Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Virulence ; 11(1): 283-294, 2020 12.
Article in English | MEDLINE | ID: mdl-32241221

ABSTRACT

There is a need for development of an effective vaccine against Francisella tularensis, as this potential bioweapon has a high mortality rate and low infectious dose when delivered via the aerosol route. Moreover, this Tier 1 agent has a history of weaponization. We engineered targeted mutations in the Type A strain F. tularensis subspecies tularensis Schu S4 in aro genes encoding critical enzymes in aromatic amino acid biosynthesis. F. tularensis Schu S4ΔaroC, Schu S4ΔaroD, and Schu S4ΔaroCΔaroD mutant strains were attenuated for intracellular growth in vitro and for virulence in vivo and, conferred protection against pulmonary wild-type (WT) F. tularensis Schu S4 challenge in the C57BL/6 mouse model. F. tularensis Schu S4ΔaroD was identified as the most promising vaccine candidate, demonstrating protection against high-dose intranasal challenge; it protected against 1,000 CFU Schu S4, the highest level of protection tested to date. It also provided complete protection against challenge with 92 CFU of a F. tularensis subspecies holarctica strain (Type B). Mice responded to vaccination with Schu S4ΔaroD with systemic IgM and IgG2c, as well as the production of a functional T cell response as measured in the splenocyte-macrophage co-culture assay. This vaccine was further characterized for dissemination, histopathology, and cytokine/chemokine gene induction at defined time points following intranasal vaccination which confirmed its attenuation compared to WT Schu S4. Cytokine, chemokine, and antibody induction patterns compared to wild-type Schu S4 distinguish protective vs. pathogenic responses to F. tularensis and elucidate correlates of protection associated with vaccination against this agent.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Vaccines/immunology , Cytokines/immunology , Francisella tularensis/genetics , Francisella tularensis/immunology , Macrophages/immunology , Animals , Bacterial Vaccines/administration & dosage , Disease Models, Animal , Female , Gene Deletion , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Vaccines, Attenuated/immunology , Virulence
2.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29311235

ABSTRACT

Francisella tularensis is a Gram-negative, facultative, intracellular coccobacillus that can infect a wide variety of hosts. In humans, F. tularensis causes the zoonosis tularemia following insect bites, ingestion, inhalation, and the handling of infected animals. The fact that a very small inoculum delivered by the aerosol route can cause severe disease, coupled with the possibility of its use as an aerosolized bioweapon, has led to the classification of Francisella tularensis as a category A select agent and has renewed interest in the formulation of a vaccine. To this end, we engineered a type A strain SchuS4 derivative containing a targeted deletion of the major facilitator superfamily (MFS) transporter fptB Based on the attenuating capacity of this deletion in the F. tularensis LVS background, we hypothesized that the deletion of this transporter would alter the intracellular replication and cytokine induction of the type A strain and attenuate virulence in the stringent C57BL/6J mouse model. Here we demonstrate that the deletion of fptB significantly alters the intracellular life cycle of F. tularensis, attenuating intracellular replication in both cell line-derived and primary macrophages and inducing a novel cytosolic escape delay. Additionally, we observed prominent differences in the in vitro cytokine profiles in human macrophage-like cells. The mutant was highly attenuated in the C57BL/6J mouse model and provided partial protection against virulent type A F. tularensis challenge. These results indicate a fundamental necessity for this nutrient transporter in the timely progression of F. tularensis through its replication cycle and in pathogenesis.


Subject(s)
Bacterial Proteins/genetics , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Host-Pathogen Interactions , Membrane Transport Proteins/genetics , Tularemia/microbiology , Animals , Bacterial Proteins/metabolism , Female , Francisella tularensis/metabolism , Gene Deletion , Humans , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Multigene Family , Sequence Deletion , Virulence
3.
PLoS One ; 11(4): e0153402, 2016.
Article in English | MEDLINE | ID: mdl-27100824

ABSTRACT

M-cells (microfold cells) are thought to be a primary conduit of intestinal antigen trafficking. Using an established neutralizing anti-RANKL (Receptor Activator of NF-κB Ligand) antibody treatment to transiently deplete M-cells in vivo, we sought to determine whether intestinal M-cells were required for the effective induction of protective immunity following oral vaccination with ΔiglB (a defined live attenuated Francisella novicida mutant). M-cell depleted, ΔiglB-vaccinated mice exhibited increased (but not significant) morbidity and mortality following a subsequent homotypic or heterotypic pulmonary F. tularensis challenge. No significant differences in splenic IFN-γ, IL-2, or IL-17 or serum antibody (IgG1, IgG2a, IgA) production were observed compared to non-depleted, ΔiglB-vaccinated animals suggesting complementary mechanisms for ΔiglB entry. Thus, we examined other possible routes of gastrointestinal antigen sampling following oral vaccination and found that ΔiglB co-localized to villus goblet cells and enterocytes. These results provide insight into the role of M-cells and complementary pathways in intestinal antigen trafficking that may be involved in the generation of optimal immunity following oral vaccination.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Intestines/cytology , Intestines/immunology , Tularemia/immunology , Tularemia/prevention & control , Vaccines, Attenuated/immunology , Animals , Female , Immunity , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukin-2/immunology , Intestines/microbiology , Mice , Mice, Inbred BALB C , Spleen/immunology , Spleen/microbiology
4.
Pathog Dis ; 73(6): ftv036, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986219

ABSTRACT

Francisella tularensis (Ft), the etiological agent of tularemia and a Tier 1 select agent, has been previously weaponized and remains a high priority for vaccine development. Ft tularensis (type A) and Ft holarctica (type B) cause most human disease. We selected six attenuating genes from the live vaccine strain (LVS; type B), F. novicida and other intracellular bacteria: FTT0507, FTT0584, FTT0742, FTT1019c (guaA), FTT1043 (mip) and FTT1317c (guaB) and created unmarked deletion mutants of each in the highly human virulent Ft strain Schu S4 (Type A) background. FTT0507, FTT0584, FTT0742 and FTT1043 Schu S4 mutants were not attenuated for virulence in vitro or in vivo. In contrast, Schu S4 gua mutants were unable to replicate in murine macrophages and were attenuated in vivo, with an i.n. LD50 > 10(5) CFU in C57BL/6 mice. However, the gua mutants failed to protect mice against lethal challenge with WT Schu S4, despite demonstrating partial protection in rabbits in a previous study. These results contrast with the highly protective capacity of LVS gua mutants against a lethal LVS challenge in mice, and underscore differences between these strains and the animal models in which they are evaluated, and therefore have important implications for vaccine development.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/genetics , Francisella tularensis/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Disease Models, Animal , Francisella tularensis/growth & development , Francisella tularensis/physiology , Gene Deletion , Genes, Bacterial , Lethal Dose 50 , Macrophages/microbiology , Mice, Inbred C57BL , Microbial Viability , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Virulence
5.
PLoS Pathog ; 10(10): e1004439, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25340543

ABSTRACT

Francisella tularensis causes the disease tularemia. Human pulmonary exposure to the most virulent form, F. tularensis subsp. tularensis (Ftt), leads to high morbidity and mortality, resulting in this bacterium being classified as a potential biothreat agent. However, a closely-related species, F. novicida, is avirulent in healthy humans. No tularemia vaccine is currently approved for human use. We demonstrate that a single dose vaccine of a live attenuated F. novicida strain (Fn iglD) protects against subsequent pulmonary challenge with Ftt using two different animal models, Fischer 344 rats and cynomolgus macaques (NHP). The Fn iglD vaccine showed protective efficacy in rats, as did a Ftt iglD vaccine, suggesting no disadvantage to utilizing the low human virulent Francisella species to induce protective immunity. Comparison of specific antibody profiles in vaccinated rat and NHP sera by proteome array identified a core set of immunodominant antigens in vaccinated animals. This is the first report of a defined live attenuated vaccine that demonstrates efficacy against pulmonary tularemia in a NHP, and indicates that the low human virulence F. novicida functions as an effective tularemia vaccine platform.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis , Immunodominant Epitopes/immunology , Tularemia/immunology , Animals , Macaca fascicularis , Mice , Models, Animal , Rats, Inbred F344 , Tularemia/mortality , Tularemia/prevention & control , Vaccination , Vaccines, Attenuated/immunology
6.
Vaccine ; 32(40): 5234-40, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25050972

ABSTRACT

Oral vaccination with the defined live attenuated Francisella novicida vaccine strain U112ΔiglB has been demonstrated to induce protective immunity against pulmonary challenge with the highly human virulent Francisella tularensis strain SCHU S4. However, this vaccination regimen requires a booster dose in mice and Exhibits 50% protective efficacy in the Fischer 344 rat model. To enhance the efficacy of this vaccine strain, we engineered U112ΔiglB to express the Salmonella typhimurium FljB flagellin D1 domain, a TLR5 agonist. The U112ΔiglB::fljB strain was highly attenuated for intracellular macrophage replication, and although the FljB protein was expressed within the cytosol, it exhibited TLR5 activation in a TLR5-expressing HEK cell line. Additionally, infection of splenocytes and lymphocytes with U112ΔiglB::fljB induced significantly greater TNF-α production than infection with U112ΔiglB. Oral vaccination with U112ΔiglB::fljB also induced significantly greater protection than U112ΔiglB against pulmonary SCHU S4 challenge in rats. The enhanced protection was accompanied by higher IgG2a production and serum-mediated reduction of Francisella infectivity. Thus, the U112ΔiglB::fljB strain may serve as a potential vaccine candidate against pneumonic tularemia.


Subject(s)
Bacterial Vaccines/immunology , Flagellin/immunology , Toll-Like Receptor 5/immunology , Tularemia/prevention & control , Animals , Antibodies, Bacterial/blood , Cell Line , Francisella tularensis , Humans , Ligands , Macrophages/microbiology , Mice, Inbred BALB C , Rats, Inbred F344 , Vaccine Potency , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...