Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Vet Res ; 55(1): 65, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773540

ABSTRACT

In 2020, a new genotype of swine H1N2 influenza virus (H1avN2-HA 1C.2.4) was identified in France. It rapidly spread within the pig population and supplanted the previously predominant H1avN1-HA 1C.2.1 virus. To characterize this new genotype which is genetically and antigenically distant from the other H1avNx viruses detected in France, an experimental study was conducted to compare the outcomes of H1avN2 and H1avN1 infections in pigs and evaluate the protection conferred by the only inactivated vaccine currently licensed in Europe containing an HA 1C (clade 1C.2.2) antigen. Infection with H1avN2 induced stronger clinical signs and earlier shedding than H1avN1. The neutralizing antibodies produced following H1avN2 infection were unable to neutralize H1avN1, and vice versa, whereas the cellular-mediated immunity cross-reacted. Vaccination slightly altered the impact of H1avN2 infection at the clinical level, but did not prevent shedding of infectious virus particles. It induced a cellular-mediated immune response towards H1avN2, but did not produce neutralizing antibodies against this virus. As in vaccinated animals, animals previously infected by H1avN1 developed a cross-reacting cellular immune response but no neutralizing antibodies against H1avN2. However, H1avN1 pre-infection induced a better protection against the H1avN2 infection than vaccination, probably due to higher levels of non-neutralizing antibodies and a mucosal immunity. Altogether, these results showed that the new H1avN2 genotype induced a severe respiratory infection and that the actual vaccine was less effective against this H1avN2-HA 1C.2.4 than against H1avN1-HA 1C.2.1, which may have contributed to the H1avN2 epizootic and dissemination in pig farms in France.


Subject(s)
Genotype , Influenza A Virus, H1N2 Subtype , Orthomyxoviridae Infections , Swine Diseases , Animals , Swine , Swine Diseases/virology , Swine Diseases/immunology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/immunology , France/epidemiology , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/immunology , Virulence , Antibodies, Neutralizing/blood , Immunity, Cellular
2.
Viruses ; 16(4)2024 04 18.
Article in English | MEDLINE | ID: mdl-38675967

ABSTRACT

Inactivated influenza A virus (IAV) vaccines help reduce clinical disease in suckling piglets, although endemic infections still exist. The objective of this study was to evaluate the detection of IAV in suckling and nursery piglets from IAV-vaccinated sows from farms with endemic IAV infections. Eight nasal swab collections were obtained from 135 two-week-old suckling piglets from four farms every other week from March to September 2013. Oral fluid samples were collected from the same group of nursery piglets. IAV RNA was detected in 1.64% and 31.01% of individual nasal swabs and oral fluids, respectively. H1N2 was detected most often, with sporadic detection of H1N1 and H3N2. Whole-genome sequences of IAV isolated from suckling piglets revealed an H1 hemagglutinin (HA) from the 1B.2.2.2 clade and N2 neuraminidase (NA) from the 2002A clade. The internal gene constellation of the endemic H1N2 was TTTTPT with a pandemic lineage matrix. The HA gene had 97.59% and 97.52% nucleotide and amino acid identities, respectively, to the H1 1B.2.2.2 used in the farm-specific vaccine. A similar H1 1B.2.2.2 was detected in the downstream nursery. These data demonstrate the low frequency of IAV detection in suckling piglets and downstream nurseries from farms with endemic infections in spite of using farm-specific IAV vaccines in sows.


Subject(s)
Farms , Influenza A virus , Influenza Vaccines , Orthomyxoviridae Infections , Phylogeny , Swine Diseases , Animals , Swine , Swine Diseases/virology , Swine Diseases/epidemiology , Swine Diseases/prevention & control , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/epidemiology , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/isolation & purification , Influenza A virus/classification , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Animals, Suckling , Vaccination/veterinary , Endemic Diseases/veterinary , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/isolation & purification , RNA, Viral/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/immunology , Genome, Viral
3.
Viruses ; 13(11)2021 10 27.
Article in English | MEDLINE | ID: mdl-34834975

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) and swine influenza A virus (swIAV) are major pathogens of the porcine respiratory disease complex, but little is known on their interaction in super-infected pigs. In this study, we investigated clinical, virological and immunological outcomes of successive infections with PRRSV-1 and H1N2 swIAV. Twenty-four specific pathogen-free piglets were distributed into four groups and inoculated either with PRRSV at study day (SD) 0, or with swIAV at SD8, or with PRRSV and swIAV one week apart at SD0 and SD8, respectively, or mock-inoculated. In PRRSV/swIAV group, the clinical signs usually observed after swIAV infection were attenuated while higher levels of anti-swIAV antibodies were measured in lungs. Concurrently, PRRSV multiplication in lungs was significantly affected by swIAV infection, whereas the cell-mediated immune response specific to PRRSV was detected earlier in blood, as compared to PRRSV group. Moreover, levels of interferon (IFN)-α measured from SD9 in the blood of super-infected pigs were lower than those measured in the swIAV group, but higher than in the PRRSV group at the same time. Correlation analyses suggested an important role of IFN-α in the two-way interference highlighted between both viral infections.


Subject(s)
Influenza A Virus, H1N2 Subtype/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Animals , Antibodies, Neutralizing , Antibodies, Viral , Immunity , Influenza A virus/immunology , Interferon-alpha , Lung/immunology , Orthomyxoviridae Infections/virology , Specific Pathogen-Free Organisms , Swine , Swine Diseases/virology
4.
J Virol ; 95(15): e0069221, 2021 07 12.
Article in English | MEDLINE | ID: mdl-33980596

ABSTRACT

Swine influenza virus (SIV) can cause respiratory illness in swine. Swine contribute to influenza virus reassortment, as avian, human, and/or swine influenza viruses can infect swine and reassort, and new viruses can emerge. Thus, it is important to determine the host antiviral responses that affect SIV replication. In this study, we examined the innate antiviral cytokine response to SIV by swine respiratory epithelial cells, focusing on the expression of interferon (IFN) and interferon-stimulated genes (ISGs). Both primary and transformed swine nasal and tracheal respiratory epithelial cells were examined following infection with field isolates. The results show that IFN and ISG expression is maximal at 12 h postinfection (hpi) and is dependent on cell type and virus genotype. IMPORTANCE Swine are considered intermediate hosts that have facilitated influenza virus reassortment events that have given rise pandemics or genetically related viruses have become established in swine. In this study, we examine the innate antiviral response to swine influenza virus in primary and immortalized swine nasal and tracheal epithelial cells, and show virus strain- and host cell type-dependent differential expression of key interferons and interferon-stimulated genes.


Subject(s)
Cytokines/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Respiratory Mucosa/immunology , Animals , Cell Line , Cytokines/immunology , Dogs , Epithelial Cells/virology , Host-Pathogen Interactions/immunology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/growth & development , Interferons/immunology , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/immunology , Respiratory Mucosa/cytology , Swine , Virus Replication/physiology
5.
Dev Comp Immunol ; 121: 104080, 2021 08.
Article in English | MEDLINE | ID: mdl-33781781

ABSTRACT

In this work, we report on two novel monoclonal antibodies, specific for porcine CD9. CD9 is a tetraspanin that is expressed on a wide variety of cells. We phenotyped porcine immune cell subsets and found that CD9 was expressed on all monocytes as well as a subset of B cells. CD9 was variably expressed on T cells, with CD4 T cells containing the highest frequency of CD9+ cells. CD9 expression positively correlated with the frequency of central memory CD4 T cells in ex vivo PBMC. Therefore, we proceeded to explore CD9 as a marker of T cell function. Here we observed that CD9 was expressed on the vast majority of long-lived influenza A virus-specific effector cells that retained the capacity for cytokine production in response to in vitro recall antigen. Therefore, the new antibodies enable the detection of a cell surface molecule with functional relevance to T cells. Considering the importance of CD9 in membrane remodelling across many cell types, they will also benefit the wider field of swine biomedical research.


Subject(s)
Immunophenotyping/methods , Memory T Cells/immunology , Orthomyxoviridae Infections/immunology , Swine/immunology , Tetraspanin 29/analysis , Animals , Antibodies, Monoclonal/metabolism , Cattle , Cell Differentiation , Cell Line , Influenza A Virus, H1N2 Subtype/immunology , Lymphocyte Activation , Memory T Cells/metabolism , Orthomyxoviridae Infections/virology , Swine/virology , Tetraspanin 29/metabolism
6.
Vet Microbiol ; 253: 108968, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33418392

ABSTRACT

Swine Influenza A virus (swIAV) poses a substantial burden to the swine industry due to its highly contagious nature, acute viral disease, and ability to cause up to 100 % morbidity. Currently, North American swine are predominately infected with three subtypes of swIAV: H1N1, H1N2, and H3N2. The ability of influenza viruses to cross both directions between humans and swine means that both human and swine-origin viruses as well as new reassortant viruses can pose a substantial public health or pandemic threat. Since the primary method of protection and control against influenza is through vaccination, more effective, new vaccine platforms need to be developed. This study uses two Canadian swIAV isolates, A/Swine/Alberta/SD0191/2016 (H1N2) [SD191] and A/Swine/Saskatchewan/SD0069/2015 (H3N2) [SD69] to design a bivalent live attenuated influenza virus vaccine (LAIV) through reverse genetics. The hemagglutinin (HA) cleavage site from both SD191-WT and SD69-WT were engineered from a trypsin-sensitive to an elastase-sensitive motif, to generate SD191-R342V and SD69-K345V, respectively. The elastase dependent SD191-R342V virus possesses a mutation from arginine to valine at amino acid (aa) 342 on HA, whereas the elastase dependent SD69-K345V virus possesses a mutation from lysine to valine at aa 345 on HA. Both elastase dependent swIAVs are completely dependent on elastase, display comparable growth properties to the wild type (WT) viruses, are genetically stable in vitro, and entirely non-virulent in pigs. Moreover, when these elastase dependent swIAVs were administered together in pigs, they were found to stimulate antibody responses and IFN-γ secreting cells, as well as prevent viral replication and lung pathology associated with WT H1N2 and H3N2 swIAV challenge. Therefore, this bivalent LAIV demonstrates the strong candidacy to protect swine against the predominant influenza subtypes in North America.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , Animals , Immunogenicity, Vaccine , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/metabolism , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/immunology , Pancreatic Elastase/metabolism , Reassortant Viruses , Reverse Genetics , Swine , Swine Diseases/immunology , Swine Diseases/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
7.
Viruses ; 12(10)2020 10 12.
Article in English | MEDLINE | ID: mdl-33053905

ABSTRACT

The surveillance of swine influenza A viruses in France revealed the emergence of an antigenic variant following deletions and mutations that are fixed in the HA-encoding gene of the European human-like reassortant swine H1N2 lineage. In this study, we compared the outcomes of the parental (H1huN2) and variant (H1huN2Δ146-147) virus infections in experimentally-inoculated piglets. Moreover, we assessed and compared the protection that was conferred by an inactivated vaccine currently licensed in Europe. Three groups of five unvaccinated or vaccinated piglets were inoculated with H1huN2 or H1huN2Δ146-147 or mock-inoculated, respectively. In unvaccinated piglets, the variant strain induced greater clinical signs than the parental virus, in relation to a higher inflammatory response that involves TNF-α production and a huge afflux of granulocytes into the lung. However, both infections led to similar levels of virus excretion and adaptive (humoral and cellular) immune responses in blood. The vaccinated animals were clinically protected from both infectious challenges and did not exhibit any inflammatory responses, regardless the inoculated virus. However, whereas vaccination prevented virus shedding in H1huN2-infected animals, it did not completely inhibit the multiplication of the variant strain, since live virus particles were detected in nasal secretions that were taken from H1huN2Δ146-147-inoculated vaccinated piglets. This difference in the level of vaccine protection was probably related to the poorer ability of the post-vaccine antibodies to neutralize the variant virus than the parental virus, even though post-vaccine cellular immunity appeared to be equally effective against both viruses. These results suggest that vaccine antigens would potentially need to be updated if this variant becomes established in Europe.


Subject(s)
Antigens, Viral/immunology , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Swine Diseases/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/genetics , France , Influenza A Virus, H1N2 Subtype/pathogenicity , Mutation/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Swine , Swine Diseases/pathology , Swine Diseases/virology , Vaccination/veterinary
8.
Microbiol Immunol ; 63(12): 517-522, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31595535

ABSTRACT

We conducted a serological survey to detect antibodies against influenza A virus (IAV) in Japanese wild boars in Kagoshima prefecture, Japan, between 2014 and 2017. Seroprevalence against a pandemic-like swine H1N1 (H1N1pdm) virus was identified in 27.1% of specimens, and 1.7% were positive for both swine H1N2 and H3N2 viruses, indicating that wild boars could play an important role in the dynamics of H1N1pdm viral dispersion in the wild. The high frequency of positive results for sera against the H1N1pdm virus suggests that cross-species IAV transmission between wild boars, livestock, and humans is a threat to veterinary and public health.


Subject(s)
Antibodies, Viral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human , Orthomyxoviridae Infections , Animals , Humans , Influenza, Human/epidemiology , Influenza, Human/virology , Japan , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Seroepidemiologic Studies , Sus scrofa/virology , Swine , Swine Diseases/virology
9.
Nat Commun ; 9(1): 4418, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356117

ABSTRACT

The response to respiratory viruses varies substantially between individuals, and there are currently no known molecular predictors from the early stages of infection. Here we conduct a community-based analysis to determine whether pre- or early post-exposure molecular factors could predict physiologic responses to viral exposure. Using peripheral blood gene expression profiles collected from healthy subjects prior to exposure to one of four respiratory viruses (H1N1, H3N2, Rhinovirus, and RSV), as well as up to 24 h following exposure, we find that it is possible to construct models predictive of symptomatic response using profiles even prior to viral exposure. Analysis of predictive gene features reveal little overlap among models; however, in aggregate, these genes are enriched for common pathways. Heme metabolism, the most significantly enriched pathway, is associated with a higher risk of developing symptoms following viral exposure. This study demonstrates that pre-exposure molecular predictors can be identified and improves our understanding of the mechanisms of response to respiratory viruses.


Subject(s)
Gene Expression/genetics , Healthy Volunteers , Heme/metabolism , Humans , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H1N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/pathogenicity , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Rhinovirus/immunology , Rhinovirus/pathogenicity
10.
PLoS One ; 13(4): e0194765, 2018.
Article in English | MEDLINE | ID: mdl-29677213

ABSTRACT

The innate immune system is paramount in the response to and clearance of influenza A virus (IAV) infection in non-immune individuals. Known factors include type I and III interferons and antiviral pathogen recognition receptors, and the cascades of antiviral and pro- and anti-inflammatory gene expression they induce. MicroRNAs (miRNAs) are increasingly recognized to participate in post-transcriptional modulation of these responses, but the temporal dynamics of how these players of the antiviral innate immune response collaborate to combat infection remain poorly characterized. We quantified the expression of miRNAs and protein coding genes in the lungs of pigs 1, 3, and 14 days after challenge with swine IAV (H1N2). Through RT-qPCR we observed a 400-fold relative increase in IFN-λ3 gene expression on day 1 after challenge, and a strong interferon-mediated antiviral response was observed on days 1 and 3 accompanied by up-regulation of genes related to the pro-inflammatory response and apoptosis. Using small RNA sequencing and qPCR validation we found 27 miRNAs that were differentially expressed after challenge, with the highest number of regulated miRNAs observed on day 3. In contrast, the number of protein coding genes found to be regulated due to IAV infection peaked on day 1. Pulmonary miRNAs may thus be aimed at fine-tuning the initial rapid inflammatory response after IAV infection. Specifically, we found five miRNAs (ssc-miR-15a, ssc-miR-18a, ssc-miR-21, ssc-miR-29b, and hsa-miR-590-3p)-four known porcine miRNAs and one novel porcine miRNA candidate-to be potential modulators of viral pathogen recognition and apoptosis. A total of 11 miRNAs remained differentially expressed 14 days after challenge, at which point the infection had cleared. In conclusion, the results suggested a role for miRNAs both during acute infection as well as later, with the potential to influence lung homeostasis and susceptibility to secondary infections in the lungs of pigs after IAV infection.


Subject(s)
Immunity, Innate/genetics , Influenza A Virus, H1N2 Subtype/immunology , Interferon-gamma/physiology , Lung/immunology , MicroRNAs/physiology , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Animals , Gene Expression Profiling , Interferon-gamma/genetics , Lung/metabolism , Orthomyxoviridae Infections/veterinary , Swine , Swine Diseases/genetics , Swine Diseases/immunology
11.
Transbound Emerg Dis ; 65(2): 465-475, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29034617

ABSTRACT

The transportation of poultry and related products for international trade contributes to transboundary pathogen spread and disease outbreaks worldwide. To prevent pathogen incursion through poultry products, many countries have regulations about animal health and poultry product quarantine. However, in Japan, animal products have been illegally introduced into the country in baggage and confiscated at the airport. Lately, the number of illegally imported poultry and the incursion risk of transboundary pathogens through poultry products have been increasing. In this study, we isolated avian influenza viruses (AIVs) from raw poultry products illegally imported to Japan by international passengers. Highly (H5N1 and H5N6) and low (H9N2 and H1N2) pathogenic AIVs were isolated from raw chicken and duck products carried by flight passengers. H5 and H9 isolates were phylogenetically closely related to viruses isolated from poultry in China, and haemagglutinin genes of H5N1 and H5N6 isolates belonged to clades 2.3.2.1c and 2.3.4.4, respectively. Experimental infections of H5 and H9 isolates in chickens and ducks demonstrated pathogenicity and tissue tropism to skeletal muscles. To prevent virus incursion by poultry products, it is important to encourage the phased cleaning based on the disease control and eradication and promote the reduction in contamination risk in animal products.


Subject(s)
Airports , Commerce , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza in Birds/virology , Poultry Products/virology , Travel , Animals , Antigens, Viral/immunology , Chickens/virology , China/epidemiology , Disease Outbreaks/prevention & control , Disease Outbreaks/veterinary , Ducks/virology , Food Microbiology , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Influenza in Birds/epidemiology , Japan , Meat/virology , Phylogeny , Poultry/virology , Poultry Diseases/epidemiology , RNA, Viral/genetics
12.
Transbound Emerg Dis ; 64(6): 2083-2092, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28181421

ABSTRACT

The aim of this study was to perform the complete genome sequence of a swine influenza A H1N2 virus strain isolated from a pig in Guanajuato, México (A/swine/Mexico/GtoDMZC01/2014) and to report its seroprevalence in 86 counties at the Central Bajio zone. To understand the evolutionary dynamics of the isolate, we undertook a phylogenetic analysis of the eight gene segments. These data revealed that the isolated virus is a reassortant H1N2 subtype, as its genes are derived from human (HA, NP, PA) and swine (M, NA, PB1, PB2 and NS) influenza viruses. Pig serum samples were analysed by the hemagglutination inhibition test, using wild H1N2 and H3N2 strains (A/swine/México/Mex51/2010 [H3N2]) as antigen sources. Positive samples to the H1N2 subtype were processed using the field-isolated H1N1 subtype (A/swine/México/Ver37/2010 [H1N1]). Seroprevalence to the H1N2 subtype was 26.74% in the sampled counties, being Jalisco the state with highest seroprevalence to this subtype (35.30%). The results herein reported demonstrate that this new, previously unregistered influenza virus subtype in México that shows internal genes from other swine viral subtypes isolated in the past 5 years, along with human virus-originated genes, is widely distributed in this area of the country.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections/veterinary , Reassortant Viruses , Animals , Hemagglutination Inhibition Tests/veterinary , Humans , Influenza A Virus, H1N2 Subtype/classification , Influenza A Virus, H1N2 Subtype/immunology , Mexico/epidemiology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Phylogeny , Seroepidemiologic Studies , Swine
13.
Vaccine ; 35(8): 1124-1131, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28117173

ABSTRACT

We have recently demonstrated the effectiveness of an influenza A virus (IAV) subunit vaccine based on biodegradable polyanhydride nanoparticles delivery in mice. In the present study, we evaluated the efficacy of ∼200nm polyanhydride nanoparticles encapsulating inactivated swine influenza A virus (SwIAV) as a vaccine to induce protective immunity against a heterologous IAV challenge in pigs. Nursery pigs were vaccinated intranasally twice with inactivated SwIAV H1N2 (KAg) or polyanhydride nanoparticle-encapsulated KAg (KAg nanovaccine), and efficacy was evaluated against a heterologous zoonotic virulent SwIAV H1N1 challenge. Pigs were monitored for fever daily. Local and systemic antibody responses, antigen-specific proliferation of peripheral blood mononuclear cells, gross and microscopic lung lesions, and virus load in the respiratory tract were compared among the groups of animals. Our pre-challenge results indicated that KAg nanovaccine induced virus-specific lymphocyte proliferation and increased the frequency of CD4+CD8αα+ T helper and CD8+ cytotoxic T cells in peripheral blood mononuclear cells. KAg nanovaccine-immunized pigs were protected from fever following SwIAV challenge. In addition, pigs immunized with the KAg nanovaccine presented with lower viral antigens in lung sections and had 6 to 8-fold reduction in nasal shedding of SwIAV four days post-challenge compared to control animals. Immunologically, increased IFN-γ secreting T lymphocyte populations against both the vaccine and challenge viruses were detected in KAg nanovaccine-immunized pigs compared to the animals immunized with KAg alone. However, in the KAg nanovaccine-immunized pigs, hemagglutination inhibition, IgG and IgA antibody responses, and virus neutralization titers were comparable to that in the animals immunized with KAg alone. Overall, our data indicated that intranasal delivery of polyanhydride-based SwIAV nanovaccine augmented antigen-specific cellular immune response in pigs, with promise to induce cross-protective immunity.


Subject(s)
Antibodies, Viral/biosynthesis , Influenza Vaccines/administration & dosage , Nanoparticles/administration & dosage , Orthomyxoviridae Infections/veterinary , Swine Diseases/prevention & control , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Helper-Inducer/drug effects , Administration, Intranasal , Animals , Cell Proliferation/drug effects , Hemagglutination Inhibition Tests , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/drug effects , Influenza A Virus, H1N2 Subtype/immunology , Influenza Vaccines/chemistry , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Lung/drug effects , Lung/immunology , Lung/virology , Nanoparticles/chemistry , Nanoparticles/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Polyanhydrides/chemistry , Polyanhydrides/metabolism , Swine , Swine Diseases/immunology , Swine Diseases/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Vaccines, Inactivated , Viral Load/drug effects
15.
J Virol ; 90(20): 9364-82, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27512056

ABSTRACT

UNLABELLED: Pigs are natural hosts for influenza A viruses and play a critical role in influenza epidemiology. However, little is known about their influenza-evoked T-cell response. We performed a thorough analysis of both the local and systemic T-cell response in influenza virus-infected pigs, addressing kinetics and phenotype as well as multifunctionality (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], and interleukin-2 [IL-2]) and cross-reactivity. A total of 31 pigs were intratracheally infected with an H1N2 swine influenza A virus (FLUAVsw) and consecutively euthanized. Lungs, tracheobronchial lymph nodes, and blood were sampled during the first 15 days postinfection (p.i.) and at 6 weeks p.i. Ex vivo flow cytometry of lung lymphocytes revealed an increase in proliferating (Ki-67(+)) CD8(+) T cells with an early effector phenotype (perforin(+) CD27(+)) at day 6 p.i. Low frequencies of influenza virus-specific IFN-γ-producing CD4(+) and CD8(+) T cells could be detected in the lung as early as 4 days p.i. On consecutive days, influenza virus-specific CD4(+) and CD8(+) T cells produced mainly IFN-γ and/or TNF-α, reaching peak frequencies around day 9 p.i., which were up to 30-fold higher in the lung than in tracheobronchial lymph nodes or blood. At 6 weeks p.i., CD4(+) and CD8(+) memory T cells had accumulated in lung tissue. These cells showed diverse cytokine profiles and in vitro reactivity against heterologous influenza virus strains, all of which supports their potential to combat heterologous influenza virus infections in pigs. IMPORTANCE: Pigs not only are a suitable large-animal model for human influenza virus infection and vaccine development but also play a central role in the emergence of new pandemic strains. Although promising candidate universal vaccines are tested in pigs and local T cells are the major correlate of heterologous control, detailed and targeted analyses of T-cell responses at the site of infection are scarce. With the present study, we provide the first detailed characterization of magnitude, kinetics, and phenotype of specific T cells recruited to the lungs of influenza virus-infected pigs, and we could demonstrate multifunctionality, cross-reactivity, and memory formation of these cells. This, and ensuing work in the pig, will strengthen the position of this species as a large-animal model for human influenza virus infection and will immediately benefit vaccine development for improved control of influenza virus infections in pigs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross Reactions/immunology , Influenza A Virus, H1N2 Subtype/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Swine Diseases/immunology , Animals , CD4-Positive T-Lymphocytes/virology , Influenza Vaccines/immunology , Interferon-gamma/immunology , Interleukin-2/immunology , Lung/virology , Swine , Swine Diseases/virology , Tumor Necrosis Factor-alpha/immunology
16.
Biologicals ; 44(4): 252-256, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27230301

ABSTRACT

A novel triple reasserted H1N2 virus A/swine/Shanghai/1/2007 (SH07) was isolated from nasal swabs of weaned pig showing clinical symptoms of coughing and sneezing. To explore the virus characteristics, mice, chickens and pigs were selected for pathogenicity study. Pigs inoculated intranasally with 10(6) TCID50 SH07 showed clinical symptoms with coughing and sneezing, but no death. The virus nuclear acid was detected in many tissues using real-time PCR, which was mainly distributed in respiratory system particularly in the lungs. The virus was low-pathogenic to chickens with 10(6) TCID50 dose inoculation either via intramuscular or intranasal routes. However virus nuclear acid detection and virus isolation confirmed that the virus can also be found in nasal and rectum. When virus was inoculated into mice by intramuscular or intranasal routes we observed 100% and 80% lethality respectively. The third generation of samples passaged on MDCK cell were SIV positive in indirect immunofluorescence assay (IFA) using antiserum against H1N2 SIV. Furthermore, the lungs of mice showed obvious lesion with interstitial pneumonia. Data in our study suggest that SH07 is preferentially pathogenic to mammals rather than birds although it is a reasserting virus with the fragments from swine, human and avian origin.


Subject(s)
Influenza A Virus, H1N2 Subtype/immunology , Influenza in Birds/immunology , Orthomyxoviridae Infections/immunology , Swine Diseases/immunology , Animals , Chickens , Dogs , Host Specificity/immunology , Humans , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/pathogenicity , Influenza in Birds/virology , Lung/immunology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , Microscopy, Fluorescence , Nasal Cavity/immunology , Nasal Cavity/virology , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Rectum/immunology , Rectum/virology , Reverse Transcriptase Polymerase Chain Reaction , Swine , Swine Diseases/virology , Virulence/immunology
17.
J Gen Virol ; 97(7): 1489-1499, 2016 07.
Article in English | MEDLINE | ID: mdl-27031847

ABSTRACT

Multiple subtypes and many antigenic variants of influenza A virus (IAV) co-circulate in swine in the USA, complicating effective use of commercial vaccines to control disease and transmission. Whole inactivated virus (WIV) vaccines may provide partial protection against IAV with substantial antigenic drift, but have been shown to induce vaccine-associated enhanced respiratory disease (VAERD) when challenged with an antigenic variant of the same haemagglutinin (HA) subtype. This study investigated the role the immune response against HA, neuraminidase (NA) and nucleoprotein (NP) may play in VAERD by reverse engineering vaccine and challenge viruses on a common backbone and using them in a series of vaccination/challenge trials. Mismatched HA between vaccine and challenge virus was necessary to induce VAERD. However, vaccines containing a matched NA abrogated the VAERD phenomenon induced by the HA mismatch and this was correlated with NA-inhibiting (NI) antibodies. Divergence between the two circulating swine N2 lineages (92 % identity) resulted in a loss of NI cross-reactivity and also resulted in VAERD with the mismatched HA. The NP lineage selected for use in the WIV vaccine strains did not affect protection or pathology. Thus the combination of HA and NA in the vaccine virus strains played a substantial role in vaccine protection versus immunopathology, suggesting that vaccines that target the HA protein alone could be more prone to VAERD due to the absence of cross-protective NI antibodies.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Orthomyxoviridae Infections/virology , Swine Diseases/virology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Neuraminidase/immunology , Nucleoproteins/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Respiratory System/virology , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control , Vaccination , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology
18.
J Med Virol ; 88(10): 1725-32, 2016 10.
Article in English | MEDLINE | ID: mdl-26950895

ABSTRACT

Current seasonal influenza vaccines require regular updates due to antigenic drift causing loss of effectiveness and therefore providing little or no protection against novel influenza A subtypes. Next generation vaccines capable of eliciting CD8(+) T cell (CTL) mediated cross-protective immunity may offer a long-term alternative strategy. However, measuring pre- and existing levels of CTL cross-protection in humans is confounded by differences in infection histories across individuals. During 2000-2003, H1N2 viruses circulated persistently in the human population for the first time and we hypothesized that the viral nucleoprotein (NP) contained novel CTL epitopes that may have contributed to the survival of the viruses. This study describes the immunogenic NP peptides of H1N1, H2N2, and H3N2 influenza viruses isolated from humans over the past century, 1918-2003, by comparing this historical dataset to reference NP peptides from H1N2 that circulated in humans during 2000-2003. Observed peptides sequences ranged from highly conserved (15%) to highly variable (12%), with variation unrelated to reported immunodominance. No unique NP peptides which were exclusive to the H1N2 viruses were noted. However, the virus had inherited the NP from a recently emerged H3N2 variant containing novel peptides, which may have assisted its persistence. Any advantage due to this novelty was subsequently lost with emergence of a newer H3N2 variant in 2003. Our approach has potential to provide insight into the population context in which influenza viruses emerge, and may help to inform immunogenic peptide selection for CTL-inducing influenza vaccines. J. Med. Virol. 88:1725-1732, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cross Protection , Epitopes, T-Lymphocyte , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Animals , Antibodies, Viral/immunology , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Nucleocapsid Proteins , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Peptides/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
19.
Sci Rep ; 6: 21812, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26893019

ABSTRACT

MicroRNAs (miRNAs) are a class of short regulatory RNA molecules which are implicated in modulating gene expression. Levels of circulating, cell-associated miRNAs in response to influenza A virus (IAV) infection has received limited attention so far. To further understand the temporal dynamics and biological implications of miRNA regulation in circulating leukocytes, we collected blood samples before and after (1, 3, and 14 days) IAV challenge of pigs. Differential expression of miRNAs and innate immune factor mRNA transcripts was analysed using RT-qPCR. A total of 20 miRNAs were regulated after IAV challenge, with the highest number of regulated miRNAs seen on day 14 after infection at which time the infection was cleared. Targets of the regulated miRNAs included genes involved in apoptosis and cell cycle regulation. Significant regulation of both miRNAs and mRNA transcripts at 14 days after challenge points to a protracted effect of IAV infection, potentially affecting the host's ability to respond to secondary infections. In conclusion, experimental IAV infection of pigs demonstrated the dynamic nature of miRNA and mRNA regulation in circulating leukocytes during and after infection, and revealed the need for further investigation of the potential immunosuppressing effect of miRNA and innate immune signaling after IAV infection.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate , Influenza A Virus, H1N2 Subtype/immunology , Influenza, Human/blood , Leukocytes, Mononuclear/immunology , Animals , Gene Expression Profiling , Humans , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/virology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , MicroRNAs/blood , MicroRNAs/genetics , RNA, Messenger/blood , RNA, Messenger/genetics , Sus scrofa , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...