Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
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
2.
J Virol ; 98(3): e0199523, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38323813

ABSTRACT

Historically, antibody reactivity to pathogens and vaccine antigens has been evaluated using serological measurements of antigen-specific antibodies. However, it is difficult to evaluate all antibodies that contribute to various functions in a single assay, such as the measurement of the neutralizing antibody titer. Bulk antibody repertoire analysis using next-generation sequencing is a comprehensive method for analyzing the overall antibody response; however, it is unreliable for estimating antigen-specific antibodies due to individual variation. To address this issue, we propose a method to subtract the background signal from the repertoire of data of interest. In this study, we analyzed changes in antibody diversity and inferred the heavy-chain complementarity-determining region 3 (CDRH3) sequences of antibody clones that were selected upon influenza virus infection in a mouse model using bulk repertoire analysis. A decrease in the diversity of the antibody repertoire was observed upon viral infection, along with an increase in neutralizing antibody titers. Using kernel density estimation of sequences in a high-dimensional sequence space with background signal subtraction, we identified several clusters of CDRH3 sequences induced upon influenza virus infection. Most of these repertoires were detected more frequently in infected mice than in uninfected control mice, suggesting that infection-specific antibody sequences can be extracted using this method. Such an accurate extraction of antigen- or infection-specific repertoire information will be a useful tool for vaccine evaluation in the future. IMPORTANCE: As specific interactions between antigens and cell-surface antibodies trigger the proliferation of B-cell clones, the frequency of each antibody sequence in the samples reflects the size of each clonal population. Nevertheless, it is extremely difficult to extract antigen-specific antibody sequences from the comprehensive bulk antibody sequences obtained from blood samples due to repertoire bias influenced by exposure to dietary antigens and other infectious agents. This issue can be addressed by subtracting the background noise from the post-immunization or post-infection repertoire data. In the present study, we propose a method to quantify repertoire data from comprehensive repertoire data. This method allowed subtraction of the background repertoire, resulting in more accurate extraction of expanded antibody repertoires upon influenza virus infection. This accurate extraction of antigen- or infection-specific repertoire information is a useful tool for vaccine evaluation.


Subject(s)
Antibodies, Viral , Orthomyxoviridae Infections , Orthomyxoviridae , Animals , Mice , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Clone Cells/cytology , Clone Cells/immunology , Complementarity Determining Regions/immunology , Influenza Vaccines/immunology , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
3.
Viruses ; 14(2)2022 02 02.
Article in English | MEDLINE | ID: mdl-35215905

ABSTRACT

Infectious salmon anaemia virus (ISAV) binds circulating Atlantic salmon erythrocytes, but the relevance of this interaction for the course of infection and development of disease remains unclear. We here characterise ISAV-erythrocyte interactions in experimentally infected Atlantic salmon and show that ISAV-binding to erythrocytes is common and precedes the development of disease. Viral RNA and infective particles were enriched in the cellular fraction of blood. While erythrocyte-associated ISAV remained infectious, erythrocytes dose-dependently limited the infection of cultured cells. Surprisingly, immunostaining of blood smears revealed expression of ISAV proteins in a small fraction of erythrocytes in one of the examined trials, confirming that ISAV can be internalised in this cell type and engage the cellular machinery in transcription and translation. However, viral protein expression in erythrocytes was rare and not required for development of disease and mortality. Furthermore, active transcription of ISAV mRNA was higher in tissues than in blood, supporting the assumption that ISAV replication predominantly takes place in endothelial cells. In conclusion, Atlantic salmon erythrocytes bind ISAV and sequester infective virus particles during infection, but do not appear to significantly contribute to ISAV replication. We discuss the implications of our findings for infection dynamics and pathogenesis of infectious salmon anaemia.


Subject(s)
Erythrocytes/virology , Fish Diseases/virology , Isavirus/physiology , Orthomyxoviridae Infections/veterinary , Salmo salar/virology , Animals , Endothelial Cells/virology , Fish Diseases/blood , Isavirus/genetics , Isavirus/isolation & purification , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , Salmo salar/blood , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics , Virion/isolation & purification , Virion/physiology , Virus Replication
4.
PLoS One ; 17(1): e0262832, 2022.
Article in English | MEDLINE | ID: mdl-35051238

ABSTRACT

Tumor progression locus 2 (Tpl2) is a serine/threonine kinase that regulates the expression of inflammatory mediators in response to Toll-like receptors (TLR) and cytokine receptors. Global ablation of Tpl2 leads to severe disease in response to influenza A virus (IAV) infection, characterized by respiratory distress, and studies in bone marrow chimeric mice implicated Tpl2 in non-hematopoietic cells. Lung epithelial cells are primary targets and replicative niches of influenza viruses; however, the specific regulation of antiviral responses by Tpl2 within lung epithelial cells has not been investigated. Herein, we show that Tpl2 is basally expressed in primary airway epithelial cells and that its expression increases in both type I and type II airway epithelial cells (AECI and AECII) in response to influenza infection. We used Nkx2.1-cre to drive Tpl2 deletion within pulmonary epithelial cells to delineate epithelial cell-specific functions of Tpl2 during influenza infection in mice. Although modest increases in morbidity and mortality were attributed to cre-dependent deletion in lung epithelial cells, no alterations in host cytokine production or lung pathology were observed. In vitro, Tpl2 inhibition within the type I airway epithelial cell line, LET1, as well as genetic ablation in primary airway epithelial cells did not alter cytokine production. Overall, these findings establish that Tpl2-dependent defects in cells other than AECs are primarily responsible for the morbidity and mortality seen in influenza-infected mice with global Tpl2 ablation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Host Microbial Interactions/genetics , Influenza A virus , MAP Kinase Kinase Kinases/metabolism , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Proto-Oncogene Proteins/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Dogs , Female , MAP Kinase Kinase Kinases/genetics , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Proto-Oncogene Proteins/genetics
5.
Bull Exp Biol Med ; 171(6): 736-740, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34705177

ABSTRACT

We studied the effect of tilorone on the dynamics of IFNα, IFNγ, and IL-1ß levels in the lung tissue and blood serum in relation to viral load in the lungs of BALB/c mice with pneumonia caused by influenza virus A/Aichi/2/68 (H3N2). Tilorone was administered per os in doses of 40, 150, and 540 µg per mouse 6, 30, and 78 h postinfection, which simulated the drug regimen used in the clinic for the treatment of influenza and acute respiratory viral infections in Russia and post-Soviet countries. Tilorone reduced viral load with the maximum amplitude (2-3 lg) after 1-2 administrations. The results of studying the dynamics of the cytokine levels in the infected animals in general support the previous hypothesis that, in repeated dosing, tilorone enhances the IFN response (compensates for its deficiency) at the early stages of acute respiratory viral infections and suppresses (damps) excessive production of IFN and proinflammatory cytokines at the later stages.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H3N2 Subtype/drug effects , Interferon Inducers/pharmacology , Lung/drug effects , Orthomyxoviridae Infections/drug therapy , Tilorone/pharmacology , Animals , Drug Administration Schedule , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/pathogenicity , Interferon-alpha/blood , Interferon-alpha/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-1beta/blood , Interleukin-1beta/immunology , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Viral Load/drug effects
6.
Front Immunol ; 12: 689436, 2021.
Article in English | MEDLINE | ID: mdl-34335593

ABSTRACT

We investigated the reaction of mouse peritoneal mast cells (MCs) in vitro after IgG-containing immune complex introduction using A/H5N1 and A/H1N1pdm09 influenza viruses as antigens. The sera of immune mice served as a source of IgG antibodies. The concentration of histamine in the supernatants was determined at 4 hours after incubation with antisera and virus. We compared the contribution of MCs to the pathogenesis of post-immunization influenza infection with A/H5N1 and A/H1N1 influenza viruses in mice. The mice were immunized parenterally with inactivated viruses and challenged with lethal doses of drift A/H5N1 and A/H1N1 influenza viruses on the 14th day after immunization. Simultaneously, half of the mice were injected intraperitoneally with a mixture of histamine receptor blockers (chloropyramine and quamatel). In in vitro experiments, the immune complex formed by A/H5N1 virus and antiserum caused a significant increase in the histamine release compared to immune serum or the virus alone. With regard to the A/H1N1 virus, such an increase was not significant. A/H1N1 immunization caused detectable HI response in mice at 12th day after immunization, in contrast to the A/H5N1 virus. After challenge of A/H5N1-immunized mice, administration of antihistamines increased the survival rate by up to 90%. When infecting the A/H1N1-immunized mice, 90% of the animals were already protected from lethal infection by day 14; the administration of histamine receptor blockers did not increase survival. Histological examination of the lungs has shown that toluidine blue staining allows to estimate the degree of MC degranulation. The possibility of in vitro activation of murine MCs by IgG-containing immune complexes has been shown. In a model of influenza infection, it was shown that the administration of histamine receptor blockers increased survival. When the protection was formed faster due to the earlier production of HI antibodies, the administration of histamine receptor blockers did not significantly affect the course of the infection. These data allow to propose that even if there are antibody-dependent MC reactions, they can be easily stopped by the administration of histamine receptor blockers.


Subject(s)
Antibodies, Viral/blood , Cell Degranulation , Histamine Release , Immunoglobulin G/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Lung/immunology , Mast Cells/immunology , Orthomyxoviridae Infections/immunology , Animals , Cell Degranulation/drug effects , Disease Models, Animal , Female , Histamine Antagonists/pharmacology , Histamine Release/drug effects , Host-Pathogen Interactions , Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Lung/drug effects , Lung/metabolism , Lung/virology , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/virology , Mice, Inbred CBA , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Severity of Illness Index , Time Factors , Vaccination
7.
Front Immunol ; 12: 692700, 2021.
Article in English | MEDLINE | ID: mdl-34335603

ABSTRACT

The highly pathogenic avian influenza (HPAI) H5N1 viruses with the capability of transmission from birds to humans have a serious impact on public health. To date, HPAI H5N1 viruses have evolved into ten antigenically distinct clades that could cause a mismatch of vaccine strains and reduce vaccine efficacy. In this study, the glycan masking and unmasking strategies on hemagglutinin antigen were used for designing two antigens: H5-dm/st2 and H5-tm/st2, and investigated for their elicited immunity using two-dose recombinant H5 (rH5) immunization and a first-dose adenovirus vector prime, followed by a second-dose rH5 protein booster immunization. The H5-dm/st2 antigen was found to elicit broadly neutralizing antibodies against different H5N1 clade/subclade viruses, as well as more stem-binding antibodies to inhibit HA-facilitated membrane fusion activity. Mice immunized with the H5-dm/st2 antigen had a higher survival rate when challenged with homologous and heterologous clades of H5N1 viruses. Mutant influenza virus replaced with the H5-dm/st2 gene generated by reverse genetics (RG) technology amplified well in MDCK cells and embryonated chicken eggs. Again, the inactivated H5N1-dm/st2 RG virus elicited more potent cross-clade neutralizing and anti-fusion antibodies in sera. Therefore, the H5N1-dm/st2 RG virus with the site-specific glycan-masking on the globular head and the glycan-unmasking on the stem region of H5 antigen can be used for further development of cross-protective H5N1 vaccines.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Broadly Neutralizing Antibodies/blood , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Immunodominant Epitopes , Immunogenicity, Vaccine , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Polysaccharides/administration & dosage , Animals , Antigens, Viral/immunology , Chick Embryo , Disease Models, Animal , Dogs , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunization , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/immunology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Polysaccharides/immunology
8.
Nat Commun ; 12(1): 1203, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619277

ABSTRACT

Influenza A virus infection in swine impacts the agricultural industry in addition to its zoonotic potential. Here, we utilize epigraph, a computational algorithm, to design a universal swine H3 influenza vaccine. The epigraph hemagglutinin proteins are delivered using an Adenovirus type 5 vector and are compared to a wild type hemagglutinin and the commercial inactivated vaccine, FluSure. In mice, epigraph vaccination leads to significant cross-reactive antibody and T-cell responses against a diverse panel of swH3 isolates. Epigraph vaccination also reduces weight loss and lung viral titers in mice after challenge with three divergent swH3 viruses. Vaccination studies in swine, the target species for this vaccine, show stronger levels of cross-reactive antibodies and T-cell responses after immunization with the epigraph vaccine compared to the wild type and FluSure vaccines. In both murine and swine models, epigraph vaccination shows superior cross-reactive immunity that should be further investigated as a universal swH3 vaccine.


Subject(s)
Algorithms , Cross Reactions/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity , Influenza A virus/immunology , Influenza Vaccines/immunology , Animals , Antibody Formation/immunology , Epitopes/immunology , Female , Humans , Influenza, Human/blood , Influenza, Human/immunology , Influenza, Human/virology , Lung/pathology , Lung/virology , Male , Mice, Inbred BALB C , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Swine , T-Lymphocytes/immunology , Vaccination , Weight Loss
9.
Zoonoses Public Health ; 68(3): 239-246, 2021 05.
Article in English | MEDLINE | ID: mdl-33576190

ABSTRACT

In January 2019, four cheetahs (Acinonyx jubatus) kept at a Japanese zoo intermittently showed respiratory signs following the incidence of seasonal influenza in animal caregivers. Respiratory materials (saliva, sputum and food tray swabs) were non-invasively collected from the four cheetahs. Although we were unable to isolate the virus, the NP gene of influenza A virus was detected in three of the cheetahs but not in the fourth cheetah that had nearly recovered. From a food tray swab which tested weakly positive by a commercial influenza detection kit, we were able to obtain the whole-genome sequence of the influenza A virus. Analysis of the genome, A/cheetah/Kanagawa/2/2019(H1N1), revealed that the virus was closely related to influenza A(H1N1)pdm09 viruses isolated from humans in Japan in the 2018-2019 winter. Production of haemagglutinin inhibition (HI) antibodies (64-128 HI) against an A(H1N1)pdm09 virus in plasma samples confirmed infection of all four cheetahs. The animals continued to produce antibodies for at least 314 days after disease onset. These findings strongly suggest that reverse zoonotic transmission of A(H1N1)pdm09 virus occurred from human to cheetah and subsequently from cheetah to cheetah in the zoo. We also show that specimens can be safely and non-invasively collected from non-domesticated animals and used to investigate respiratory infectious diseases.


Subject(s)
Acinonyx , Influenza A Virus, H1N1 Subtype/isolation & purification , Orthomyxoviridae Infections/veterinary , Animals , Animals, Zoo , Antibodies, Viral/blood , Antigens, Viral/isolation & purification , Cluster Analysis , Environmental Microbiology , Female , Japan/epidemiology , Male , Nasopharynx/virology , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Saliva/virology
10.
J Vet Diagn Invest ; 32(6): 887-891, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33025860

ABSTRACT

Highly pathogenic influenza A viruses (IAVs) cause substantial damage to the poultry industry. A simple and quick testing method is required for strict control of this infectious agent. The fluorescence polarization immunoassay (FPIA) is a rapid test based on antigen-antibody binding, which can detect antigen-specific antibody in the infected animal samples within a few minutes. FPIA is a one-step reaction assay that does not require a secondary antibody and complicated steps. We evaluated the usefulness of FPIA for the detection of anti-IAV antibodies, including those against internal proteins and H5 subtype HA, in sera. In the FPIA using fluorescent peptides of internal NP and M1 proteins, millipolarization units (MPUs), which increase depending on the amount of antibody, were higher in antibody-positive sera than in antibody-negative sera. Moreover, in FPIA using fluorescent recombinant H5 subtype HA proteins, anti-H5 serum gave the highest MPUs among the antisera raised in goats against individual H1-H15 subtype IAVs. Our results support the utility of FPIA for the detection of anti-IAV antibodies, especially the anti-H5 antibody.


Subject(s)
Antibodies, Viral/blood , Chickens/blood , Fluorescence Polarization Immunoassay/veterinary , Goats/blood , Influenza A virus/immunology , Orthomyxoviridae Infections/veterinary , Animals , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae Infections/virology , Time Factors
11.
Nutrients ; 12(7)2020 Jul 05.
Article in English | MEDLINE | ID: mdl-32635656

ABSTRACT

Vitamin D is a fat-soluble vitamin that is metabolized by the liver into 25-hydroxyvitamin D [25(OH)D] and then by the kidney into 1,25-dihydroxyvitamin D [1,25(OH)2D], which activates the vitamin D receptor expressed in various cells, including immune cells, for an overall immunostimulatory effect. Here, to investigate whether oral supplementation of 25-hydroxyvitamin D3 [25(OH)D3], a major form of vitamin D metabolite 25(OH)D, has a prophylactic effect on influenza A virus infection, mice were fed a diet containing a high dose of 25(OH)D3 and were challenged with the influenza virus. In the lungs of 25(OH)D3-fed mice, the viral titers were significantly lower than in the lungs of standardly fed mice. Additionally, the proinflammatory cytokines IL-5 and IFN-γ were significantly downregulated after viral infection in 25(OH)D3-fed mice, while anti-inflammatory cytokines were not significantly upregulated. These results indicate that 25(OH)D3 suppresses the production of inflammatory cytokines and reduces virus replication and clinical manifestations of influenza virus infection in a mouse model.


Subject(s)
Calcifediol/administration & dosage , Dietary Supplements , Influenza A virus/drug effects , Orthomyxoviridae Infections/prevention & control , Vitamins/administration & dosage , Animals , Cytokines/blood , Disease Models, Animal , Down-Regulation/drug effects , Interferon-gamma/blood , Interleukin-5/blood , Lung/virology , Mice , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , Up-Regulation/drug effects , Virus Replication/drug effects
12.
Int Immunopharmacol ; 84: 106521, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32315950

ABSTRACT

Platelets have been proved to exacerbate influenza infection and its complications. Inhibition of platelet activation may be a feasible method for preventing severe infection and secondary acute lung injury (ALI). Isofraxidin (IFD) is a natural coumarin isolated from the plants Sarcandra glabra and Siberian ginseng, and exerts anticancer, antioxidant and antiinflammatory effects. In the present study, we examined the therapeutic effects of IFD in ADP- or arachidonic acid (AA)-induced platelet aggregation model and in influenza A virus (IAV)-induced ALI mouse model. The results showed that IFD significantly inhibited platelet aggregation induced by ADP and AA in vitro in a concentration-dependent manner as well as the release of soluble P-selectin and platelet factor 4. Moreover, IFD significantly relieved IAV-induced lung inflammation, reduced the expressions of platelet activation biomarkers (P-selectin and CD61), decreased the serum levels of TNF-α, IL-1ß, IL-6 and MIP-2, suppressed peripheral platelet aggregation and prolonged the survival time of infected mice. The western blotting results also demonstrated that IFD reduced the phosphorylation levels of PI3K, AKT and p38 in the activated platelets stimulated by ADP and IAV infection. But IFD did not have any effects on IAV replication. It indicated that IFD ameliorated IAV-induced severe lung damage and lethal infection by suppressing platelet aggregation via regulating PI3K/AKT and MAPK pathways.


Subject(s)
Acute Lung Injury/drug therapy , Alphainfluenzavirus , Anti-Inflammatory Agents/therapeutic use , Coumarins/therapeutic use , Orthomyxoviridae Infections/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Acute Lung Injury/blood , Acute Lung Injury/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Coumarins/pharmacology , Cytokines/blood , Dogs , Inflammation , Madin Darby Canine Kidney Cells , Male , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/metabolism , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley
13.
J Infect Dis ; 222(5): 820-831, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32246148

ABSTRACT

BACKGROUND: Influenza A virus (IAV) causes a wide range of extrarespiratory complications. However, the role of host factors in these complications of influenza virus infection remains to be defined. METHODS: Here, we sought to use transcriptional profiling, virology, histology, and echocardiograms to investigate the role of a high-fat diet in IAV-associated cardiac damage. RESULTS: Transcriptional profiling showed that, compared to their low-fat counterparts (LF mice), mice fed a high-fat diet (HF mice) had impairments in inflammatory signaling in the lung and heart after IAV infection. This was associated with increased viral titers in the heart, increased left ventricular mass, and thickening of the left ventricular wall in IAV-infected HF mice compared to both IAV-infected LF mice and uninfected HF mice. Retrospective analysis of clinical data revealed that cardiac complications were more common in patients with excess weight, an association which was significant in 2 out of 4 studies. CONCLUSIONS: Together, these data provide the first evidence that a high-fat diet may be a risk factor for the development of IAV-associated cardiovascular damage and emphasizes the need for further clinical research in this area.


Subject(s)
Diet, High-Fat , Heart Diseases/virology , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/complications , Animals , Body Mass Index , Body Weight , Cytokines/blood , Cytokines/genetics , Echocardiography , Female , Gene Expression Profiling , Heart/virology , Heart Diseases/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Influenza, Human/complications , Interferon Regulatory Factor-7/genetics , Interleukin-1beta/genetics , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , RNA, Viral/metabolism , Risk Factors , Signal Transduction/genetics , Ubiquitins/genetics
14.
Methods Mol Biol ; 2123: 401-412, 2020.
Article in English | MEDLINE | ID: mdl-32170706

ABSTRACT

Serologic tests for equine influenza virus (EIV) antibodies are used for many purposes, including retrospective diagnosis, subtyping of virus isolates, antigenic comparison of different virus strains, and measurement of immune responses to EIV vaccines. The hemagglutination inhibition (HI) assay, single radial hemolysis (SRH), and serum micro-neutralization tests are the most widely used for these purposes and are described here. The presence of inhibitors of hemagglutination in equine serum complicates interpretation of HI assay results, and there are alternative protocols (receptor-destroying enzyme, periodate, trypsin-periodate) for their removal. With the EIV H3N8 strains in particular, equine antibody titers may be magnified by pre-treating the HI test antigen with Tween-80 and ether. The SRH assay offers stronger correlations between serum antibody titers and protection from disease. Other tests are sometimes used for specialized purposes such as the neuraminidase-inhibition assay for subtyping, or ELISA for measuring different specific antibody isotypes, and are not described here.


Subject(s)
Horses/blood , Horses/virology , Influenza A Virus, H3N8 Subtype/physiology , Serologic Tests/methods , Animals , Dogs , Hemagglutination Inhibition Tests , Hemolysis , Horse Diseases/blood , Horse Diseases/virology , Madin Darby Canine Kidney Cells , Neutralization Tests , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Serum/metabolism
15.
Front Immunol ; 11: 604913, 2020.
Article in English | MEDLINE | ID: mdl-33603740

ABSTRACT

We have used the pig, a large natural host animal for influenza with many physiological similarities to humans, to characterize αß, γδ T cell and antibody (Ab) immune responses to the 2009 pandemic H1N1 virus infection. We evaluated the kinetic of virus infection and associated response in inbred Babraham pigs with identical MHC (Swine Leucocyte Antigen) and compared them to commercial outbred animals. High level of nasal virus shedding continued up to days 4 to 5 post infection followed by a steep decline and clearance of virus by day 9. Adaptive T cell and Ab responses were detectable from days 5 to 6 post infection reaching a peak at 9 to 14 days. γδ T cells produced cytokines ex vivo at day 2 post infection, while virus reactive IFNγ producing γδ T cells were detected from day 7 post infection. Analysis of NP tetramer specific and virus specific CD8 and CD4 T cells in blood, lung, lung draining lymph nodes, and broncho-alveolar lavage (BAL) showed clear differences in cytokine production between these tissues. BAL contained the most highly activated CD8, CD4, and γδ T cells producing large amounts of cytokines, which likely contribute to elimination of virus. The weak response in blood did not reflect the powerful local lung immune responses. The immune response in the Babraham pig following H1N1pdm09 influenza infection was comparable to that of outbred animals. The ability to utilize these two swine models together will provide unparalleled power to analyze immune responses to influenza.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/virology , T-Lymphocyte Subsets/virology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cytokines/metabolism , Disease Models, Animal , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Host-Pathogen Interactions , Inbreeding , Influenza A Virus, H1N1 Subtype/pathogenicity , Kinetics , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Species Specificity , Sus scrofa , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Viral Load , Virus Shedding
16.
Anat Histol Embryol ; 49(5): 606-619, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31793053

ABSTRACT

Every year, influenza viruses spread around the world, infecting the respiratory systems of countless humans and animals, causing illness and even death. Severe influenza infection is associated with pulmonary epithelial damage and endothelial dysfunction leading to acute lung injury (ALI). There is evidence that an aggressive cytokine storm and cell damage in lung capillaries as well as endothelial/platelet interactions contribute to vascular leakage, pro-thrombotic milieu and infiltration of immune effector cells. To date, treatments for ALI caused by influenza are limited to antiviral drugs, active ventilation or further symptomatic treatments. In this review, we summarize the mechanisms of influenza-mediated pathogenesis, permissive animal models and histopathological changes of lung tissue in both mice and men and compare it with histological and electron microscopic data from our own group. We highlight the molecular and cellular interactions between pulmonary endothelium and platelets in homeostasis and influenza-induced pathogenesis. Finally, we discuss novel therapeutic targets on platelets/endothelial interaction to reduce or resolve ALI.


Subject(s)
Blood Platelets/physiology , Endothelium/physiology , Influenza, Human/blood , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae/genetics , Animals , Blood Platelets/metabolism , Disease Models, Animal , Humans , Influenza, Human/pathology , Orthomyxoviridae/classification , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/pathology , Platelet Activation , Pulmonary Alveoli/pathology
17.
Int J Nanomedicine ; 14: 9337-9349, 2019.
Article in English | MEDLINE | ID: mdl-31819435

ABSTRACT

BACKGROUND: Enzyme-linked immunosorbent assay (ELISA) is a common method for diagnosing swine influenza. However, the production of classical antibodies is both costly and time-consuming. As a promising alternative diagnostic tool, single-domain antibodies (sdAbs) offer the advantages of simpler and faster generation, good stability and solubility, and high affinity and specificity. METHODS: Phage display technology was used to isolate sdAbs against the SIV-NP protein from a camel VHH library. The sdAb5 was fused to the biotin acceptor peptide (BAP) and a His-Tag for its expression as monomeric and site-specific biotinylation in E.coli to develop an sdAb-based blocking ELISA (sdAb-ELISA). In the sdAb-ELISA, the anti-SIV antibodies from swine samples were used to block the binding between the biotinylated sdAb5 and SIV-NP protein coated on the ELISA plate. The specificity, sensitivity, and reproducibility of sdAb-ELISA were determined. In addition, consistency among sdAb-ELISA, commercial ELISA kit, and Western blot was evaluated. RESULTS: Six SIV-NP-specific sdAbs were isolated, among which sdAb5 was identified as a dominant sdAb with higher reactivity. The cut-off value of biotinylated sdAb5-based bELISA was determined to be 29.8%. Compared with the positive reference serum against five different types of swine viruses, the developed sdAb-ELISA showed 100% specificity. The detection limit of sdAb-ELISA was 1:160 in an anti-SIV positive reference serum, which is lower than that of the commercial ELISA kit (1:20). In 78 diluted anti-SIV positive serum (1:80), 21 and 42 samples were confirmed as positive by the commercial ELISA kit and sdAb-ELISA, respectively. The coefficients of variation of intra- and inter-assay were 1.79-4.57% and 5.54-9.98%, respectively. The sdAb-ELISA and commercial ELISA kit showed a consistency of 94.17% in clinical swine serum samples. Furthermore, the coincidence rate was 96.67% between the results detected by sdAb-ELISA and Western blot. CONCLUSION: A specific, sensitive, and reproducible sdAb-ELISA was successfully developed, which offers a new, promising method to detect anti-SIV antibodies in swine serum.


Subject(s)
Antibodies, Viral/immunology , Enzyme-Linked Immunosorbent Assay/methods , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/immunology , Single-Domain Antibodies/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Biotin/chemistry , Biotinylation , Camelus , Nucleoproteins/isolation & purification , Nucleoproteins/metabolism , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , Peptide Library , Reproducibility of Results , Single-Domain Antibodies/chemistry , Swine
18.
ACS Synth Biol ; 8(11): 2472-2482, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31565926

ABSTRACT

Hemagglutinin (HA) is the major surface antigen of influenza virus and the most promising influenza vaccine immunogen. In 2013, the devastating H7N9 influenza virus was identified in China, which induced high mortality. The HA of this virus (H7) is relatively unstable, making it challenging to produce an effective vaccine. To improve the stability of HA protein from H7N9 influenza virus for better vaccine antigens without impairing immunogenicity, we recombined the HA from H7N9 (H7) with a more stable HA from H3N2 (H3) by structure-guided recombination, resulting in six chimeric HAs, FrA-FrF. Two of these chimeric HAs, FrB and FrC, exhibited proper hemagglutination activity and presented improved thermal stability compared to the original H7. Mice immunized with FrB and FrC elicited H7-specific antibodies comparable to those induced by parental H7, and the antisera collected from these immunized mice successfully inhibited H7N9 infection in a microneutralization assay. These results suggest that our structural-recombination approach can create stabilizing chimeric antigens while maintaining proper immunogenicity, which may not only benefit the construction of more stable HA vaccines to fight against H7N9 infection, but also facilitate effective vaccine improvements for other influenza viruses or infectious pathogens. In addition, this study also demonstrates the potential for better engineering of multimeric protein complexes like HA to achieve improved function, which are often immunologically or pharmaceutically important but difficult to modify.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Hemagglutinins/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/therapy , Recombinant Fusion Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Female , Immunization/methods , Immunogenicity, Vaccine , Immunoglobulin G/blood , Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H7N9 Subtype/chemistry , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , Protein Stability , Recombination, Genetic , Treatment Outcome
19.
Sci Rep ; 9(1): 15128, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31641151

ABSTRACT

Enterotoxin-based adjuvants including cholera toxin and heat-labile toxin (LT) are powerful manipulators of mucosal immunity; however, past clinical trials identified unacceptable neurological toxicity when LT or mutant AB5 adjuvant proteins were added to intranasal vaccines. Here, we examined the isolated enzymatic A1 domain of LT (LTA1) for intranasal safety and efficacy in combination with influenza (flu) vaccination. LTA1-treated mice exhibited no neurotoxicity, as measured by olfactory system testing and H&E staining of nasal tissue in contrast with cholera toxin. In vaccination studies, intranasal LTA1 enhanced immune responses to inactivated virus antigen and subsequent protection against H1N1 flu challenge in mice (8-week or 24-months). In addition, lung H1N1 viral titers post-challenge correlated to serum antibody responses; however, enhanced protection was also observed in µMT mice lacking B-cells while activation and recruitment of CD4 T-cells into the lung was apparent. Thus, we report that LTA1 protein is a novel, safe and effective enterotoxin adjuvant that improves protection of an intranasal flu vaccination by a mechanism that does not appear to require B-cells.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aging/immunology , B-Lymphocytes/immunology , Enterotoxins/administration & dosage , Lymphocyte Depletion , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antibodies/blood , Antibody Formation/drug effects , B-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Enterotoxins/immunology , Enterotoxins/toxicity , Female , Immunity, Mucosal/drug effects , Immunization , Inflammation/pathology , Influenza A Virus, H1N1 Subtype/immunology , Lung/virology , Lymphocyte Activation/drug effects , Mast Cells/drug effects , Mast Cells/pathology , Mice, Inbred C57BL , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology
20.
J Exp Med ; 216(8): 1828-1842, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31196981

ABSTRACT

Mg2+ is required at micromolar concentrations as a cofactor for ATP, enzymatic reactions, and other biological processes. We show that decreased extracellular Mg2+ reduced intracellular Mg2+ levels and impaired the Ca2+ flux, activation marker up-regulation, and proliferation after T cell receptor (TCR) stimulation. Reduced Mg2+ specifically impairs TCR signal transduction by IL-2-inducible T cell kinase (ITK) due to a requirement for a regulatory Mg2+ in the catalytic pocket of ITK. We also show that altered catalytic efficiency by millimolar changes in free basal Mg2+ is an unrecognized but conserved feature of other serine/threonine and tyrosine kinases, suggesting a Mg2+ regulatory paradigm of kinase function. Finally, a reduced serum Mg2+ concentration in mice causes an impaired CD8+ T cell response to influenza A virus infection, reduces T cell activation, and exacerbates morbidity. Thus, Mg2+ directly regulates the active site of specific kinases during T cell responses, and maintaining a high serum Mg2+ concentration is important for antiviral immunity in otherwise healthy animals.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Magnesium/pharmacology , Orthomyxoviridae Infections/immunology , Protein-Tyrosine Kinases/metabolism , Animals , Biocatalysis/drug effects , Blood Donors , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Calcium/metabolism , Catalytic Domain/drug effects , Cells, Cultured , Humans , Lymphocyte Activation/drug effects , Magnesium/blood , Magnesium/chemistry , Male , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , Osmolar Concentration , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/chemistry , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL