, H3N2 Subtype/immunology , Humans
10.
Cell Mol Life Sci ; 81(1): 355, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39158695

ABSTRACT

Caspase-8, an aspartate-specific cysteine protease that primarily functions as an initiator caspase to induce apoptosis, can downregulate innate immunity in part by cleaving RIPK1 and IRF3. However, patients with caspase-8 mutations or deficiency develop immunodeficiency and are prone to viral infections. The molecular mechanism underlying this controversy remains unknown. Whether caspase-8 enhances or suppresses antiviral responses against influenza A virus (IAV) infection remains to be determined. Here, we report that caspase-8 is readily activated in A549 and NL20 cells infected with the H5N1, H5N6, and H1N1 subtypes of IAV. Surprisingly, caspase-8 deficiency and two caspase-8 inhibitors, Z-VAD and Z-IETD, do not enhance but rather downregulate antiviral innate immunity, as evidenced by decreased TBK1, IRF3, IκBα, and p65 phosphorylation, decreased IL-6, IFN-ß, MX1, and ISG15 gene expression; and decreased IFN-ß production but increased virus replication. Mechanistically, caspase-8 cleaves and inactivates CYLD, a tumor suppressor that functions as a deubiquitinase. Caspase-8 inhibition suppresses CYLD cleavage, RIG-I and TAK1 ubiquitination, and innate immune signaling. In contrast, CYLD deficiency enhances IAV-induced RIG-I and TAK1 ubiquitination and innate antiviral immunity. Neither caspase-3 deficiency nor treatment with its inhibitor Z-DEVD affects CYLD cleavage or antiviral innate immunity. Our study provides evidence that caspase-8 activation in two human airway epithelial cell lines does not silence but rather enhances innate immunity by inactivating CYLD.


Subject(s)
Caspase 8 , DEAD Box Protein 58 , Deubiquitinating Enzyme CYLD , Immunity, Innate , Influenza A virus , Influenza, Human , MAP Kinase Kinase Kinases , Ubiquitination , Humans , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzyme CYLD/genetics , Caspase 8/metabolism , Caspase 8/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/immunology , Influenza A virus/immunology , DEAD Box Protein 58/metabolism , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Influenza, Human/immunology , Influenza, Human/virology , A549 Cells , Animals , Signal Transduction/immunology , Receptors, Immunologic
11.
Emerg Microbes Infect ; 13(1): 2399268, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39207215

ABSTRACT

High pathogenicity avian influenza (HPAI) virus H5N1 first emerged in Bangladesh in 2007. Despite the use of vaccines in chickens since 2012 to control HPAI, HPAI H5Nx viruses have continued to infect poultry, and wild birds, resulting in notable mass mortalities in house crows (Corvus splendens). The first HPAI H5Nx viruses in Bangladesh belonged to clade 2.2.2, followed by clade 2.3.4.2 and 2.3.2.1 viruses in 2011. After the implementation of chicken vaccination in 2012, these viruses were mostly replaced by clade 2.3.2.1a viruses and more recently clade 2.3.4.4b and h viruses. In this study, we reconstruct the phylogenetic history of HPAI H5Nx viruses in Bangladesh to evaluate the role of major host species in the maintenance and evolution of HPAI H5Nx virus in Bangladesh and reveal the role of heavily impacted crows in virus epidemiology. Epizootic waves caused by HPAI H5N1 and H5N6 viruses amongst house crows occurred annually in winter. Bayesian phylodynamic analysis of clade 2.3.2.1a revealed frequent bidirectional viral transitions between domestic ducks, chickens, and house crows that was markedly skewed towards ducks; domestic ducks might be the source, or reservoir, of HPAI H5Nx in Bangladesh, as the number of viral transitions from ducks to chickens and house crows was by far more numerous than the other transitions. Our results suggest viral circulation in domestic birds despite vaccination, with crow epizootics acting as a sentinel. The vaccination strategy needs to be updated to use more effective vaccinations, assess vaccine efficacy, and extension of vaccination to domestic ducks, the key reservoir.


Subject(s)
Chickens , Disease Reservoirs , Ducks , Influenza in Birds , Phylogeny , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Influenza in Birds/prevention & control , Ducks/virology , Bangladesh/epidemiology , Disease Reservoirs/virology , Chickens/virology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/isolation & purification , Crows/virology , Animals, Wild/virology , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza A virus/classification , Influenza A virus/immunology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Poultry Diseases/prevention & control
12.
mBio ; 15(9): e0066824, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39105586

ABSTRACT

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


Subject(s)
COVID-19 Vaccines , COVID-19 , Influenza Vaccines , SARS-CoV-2 , mRNA Vaccines , Animals , COVID-19/prevention & control , COVID-19/immunology , Mice , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Humans , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/administration & dosage , Mice, Inbred BALB C , Female , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A virus/immunology , Influenza A virus/genetics
13.
PLoS Pathog ; 20(8): e1012498, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39178311

ABSTRACT

Influenza infections result in a significant number of severe illnesses annually, many of which are complicated by secondary bacterial super-infection. Primary influenza infection has been shown to increase susceptibility to secondary methicillin-resistant Staphylococcus aureus (MRSA) infection by altering the host immune response, leading to significant immunopathology. Type III interferons (IFNs), or IFNλs, have gained traction as potential antiviral therapeutics due to their restriction of viral replication without damaging inflammation. The role of IFNλ in regulating epithelial biology in super-infection has recently been established; however, the impact of IFNλ on immune cells is less defined. In this study, we infected wild-type and IFNLR1-/- mice with influenza A/PR/8/34 followed by S. aureus USA300. We demonstrated that global IFNLR1-/- mice have enhanced bacterial clearance through increased uptake by phagocytes, which was shown to be cell-intrinsic specifically in myeloid cells in mixed bone marrow chimeras. We also showed that depletion of IFNLR1 on CX3CR1 expressing myeloid immune cells, but not neutrophils, was sufficient to significantly reduce bacterial burden compared to mice with intact IFNLR1. These findings provide insight into how IFNλ in an influenza-infected lung impedes bacterial clearance during super-infection and show a direct cell intrinsic role for IFNλ signaling on myeloid cells.


Subject(s)
Mice, Knockout , Orthomyxoviridae Infections , Phagocytes , Superinfection , Animals , Mice , Phagocytes/immunology , Orthomyxoviridae Infections/immunology , Superinfection/immunology , Superinfection/microbiology , Mice, Inbred C57BL , Staphylococcal Infections/immunology , Receptors, Interferon/metabolism , Receptors, Interferon/genetics , Interferon Lambda , Interferons/metabolism , Interferons/immunology , Influenza A virus/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Lung/immunology , Lung/virology , Lung/microbiology , Interleukins
14.
Sci Immunol ; 9(98): eado1227, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093958

ABSTRACT

The lung is constantly exposed to airborne pathogens and particles that can cause alveolar damage. Hence, appropriate repair responses are essential for gas exchange and life. Here, we deciphered the spatiotemporal trajectory and function of an atypical population of macrophages after lung injury. Post-influenza A virus (IAV) infection, short-lived monocyte-derived Ly6G-expressing macrophages (Ly6G+ Macs) were recruited to the alveoli of lung perilesional areas. Ly6G+ Macs engulfed immune cells, exhibited a high metabolic potential, and clustered with alveolar type 2 epithelial cells (AT2s) in zones of active epithelial regeneration. Ly6G+ Macs were partially dependent on granulocyte-macrophage colony-stimulating factor and interleukin-4 receptor signaling and were essential for AT2-dependent alveolar regeneration. Similar macrophages were recruited in other models of injury and in the airspaces of lungs from patients with suspected pneumonia. This study identifies perilesional alveolar Ly6G+ Macs as a spatially restricted, short-lived macrophage subset promoting epithelial regeneration postinjury, thus representing an attractive therapeutic target for treating lung damage.


Subject(s)
Antigens, Ly , Lung Injury , Macrophages, Alveolar , Mice, Inbred C57BL , Regeneration , Animals , Antigens, Ly/metabolism , Antigens, Ly/immunology , Mice , Regeneration/immunology , Lung Injury/immunology , Macrophages, Alveolar/immunology , Male , Humans , Female , Orthomyxoviridae Infections/immunology , Pulmonary Alveoli/immunology , Influenza A virus/immunology , Influenza A virus/physiology
15.
Emerg Microbes Infect ; 13(1): 2387450, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39129565

ABSTRACT

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


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes , Influenza A virus , Macrophages , Orthomyxoviridae Infections , Animals , Influenza A virus/immunology , Influenza A virus/genetics , Mice , Macrophages/immunology , Macrophages/virology , CD8-Positive T-Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , CD4-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Single-Cell Analysis/methods , RNA-Seq/methods , Single-Cell Gene Expression Analysis
16.
Int J Mol Sci ; 25(16)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39201783

ABSTRACT

This study comprehensively evaluated the DNA/RNA Defend Pro (DRDP) sample collection buffer, designed to inactivate and stabilize patient samples. The primary objectives were to assess DRDP's efficacy in ensuring sample stability, facilitating extraction-free polymerase chain reaction (PCR), and ensuring compatibility with rapid antigen testing (RAT). Ninety-five diagnostic nasopharyngeal swab samples tested for influenza virus (influenza A), respiratory syncytial virus (RSV A), and/or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were 10-fold diluted with DRDP and anonymized. Initial characterization and retesting of these samples using cobas Liat confirmed 88 samples as positive, validating the presence of viral targets. Results from rapid antigen testing showed lower sensitivity compared to nucleic acid amplification testing (NAAT) but maintained perfect specificity, with 40 out of 88 positive samples by cobas Liat also testing positive for RAT. Direct RT-qPCR of DRDP-diluted samples demonstrated robust compatibility, with 72 out of 88 samples positive for cobas Liat also testing positive by direct RT-qPCR. Non-concordant results could be explained by the 200-fold lower input of extraction-free NAAT. Stability testing involved incubating 31 positive samples at 4 °C, 20 °C, and 37 °C for 7 days, with extraction-free NAAT. DRDP guaranteed viral RNA stability at all temperatures for influenza A, SARS-CoV-2, and RSV A, showing stability up to 7 days at 4 °C. In conclusion, DRDP is an effective stabilizing medium compatible with direct RT-qPCR and rapid antigen testing and shows great potential for optimizing diagnostic processes, particularly in resource-limited or time-sensitive scenarios.


Subject(s)
Nasopharynx , SARS-CoV-2 , Specimen Handling , Nasopharynx/virology , Humans , Specimen Handling/methods , SARS-CoV-2/isolation & purification , SARS-CoV-2/immunology , SARS-CoV-2/genetics , RNA, Viral/analysis , RNA, Viral/isolation & purification , RNA, Viral/genetics , Buffers , Influenza A virus/isolation & purification , Influenza A virus/immunology , Influenza A virus/genetics , Sensitivity and Specificity , COVID-19/diagnosis , COVID-19/virology , Antigens, Viral/analysis , Influenza, Human/diagnosis , Influenza, Human/virology
17.
J Immunol ; 213(8): 1170-1186, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39212406

ABSTRACT

We demonstrate the role of signaling via the glucocorticoid receptor, NR3C1, in differentiation of CD8+ T cell memory. Pharmacological inhibition as well as the short hairpin RNA-mediated knockdown of the receptor hindered memory transition and limited the homeostatic turnover of the activated CD8+ T cells. Dexamethasone exposure of CD8+ T cells expanded during a resolving infection with influenza A virus or a γ-herpesvirus promoted conversion of effector cells into memory cells by modulating cellular metabolism and lowering the accumulation of reactive oxygen species. Reduced reactive oxygen species levels in the responding effector cells upregulated Bcl2 and enhanced survival. The generated virus-specific memory CD8+ T cells were efficiently recalled following challenge of animals with a secondary infection to control it better. The memory-enhancing effect was predominantly evident at low doses of dexamethasone. Therefore, controlled glucocorticoid signaling within the effector CD8+ T cells is crucial for optimal memory differentiation.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Differentiation , Dexamethasone , Glucocorticoids , Immunologic Memory , Receptors, Glucocorticoid , Animals , Mice , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Glucocorticoids/pharmacology , Dexamethasone/pharmacology , Cell Differentiation/immunology , Influenza A virus/immunology , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Signal Transduction/immunology , Memory T Cells/immunology , Reactive Oxygen Species/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Herpesviridae Infections/immunology
18.
J Virol ; 98(9): e0079624, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39115433

ABSTRACT

Host cells have evolved an intricate regulatory network to fine tune the type-I interferon responses. However, the full picture of this regulatory network remains to be depicted. In this study, we found that knock out of zinc-finger CCHC-type containing protein 8 (ZCCHC8) impairs the replication of influenza A virus (IAV), Sendai virus (Sev), Japanese encephalitis virus (JEV), and vesicular stomatitis virus (VSV). Further investigation unveiled that ZCCHC8 suppresses the type-I interferon responses by targeting the interferon regulatory factor 3 (IRF3) signaling pathway. Mechanistically, ZCCHC8 associates with phosphorylated IRF3 and disrupts the interaction of IRF3 with the co-activator CREB-binding protein (CBP). Additionally, the direct binding of ZCCHC8 with the IFN-stimulated response element (ISRE) impairs the ISRE-binding of IRF3. Our study contributes to the comprehensive understanding for the negative regulatory network of the type-I interferon responses and provides valuable insights for the control of multiple viruses from a host-centric perspective.IMPORTANCEThe innate immune responses serve as the initial line of defense against invading pathogens and harmful substances. Negative regulation of the innate immune responses plays an essential role in avoiding auto-immune diseases and over-activated immune responses. Hence, the comprehensive understanding of the negative regulation network for innate immune responses could provide novel therapeutic insights for the control of viral infections and immune dysfunction. In this study, we report that ZCCHC8 negatively regulates the type-I interferon responses. We illustrate that ZCCHC8 impedes the IRF3-CBP association by interacting with phosphorylated IRF3 and competes with IRF3 for binding to ISRE. Our study demonstrates the role of ZCCHC8 in the replication of multiple RNA viruses and contributes to a deeper understanding of the negative regulation system for the type-I interferon responses.


Subject(s)
CREB-Binding Protein , Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Type I , Sendai virus , Signal Transduction , Virus Replication , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Humans , HEK293 Cells , Sendai virus/physiology , Sendai virus/genetics , CREB-Binding Protein/metabolism , CREB-Binding Protein/genetics , RNA Viruses/physiology , RNA Viruses/immunology , RNA Viruses/genetics , Animals , A549 Cells , Influenza A virus/physiology , Influenza A virus/immunology , Phosphorylation , Host-Pathogen Interactions , Vesiculovirus/physiology , Encephalitis Virus, Japanese/physiology , Encephalitis Virus, Japanese/immunology
19.
Nat Commun ; 15(1): 6802, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122688

ABSTRACT

Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.


Subject(s)
Administration, Intranasal , Escherichia coli , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Probiotics , Animals , Escherichia coli/genetics , Probiotics/administration & dosage , Female , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Mice , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza A virus/immunology , Influenza A virus/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Immunity, Innate , Immunity, Mucosal , Humans , Antibodies, Viral/immunology , Viroporin Proteins
20.
Sci Adv ; 10(33): eado4313, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39141734

ABSTRACT

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


Subject(s)
CD8-Positive T-Lymphocytes , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Influenza A virus/immunology , Humans , Lymphocyte Activation/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Optical Tweezers
SELECTION OF CITATIONS
SEARCH DETAIL