Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
PLoS One ; 19(5): e0294998, 2024.
Article in English | MEDLINE | ID: mdl-38713688

ABSTRACT

Tularemia is a zoonotic disease caused by the facultative intracellular gram-negative bacterium Francisella tularensis. F. tularensis has a very low infection dose by the aerosol route which can result in an acute, and potentially lethal, infection in humans. Consequently, it is classified as a Category A bioterrorism agent by the US Centers for Disease Control (CDC) and is a pathogen of concern for the International Biodefence community. There are currently no licenced tularemia vaccines. In this study we report on the continued assessment of a tularemia subunit vaccine utilising ß-glucan particles (GPs) as a vaccine delivery platform for immunogenic F. tularensis antigens. Using a Fischer 344 rat infection model, we demonstrate that a GP based vaccine comprising the F. tularensis lipopolysaccharide antigen together with the protein antigen FTT0814 provided partial protection of F344 rats against an aerosol challenge with a high virulence strain of F. tularensis, SCHU S4. Inclusion of imiquimod as an adjuvant failed to enhance protective efficacy. Moreover, the level of protection afforded was dependant on the challenge dose. Immunological characterisation of this vaccine demonstrated that it induced strong antibody immunoglobulin responses to both polysaccharide and protein antigens. Furthermore, we demonstrate that the FTT0814 component of the GP vaccine primed CD4+ and CD8+ T-cells from immunised F344 rats to express interferon-γ, and CD4+ cells to express interleukin-17, in an antigen specific manner. These data demonstrate the development potential of this tularemia subunit vaccine and builds on a body of work highlighting GPs as a promising vaccine platform for difficult to treat pathogens including those of concern to the bio-defence community.


Subject(s)
Bacterial Vaccines , Disease Models, Animal , Francisella tularensis , Rats, Inbred F344 , Tularemia , Vaccines, Subunit , Animals , Tularemia/prevention & control , Tularemia/immunology , Rats , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Francisella tularensis/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Glucans/immunology , Glucans/pharmacology , T-Lymphocytes/immunology , Female , Antigens, Bacterial/immunology
2.
Infect Immun ; 92(3): e0045523, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38289122

ABSTRACT

Melioidosis is a disease that is difficult to treat due to the causative organism, Burkholderia pseudomallei being inherently antibiotic resistant and it having the ability to invade, survive, and replicate in an intracellular environment. Combination therapy approaches are routinely being evaluated in animal models with the aim of improving the level of protection and clearance of colonizing bacteria detected. In this study, a subunit vaccine layered with the antibiotic finafloxacin was evaluated in vivo against an inhalational infection with B. pseudomallei in Balb/c mice. Groups of mice vaccinated, infected, and euthanized at antibiotic initiation had a reduced bacterial load compared to those that had not been immunized. In addition, the subunit vaccine provided a synergistic effect when it was delivered with a CpG ODN and finafloxacin was initiated at 48 h post-challenge. Vaccination was also shown to improve the outcome, in a composite measure of survival and clearance. In summary, layering a subunit vaccine with the antibiotic finafloxacin is a promising therapeutic alternative for use in the treatment of B. pseudomallei infections.


Subject(s)
Burkholderia pseudomallei , Melioidosis , Animals , Mice , Mice, Inbred BALB C , Melioidosis/drug therapy , Melioidosis/prevention & control , Anti-Bacterial Agents/therapeutic use , Vaccination , Vaccines, Subunit , Disease Models, Animal
3.
Sci Rep ; 9(1): 7225, 2019 05 10.
Article in English | MEDLINE | ID: mdl-31076594

ABSTRACT

With the rise of antimicrobial resistance, novel ways to treat bacterial infections are required and the use of predatory bacteria may be one such approach. Bdellovibrio species have been shown in vitro to predate on a wide range of other Gram-negative bacteria, including CDC category A/B pathogens such as Yersinia pestis. The data reported here show that treatment of SKH-1 mice with Bdellovibrio bacteriovorus HD100 provided significant protection from a lethal challenge of Yersinia pestis CO92. This is the first report of protection conferred by predation in vivo against a systemic pathogen challenge. However, this protective effect was not observed in a preliminary study with Balb/c mice. Therefore the effects of the predatory bacteria are complex and may be dependent on immune status/genetics of the host. Overall, predatory bacteria may have utility as a therapeutic modality but further work is required to understand the predator-host interaction.


Subject(s)
Bdellovibrio bacteriovorus/physiology , Plague/prevention & control , Yersinia pestis/pathogenicity , Animals , Disease Models, Animal , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Optical Imaging , Phagocytosis , Plague/microbiology , Plague/pathology
4.
Int J Antimicrob Agents ; 54(1): 85-88, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31029736

ABSTRACT

Disulfiram (DSF) can help treat alcohol dependency by inhibiting aldehyde dehydrogenase (ALDH). Genomic analysis revealed that Francisella tularensis, the causative agent of tularemia, has lost all but one ALDH-like domain and that this domain retains the target of DSF. In this study, minimum inhibitory concentration (MIC) assays demonstrated that both DSF and its primary metabolite diethyldithiocarbamate (DDC) have strong antimicrobial activity against F. tularensis strain SCHU S4, with the MIC of DSF determined as 2 µg/mL in comparison with 8 µg/mL for DDC. The activity of DSF was further confirmed using an in vitro human macrophage infection assay. Francisella tularensis bacteria in DSF-treated cells were reduced in comparison with untreated and DDC-treated cells, comparable with that observed in doxycycline-treated cells. This suggests that DSF may be suitable for further investigation as an in vivo therapy for tularemia.


Subject(s)
Acetaldehyde Dehydrogenase Inhibitors/pharmacology , Alcohol Deterrents/pharmacology , Anti-Bacterial Agents/pharmacology , Disulfiram/pharmacology , Francisella tularensis/drug effects , Francisella tularensis/growth & development , Bacterial Load , Humans , Microbial Sensitivity Tests , Monocytes/drug effects , Monocytes/microbiology , THP-1 Cells
5.
PLoS One ; 13(10): e0200213, 2018.
Article in English | MEDLINE | ID: mdl-30296254

ABSTRACT

Francisella tularensis is an intracellular pathogen causing the disease tularemia, and an organism of concern to biodefence. There is no licensed vaccine available. Subunit approaches have failed to induce protection, which requires both humoral and cellular immune memory responses, and have been hampered by a lack of understanding as to which antigens are immunoprotective. We undertook a preliminary in silico analysis to identify candidate protein antigens. These antigens were then recombinantly expressed and encapsulated into glucan particles (GPs), purified Saccharomyces cerevisiae cell walls composed primarily of ß-1,3-glucans. Immunological profiling in the mouse was used to down-selection to seven lead antigens: FTT1043 (Mip), IglC, FTT0814, FTT0438, FTT0071 (GltA), FTT0289, FTT0890 (PilA) prior to transitioning their evaluation to a Fischer 344 rat model for efficacy evaluation. F344 rats were vaccinated with the GP protein antigens co-delivered with GP-loaded with Francisella LPS. Measurement of cell mediated immune responses and computational epitope analysis allowed down-selection to three promising candidates: FTT0438, FTT1043 and FTT0814. Of these, a GP vaccine delivering Francisella LPS and the FTT0814 protein was able to induce protection in rats against an aerosol challenge of F. tularensis SchuS4, and reduced organ colonisation and clinical signs below that which immunisation with a GP-LPS alone vaccine provided. This is the first report of a protein supplementing protection induced by LPS in a Francisella vaccine. This paves the way for developing an effective, safe subunit vaccine for the prevention of inhalational tularemia, and validates the GP platform for vaccine delivery where complex immune responses are required for prevention of infections by intracellular pathogens.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis , Glucans/chemistry , Tularemia/prevention & control , Animals , Coculture Techniques , Glucans/administration & dosage , Immunity, Cellular , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Saccharomyces cerevisiae , Tularemia/immunology , Vaccines, Attenuated/immunology , Vaccines, Subunit/immunology
6.
Antimicrob Agents Chemother ; 60(12): 7206-7215, 2016 12.
Article in English | MEDLINE | ID: mdl-27671061

ABSTRACT

Bioluminescence imaging (BLI) enables real-time, noninvasive tracking of infection in vivo and longitudinal infection studies. In this study, a bioluminescent Francisella tularensis strain, SCHU S4-lux, was used to develop an inhalational infection model in BALB/c mice. Mice were infected intranasally, and the progression of infection was monitored in real time using BLI. A bioluminescent signal was detectable from 3 days postinfection (3 dpi), initially in the spleen and then in the liver and lymph nodes, before finally becoming systemic. The level of bioluminescent signal correlated with bacterial numbers in vivo, enabling noninvasive quantification of bacterial burdens in tissues. Treatment with levofloxacin (commencing at 4 dpi) significantly reduced the BLI signal. Furthermore, BLI was able to distinguish noninvasively between different levofloxacin treatment regimens and to identify sites of relapse following treatment cessation. These data demonstrate that BLI and SCHU S4-lux are suitable for the study of F. tularensis pathogenesis and the evaluation of therapeutics for tularemia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Francisella tularensis/drug effects , Francisella tularensis/growth & development , Tularemia/drug therapy , Tularemia/pathology , Animals , Disease Models, Animal , Female , Francisella tularensis/metabolism , Levofloxacin/pharmacology , Liver/microbiology , Luminescent Measurements , Lymph Nodes/microbiology , Mice , Mice, Inbred BALB C , Spleen/microbiology , Tularemia/microbiology
7.
PLoS One ; 11(7): e0158575, 2016.
Article in English | MEDLINE | ID: mdl-27391310

ABSTRACT

Toll-like receptors (TLRs) recognise invading pathogens and mediate downstream immune signalling via Toll/IL-1 receptor (TIR) domains. TIR domain proteins (Tdps) have been identified in multiple pathogenic bacteria and have recently been implicated as negative regulators of host innate immune activation. A Tdp has been identified in Bacillus anthracis, the causative agent of anthrax. Here we present the first study of this protein, designated BaTdp. Recombinantly expressed and purified BaTdp TIR domain interacted with several human TIR domains, including that of the key TLR adaptor MyD88, although BaTdp expression in cultured HEK293 cells had no effect on TLR4- or TLR2- mediated immune activation. During expression in mammalian cells, BaTdp localised to microtubular networks and caused an increase in lipidated cytosolic microtubule-associated protein 1A/1B-light chain 3 (LC3), indicative of autophagosome formation. In vivo intra-nasal infection experiments in mice showed that a BaTdp knockout strain colonised host tissue faster with higher bacterial load within 4 days post-infection compared to the wild type B. anthracis. Taken together, these findings indicate that BaTdp does not play an immune suppressive role, but rather, its absence increases virulence. BaTdp present in wild type B. anthracis plausibly interact with the infected host cell, which undergoes autophagy in self-defence.


Subject(s)
Autophagy/physiology , Bacillus anthracis/metabolism , Bacterial Proteins/metabolism , Microtubules/metabolism , Animals , Anthrax/microbiology , Autophagy/genetics , Bacillus anthracis/genetics , Bacterial Proteins/genetics , Cell Line , Cell Survival/physiology , Female , HEK293 Cells , Humans , Mice , Microscopy, Confocal , Phylogeny , Protein Binding , Signal Transduction , Virulence/genetics , Virulence/physiology
8.
J Immunol Res ; 2014: 237043, 2014.
Article in English | MEDLINE | ID: mdl-25333059

ABSTRACT

Understanding the interactions between host and pathogen is important for the development and assessment of medical countermeasures to infectious agents, including potential biodefence pathogens such as Bacillus anthracis, Ebola virus, and Francisella tularensis. This review focuses on technological advances which allow this interaction to be studied in much greater detail. Namely, the use of "omic" technologies (next generation sequencing, DNA, and protein microarrays) for dissecting the underlying host response to infection at the molecular level; optical imaging techniques (flow cytometry and fluorescence microscopy) for assessing cellular responses to infection; and biophotonic imaging for visualising the infectious disease process. All of these technologies hold great promise for important breakthroughs in the rational development of vaccines and therapeutics for biodefence agents.


Subject(s)
Flow Cytometry/methods , High-Throughput Nucleotide Sequencing/methods , Microarray Analysis/methods , Microscopy, Fluorescence/methods , Bacillus anthracis/immunology , Bacillus anthracis/physiology , Ebolavirus/immunology , Ebolavirus/physiology , Francisella tularensis/immunology , Francisella tularensis/physiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune System/immunology , Immune System/microbiology , Immune System/virology
9.
Protein Pept Lett ; 21(4): 374-81, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24164262

ABSTRACT

Proline-rich antibacterial peptides protect experimental animals from bacterial challenge even if they are unable to kill the microorganisms in vitro. Their major in vivo modes of action are inhibition of bacterial protein folding and immunostimulation. Here we investigated whether the proline-rich antibacterial peptide dimer A3-APO was able to inhibit Bacillus cereus enterotoxin production in vitro and restrict the proliferation of lethal toxin-induced Bacillus anthracis replication in mouse macrophages. After 24 h incubation, peptide A3-APO and its single chain metabolite reduced the amount of properly folded B. cereus diarrhoeal enterotoxin production in a concentration-dependent manner leading to only 10-25% of the original amount of toxin detectable by a conformation-sensitive immunoassay. Likewise, after 4 h incubation, A3-APO restricted the proliferation of B. anthracis in infected macrophages by 40-45% compared to untreated cells both intracellularly and in the extracellular cell culture milieu. Although the peptide had a minimal inhibitory concentration of >512 mg/L against B. anthracis in vitro, in systemic mouse challenge models it improved survival by 20- 37%, exhibiting statistically significant cumulative efficacy when administered at 3x5 mg/kg intraperitoneally or intramuscularly. We hypothesize that the activity in isolated murine macrophages and in vivo is due to deactivation of bacterial toxins. Bacterial protein folding inhibition in synergy with other types of antimicrobial modes offers a remarkable novel strategy in combating resistant or life-threatening infections.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/drug effects , Bacterial Toxins/antagonists & inhibitors , Peptides/therapeutic use , Amino Acid Sequence , Animals , Anthrax/microbiology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/cytology , Bacillus anthracis/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Cell Line , Enterotoxins/antagonists & inhibitors , Enterotoxins/chemistry , Enterotoxins/metabolism , Female , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemistry , Peptides/pharmacology , Protein Folding/drug effects
10.
Peptides ; 43: 96-101, 2013 May.
Article in English | MEDLINE | ID: mdl-23500517

ABSTRACT

Early activation of the innate immune response is important for protection against infection with Francisella tularensis live vaccine strain (LVS) in mice. The human cathelicidin antimicrobial peptide LL-37 is known to have immunomodulatory properties, and therefore exogenously administered LL-37 may be suitable as an early post-exposure therapy to protect against LVS infection. LL-37 has been evaluated for immunostimulatory activity in uninfected mice and for activity against LVS in macrophage assays and protective efficacy when administered post-challenge in a mouse model of respiratory tularemia. Increased levels of pro-inflammatory cytokine IL-6, chemokines monocyte chemoattractant protein 1 (MCP-1) and CXCL1 with increased neutrophil influx into the lungs were observed in uninfected mice after intranasal administration of LL-37. Following LVS challenge, LL-37 administration resulted in increased IL-6, IL-12 p70, IFNγ and MCP-1 production, a slowing of LVS growth in the lung, and a significant extension of mean time to death compared to control mice. However, protection was transient, with the LL-37 treated mice eventually succumbing to infection. As this short course of nasally delivered LL-37 was moderately effective at overcoming the immunosuppressive effects of LVS infection this suggests that a more sustained treatment regimen may be an effective therapy against this pathogen.


Subject(s)
Antimicrobial Cationic Peptides/therapeutic use , Tularemia/drug therapy , Administration, Intranasal , Animals , Antimicrobial Cationic Peptides/administration & dosage , Antimicrobial Cationic Peptides/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Francisella tularensis/drug effects , Francisella tularensis/immunology , Immunomodulation , Macrophages/drug effects , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Structure-Activity Relationship , Tularemia/immunology , Cathelicidins
11.
Microbiology (Reading) ; 158(Pt 6): 1593-1606, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22403187

ABSTRACT

The Toll/interleukin (IL)-1 receptor (TIR) domain is an essential component of eukaryotic innate immune signalling pathways. Interaction between TIR domains present in Toll-like receptors and associated adaptors initiates and propagates an immune signalling cascade. Proteins containing TIR domains have also been discovered in bacteria. Studies have subsequently shown that these proteins are able to modulate mammalian immune signalling pathways dependent on TIR interactions and that this may represent an evasion strategy for bacterial pathogens. Here, we investigate a TIR domain protein from the highly virulent bacterium Yersinia pestis, the causative agent of plague. When overexpressed in vitro this protein is able to downregulate IL-1ß- and LPS-dependent signalling to NFκB and to interact with the TIR adaptor protein MyD88. This interaction is dependent on a single proline residue. However, a Y. pestis knockout mutant lacking the TIR domain protein was not attenuated in virulence in a mouse model of bubonic plague. Minor alterations in the host cytokine response to the mutant were indicated, suggesting a potential subtle role in pathogenesis. The Y. pestis mutant also showed increased auto-aggregation and reduced survival in high-salinity conditions, phenotypes which may contribute to pathogenesis or survival.


Subject(s)
Bacterial Proteins/metabolism , Interleukin-1/metabolism , Plague/metabolism , Plague/microbiology , Toll-Like Receptors/metabolism , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Plague/genetics , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Signal Transduction , Toll-Like Receptors/genetics , Virulence , Yersinia pestis/chemistry , Yersinia pestis/genetics
12.
Microbes Infect ; 14(7-8): 584-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22370220

ABSTRACT

In this paper we evaluate the role of human γδ T cells in control of Francisella tularensis infection. Using an in vitro model of infection, a reduction in bacterial numbers was detected in the presence of human γδ T cells for both attenuated LVS and virulent SCHU S4 strains of F. tularensis. Antibody neutralisation of IFN-γ caused an increase in survival of F. tularensis LVS suggesting that γδ T cell-mediated control of F. tularensis infection is partially mediated by IFN-γ.


Subject(s)
Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Tularemia/immunology , Tularemia/microbiology , Cells, Cultured , Francisella tularensis/classification , Humans , Receptors, Interferon/metabolism , Species Specificity , Interferon gamma Receptor
13.
Vaccine ; 28(41): 6740-8, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20691267

ABSTRACT

Studies have confirmed the key role of Bacillus anthracis protective antigen (PA) in the US and UK human anthrax vaccines. However, given the tripartite nature of the toxin, other components, including lethal factor (LF), are also likely to contribute to protection. We examined the antibody and T cell responses to PA and LF in human volunteers immunized with the UK anthrax vaccine (AVP). Individual LF domains were assessed for immunogenicity in mice when given alone or with PA. Based on the results obtained, a novel fusion protein comprising D1 of LF and the host cell-binding domain of PA (D4) was assessed for protective efficacy. Murine protection studies demonstrated that both full-length LF and D1 of LF conferred complete protection against a lethal intraperitoneal challenge with B. anthracis STI spores. Subsequent studies with the LFD1-PAD4 fusion protein showed a similar level of protection. LF is immunogenic in humans and is likely to contribute to the protection stimulated by AVP. A single vaccine comprising protective regions from LF and PA would simplify production and confer a broader spectrum of protection than that seen with PA alone.


Subject(s)
Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacillus anthracis/immunology , Female , Humans , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology
14.
J Med Microbiol ; 59(Pt 12): 1415-1427, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20798216

ABSTRACT

Deposition of Bacillus anthracis endospores within either the lungs or nasal passages of A/J mice after aerosol exposure was influenced by different particle sized aerosols and resulted in different infection kinetics. The infection resulting from the inhalation of endospores within a 12 µm particle aerosol was prolonged compared to that from a 1 µm particle aerosol with a mean time-to-death of 161 ± 16.1 h and 101.6 ± 10.4 h, respectively. Inhalation of endospores within 1 µm or 12 µm particle aerosols resulted in a median lethal dose of 2432 and 7656 c.f.u., respectively. Initial involvement of the upper respiratory tract lymph nodes was observed in 75-83% of mice exposed to either the 1 µm or 12 µm particle inhalational infections. Lung deposition was significantly greater after inhalation of the 1 µm particle aerosol with pronounced involvement of the mediastinal lymph node. Gastrointestinal involvement was observed only in mice exposed to 12 µm particle aerosols where bacteriological and histopathological analysis indicated primary gastritis (17%), activation of the Peyer's patches (72%) and colonization and necrosis of the mesenteric lymph nodes (67%). Terminal disease was characterized by bacteraemia in both inhalational infections with preferential dissemination to spleen, liver, kidneys and thymus. Immunization with 1 µg recombinant protective antigen vaccine was equally efficacious against B. anthracis infections arising from the inhalation of 1 and 12 µm particle aerosols, providing 73-80% survival under a suboptimum immunization schedule.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Anthrax/transmission , Bacillus anthracis/physiology , Administration, Inhalation , Aerosols , Animals , Disease Models, Animal , Female , Mice , Particle Size
15.
Vaccine ; 27(21): 2775-80, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19366573

ABSTRACT

Yersinia pestis is the causative agent of plague. Naturally occurring cases of the disease and the potential use of Y. pestis as a bioweapon fuel the need for efficacious vaccines. The most recent plague vaccine is a killed whole cell preparation that is expensive to manufacture and its side effects are common. The protective antigens F1 and V have been identified and are currently being developed as a combined subunit vaccine. Protective epitopes of the V antigen have previously been shown to reside in the central part of the protein. In order to identify the minimum protective fragment of the V antigen that can provide protection against plague, the structures of several small fragments of the antigen were modelled in silico and recombinant proteins were produced. These fragments were probed for the retention of a protective epitope using a protective monoclonal antibody and protection against Y. pestis in mice was determined. The smallest protective fragment of V antigen identified comprised amino acids 135-262. Finally the ability of this fragment to confer protection when given in the context of a DNA vaccine was confirmed.


Subject(s)
Antigens, Bacterial/immunology , Peptide Fragments/immunology , Plague/prevention & control , Pore Forming Cytotoxic Proteins/immunology , Yersinia pestis/immunology , Animals , Computer Simulation , Female , Immunoassay , Mice , Mice, Inbred BALB C , Models, Molecular , Peptide Fragments/chemistry , Plague/immunology , Protein Structure, Tertiary
16.
Infect Immun ; 75(11): 5425-33, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17646360

ABSTRACT

The unpredictable nature of bioterrorism and the absence of real-time detection systems have highlighted the need for an efficient postexposure therapy for Bacillus anthracis infection. One approach is passive immunization through the administration of antibodies that mitigate the biological action of anthrax toxin. We isolated and characterized two protective fully human monoclonal antibodies with specificity for protective antigen (PA) and lethal factor (LF). These antibodies, designated IQNPA (anti-PA) and IQNLF (anti-LF), were developed as hybridomas from individuals immunized with licensed anthrax vaccine. The effective concentration of IQNPA that neutralized 50% of the toxin in anthrax toxin neutralization assays was 0.3 nM, while 0.1 nM IQNLF neutralized the same amount of toxin. When combined, the antibodies had additive neutralization efficacy. IQNPA binds to domain IV of PA containing the host cell receptor binding site, while IQNLF recognizes domain I containing the PA binding region in LF. A single 180-mug dose of either antibody given to A/J mice 2.5 h before challenge conferred 100% protection against a lethal intraperitoneal spore challenge with 24 50% lethal doses [LD50s] of B. anthracis Sterne and against rechallenge on day 20 with a more aggressive challenge dose of 41 LD50s. Mice treated with either antibody and infected with B. anthracis Sterne developed detectable murine anti-PA and anti-LF immunoglobulin G antibody responses by day 17 that were dependent on which antibody the mice had received. Based on these results, IQNPA and IQNLF act independently during prophylactic anthrax treatment and do not interfere with the establishment of endogenous immunity.


Subject(s)
Anthrax/drug therapy , Anthrax/prevention & control , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Antitoxins/pharmacology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Animals , Anthrax/immunology , Antibodies, Bacterial/blood , Female , Humans , Hybridomas , Immunization, Passive , Immunoglobulin G/blood , Inhibitory Concentration 50 , Mice , Survival Analysis
17.
Proteomics ; 7(13): 2172-83, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17533643

ABSTRACT

Stimulation of protective immune responses against intracellular pathogens is difficult to achieve using non-replicating vaccines. BALB/c mice immunized by intramuscular injection with killed Francisella tularensis (live vaccine strain) adjuvanted with preformed immune stimulating complexes admixed with CpG, were protected when systemically challenged with a highly virulent strain of F. tularensis (Schu S4). Serum from immunized mice was used to probe a whole proteome microarray in order to identify immunodominant antigens. Eleven out of the top 12 immunodominant antigens have been previously described as immunoreactive in F. tularensis. However, 31 previously unreported immunoreactive antigens were revealed using this approach. Twenty four (50%) of the ORFs on the immunodominant hit list belonged to the category of surface or membrane associated proteins compared to only 22% of the entire proteome. There were eight hypothetical protein hits and eight hits from proteins associated with different aspects of metabolism. The chip also allowed us to readily determine the IgG subclass bias, towards individual or multiple antigens, in protected and unprotected animals. These data give insight into the protective immune response and have potentially important implications for the rational design of non-living vaccines for tularemia and other intracellular pathogens.


Subject(s)
Francisella tularensis/immunology , Immunodominant Epitopes/analysis , Protein Array Analysis/methods , Proteomics/methods , Adjuvants, Immunologic , Aluminum Hydroxide/immunology , Animals , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Female , Francisella tularensis/metabolism , ISCOMs/immunology , Immunodominant Epitopes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/immunology , Proteome/immunology , Proteome/metabolism , Spleen/cytology , Spleen/immunology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tularemia/immunology , Tularemia/microbiology , Tularemia/prevention & control , Vaccination
18.
Vaccine ; 25(2): 346-55, 2007 Jan 04.
Article in English | MEDLINE | ID: mdl-17007969

ABSTRACT

Protective immunity to anthrax can be achieved by antibodies raised against the secreted protective antigen (PA) and this forms the basis of the current acellular vaccines for human use. Bacillus subtilis spores have previously been used for delivery of heterologous antigens by the oral and nasal routes and their intrinsic heat-stability make them attractive vaccine vehicles. In this study we have expressed PA, or segments of PA, in B. subtilis using two strategies. First, display on the spore coat, and second, in the germinated spore (or vegetative cell). Using parenteral delivery we show that recombinant spores can be used to confer protective immunity in a murine model using an in vitro toxin neutralization assay and a challenge experiment with the latter showing protection to 100 median lethal dose of B. anthracis spores. PA must be secreted from the live bacterium or alternatively displayed on the spore surface to confer protective immunity. Intracellular expression of PA failed to confer protective immunity. The highest levels of protective immunity were achieved when PA was displayed on the spore surface as well as in the germinating spore.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Vaccines, Synthetic/immunology , Animals , Antibodies, Bacterial/blood , Female , Immunization , Immunoglobulin G/blood , Immunoglobulin G/classification , Mice , Mice, Inbred BALB C , Neutralization Tests , Spores, Bacterial
19.
Infect Immun ; 74(12): 6624-31, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16982834

ABSTRACT

Caf1, a chaperone-usher protein from Yersinia pestis, is a major protective antigen in the development of subunit vaccines against plague. However, recombinant Caf1 forms polymers of indeterminate size. We report the conversion of Caf1 from a polymer to a monomer by circular permutation of the gene. Biophysical evaluation confirmed that the engineered Caf1 was a folded monomer. We compared the immunogenicity of the engineered monomer with polymeric Caf1 in antigen presentation assays to CD4 T-cell hybridomas in vitro, as well as in the induction of antibody responses and protection against subcutaneous challenge with Y. pestis in vivo. In C57BL/6 mice, for which the major H-2(b)-restricted immunodominant CD4 T-cell epitopes were intact in the engineered monomer, immunogenicity and protective efficacy were preserved, although antibody titers were decreased 10-fold. Disruption of an H-2(d)-restricted immunodominant CD4 T-cell epitope during circular permutation resulted in a compromised T-cell response, a low postvaccination antibody titer, and a lack of protection of BALB/c mice. The use of circular permutation in vaccine design has not been reported previously.


Subject(s)
Bacterial Proteins/immunology , Immunodominant Epitopes/immunology , Plague Vaccine/immunology , Plague/prevention & control , Protein Engineering , Yersinia pestis/immunology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biological Assay , CD4-Positive T-Lymphocytes/immunology , Hybridomas/immunology , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mutation , Plague Vaccine/genetics , Protein Conformation , Protein Folding , Solutions/chemistry
20.
Immunology ; 119(3): 385-92, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16919002

ABSTRACT

We mapped mouse CD4 T-cell epitopes located in three structurally distinct regions of the V antigen of Yersinia pestis. T-cell hybridomas specific for epitopes from each region were generated to study the mechanisms of processing and presentation of V antigen by bone-marrow-derived macrophages. All three epitopes required uptake and/or processing from V antigen as well as presentation to T cells by newly synthesized major histocompatibility complex (MHC) class II molecules over a time period of 3-4 hr. Sensitivity to inhibitors showed a dependence on low pH and cysteine, serine and metalloproteinase, but not aspartic proteinase, activity. The data indicate that immunodominant epitopes from all three structural regions of V antigen were presented preferentially by the classical MHC class II-restricted presentation pathway. The requirement for processing by the co-ordinated activity of several enzyme families is consistent with the buried location of the epitopes in each region of V antigen. Understanding the structure-function relationship of multiple immunodominant epitopes of candidate subunit vaccines is necessary to inform choice of adjuvants for vaccine delivery. In the case of V antigen, adjuvants designed to target it to lysosomes are likely to induce optimal responses to multiple protective T-cell epitopes.


Subject(s)
Antigen Presentation/immunology , Antigens, Bacterial/immunology , Histocompatibility Antigens Class II/immunology , Pore Forming Cytotoxic Proteins/immunology , Yersinia pestis/immunology , Animals , Bone Marrow/immunology , CD4-Positive T-Lymphocytes/immunology , Epitope Mapping/methods , Epitopes, T-Lymphocyte/analysis , Female , Immunodominant Epitopes/analysis , Macrophages/immunology , Mice , Mice, Inbred Strains , Plague Vaccine/immunology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...