Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 11: 582833, 2020.
Article in English | MEDLINE | ID: mdl-33193394

ABSTRACT

New evidence has been emerging that antibodies can be protective in various experimental models of tuberculosis. Here, we report on protection against multidrug-resistant Mycobacterium tuberculosis (MDR-TB) infection using a combination of the human monoclonal IgA 2E9 antibody against the alpha-crystallin (Acr, HspX) antigen and mouse interferon-gamma in mice transgenic for the human IgA receptor, CD89. The effect of the combined mucosal IgA and IFN-γ; treatment was strongest (50-fold reduction) when therapy was applied at the time of infection, but a statistically significant reduction of lung bacterial load was observed even when the therapy was initiated once the infection had already been established. The protection involving enhanced phagocytosis and then neutrophil mediated killing of infected cells was IgA isotype mediated, because treatment with an IgG version of 2E9 antibody was not effective in human IgG receptor CD64 transgenic mice. The Acr antigen specificity of IgA antibodies for protection in humans has been indicated by their elevated serum levels in latent tuberculosis unlike the lack of IgA antibodies against the virulence-associated MPT64 antigen. Our results represent the first evidence for potential translation of mucosal immunotherapy for the management of MDR-TB.


Subject(s)
Interferon-gamma/therapeutic use , Lung/immunology , Mycobacterium tuberculosis/physiology , Neutrophils/immunology , Respiratory Mucosa/immunology , Tuberculosis/therapy , Animals , Antibodies, Monoclonal/metabolism , Antigens, Bacterial/immunology , Antigens, CD/genetics , Antigens, CD/metabolism , Bacterial Load , Bacterial Proteins/immunology , Drug Resistance, Multiple , Humans , Immunoglobulin A/metabolism , Lung/microbiology , Mice , Mice, Transgenic , Mycobacterium tuberculosis/pathogenicity , Phagocytosis , Receptors, Fc/genetics , Receptors, Fc/metabolism , Receptors, IgG/genetics , THP-1 Cells , U937 Cells , alpha-Crystallins/immunology
2.
Front Immunol ; 10: 1349, 2019.
Article in English | MEDLINE | ID: mdl-31293568

ABSTRACT

Liposomes have been long considered as a vaccine delivery system but this technology remains to be fully utilized. Here, we describe a novel liposome-based subunit vaccine formulation for tuberculosis (TB) based on phosphatidylserine encapsulating two prominent TB antigens, Ag85B, and ESAT-6. We show that the resulting liposomes (Lipo-AE) are stable upon storage and can be readily taken up by antigen presenting cells and that their antigenic cargo is delivered and processed within endosomal cell compartments. The Lipo-AE vaccine formulation combined with the PolyIC adjuvant induced a mixed Th1/Th17-Th2 immune response to Ag85B but only a weak response to ESAT-6. An immunization regimen based on systemic delivery followed by mucosal boost with Lipo-AE resulted in the accumulation of resident memory T cells in the lungs. Most importantly though, when Lipo-AE vaccine candidate was administered to BCG-immunized mice subsequently challenged with low dose aerosol Mycobacterium tuberculosis, we observed a significant reduction of the bacterial load in the lungs and spleen compared to BCG alone. We therefore conclude that the immunization with mycobacterial antigens delivered by phosphatidylserine based liposomes in combination with Poly:IC adjuvant may represent a novel BCG boosting vaccination strategy.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , BCG Vaccine/immunology , Bacterial Proteins/immunology , Liposomes/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Load , Drug Delivery Systems , Female , Humans , Immunologic Memory/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Phosphatidylserines/immunology , Poly I-C/immunology , Spleen/microbiology , T-Lymphocytes, Helper-Inducer/immunology , Vaccination , Vaccines, Subunit/immunology
3.
Front Immunol ; 9: 346, 2018.
Article in English | MEDLINE | ID: mdl-29593708

ABSTRACT

Tuberculosis (TB) is the most deadly infectious disease in existence, and the only available vaccine, Bacillus Calmette-Guérin (BCG), is almost a century old and poorly protective. The immunological complexity of TB, coupled with rising resistance to antimicrobial therapies, necessitates a pipeline of diverse novel vaccines. Here, we show that Bacillus subtilis spores can be coated with a fusion protein 1 ("FP1") consisting of Mycobacterium tuberculosis (Mtb) antigens Ag85B, ACR, and HBHA. The resultant vaccine, Spore-FP1, was tested in a murine low-dose Mtb aerosol challenge model. Mice were primed with subcutaneous BCG, followed by mucosal booster immunizations with Spore-FP1. We show that Spore-FP1 enhanced pulmonary control of Mtb, as evidenced by reduced bacterial burdens in the lungs. This was associated with elevated antigen-specific IgG and IgA titers in the serum and lung mucosal surface, respectively. Spore-FP1 immunization generated superior antigen-specific memory T-cell proliferation in both CD4+ and CD8+ compartments, alongside bolstered Th1-, Th17-, and Treg-type cytokine production, compared to BCG immunization alone. CD69+CD103+ tissue resident memory T-cells (Trm) were found within the lung parenchyma after mucosal immunization with Spore-FP1, confirming the advantages of mucosal delivery. Our data show that Spore-FP1 is a promising new TB vaccine that can successfully augment protection and immunogenicity in BCG-primed animals.


Subject(s)
Antigens, Bacterial , Bacillus subtilis , Drug Delivery Systems , Immunity, Mucosal/drug effects , Mycobacterium bovis/immunology , Spores, Bacterial , Tuberculosis Vaccines , Tuberculosis/prevention & control , Administration, Inhalation , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/pharmacology , Bacillus subtilis/genetics , Bacillus subtilis/immunology , Female , Immunization, Secondary , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Spores, Bacterial/genetics , Spores, Bacterial/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tuberculosis/immunology , Tuberculosis Vaccines/genetics , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/pharmacokinetics
4.
Plant Biotechnol J ; 16(7): 1283-1294, 2018 07.
Article in English | MEDLINE | ID: mdl-29223138

ABSTRACT

Dengue is a major global disease requiring improved treatment and prevention strategies. The recently licensed Sanofi Pasteur Dengvaxia vaccine does not protect children under the age of nine, and additional vaccine strategies are thus needed to halt this expanding global epidemic. Here, we employed a molecular engineering approach and plant expression to produce a humanized and highly immunogenic poly-immunoglobulin G scaffold (PIGS) fused to the consensus dengue envelope protein III domain (cEDIII). The immunogenicity of this IgG Fc receptor-targeted vaccine candidate was demonstrated in transgenic mice expressing human FcγRI/CD64, by induction of neutralizing antibodies and evidence of cell-mediated immunity. Furthermore, these molecules were able to prime immune cells from human adenoid/tonsillar tissue ex vivo as evidenced by antigen-specific CD4+ and CD8+ T-cell proliferation, IFN-γ and antibody production. The purified polymeric fraction of dengue PIGS (D-PIGS) induced stronger immune activation than the monomeric form, suggesting a more efficient interaction with the low-affinity Fcγ receptors on antigen-presenting cells. These results show that the plant-expressed D-PIGS have the potential for translation towards a safe and easily scalable single antigen-based tetravalent dengue vaccine.


Subject(s)
Dengue Vaccines/immunology , Genetic Engineering , Receptors, Polymeric Immunoglobulin/genetics , Recombinant Fusion Proteins/genetics , Adenoids/immunology , Adjuvants, Immunologic/genetics , Animals , Antibodies, Neutralizing/immunology , Dengue Vaccines/genetics , Female , Genetic Engineering/methods , Humans , Immunity, Cellular , Immunoglobulin G/immunology , Male , Mice , Mice, Transgenic , Palatine Tonsil/immunology , Plants, Genetically Modified , Receptors, IgG/immunology , Receptors, Polymeric Immunoglobulin/immunology , Recombinant Fusion Proteins/immunology , Nicotiana/genetics
5.
Cytokine ; 99: 154-162, 2017 11.
Article in English | MEDLINE | ID: mdl-28917204

ABSTRACT

IL-4 is a pleiotropic cytokine that is highly Th2 polarizing. The ratio of IL-4 and its splice variant IL-4Δ2 observed in human health and disease suggests a role for both isoforms. In the present study, the biological function of murine IL-4Δ2 and the potential mechanism of action were studied. We report for the first time the generation of a functional, recombinant murine IL-4Δ2 form which is suggestive of its possible biological role in this species. Recombinant murine IL-4Δ2 inhibited IL-4 mediated cellular processes in macrophages and lymphocytes. Specifically, (i) it reversed IL-4 mediated inhibition of IFN-γ induced nitric oxide release by macrophages, (ii) inhibited IL-4 mediated induction of T cell proliferation, and (iii) prevented IL-4 stimulation of IgE synthesis by B cells. However, IL-4Δ2 did not compete with IL-4 for IL-4Rα binding and did not interfere with the downstream STAT-6 phosphorylation in T cells, suggesting an alternative mechanism for its antagonism of specific IL4-driven effects. These findings suggest that the mouse is a suitable experimental model for studies of the biology of IL-4 and its alternative splice variant.


Subject(s)
Alternative Splicing/genetics , Down-Regulation/genetics , Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , STAT6 Transcription Factor/metabolism , Alternative Splicing/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Down-Regulation/drug effects , Immunoglobulin E/biosynthesis , Interferon-gamma/metabolism , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Phosphorylation/drug effects , Recombinant Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
6.
Plant Biotechnol J ; 12(7): 840-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24629003

ABSTRACT

Progress with protein-based tuberculosis (TB) vaccines has been limited by poor availability of adjuvants suitable for human application. Here, we developed and tested a novel approach to molecular engineering of adjuvanticity that circumvents the need for exogenous adjuvants. Thus, we generated and expressed in transgenic tobacco plants the recombinant immune complexes (RICs) incorporating the early secreted Ag85B and the latency-associated Acr antigen of Mycobacterium tuberculosis, genetically fused as a single polypeptide to the heavy chain of a monoclonal antibody to Acr. The RICs were formed by virtue of the antibody binding to Acr from adjacent molecules, thus allowing self-polymerization of the complexes. TB-RICs were purified from the plant extracts and shown to be biologically active by demonstrating that they could bind to C1q component of the complement and also to the surface of antigen-presenting cells. Mice immunized with BCG and then boosted with two intranasal immunizations with TB-RICs developed antigen-specific serum IgG antibody responses with mean end-point titres of 1 : 8100 (Acr) and 1 : 24 300 (Ag85B) and their splenocytes responded to in vitro stimulation by producing interferon gamma. 25% of CD4+ proliferating cells simultaneously produced IFN-γ, IL-2 and TNF-α, a phenotype that has been linked with protective immune responses in TB. Importantly, mucosal boosting of BCG-immunized mice with TB-RICs led to a reduced M. tuberculosis infection in their lungs from log10 mean = 5.69 ± 0.1 to 5.04 ± 0.2, which was statistically significant. We therefore propose that the plant-expressed TB-RICs represent a novel molecular platform for developing self-adjuvanting mucosal vaccines.


Subject(s)
Adjuvants, Immunologic/biosynthesis , Antigen-Antibody Complex/metabolism , Mycobacterium tuberculosis/immunology , Nicotiana/genetics , Tuberculosis Vaccines/immunology , Adjuvants, Immunologic/metabolism , Administration, Intranasal , Animals , Antibody Formation , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cloning, Molecular , Humans , Interleukin-2/metabolism , Mice , Plants, Genetically Modified/metabolism , Nicotiana/metabolism , Tuberculosis Vaccines/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
7.
Eur J Immunol ; 44(2): 440-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24214530

ABSTRACT

Mucosal boosting of BCG-immunised individuals with a subunit tuberculosis (TB) vaccine would be highly desirable, considering that the lungs are the principal port of entry for Mycobacterium tuberculosis (MTB) and the site of the primary infection and reactivation. However, the main roadblock for subunit TB vaccine development is the lack of suitable adjuvants that could induce robust local and systemic immune responses. Here, we describe a novel vaccine delivery system that was designed to mimic, in part, the MTB pathogen itself. The surface of yellow carnauba wax nanoparticles was coated with the highly immunogenic Ag85B Ag of MTB and they were directed to the alveolar epithelial surfaces by the incorporation of the heparin-binding hemagglutinin adhesion (HBHA) protein. Our results showed that the i.n. immunisation of BCG-primed BALB/c mice with nanoparticles adsorbed with Ag85B-HBHA (Nano-AH vaccine) induced robust humoral and cellular immune responses and IFN-γ production, and multifunctional CD4⁺ T cells expressing IFN-γ, IL-2 and TNF-α. Mice challenged with H37Rv MTB had a significantly reduced bacterial load in their lungs when compared with controls immunised with BCG alone. We therefore conclude that this immunisation approach is an effective means of boosting the BCG-induced anti-TB immunity.


Subject(s)
Antigens, Bacterial/immunology , Mycobacterium tuberculosis/immunology , Nanoparticles/administration & dosage , Pulmonary Alveoli/immunology , Respiratory Mucosa/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Acyltransferases/genetics , Acyltransferases/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Animals , Antigens, Bacterial/genetics , BCG Vaccine/immunology , Bacterial Load/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cell Proliferation , Epithelial Cells/immunology , Epithelial Cells/microbiology , Female , Interferon-gamma/immunology , Interleukin-2/immunology , Lectins/genetics , Lectins/immunology , Mice , Mice, Inbred BALB C , Pulmonary Alveoli/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Respiratory Mucosa/microbiology , Tuberculosis/microbiology , Tuberculosis Vaccines/genetics , Tumor Necrosis Factor-alpha/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...