Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
PLoS One ; 15(8): e0237218, 2020.
Article in English | MEDLINE | ID: mdl-32760143

ABSTRACT

Influenza is an infectious respiratory illness caused by influenza viruses. Despite yearly updates, the efficacy of influenza vaccines is significantly curtailed by the virus antigenic drift and antigenic shift. These constant changes to the influenza virus make-up also challenge the development of a universal flu vaccine, which requires conserved antigenic regions shared by influenza viruses of different subtypes. We propose that it is possible to bypass these challenges by the development of an influenza vaccine based on conserved proteins delivered in an adjuvanted nanoparticle system. In this study, we generated influenza nanoparticle constructs using trimethyl chitosan nanoparticles (TMC nPs) as the carrier of recombinant influenza hemagglutinin subunit 2 (HA2) and nucleoprotein (NP). The purified HA2 and NP recombinant proteins were encapsulated into TMC nPs to form HA2-TMC nPs and NP-TMC nPs, respectively. Primary human intranasal epithelium cells (HNEpCs) were used as an in vitro model to measure immunity responses. HA2-TMC nPs, NP-TMC nPs, and HA2-NP-TMC nPs (influenza nanoparticle constructs) showed no toxicity in HNEpCs. The loading efficiency of HA2 and NP into the TMC nPs was 97.9% and 98.5%, respectively. HA2-TMC nPs and NP-TMC nPs more efficiently delivered HA2 and NP proteins to HNEpCs than soluble HA2 and NP proteins alone. The induction of various cytokines and chemokines was more evident in influenza nanoparticle construct-treated HNEpCs than in soluble protein-treated HNEpCs. In addition, soluble factors secreted by influenza nanoparticle construct-treated HNEpCs significantly induced MoDCs maturation markers (CD80, CD83, CD86 and HLA-DR), as compared to soluble factors secreted by protein-treated HNEpCs. HNEpCs treated with the influenza nanoparticle constructs significantly reduced influenza virus replication in an in vitro challenge assay. The results indicate that TMC nPs can be used as influenza vaccine adjuvants and carriers capable of delivering HA2 and NP proteins to HNEpCs.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chitosan/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/pharmacology , Influenza, Human/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Cell Line , Cells, Cultured , Chitosan/administration & dosage , Dogs , Drug Carriers/administration & dosage , Drug Carriers/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Humans , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Nanoparticles/administration & dosage , Nucleocapsid Proteins , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , RNA-Binding Proteins/administration & dosage , RNA-Binding Proteins/pharmacology , Viral Core Proteins/administration & dosage , Viral Core Proteins/pharmacology
2.
JCI Insight ; 5(10)2020 05 21.
Article in English | MEDLINE | ID: mdl-32434990

ABSTRACT

Protein-based, self-assembling nanoparticles elicit superior immunity compared with soluble protein vaccines, but the immune mechanisms underpinning this effect remain poorly defined. Here, we investigated the immunogenicity of a prototypic ferritin-based nanoparticle displaying influenza hemagglutinin (HA) in mice and macaques. Vaccination of mice with HA-ferritin nanoparticles elicited higher serum antibody titers and greater protection against experimental influenza challenge compared with soluble HA protein. Germinal centers in the draining lymph nodes were expanded and persistent following HA-ferritin vaccination, with greater deposition of antigen that colocalized with follicular dendritic cells. Our findings suggest that a highly ordered and repetitive antigen array may directly drive germinal centers through a B cell-intrinsic mechanism that does not rely on ferritin-specific T follicular helper cells. In contrast to mice, enhanced immunogenicity of HA-ferritin was not observed in pigtail macaques, where antibody titers and lymph node immunity were comparable to soluble vaccination. An improved understanding of factors that drive nanoparticle vaccine immunogenicity in small and large animal models will facilitate the clinical development of nanoparticle vaccines for broad and durable protection against diverse pathogens.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Hemagglutinin Glycoproteins, Influenza Virus , Immunologic Memory/drug effects , Influenza Vaccines , Nanoparticles , Animals , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Macaca nemestrina , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , T-Lymphocytes, Helper-Inducer/immunology
3.
Vopr Virusol ; 65(3): 150-158, 2020 Jul 22.
Article in Russian | MEDLINE | ID: mdl-33533217

ABSTRACT

INTRODUCTION: Influenza is a severe viral disease, a frequent complication of which is a secondary bacterial pneumonia. Influenza vaccines prevent secondary bacterial complications. Virus-like particles are one of the promising areas for the development of new vaccines. The aim of this work is to study the correlation of the pathomorphological characteristics of the lungs with clinical, virological, and microbiological markers of the disease at vaccination with virus-like particles (VLPs), containing hemagglutinin (HA) of influenza virus (HA-Gag-VLPs) in a murine model of secondary bacterial pneumonia induced by S. pneumoniae after influenza infection. MATERIAL AND METHODS: BALB/c mice were vaccinated with VLPs containing influenza HA. After 21 days, mice were infected with two strains of influenza viruses, homologous and non-homologous, and 5 days after viral infection, were infected with S. pneumoniae. The vaccination effect was evaluated by morphological, virological (titer of the virus in the lungs) and microbiological (titer of bacteria in the lungs) data, and was confirmed by clinical data (survival, change in body weight). RESULTS: Immunization with HA-Gag-VLPs, followed by infection with a homologous influenza virus and S. pneumoniae, reduced the area of foci of inflammation, inhibited the replication of the virus and bacteria in the lungs, and also protected animals from death and reduced their weight loss. Immunization with HA-Gag-VLPs upon infection with a heterologous strain and S. pneumoniae did not affect these criteria. CONCLUSION: The immunization with HA-Gag-VLPs prevented the viral replication, providing a reduction of S. pneumoniae titer and the degree of lung damage, protecting animals from the disease in a murine model of secondary bacterial pneumonia, induced by S. pneumoniae, after influenza infection with homologous strain of the virus.


Subject(s)
Influenza, Human/drug therapy , Lung/drug effects , Pneumonia, Bacterial/drug therapy , Vaccines, Virus-Like Particle/pharmacology , Animals , Antibodies, Viral/blood , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/complications , Influenza, Human/pathology , Influenza, Human/virology , Lung/virology , Mice , Mice, Inbred BALB C , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/virology
4.
Front Immunol ; 11: 606518, 2020.
Article in English | MEDLINE | ID: mdl-33414788

ABSTRACT

Radioprotective 105 (RP105) (also termed CD180) is an orphan and unconventional Toll-like receptor (TLR) that lacks an intracellular signaling domain. The agonistic anti-RP105 monoclonal antibody (mAb) can cross-link RP105 on B cells, resulting in the proliferation and activation of B cells. Anti-RP105 mAb also has a potent adjuvant effect, providing higher levels of antigen-specific antibodies compared to alum. However, adjuvanticity is required for the covalent link between anti-RP105 mAb and the antigen. This is a possible obstacle to immunization due to the link between anti-RP105 mAb and some antigens, especially multi-transmembrane proteins. We have previously succeeded in inducing rapid and potent recombinant mAbs in mice using antibody gene-based delivery. To simplify the covalent link between anti-RP105 mAb and antigens, we generated genetic constructs of recombinant anti-RP105 mAb (αRP105) bound to the transmembrane domain of the IgG-B cell receptor (TM) (αRP105-TM), which could enable the anti-RP105 mAb to link the antigen via the cell membrane. We confirmed the expression of αRP105-TM and the antigen hemagglutinin, which is a membrane protein of the influenza virus, on the same cell. We also found that αRP105-TM could activate splenic B cells, including both mature and immature cells, depending on the cell surface RP105 in vitro. To evaluate the adjuvanticity of αRP105-TM, we conducted DNA immunization in mice with the plasmids encoding αRP105-TM and hemagglutinin, followed by challenge with an infection of a lethal dose of an influenza virus. We then obtained partially but significantly hemagglutinin-specific antibodies and observed protective effects against a lethal dose of influenza virus infection. The current αRP105-TM might provide adjuvanticity for a vaccine via a simple preparation of the expression plasmids encoding αRP105-TM and of that encoding the target antigen.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD/metabolism , B-Lymphocytes/drug effects , Cell Membrane/drug effects , Gene Transfer Techniques , Genetic Vectors , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza Vaccines/pharmacology , Orthomyxoviridae Infections/prevention & control , Spleen/drug effects , Adjuvants, Immunologic/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Surface/genetics , Antigens, Surface/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Proliferation/drug effects , Coculture Techniques , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Hybridomas , Immunization , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Lymphocyte Activation/drug effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Rats , Receptors, IgG/genetics , Receptors, IgG/immunology , Spleen/immunology , Spleen/metabolism , Vaccines, DNA/pharmacology
5.
J Immunol Methods ; 470: 20-26, 2019 07.
Article in English | MEDLINE | ID: mdl-31028753

ABSTRACT

This study aimed to investigate whether the human antigen presenting cells (APCs) can process and present Salmonella expressing H7N9 hemagglutinin (Sal-HA), neuraminidase (Sal-NA) or M2 ectodomain (Sal-M2e) to T cells and subsequently activate CD4+ T cell responses in vitro. In this study, APCs generated from human peripheral blood mononuclear cells (PBMCs) were first treated with mitomycin-C, followed by stimulation with Sal-HA, Sal-M2e, Sal-NA or Salmonella alone for 24 h. Subsequently, stimulated APCs were coincubated with untreated PBMCs (1:10) of the same individual for 24 or 72 h and then analysed for cytokine induction and T cell proliferations by qRT-PCR assay and flow cytometry, respectively. Our results demonstrated that APCs stimulated with Sal-HA, Sal-M2e or Sal-NA induced significantly (p < .05) higher CD3+CD4+ T cell proliferations compared to the APCs treated with Salmonella alone. Our data further revealved that APCs treated with Sal-HA induced significantly (p < .05) higher CD3+CD4+ T cell responses compared to the APCs treated with either Sal-M2e or Sal-NA, which both induced almost comparable levels. The T cell proliferation responses were further measured by lymphocyte proliferation assay and the results showed that Sal-HA and Sal-M2e stimulated APCs induced significantly (p < .05) higher proliferations in T cells compared to the APCs stimulated with either Sal-NA or Salmonella alone. With respect to cytokine inductions, APCs treated with either Sal-HA or Sal-M2e induced significantly (p < .05) higher mRNA transcription levels of proinflammatory (IL-1ß, IL-6, IL-12 and IL-23), Th1 (IFN-γ), Th17 (IL-17 and IL-21) and Th2 (IL-10 and TGF-ß) cytokines in T cells compared to Sal-NA or Salmonella alone treated APCs. In conclusion, we show that Salmonella system can efficiently deliver vaccine antigens to APCs and is, thus, capable to elicit heterologous antigen-specific adaptive immunity.


Subject(s)
Antigen-Presenting Cells/drug effects , Antigens, Viral/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Neuraminidase/pharmacology , Salmonella typhimurium/genetics , Viral Matrix Proteins/pharmacology , Animals , Antigen Presentation/drug effects , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Coculture Techniques , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunity, Cellular/drug effects , Influenza A Virus, H7N9 Subtype/chemistry , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/prevention & control , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Mitomycin/pharmacology , Neuraminidase/genetics , Neuraminidase/immunology , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Salmonella typhimurium/immunology , Transforming Growth Factor beta/biosynthesis , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
6.
PLoS One ; 14(3): e0212431, 2019.
Article in English | MEDLINE | ID: mdl-30822308

ABSTRACT

Swine influenza viruses (SIVs), the causal agents of swine influenza, are not only important to control due to the economic losses in the swine industry, but also can be pandemic pathogens. Vaccination is one of the most relevant strategies to control and prevent influenza infection. Current human vaccines against influenza induce strain-specific immunity and annual update is required due to the virus antigenic shift phenomena. Previously, our group has reported the use of conserved hemagglutinin peptides (HA-peptides) derived from H1-influenza virus as a potential multivalent vaccine candidate. Immunization of swine with these HA-peptides elicited antibodies that recognized and neutralized heterologous influenza viruses in vitro and demonstrated strong hemagglutination-inhibiting activity. In the present work, we cloned one HA-peptide (named NG34) into a plasmid fused with cytotoxic T lymphocyte-associated antigen (CTLA4) which is a molecule that modifies T cell activation and with an adjuvant activity interfering with the adaptive immune response. The resulting plasmid, named pCMV-CTLA4-Ig-NG34, was administered twice to animals employing a needle-free delivery approach. Two studies were carried out to test the efficacy of pCMV-CTLA4-Ig-NG34 as a potential swine influenza vaccine, one in seronegative and another in seropositive pigs against SIV. The second one was aimed to evaluate whether pCMV-CTLA4-Ig-NG34 vaccination would overcome maternally derived antibodies (MDA). After immunization, all animals were intranasally challenged with an H3N2 influenza strain. A complete elimination or significant reduction in the viral shedding was observed within the first week after the challenge in the vaccinated animals from both studies. In addition, no challenged heterologous virus load was detected in the airways of vaccinated pigs. Overall, it is suggested that the pCMV-CTLA4-Ig-NG34 vaccine formulation could potentially be used as a multivalent vaccine against influenza viruses.


Subject(s)
Abatacept , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines , Orthomyxoviridae Infections , Peptides , Swine Diseases , Vaccines, DNA , Virus Shedding , Abatacept/genetics , Abatacept/immunology , Abatacept/pharmacology , Animals , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Madin Darby Canine Kidney Cells , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Peptides/genetics , Peptides/immunology , Peptides/pharmacology , Plasmids/genetics , Plasmids/immunology , Plasmids/pharmacology , Swine , Swine Diseases/genetics , Swine Diseases/immunology , Swine Diseases/prevention & control , Vaccination , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology , Virus Shedding/drug effects , Virus Shedding/genetics , Virus Shedding/immunology
7.
Nanomedicine ; 17: 223-235, 2019 04.
Article in English | MEDLINE | ID: mdl-30695729

ABSTRACT

Hemagglutinin (HA) displayed on a ferritin nano-cage has been shown to be effective in generating a potent immune response against a broad range of influenza infections. Here, we showed that conjugation of flagellin together with HA to the exterior surface of the ferritin cage greatly enhanced not only the humoral immune response in mice but also antigen-specific T cell responses that include Th1 cytokine secretion. The effect of flagellin remained essentially unchanged when the molar ratio of flagellin to HA was reduced from 1:1 to 1:3. Injection of the ferritin-HA-flagellin cage provided protection against lethal virus challenge in mice. We used a small immunoglobulin fragment VL12.3 as a convenient method for attaching HA and flagellin to the ferritin cage. This attachment method can be used for rapid screening of a variety of protein cages and nano-assemblies to identify the most suitable carrier and adjuvant proteins for the target antigen.


Subject(s)
Adjuvants, Immunologic/chemistry , Ferritins/chemistry , Flagellin/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A virus/chemistry , Salmonella typhimurium/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Cell Line , Female , Ferritins/pharmacology , Flagellin/pharmacology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Mice , Mice, Inbred BALB C , Nanostructures/chemistry
8.
Front Immunol ; 9: 2060, 2018.
Article in English | MEDLINE | ID: mdl-30271406

ABSTRACT

Due to the high risk of an outbreak of pandemic influenza, the development of a broadly protective universal influenza vaccine is highly warranted. The design of such a vaccine has attracted attention and much focus has been given to nanoparticle-based influenza vaccines which can be administered intranasally. This is particularly interesting since, contrary to injectable vaccines, mucosal vaccines elicit local IgA and lung resident T cell immunity, which have been found to correlate with stronger protection in experimental models of influenza virus infections. Also, studies in human volunteers have indicated that pre-existing CD4+ T cells correlate well to increased resistance against infection. We have previously developed a fusion protein with 3 copies of the ectodomain of matrix protein 2 (M2e), which is one of the most explored conserved influenza A virus antigens for a broadly protective vaccine known today. To improve the protective ability of the self-adjuvanting fusion protein, CTA1-3M2e-DD, we incorporated it into porous maltodextrin nanoparticles (NPLs). This proof-of-principle study demonstrates that the combined vaccine vector given intranasally enhanced immune protection against a live challenge infection and reduced the risk of virus transmission between immunized and unimmunized individuals. Most importantly, immune responses to NPLs that also contained recombinant hemagglutinin (HA) were strongly enhanced in a CTA1-enzyme dependent manner and we achieved broadly protective immunity against a lethal infection with heterosubtypic influenza virus. Immune protection was mediated by enhanced levels of lung resident CD4+ T cells as well as anti-HA and -M2e serum IgG and local IgA antibodies.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza A virus/immunology , Influenza Vaccines/pharmacology , Orthomyxoviridae Infections , Viral Matrix Proteins/pharmacology , Animals , CD4-Positive T-Lymphocytes , Female , Male , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Porosity , Recombinant Fusion Proteins/pharmacology
9.
PLoS One ; 13(8): e0201429, 2018.
Article in English | MEDLINE | ID: mdl-30138320

ABSTRACT

BACKGROUND: Influenza infection could be more effectively controlled if a multi-purpose vaccine with the ability to induce responses against most, or all, influenza A subtypes could be generated. Conserved viral proteins are a promising basis for the creation of a broadly protective vaccine. In the present study, the immunogenicity and protective properties of three recombinant proteins (vaccine candidates), comprising conserved viral proteins fused with bacterial flagellin, were compared. METHODS: Balb/c mice were immunized intranasally with recombinant proteins comprising either one viral protein (the ectodomain of the M2 protein, 'M2e') or two viral proteins (M2e and the hemagglutinin second subunit 'HA2' epitope) genetically fused with flagellin. Further, two different consensus variants of HA2 were used. Therefore, three experimental positives were used in addition to the negative control (Flg-his). The mucosal, humoral, and T-cell immune responses to these constructs were evaluated. RESULT: We have demonstrated that insertion of the HA2 consensus polypeptide (aa 76-130), derived from either the first (HA2-1) or second (HA2-2) virus phylogenetic group, into the recombinant Flg4M2e protein significantly enhanced its immunogenicity and protective properties. Intranasal administration of the vaccine candidates (Flg-HA2-2-4M2e or Flg-HA2-1-4M2e) induced considerable mucosal and systemic responses directed at both the M2e-protein and, in general, the influenza A virus. However, the immune response elicited by the Flg-HA2-1-4M2e protein was weaker than the one generated by Flg-HA2-2-4M2e. These recombinant proteins containing both viral peptides provide complete protection from lethal challenge with various influenza viruses: A/H3N2; A/H2N2; and A/H5N1. CONCLUSION: This study demonstrates that the intranasal administration of Flg-HA2-2-4M2e recombinant protein induces a strong immune response which provides broad protection against various influenza viruses. This construct is therefore a strong candidate for development as a universal vaccine.


Subject(s)
Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Peptides/immunology , Animals , Epitopes/pharmacology , Female , Filaggrin Proteins , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Peptides/pharmacology
10.
PLoS One ; 13(6): e0195903, 2018.
Article in English | MEDLINE | ID: mdl-29897914

ABSTRACT

Seasonal influenza is a contagious respiratory illness that annually affects millions of people worldwide. To identify currently circulating influenza virus subtypes, the Centers for Disease Control and Prevention's International Reagent Resource distributes the World Health Organization (WHO) influenza reagent kits, which are used globally by testing laboratories for influenza surveillance. The data generated by the kits aid in strain selection for the influenza vaccine each season. The use of animals to produce high quality and quantities of antibodies is critical to the production of these kits. In this study, we assessed the effects and efficacy of repeated sampling from automated plasmapheresis in goats. Analysis of blood samples demonstrated that repeated automated plasmapheresis procedures did not adversely affect the immediate or long-term health of goats. Further, our results indicate that repeated plasmapheresis in goats was capable of generating 2 liters of antibody-rich plasma per goat per week. This volume is sufficient to produce enough WHO influenza kits to conduct over 1 million tests. Thus, we have shown that the rapid production of plasma in goats can positively impact the public health preparedness and response to influenza.


Subject(s)
Antibodies, Viral/blood , Antibodies, Viral/isolation & purification , Goats/blood , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza Vaccines/pharmacology , Plasmapheresis , Vaccination , Animals , Antibodies, Viral/immunology , Goats/immunology , Influenza Vaccines/immunology
11.
Biochem Biophys Res Commun ; 467(4): 766-70, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26478433

ABSTRACT

Health policy precludes neonatal vaccination against influenza. Hence, morbidity and mortality are high under 6 months of age. Lactoferrin may activate diminished numbers of dysfunctional dendritic cells and reverse neonatal vaccine failures. Aluminum hydroxide/ALUM recruits neutrophils that secrete lactoferrin at deposition sites of antigen. We theorized lactoferrin + influenza antigen initiates an equivalent antibody response compared to ALUM. Three-day-old mice received subcutaneously 30 µg of H1N1 hemagglutinin + 200 µg of bovine lactoferrin versus hemagglutinin + ALUM. Controls received hemagglutinin, lactoferrin, or ALUM. After 21 days, sera measured anti-H1N1 (ELISA) and neutralizing antibody (plaque assays). ELISA detected equal antibody production with lactoferrin + hemagglutinin compared to hemagglutinin + ALUM; both sera also neutralized H1N1 virus at a 1:20 dilution (p < 0.01). Controls had no anti-H1N1 antibody. Neonates given lactoferrin had no anaphylaxis when challenged four weeks later. Lactoferrin is a safe and effective adjuvant for inducing antibody against influenza in neonates.


Subject(s)
Adjuvants, Immunologic/pharmacology , Lactoferrin/pharmacology , Orthomyxoviridae Infections/immunology , Vaccination/methods , Aluminum Hydroxide/immunology , Aluminum Hydroxide/pharmacology , Animals , Animals, Newborn , Cattle , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza A Virus, H1N1 Subtype/pathogenicity , Lactoferrin/immunology , Madin Darby Canine Kidney Cells/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
12.
J Immunol ; 193(5): 2416-26, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25057007

ABSTRACT

Dendritic cells are major APCs that can efficiently prime immune responses. However, the roles of skin-resident Langerhans cells (LCs) in eliciting immune responses have not been fully understood. In this study, we demonstrate for the first time, to our knowledge, that LCs in cynomolgus macaque skin are capable of inducing antiviral-specific immune responses in vivo. Targeting HIV-Gag or influenza hemagglutinin Ags to skin LCs using recombinant fusion proteins of anti-Langerin Ab and Ags resulted in the induction of the viral Ag-specific responses. We further demonstrated that such Ag-specific immune responses elicited by skin LCs were greatly enhanced by TLR ligands, polyriboinosinic polyribocytidylic acid, and R848. These enhancements were not due to the direct actions of TLR ligands on LCs, but mainly dependent on TNF-α secreted from macrophages and neutrophils recruited to local tissues. Skin LC activation and migration out of the epidermis are associated with macrophage and neutrophil infiltration into the tissues. More importantly, blocking TNF-α abrogated the activation and migration of skin LCs. This study highlights that the cross-talk between innate immune cells in local tissues is an important component for the establishment of adaptive immunity. Understanding the importance of local immune networks will help us to design new and effective vaccines against microbial pathogens.


Subject(s)
HIV-1/immunology , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza A virus/immunology , Langerhans Cells/immunology , Skin/immunology , Tumor Necrosis Factor-alpha/immunology , gag Gene Products, Human Immunodeficiency Virus/pharmacology , Adaptive Immunity/drug effects , Adaptive Immunity/physiology , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Imidazoles/pharmacology , Macaca mulatta , Macrophages/immunology , Neutrophils/immunology , Poly I/pharmacology , gag Gene Products, Human Immunodeficiency Virus/immunology
13.
J Pharm Sci ; 103(3): 821-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24425059

ABSTRACT

The recombinant hemagglutinin (rHA)-based influenza vaccine Flublok® has recently been approved in the United States as an alternative to the traditional egg-derived flu vaccines. Flublok is a purified vaccine with a hemagglutinin content that is threefold higher than standard inactivated influenza vaccines. When rHA derived from an H3N2 influenza virus was expressed, purified, and stored for 1 month, a rapid loss of in vitro potency (∼50%) was observed as measured by the single radial immunodiffusion (SRID) assay. A comprehensive characterization of the rHA protein antigen was pursued to identify the potential causes and mechanisms of this potency loss. In addition, the biophysical and chemical stability of the rHA in different formulations and storage conditions was evaluated over time. Results demonstrate that the potency loss over time did not correlate with trends in changes to the higher order structure or hydrodynamic size of the rHA. The most likely mechanism for the early loss of potency was disulfide-mediated cross-linking of rHA, as the formation of non-native disulfide-linked multimers over time correlated well with the observed potency loss. Furthermore, a loss of free thiol content, particularly in specific cysteine residues in the antigen's C-terminus, was correlated with potency loss measured by SRID.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H3N2 Subtype/metabolism , Influenza Vaccines/chemistry , Chemical Phenomena , Cysteine/analysis , Cysteine/chemistry , Cystine/analysis , Cystine/chemistry , Drug Stability , Drug Storage , Excipients/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Hydrodynamics , Immunodiffusion , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/genetics , Influenza Vaccines/metabolism , Influenza Vaccines/pharmacology , Octoxynol/chemistry , Oxidation-Reduction , Peptide Mapping , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spectroscopy, Fourier Transform Infrared , Temperature , Thioglycolates/chemistry
14.
Proc Natl Acad Sci U S A ; 111(7): 2476-81, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24469815

ABSTRACT

The 2009 H1N1 pandemic and recent human cases of H5N1, H7N9, and H6N1 in Asia highlight the need for a universal influenza vaccine that can provide cross-strain or even cross-subtype protection. Here, we show that recombinant monoglycosylated hemagglutinin (HAmg) with an intact protein structure from either seasonal or pandemic H1N1 can be used as a vaccine for cross-strain protection against various H1N1 viruses in circulation from 1933 to 2009 in mice and ferrets. In the HAmg vaccine, highly conserved sequences that were originally covered by glycans in the fully glycosylated HA (HAfg) are exposed and thus, are better engulfed by dendritic cells (DCs), stimulated better DC maturation, and induced more CD8+ memory T cells and IgG-secreting plasma cells. Single B-cell RT-PCR followed by sequence analysis revealed that the HAmg vaccine activated more diverse B-cell repertoires than the HAfg vaccine and produced antibodies with cross-strain binding ability. In summary, the HAmg vaccine elicits cross-strain immune responses that may mitigate the current need for yearly reformulation of strain-specific inactivated vaccines. This strategy may also map a new direction for universal vaccine design.


Subject(s)
Drug Design , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Immunity, Cellular/immunology , Influenza Vaccines/pharmacology , Influenza, Human/prevention & control , Orthomyxoviridae/immunology , Animals , Base Sequence , Chromatography, Liquid , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Ferrets , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology , Species Specificity , Tandem Mass Spectrometry
15.
Vopr Virusol ; 58(3): 25-7, 2013.
Article in Russian | MEDLINE | ID: mdl-24006629

ABSTRACT

The modern influenza virus subtypes H3N2, H5N1, and H1N1 reduced the metabolism of the endothelial cells within the range from 20% to 60% (compared with control). The degree of the activity of the dehydrogenase reduction depended on the dose of virus and time of virus reproduction. HA and NA also actively reduced the metabolism of the cells ranging from 5% to 60%, depending on the concentration of the proteins and time of their impact on cells. Neuraminidase was more active than hemagglutinin in the MTT test (at concentration 50 microg protein/ml).


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/growth & development , Neuraminidase/pharmacology , Biomarkers/metabolism , Cell Line , Cell Survival/drug effects , Endothelial Cells/cytology , Endothelial Cells/virology , Endothelium, Vascular/cytology , Endothelium, Vascular/virology , Hemagglutinin Glycoproteins, Influenza Virus/isolation & purification , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Neuraminidase/isolation & purification , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Tetrazolium Salts , Thiazoles
16.
FASEB J ; 26(12): 4886-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22889831

ABSTRACT

Major histocompatibility complex (MHC) class II-associated antigen presentation involves an array of interacting molecules. CD74, the cell surface isoform of the MHC class II-associated invariant chain, is one such molecule; its role remains poorly defined. To address this, we have employed a high-resolution single-particle imaging method for quantifying the colocalization of CD74 with human leukocyte antigen (HLA)-DR molecules on human fibroblast cells known for their capacity to function as antigen-presenting cells. We have also examined whether the colocalization induces internalization of HLA-DR using HA(307-319), a "universal" peptide that binds specifically to the peptide-binding groove of all HLA-DR molecules, irrespective of their alleles. We have determined that 25 ± 1.3% of CD74 and 17 ± 0.3% of HLA-DR are colocalized, and the association of CD74 with HLA-DR and the internalization of HLA-DR are both inhibited by HA(307-319). A similar inhibition of HLA-DR internalization was observed in freshly isolated monocyte-derived dendritic cells. A key role of CD74 is to translocate HLA-DR molecules to early endosomes for reloading with peptides prior to recycling to the cell surface. We conclude that CD74 regulates the balance of peptide-occupied and peptide-free forms of MHC class II at the cell surface.


Subject(s)
Antigen-Presenting Cells/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Membrane/metabolism , HLA-DR Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Microscopy, Fluorescence/methods , Adult , Algorithms , Antigens, Differentiation, B-Lymphocyte/genetics , Cell Line , Cells, Cultured , Dendritic Cells/metabolism , Endocytosis/drug effects , Flow Cytometry , Fluorescence Resonance Energy Transfer , HLA-DR Antigens/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Histocompatibility Antigens Class II/genetics , Humans , Imaging, Three-Dimensional , Microscopy, Confocal , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Binding/drug effects
17.
Sci Signal ; 5(212): ra16, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22355189

ABSTRACT

The threat of a new influenza pandemic has existed since 1997, when the highly pathogenic H5N1 strain of avian influenza A virus infected humans in Hong Kong and spread across Asia, where it continued to infect poultry and people. The human mortality rate of H5N1 infection is about 60%, whereas that of seasonal H1N1 infection is less than 0.1%. The high mortality rate associated with H5N1 infection is predominantly a result of respiratory failure caused by acute lung injury; however, how viral infection contributes to this disease pathology is unclear. Here, we used electron microscopy to show the accumulation of autophagosomes in H5N1-infected lungs from a human cadaver and mice, as well as in infected A549 human epithelial lung cells. We also showed that H5N1, but not seasonal H1N1, induced autophagic cell death in alveolar epithelial cells through a pathway involving the kinase Akt, the tumor suppressor protein TSC2, and the mammalian target of rapamycin. Additionally, we suggest that the hemagglutinin protein of H5N1 may be responsible for stimulating autophagy. When applied prophylactically, reagents that blocked virus-induced autophagic signaling substantially increased the survival rate of mice and substantially ameliorated the acute lung injury and mortality caused by H5N1 infection. We conclude that the autophagic cell death of alveolar epithelial cells likely plays a crucial role in the high mortality rate of H5N1 infection, and we suggest that autophagy-blocking agents might be useful as prophylactics and therapeutics against infection of humans by the H5N1 virus.


Subject(s)
Autophagy/physiology , Influenza A Virus, H5N1 Subtype , Influenza, Human/pathology , Lung/ultrastructure , Signal Transduction/physiology , Analysis of Variance , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Blotting, Western , Cell Line , DNA Primers/genetics , Epithelial Cells/physiology , Gene Knockdown Techniques , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Humans , Lung/virology , Mice , Mice, Inbred BALB C , Microscopy, Electron , Microtubule-Associated Proteins/genetics , Oncogene Protein v-akt/metabolism , Phagosomes/pathology , Real-Time Polymerase Chain Reaction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
18.
Cell Mol Immunol ; 8(4): 348-58, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21383676

ABSTRACT

There has been an increase in interest in the use of altered peptides as antigen-specific therapeutic agents in autoimmune diseases. Here we investigated the inhibitory effect and possible mechanism of an altered influenza virus haemagglutinin (HA)-derived peptide in collagen-induced arthritis (CIA). CIA was induced in DBA/1 mice by immunisation with type II collagen (CII). Altered HA308-317, wild-type HA308-317 or irrelevant peptide was administered intranasally beginning from arthritis onset. Clinical and histological scores were assessed, and cytokine levels in the serum or supernatants from splenocytes were determined. The percentages of Th1 and Th2 cells in response to different peptides were analysed by FACS both in vivo and in vitro. Our results showed that intranasal administration of altered HA308-317 peptide significantly ameliorated CIA. The therapeutic effect of altered HA308-317 peptide was associated with a substantial decrease in production of interferon (IFN)-γ, interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1, anti-CII IgG, IgG1 and IgG2a antibodies, and an markedly increase in production of IL-10 and IL-4 in serum or supernatants from splenocytes treated with altered HA308-317 peptide. The percentage of Th2 (CD4(+)IL-4(+)) cells was upregulated significantly by altered HA308-317 peptide with a decreased percentage of Th1 (T helper 1; CD4(+)INF-γ(+)) cells both in vivo and in vitro. These findings suggest that altered HA308-317 peptide might be a promising candidate for rheumatoid arthritis (RA) treatment.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Administration, Intranasal , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Collagen Type II/immunology , Collagen Type II/metabolism , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Male , Mice , Mice, Inbred DBA , Orthomyxoviridae/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Th1 Cells/metabolism , Th2 Cells/metabolism
19.
J Immunol ; 183(5): 2915-20, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19667099

ABSTRACT

Despite its use widely as a therapeutic agent, and proposed use as vaccine adjuvant, the effect of IFNalpha on T cell function is poorly understood. As a pleiotropic innate cytokine produced rapidly in response to pathogens, it is well placed to impinge on specific immune responses. The aim of this study was to examine the impact of IFNalpha on the function of human memory CD4(+) T cells using the recall Ags purified protein derivative, tetanus toxoid, and hemagglutinin. IFNalpha administered either in vivo or added exogenously in vitro tended to enhance proliferative responses of purified protein derivative-specific T cells in marked contrast to the other cognate populations whose responses were often diminished. Purifying the memory CD4(+)CD45RO(+) T cells confirmed IFNalpha acted directly on these cells and not via an intermediate. The T cells could be divided into two broad categories depending on how IFNalpha effected their responses to cognate Ag: 1) enhanced proliferation and a striking increase in IFNgamma-production compared with smaller increases in IL-10 (increased ratio of IFNgamma:IL-10), and 2) neutral or diminished proliferation coupled with a smaller increase in IFNgamma relative to the increase in IL-10 (reduced IFNgamma:IL-10 ratio). IFNalpha has a role in modifying memory T cell responses when they are exposed to cognate Ag and may be important in vaccination strategies designed to augment particular Th memory responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-alpha/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , Humans , Immunologic Memory/drug effects , Injections, Subcutaneous , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Tetanus Toxoid/pharmacology , Tuberculin/pharmacology
20.
Biochim Biophys Acta ; 1788(2): 450-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18976631

ABSTRACT

Little is known about the mechanisms of cell-cell fusion in development and diseases and, especially, about fusion stages downstream of an opening of nascent fusion pore(s). Earlier works on different cell-cell fusion reactions have indicated that cytoskeleton plays important role in syncytium formation. However, due to complexity of these reactions and multifaceted contributions of cytoskeleton in cell physiology, it has remained unclear whether cytoskeleton directly drives fusion pore expansion or affects preceding fusion stages. Here we explore cellular reorganization associated with fusion pore expansion in syncytium formation using relatively simple experimental system. Fusion between murine embryonic fibroblasts NIH3T3-based cells is initiated on demand by well-characterized fusogen influenza virus hemagglutinin. We uncouple early fusion stages dependent on protein fusogens from subsequent fusion pore expansion stage and establish that the transition from local fusion to syncytium requires metabolic activity of living cells. Effective syncytium formation for cells with disorganized actin and microtubule cytoskeleton argues against hypothesis that cytoskeleton drives fusion expansion.


Subject(s)
Cytoskeleton/drug effects , Cytoskeleton/metabolism , Giant Cells/drug effects , Giant Cells/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Actins/metabolism , Animals , Giant Cells/cytology , Hydrogen-Ion Concentration , Mice , NIH 3T3 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...