Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.243
Filter
1.
Nat Commun ; 15(1): 5593, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961067

ABSTRACT

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.


Subject(s)
Antibodies, Viral , Cross Reactions , Influenza A Virus, H3N2 Subtype , Influenza, Human , Neuraminidase , Pandemics , Neuraminidase/immunology , Neuraminidase/genetics , Animals , Humans , Antibodies, Viral/immunology , Antibodies, Viral/blood , Influenza A Virus, H3N2 Subtype/immunology , Female , Cross Reactions/immunology , Mice , Influenza, Human/immunology , Influenza, Human/epidemiology , Influenza, Human/virology , Aged , Influenza A Virus, H2N2 Subtype/immunology , Influenza A Virus, H2N2 Subtype/genetics , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/veterinary , Birds/virology , Middle Aged , Influenza in Birds/epidemiology , Influenza in Birds/immunology , Influenza in Birds/virology , Influenza A Virus, H9N2 Subtype/immunology , Adult , Viral Proteins/immunology , Viral Proteins/genetics
2.
Vet Res ; 55(1): 86, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970119

ABSTRACT

H7N9 subtype avian influenza viruses (AIVs) cause 1567 human infections and have high mortality, posing a significant threat to public health. Previously, we reported that two avian-derived H7N9 isolates (A/chicken/Eastern China/JTC4/2013 and A/chicken/Eastern China/JTC11/2013) exhibit different pathogenicities in mice. To understand the genetic basis for the differences in virulence, we constructed a series of mutant viruses based on reverse genetics. We found that the PB2-E627K mutation alone was not sufficient to increase the virulence of H7N9 in mice, despite its ability to enhance polymerase activity in mammalian cells. However, combinations with PB1-V719M and/or PA-N444D mutations significantly enhanced H7N9 virulence. Additionally, these combined mutations augmented polymerase activity, thereby intensifying virus replication, inflammatory cytokine expression, and lung injury, ultimately increasing pathogenicity in mice. Overall, this study revealed that virulence in H7N9 is a polygenic trait and identified novel virulence-related residues (PB2-627K combined with PB1-719M and/or PA-444D) in viral ribonucleoprotein (vRNP) complexes. These findings provide new insights into the molecular mechanisms underlying AIV pathogenesis in mammals, with implications for pandemic preparedness and intervention strategies.


Subject(s)
Influenza A Virus, H7N9 Subtype , Mutation , Orthomyxoviridae Infections , Viral Proteins , Animals , Mice , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/physiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Virulence , Female , Viral Proteins/genetics , Viral Proteins/metabolism , Mice, Inbred BALB C , Virus Replication
3.
Science ; 385(6705): 123, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38991057

ABSTRACT

The relentless march of a highly pathogenic avian influenza virus (HPAIV) strain, known as H5N1, to become an unprecedented panzootic continues unchecked. The leap of H5N1 clade 2.3.4.4b from Eurasia and Africa to North America in 2021 and its further spread to South America and the Antarctic have exposed new avian and mammalian populations to the virus and led to outbreaks on an unrivaled scale. The virus has infected wild birds across vast geographic regions and caused wildlife deaths in some of the world's most biodiverse ecosystems. Hundreds of millions of poultry have died or been culled, affecting global food security in some of the world's poorest regions. Numerous mammalian species, including sea lions and fur animals, have been infected. Outbreaks in dairy cows in the United States have been occurring for months, seemingly unchecked in most affected states. Why is there not a greater sense of urgency to control these infections?


Subject(s)
Cattle Diseases , Disease Outbreaks , Influenza A Virus, H5N1 Subtype , Influenza in Birds , Orthomyxoviridae Infections , Animals , Cattle , Humans , Birds/virology , Cattle Diseases/epidemiology , Cattle Diseases/prevention & control , Cattle Diseases/virology , Disease Outbreaks/prevention & control , Disease Outbreaks/veterinary , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/epidemiology , Influenza in Birds/transmission , Influenza in Birds/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Poultry/virology , United States/epidemiology
4.
Virol J ; 21(1): 151, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965616

ABSTRACT

BACKGROUND: The canine influenza virus (CIV) outbreak has garnered considerable attention as it poses a significant threat to dog health. During the H3N2 CIV evolution in beagles, the virus formed a new clade after 2019 and gradually became more adaptable to other mammals. Therefore, successfully elucidating the biological characteristics and constructing a canine influenza infection model is required for CIV characterization. METHODS: We performed genetic analyses to examine the biological characteristics and infection dynamics of CIV. RESULTS: The genotype of our H3N2 CIV strain (from 2019 in Shanghai) belonged to the 5.1 clade, which is now prevalent in China. Using MDCK cells, we investigated viral cytopathic effects. Virus size and morphology were observed using transmission electron microscopy. Beagles were also infected with 104, 105, and 106 50% egg-infectious doses (EID50). When compared with the other groups, the 106 EID50 group showed the most obvious clinical symptoms, the highest virus titers, and typical lung pathological changes. Our results suggested that the other two treatments caused mild clinical manifestations and pathological changes. Subsequently, CIV distribution in the 106 EID50 group was detected by hematoxylin and eosin (H&E) and immunofluorescence (IF) staining, which indicated that CIV primarily infected the lungs. CONCLUSIONS: The framework established in this study will guide further CIV prevention strategies.


Subject(s)
Dog Diseases , Genotype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , Animals , Dogs , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/pathology , Dog Diseases/virology , Madin Darby Canine Kidney Cells , China/epidemiology , Lung/virology , Lung/pathology , Phylogeny , Viral Load , Disease Models, Animal
5.
Emerg Microbes Infect ; 13(1): 2368202, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38970562

ABSTRACT

Influenza A viruses (IAV) impose significant respiratory disease burdens in both swine and humans worldwide, with frequent human-to-swine transmission driving viral evolution in pigs and highlighting the risk at the animal-human interface. Therefore, a comprehensive One Health approach (interconnection among human, animal, and environmental health) is needed for IAV prevention, control, and response. Animal influenza genomic surveillance remains limited in many Latin American countries, including Colombia. To address this gap, we genetically characterized 170 swine specimens from Colombia (2011-2017). Whole genome sequencing revealed a predominance of pandemic-like H1N1 lineage, with a minority belonging to H3N2 and H1N2 human seasonal-like lineage and H1N1 early classical swine lineages. Significantly, we have identified reassortant and recombinant viruses (H3N2, H1N1) not previously reported in Colombia. This suggests a broad genotypic viral diversity, likely resulting from reassortment between classical endemic viruses and new introductions established in Colombia's swine population (e.g. the 2009 H1N1 pandemic). Our study highlights the importance of a One Health approach in disease control, particularly in an ecosystem where humans are a main source of IAV to swine populations, and emphasizes the need for continued surveillance and enhanced biosecurity measures. The co-circulation of multiple subtypes in regions with high swine density facilitates viral exchange, underscoring the importance of monitoring viral evolution to inform vaccine selection and public health policies locally and globally.


Subject(s)
Evolution, Molecular , Genetic Variation , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , Phylogeny , Swine Diseases , Animals , Swine , Colombia/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/epidemiology , Swine Diseases/virology , Swine Diseases/epidemiology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/isolation & purification , One Health , Humans , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Whole Genome Sequencing , Genome, Viral , Epidemiological Monitoring , Reassortant Viruses/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/classification , Influenza, Human/virology , Influenza, Human/epidemiology
6.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38968329

ABSTRACT

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.


Subject(s)
CRISPR-Cas Systems , Influenza, Human , RNA Stability , RNA, Messenger , RNA, Viral , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Humans , Influenza, Human/virology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections/virology , Antiviral Agents/pharmacology , Dogs , Cricetinae , Viral Proteins/genetics , Viral Proteins/metabolism , Mesocricetus , Madin Darby Canine Kidney Cells
7.
PLoS Pathog ; 20(7): e1011910, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39024231

ABSTRACT

The pig is a natural host for influenza viruses and integrally involved in virus evolution through interspecies transmissions between humans and swine. Swine have many physiological, anatomical, and immunological similarities to humans, and are an excellent model for human influenza. Here, we employed single cell RNA-sequencing (scRNA-seq) and flow cytometry to characterize the major leukocyte subsets in bronchoalveolar lavage (BAL), twenty-one days after H1N1pdm09 infection or respiratory immunization with an adenoviral vector vaccine expressing hemagglutinin and nucleoprotein with or without IL-1ß. Mapping scRNA-seq clusters from BAL onto those previously described in peripheral blood facilitated annotation and highlighted differences between tissue resident and circulating immune cells. ScRNA-seq data and functional assays revealed lasting impacts of immune challenge on BAL populations. First, mucosal administration of IL-1ß reduced the number of functionally active Treg cells. Second, influenza infection upregulated IFI6 in BAL cells and decreased their susceptibility to virus replication in vitro. Our data provide a reference map of porcine BAL cells and reveal lasting immunological consequences of influenza infection and respiratory immunization in a highly relevant large animal model for respiratory virus infection.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Lung , Orthomyxoviridae Infections , Single-Cell Analysis , Animals , Swine , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Lung/immunology , Lung/virology , Influenza Vaccines/immunology , Influenza A Virus, H1N1 Subtype/immunology , Immunization , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology
8.
Nat Commun ; 15(1): 6053, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025863

ABSTRACT

Respiratory viral infections cause morbidity and mortality worldwide. Despite the success of vaccines, vaccination efficacy is weakened by the rapid emergence of viral variants with immunoevasive properties. The development of an off-the-shelf, effective, and safe therapy against respiratory viral infections is thus desirable. Here, we develop NanoSTING, a nanoparticle formulation of the endogenous STING agonist, 2'-3' cGAMP, to function as an immune activator and demonstrate its safety in mice and rats. A single intranasal dose of NanoSTING protects against pathogenic strains of SARS-CoV-2 (alpha and delta VOC) in hamsters. In transmission experiments, NanoSTING reduces the transmission of SARS-CoV-2 Omicron VOC to naïve hamsters. NanoSTING also protects against oseltamivir-sensitive and oseltamivir-resistant strains of influenza in mice. Mechanistically, NanoSTING upregulates locoregional interferon-dependent and interferon-independent pathways in mice, hamsters, as well as non-human primates. Our results thus implicate NanoSTING as a broad-spectrum immune activator for controlling respiratory virus infection.


Subject(s)
Administration, Intranasal , Nanoparticles , SARS-CoV-2 , Animals , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice , Cricetinae , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Disease Models, Animal , Humans , Membrane Proteins/agonists , Membrane Proteins/metabolism , Female , Nucleotides, Cyclic/pharmacology , Rats , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/drug therapy , Male , Antiviral Agents/pharmacology , Antiviral Agents/administration & dosage , Mice, Inbred C57BL
9.
Virology ; 597: 110162, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38955082

ABSTRACT

There is an urgent need for influenza vaccines that offer broad cross-protection. The highly conserved ectodomain of the influenza matrix protein 2 (M2e) is a promising candidate; however, its low immunogenicity can be addressed. In this study, we developed influenza vaccines using the Lumazine synthase (LS) platform. The primary objective of this study was to determine the protective potential of M2e proteins expressed on Lumazine synthase (LS) nanoparticles. M2e-LS proteins, produced through the E. coli system, spontaneously assemble into nanoparticles. The study investigated the efficacy of the M2e-LS nanoparticle vaccine in mice. Mice immunized with M2e-LS nanoparticles exhibited significantly higher levels of intracellular cytokines than those receiving soluble M2e proteins. The M2e-LS protein exhibited robust immunogenicity and provided 100% protection against cross-clade influenza.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Multienzyme Complexes , Nanoparticles , Orthomyxoviridae Infections , Viral Matrix Proteins , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Nanoparticles/chemistry , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Female , Mice, Inbred BALB C , Antibodies, Viral/immunology , Cytokines/metabolism , Cross Protection/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Escherichia coli/genetics , Escherichia coli/metabolism , Viroporin Proteins
10.
PLoS One ; 19(7): e0301664, 2024.
Article in English | MEDLINE | ID: mdl-38985719

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing , Camelids, New World , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections , Single-Domain Antibodies , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Single-Domain Antibodies/immunology , Antibodies, Neutralizing/immunology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Camelids, New World/immunology , Antibodies, Viral/immunology , Female , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Epitopes/immunology , Dogs , Mice, Inbred BALB C
11.
J Med Virol ; 96(7): e29768, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38978388

ABSTRACT

The vagus nerve circuit, operating through the alpha-7 nicotinic acetylcholine receptor (α7 nAChR), regulates the inflammatory response by influencing immune cells. However, the role of vagal-α7 nAChR signaling in influenza virus infection is unclear. In particular, does vagal-α7 nAChR signaling impact the infection of alveolar epithelial cells (AECs), the primary target cells of influenza virus? Here, we demonstrated a distinct role of α7 nAChR in type II AECs compared to its role in immune cells during influenza infection. We found that deletion of Chrna7 (encoding gene of α7 nAChR) in type II AECs or disruption of vagal circuits reduced lung influenza infection and protected mice from influenza-induced lung injury. We further unveiled that activation of α7 nAChR enhanced influenza infection through PTP1B-NEDD4L-ASK1-p38MAPK pathway. Mechanistically, activation of α7 nAChR signaling decreased p38MAPK phosphorylation during infection, facilitating the nuclear export of influenza viral ribonucleoproteins and thereby promoting infection. Taken together, our findings reveal a mechanism mediated by vagal-α7 nAChR signaling that promotes influenza viral infection and exacerbates disease severity. Targeting vagal-α7 nAChR signaling may offer novel strategies for combating influenza virus infections.


Subject(s)
Lung , Orthomyxoviridae Infections , Signal Transduction , Vagus Nerve , alpha7 Nicotinic Acetylcholine Receptor , Animals , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Vagus Nerve/metabolism , Mice , Orthomyxoviridae Infections/virology , Lung/virology , Lung/pathology , Mice, Inbred C57BL , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/metabolism , Humans , Mice, Knockout
12.
Curr Microbiol ; 81(9): 267, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39003673

ABSTRACT

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.


Subject(s)
Cytokines , Disease Models, Animal , Feces , Gastrointestinal Microbiome , Germ-Free Life , Immunity, Mucosal , Influenza A Virus, H1N1 Subtype , Animals , Swine , Feces/microbiology , Feces/virology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Cytokines/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Viral Load , Infant , Influenza, Human/immunology , Influenza, Human/microbiology , Influenza, Human/virology
13.
BMC Vet Res ; 20(1): 285, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956597

ABSTRACT

Clade 2.3.4.4b highly pathogenic avian influenza (HPAI) H5N1 virus was detected in the South American sea lions found dead in Santa Catarina, Brazil, in October 2023. Whole genome sequencing and comparative phylogenetic analysis were conducted to investigate the origin, genetic diversity, and zoonotic potentials of the H5N1 viruses. The H5N1 viruses belonged to the genotype B3.2 of clade 2.3.4.4b H5N1 virus, which was identified in North America and disseminated to South America. They have acquired new amino acid substitutions related to mammalian host affinity. Our study provides insights into the genetic landscape of HPAI H5N1 viruses in Brazil, highlighting the continuous evolutionary processes contributing to their possible adaptation to mammalian hosts.


Subject(s)
Influenza A Virus, H5N1 Subtype , Phylogeny , Sea Lions , Whole Genome Sequencing , Animals , Sea Lions/virology , Brazil , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/classification , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Genome, Viral , Genotype , Genetic Variation
15.
Nat Commun ; 15(1): 5025, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871701

ABSTRACT

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.


Subject(s)
Ferrets , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H1N2 Subtype , Influenza, Human , Orthomyxoviridae Infections , Pandemics , Animals , Ferrets/virology , Humans , Swine , Influenza, Human/virology , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza, Human/blood , Influenza, Human/transmission , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Swine Diseases/virology , Swine Diseases/epidemiology , Swine Diseases/immunology , Swine Diseases/transmission , Swine Diseases/blood , Female , Virus Shedding , Male , Adult , Virus Replication
16.
Euro Surveill ; 29(25)2024 Jun.
Article in English | MEDLINE | ID: mdl-38904109

ABSTRACT

Highly pathogenic avian influenza (HPAI) has caused widespread mortality in both wild and domestic birds in Europe 2020-2023. In July 2023, HPAI A(H5N1) was detected on 27 fur farms in Finland. In total, infections in silver and blue foxes, American minks and raccoon dogs were confirmed by RT-PCR. The pathological findings in the animals include widespread inflammatory lesions in the lungs, brain and liver, indicating efficient systemic dissemination of the virus. Phylogenetic analysis of Finnish A(H5N1) strains from fur animals and wild birds has identified three clusters (Finland I-III), and molecular analyses revealed emergence of mutations known to facilitate viral adaptation to mammals in the PB2 and NA proteins. Findings of avian influenza in fur animals were spatially and temporally connected with mass mortalities in wild birds. The mechanisms of virus transmission within and between farms have not been conclusively identified, but several different routes relating to limited biosecurity on the farms are implicated. The outbreak was managed in close collaboration between animal and human health authorities to mitigate and monitor the impact for both animal and human health.


Subject(s)
Animals, Wild , Charadriiformes , Disease Outbreaks , Influenza A Virus, H5N1 Subtype , Influenza in Birds , Phylogeny , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Finland/epidemiology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/isolation & purification , Animals, Wild/virology , Charadriiformes/virology , Disease Outbreaks/veterinary , Farms , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/epidemiology , Foxes/virology , Birds/virology , Mink/virology
17.
Front Immunol ; 15: 1361323, 2024.
Article in English | MEDLINE | ID: mdl-38835763

ABSTRACT

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.


Subject(s)
Antibodies, Viral , Ferritins , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Mice, Inbred BALB C , Nanoparticles , Orthomyxoviridae Infections , Animals , Influenza A Virus, H1N1 Subtype/immunology , Ferritins/immunology , Influenza Vaccines/immunology , Swine , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/virology , Female , Nanovaccines
19.
Virology ; 596: 110124, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838475

ABSTRACT

While mammals can be infected by influenza A virus either sporadically or with well adapted lineages, aquatic birds are the natural reservoir of the pathogen. So far most of the knowledge on influenza virus dynamics was however gained on mammalian models. In this study, we infected turkeys using a low pathogenic avian influenza virus and determined the infection dynamics with a target-cell limited model. Results showed that turkeys had a different set of infection characteristics, compared with humans and ponies. The viral clearance rates were similar between turkeys and ponies but higher than that in humans. The cell death rates and cell to cell transmission rates were similar between turkeys and humans but higher than those in ponies. Overall, this study indicated the variations of within-host dynamics of influenza infection in avian, humans, and other mammalian systems.


Subject(s)
Influenza A Virus, H9N2 Subtype , Influenza in Birds , Turkeys , Animals , Turkeys/virology , Influenza in Birds/virology , Influenza in Birds/transmission , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza A Virus, H9N2 Subtype/physiology , Humans , Horses , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/transmission , Influenza, Human/virology
20.
Nat Microbiol ; 9(7): 1764-1777, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38849624

ABSTRACT

Influenza virus infection is initiated by the attachment of the viral haemagglutinin (HA) protein to sialic acid receptors on the host cell surface. Most virus particles enter cells through clathrin-mediated endocytosis (CME). However, it is unclear how viral binding signals are transmitted through the plasma membrane triggering CME. Here we found that metabotropic glutamate receptor subtype 2 (mGluR2) and potassium calcium-activated channel subfamily M alpha 1 (KCa1.1) are involved in the initiation and completion of CME of influenza virus using an siRNA screen approach. Influenza virus HA directly interacted with mGluR2 and used it as an endocytic receptor to initiate CME. mGluR2 interacted and activated KCa1.1, leading to polymerization of F-actin, maturation of clathrin-coated pits and completion of the CME of influenza virus. Importantly, mGluR2-knockout mice were significantly more resistant to different influenza subtypes than the wild type. Therefore, blocking HA and mGluR2 interaction could be a promising host-directed antiviral strategy.


Subject(s)
Endocytosis , Mice, Knockout , Receptors, Metabotropic Glutamate , Animals , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/genetics , Mice , Humans , Virus Internalization , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Clathrin/metabolism , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/metabolism , HEK293 Cells , Actins/metabolism , Dogs , Madin Darby Canine Kidney Cells , Receptors, Virus/metabolism , Receptors, Virus/genetics , Influenza, Human/virology , Influenza, Human/metabolism , Orthomyxoviridae/physiology , Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL