Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1238391, 2023.
Article in English | MEDLINE | ID: mdl-37781364

ABSTRACT

Francisella tularensis is the etiological agent of the potentially severe infection tularemia. An existing F: tularensis vaccine, the live vaccine strain (LVS), has been used to protect at-risk personnel, but it is not licensed in any country and it has limited efficacy. Therefore, there is a need of a new, efficacious vaccine. The aim of the study was to perform a detailed analysis of the characteristics of the human immune response to F. tularensis, since this will generate crucial knowledge required to develop new vaccine candidates. Nine individuals were administered the LVS vaccine and peripheral blood mononuclear cells (PBMC) were collected before and at four time points up to one year after vaccination. The properties of the PBMC were characterized by flow cytometry analysis of surface markers and intracellular cytokine staining. In addition, the cytokine content of supernatants from F. tularensis-infected PBMC cultures was determined and the protective properties of the supernatants investigated by adding them to cultures with infected monocyte-derived macrophages (MDM). Unlike before vaccination, PBMC collected at all four time points after vaccination demonstrated F. tularensis-specific cell proliferation, cytokine secretion and cytokine-expressing memory cells. A majority of 17 cytokines were secreted at higher levels by PBMC collected at all time points after vaccination than before vaccination. A discriminative analysis based on IFN-γ and IL-13 secretion correctly classified samples obtained before and after vaccination. Increased expression of IFN-γ, IL-2, and MIP-1ß were observed at all time points after vaccination vs. before vaccination and the most significant changes occurred among the CD4 transient memory, CD8 effector memory, and CD8 transient memory T-cell populations. Growth restriction of the highly virulent F. tularensis strain SCHU S4 in MDM was conferred by supernatants and protection correlated to levels of IFN-γ, IL-2, TNF, and IL-17. The findings demonstrate that F. tularensis vaccination induces long-term T-cell reactivity, including TEM and TTM cell populations. Individual cytokine levels correlated with the degree of protection conferred by the supernatants. Identification of such memory T cells and effector mechanisms provide an improved understanding of the protective mechanisms against F. tularensis. mechanisms against F. tularensis.


Subject(s)
Francisella tularensis , Humans , Leukocytes, Mononuclear , Interleukin-2/metabolism , Bacterial Vaccines , Cytokines/metabolism , Immunity
2.
Cell Rep ; 35(11): 109247, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133919

ABSTRACT

The outer membrane protects Gram-negative bacteria from the host environment. Lipopolysaccharide (LPS), a major outer membrane constituent, has distinct components (lipid A, core, O-antigen) generated by specialized pathways. In this study, we describe the surprising convergence of these pathways through FlmX, an uncharacterized protein in the intracellular pathogen Francisella. FlmX is in the flippase family, which includes proteins that traffic lipid-linked envelope components across membranes. flmX deficiency causes defects in lipid A modification, core remodeling, and O-antigen addition. We find that an F. tularensis mutant lacking flmX is >1,000,000-fold attenuated. Furthermore, FlmX is required to resist the innate antimicrobial LL-37 and the antibiotic polymyxin. Given FlmX's central role in LPS modification and its conservation in intracellular pathogens Brucella, Coxiella, and Legionella, FlmX may represent a novel drug target whose inhibition could cripple bacterial virulence and sensitize bacteria to innate antimicrobials and antibiotics.


Subject(s)
Bacterial Proteins/metabolism , Francisella/metabolism , Francisella/pathogenicity , Lipopolysaccharides/metabolism , Animals , Antimicrobial Cationic Peptides/pharmacology , DNA Transposable Elements/genetics , Escherichia coli/metabolism , Female , Francisella/genetics , Galactosamine/metabolism , Gene Expression Regulation, Bacterial , Immunity, Innate/drug effects , Immunity, Innate/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , O Antigens/metabolism , Polymyxin B/pharmacology , Virulence/genetics
3.
Plasmid ; 115: 102564, 2021 05.
Article in English | MEDLINE | ID: mdl-33610608

ABSTRACT

Francisella tularensis is a Gram-negative intracellular pathogen causing tularemia. A number of its potential virulence factors have been identified, but their biology and functions are not precisely known. Understanding the biological and immunological functions of these proteins requires adequate genetic tools for homologous and heterologous expression of cloned genes, maintaining both original structure and post-translational modifications. Here, we report the construction of a new multipurpose shuttle plasmid - pEVbr - which can be used for high-level expression in F. tularensis. The pEVbr plasmid has been constructed by modifying the TetR-regulated expression vector pEDL17 (LoVullo, 2012) that includes (i) a strong F. tularensis bfr promoter, and (ii) two tet operator sequences cloned into the promoter. The cloned green fluorescent protein (GFP), used as a reporter, demonstrated almost undetectable basal expression level under uninduced conditions and a highly dynamic dose-dependent response to the inducer. The utility of the system was further confirmed by cloning the gapA and FTT_1676 genes into the pEVbr vector and quantifying proteins expression in F. tularensis LVS, as well as by studying post-translational modification of the cloned genes. This study demonstrates that high levels of recombinant native-like Francisella proteins can be produced in Francisella cells. Hence, this system may be beneficial for the analysis of protein function and the development of new treatments and vaccines.


Subject(s)
Francisella tularensis , Tularemia , Francisella tularensis/genetics , Humans , Plasmids/genetics , Recombinant Proteins/genetics , Tetracycline/pharmacology
4.
Appl Environ Microbiol ; 87(6)2021 02 26.
Article in English | MEDLINE | ID: mdl-33397692

ABSTRACT

Francisella tularensis, the causative agent of the zoonotic disease tularemia, can cause seasonal outbreaks of acute febrile illness in humans with disease peaks in late summer to autumn. Interestingly, its mechanisms for environmental persistence between outbreaks are poorly understood. One hypothesis is that F. tularensis forms biofilms in aquatic environments. We utilized two fully virulent wild-type strains: FSC200 (Francisella tularensis subsp. holarctica) and Schu S4 (Francisella tularensis subsp. tularensis) and three control strains, the attenuated live vaccine strain (LVS; F. tularensis subsp. holarctica), a Schu S4 ΔwbtI mutant that is documented to form biofilms, and the low-virulence strain U112 of the closely related species Francisella novicida Strains were incubated in saline solution (0.9% NaCl) microcosms for 24 weeks at both 4°C and 20°C, whereupon viability and biofilm formation were measured. These temperatures were selected to approximate winter and summer temperatures of fresh water in Scandinavia, respectively. U112 and Schu S4 ΔwbtI formed biofilms, but F. tularensis strains FSC200 and Schu S4 and the LVS did not. All strains exhibited prolonged viability at 4°C compared to 20°C. U112 and FSC200 displayed remarkable long-term persistence at 4°C, with only 1- and 2-fold log reductions, respectively, of viable cells after 24 weeks. Schu S4 exhibited lower survival, yielding no viable cells by week 20. At 24 weeks, cells from FSC200, but not from Schu S4, were still fully virulent in mice. Taken together, these results demonstrate biofilm-independent, long-term survival of pathogenic F. tularensis subsp. holarctica in conditions that mimic overwinter survival in aquatic environments.IMPORTANCE Tularemia, a disease caused by the environmental bacterium Francisella tularensis, is characterized by acute febrile illness. F. tularensis is highly infectious: as few as 10 organisms can cause human disease. Tularemia is not known to be spread from person to person. Rather, all human infections are independently acquired from the environment via the bite of blood-feeding arthropods, ingestion of infected food or water, or inhalation of aerosolized bacteria. Despite the environmental origins of human disease events, the ecological factors governing the long-term persistence of F. tularensis in nature between seasonal human outbreaks are poorly understood. The significance of our research is in identifying conditions that promote long-term survival of fully virulent F. tularensis outside a mammalian host or insect vector. These conditions are similar to those found in natural aquatic environments in winter and provide important new insights on how F. tularensis may persist long-term in the environment.


Subject(s)
Francisella tularensis , Fresh Water/microbiology , Animals , Female , Francisella tularensis/pathogenicity , Francisella tularensis/physiology , Mice, Inbred C57BL , Temperature , Tularemia , Virulence
5.
Front Cell Infect Microbiol ; 10: 594063, 2020.
Article in English | MEDLINE | ID: mdl-33363054

ABSTRACT

Francisella tularensis is a Select Agent that causes the severe disease tularemia in humans and many animal species. The bacterium demonstrates rapid intracellular replication, however, macrophages can control its replication if primed and activation with IFN-γ is known to be essential, although alone not sufficient, to mediate such control. To further investigate the mechanisms that control intracellular F. tularensis replication, an in vitro co-culture system was utilized containing splenocytes obtained from naïve or immunized C57BL/6 mice as effectors and infected bone marrow-derived wild-type or chromosome-3-deficient guanylate-binding protein (GBP)-deficient macrophages. Cells were infected either with the F. tularensis live vaccine strain (LVS), the highly virulent SCHU S4 strain, or the surrogate for F. tularensis, F. novicida. Regardless of strain, significant control of the bacterial replication was observed in co-cultures with wild-type macrophages and immune splenocytes, but not in cultures with immune splenocytes and GBPchr3-deficient macrophages. Supernatants demonstrated very distinct, infectious agent-dependent patterns of 23 cytokines, whereas the cytokine patterns were only marginally affected by the presence or absence of GBPs. Levels of a majority of cytokines were inversely correlated to the degree of control of the SCHU S4 and LVS infections, but this was not the case for the F. novicida infection. Collectively, the co-culture assay based on immune mouse-derived splenocytes identified a dominant role of GBPs for the control of intracellular replication of various F. tularensis strains, regardless of their virulence, whereas the cytokine patterns markedly were dependent on the infectious agents, but less so on GBPs.


Subject(s)
Francisella tularensis , Tularemia , Animals , Bacterial Vaccines , Carrier Proteins , Coculture Techniques , Disease Models, Animal , GTP-Binding Proteins , Mice , Mice, Inbred C57BL
6.
Pathogens ; 9(5)2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32365846

ABSTRACT

Francisella tularensis causes the severe disease tularemia. In the present study, the aim was to identify correlates of protection in the rat co-culture model by investigating the immune responses using two vaccine candidates conferring distinct degrees of protection in rat and mouse models. The immune responses were characterized by use of splenocytes from naïve or Live vaccine strain- (LVS) or ∆clpB/∆wbtC-immunized Fischer 344 rats as effectors and bone marrow-derived macrophages infected with the highly virulent strain SCHU S4. A complex immune response was elicited, resulting in cytokine secretion, nitric oxide production, and efficient control of the intracellular bacterial growth. Addition of LVS-immune splenocytes elicited a significantly better control of bacterial growth than ∆clpB/∆wbtC splenocytes. This mirrored the efficacy of the vaccine candidates in the rat model. Lower levels of IFN-γ, TNF, fractalkine, IL-2, and nitrite were present in the co-cultures with ∆clpB/∆wbtC splenocytes than in those with splenocytes from LVS-immunized rats. Nitric oxide was found to be a correlate of protection, since the levels inversely correlated to the degree of protection and inhibition of nitric oxide production completely reversed the growth inhibition of SCHU S4. Overall, the results demonstrate that the co-culture assay with rat-derived cells is a suitable model to identify correlates of protection against highly virulent strains of F. tularensis.

7.
PLoS Pathog ; 16(4): e1008466, 2020 04.
Article in English | MEDLINE | ID: mdl-32275693

ABSTRACT

Francisella tularensis, a highly infectious, intracellular bacterium possesses an atypical type VI secretion system (T6SS), which is essential for its virulence. The chaperone ClpB, a member of the Hsp100/Clp family, is involved in Francisella T6SS disassembly and type VI secretion (T6S) is impaired in its absence. We asked if the role of ClpB for T6S was related to its prototypical role for the disaggregation activity. The latter is dependent on its interaction with the DnaK/Hsp70 chaperone system. Key residues of the ClpB-DnaK interaction were identified by molecular dynamic simulation and verified by targeted mutagenesis. Using such targeted mutants, it was found that the F. novicida ClpB-DnaK interaction was dispensable for T6S, intracellular replication, and virulence in a mouse model, although essential for handling of heat shock. Moreover, by mutagenesis of key amino acids of the Walker A, Walker B, and Arginine finger motifs of each of the two Nucleotide-Binding Domains, their critical roles for heat shock, T6S, intracellular replication, and virulence were identified. In contrast, the N-terminus was dispensable for heat shock, but required for T6S, intracellular replication, and virulence. Complementation of the ΔclpB mutant with a chimeric F. novicida ClpB expressing the N-terminal of Escherichia coli, led to reconstitution of the wild-type phenotype. Collectively, the data demonstrate that the ClpB-DnaK interaction does not contribute to T6S, whereas the N-terminal and NBD domains displayed critical roles for T6S and virulence.


Subject(s)
Endopeptidase Clp/metabolism , Francisella tularensis/physiology , HSP70 Heat-Shock Proteins/metabolism , Animals , Bacterial Proteins/metabolism , Endopeptidase Clp/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Francisella tularensis/genetics , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , HSP70 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Response , Mice , Mice, Inbred C57BL , Molecular Chaperones/metabolism , Molecular Dynamics Simulation , Type VI Secretion Systems/metabolism , Virulence/physiology
8.
Sci Rep ; 8(1): 11324, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054549

ABSTRACT

Francisella tularensis, a highly infectious, intracellular bacterium possesses an atypical type VI secretion system (T6SS), which is essential for the virulence of the bacterium. Recent data suggest that the HSP100 family member, ClpB, is involved in T6SS disassembly in the subspecies Francisella novicida. Here, we investigated the role of ClpB for the function of the T6SS and for phenotypic characteristics of the human pathogenic subspecies holarctica and tularensis. The ∆clpB mutants of the human live vaccine strain, LVS, belonging to subspecies holarctica, and the highly virulent SCHU S4 strain, belonging to subspecies tularensis, both showed extreme susceptibility to heat shock and low pH, severely impaired type VI secretion (T6S), and significant, but impaired intracellular replication compared to the wild-type strains. Moreover, they showed essentially intact phagosomal escape. Infection of mice demonstrated that both ΔclpB mutants were highly attenuated, but the SCHU S4 mutant showed more effective replication than the LVS strain. Collectively, our data demonstrate that ClpB performs multiple functions in the F. tularensis subspecies holarctica and tularensis and its function is important for T6S, intracellular replication, and virulence.


Subject(s)
Endopeptidase Clp/genetics , Francisella tularensis/genetics , Tularemia/genetics , Type VI Secretion Systems/deficiency , Animals , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Cytoplasm/genetics , Cytoplasm/microbiology , Disease Models, Animal , Francisella tularensis/classification , Francisella tularensis/pathogenicity , Humans , Macrophages/microbiology , Mice , Species Specificity , Tularemia/microbiology , Type VI Secretion Systems/genetics
9.
Article in English | MEDLINE | ID: mdl-29468144

ABSTRACT

Cell-mediated immunity (CMI) is normally required for efficient protection against intracellular infections, however, identification of correlates is challenging and they are generally lacking. Francisella tularensis is a highly virulent, facultative intracellular bacterium and CMI is critically required for protection against the pathogen, but how this is effectuated in humans is poorly understood. To understand the protective mechanisms, we established an in vitro co-culture assay to identify how control of infection of F. tularensis is accomplished by human cells and hypothesized that the model will mimic in vivo immune mechanisms. Non-adherent peripheral blood mononuclear cells (PBMCs) were expanded with antigen and added to cultures with adherent PBMC infected with the human vaccine strain, LVS, or the highly virulent SCHU S4 strain. Intracellular numbers of F. tularensis was followed for 72 h and secreted and intracellular cytokines were analyzed. Addition of PBMC expanded from naïve individuals, i.e., those with no record of immunization to F. tularensis, generally resulted in little or no control of intracellular bacterial growth, whereas addition of PBMC from a majority of F. tularensis-immune individuals executed static and sometimes cidal effects on intracellular bacteria. Regardless of infecting strain, statistical differences between the two groups were significant, P < 0.05. Secretion of 11 cytokines was analyzed after 72 h of infection and significant differences with regard to secretion of IFN-γ, TNF, and MIP-1ß was observed between immune and naïve individuals for LVS-infected cultures. Also, in LVS-infected cultures, CD4 T cells from vaccinees, but not CD8 T cells, showed significantly higher expression of IFN-γ, MIP-1ß, TNF, and CD107a than cells from naïve individuals. The co-culture system appears to identify correlates of immunity that are relevant for the understanding of mechanisms of the protective host immunity to F. tularensis.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/physiology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Tularemia/immunology , Tularemia/microbiology , Antigens, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Biomarkers , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Host-Pathogen Interactions/immunology , Humans , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Tularemia/prevention & control
10.
PLoS Pathog ; 13(10): e1006630, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28968459

ABSTRACT

Guanylate binding proteins (GBPs) are interferon-inducible proteins involved in the cell-intrinsic immunity against numerous intracellular pathogens. The molecular mechanisms underlying the potent antibacterial activity of GBPs are still unclear. GBPs have been functionally linked to the NLRP3, the AIM2 and the caspase-11 inflammasomes. Two opposing models are currently proposed to explain the GBPs-inflammasome link: i) GBPs would target intracellular bacteria or bacteria-containing vacuoles to increase cytosolic PAMPs release ii) GBPs would directly facilitate inflammasome complex assembly. Using Francisella novicida infection, we investigated the functional interactions between GBPs and the inflammasome. GBPs, induced in a type I IFN-dependent manner, are required for the F. novicida-mediated AIM2-inflammasome pathway. Here, we demonstrate that GBPs action is not restricted to the AIM2 inflammasome, but controls in a hierarchical manner the activation of different inflammasomes complexes and apoptotic caspases. IFN-γ induces a quantitative switch in GBPs levels and redirects pyroptotic and apoptotic pathways under the control of GBPs. Furthermore, upon IFN-γ priming, F. novicida-infected macrophages restrict cytosolic bacterial replication in a GBP-dependent and inflammasome-independent manner. Finally, in a mouse model of tularemia, we demonstrate that the inflammasome and the GBPs are two key immune pathways functioning largely independently to control F. novicida infection. Altogether, our results indicate that GBPs are the master effectors of IFN-γ-mediated responses against F. novicida to control antibacterial immune responses in inflammasome-dependent and independent manners.


Subject(s)
Francisella tularensis/immunology , GTP-Binding Proteins/immunology , Inflammasomes/immunology , Interferon-gamma/immunology , Tularemia/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Francisella , Gene Knockdown Techniques , Gram-Negative Bacterial Infections/immunology , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Article in English | MEDLINE | ID: mdl-27933275

ABSTRACT

Francisella tularensis is a highly virulent intracellular bacterium and cell-mediated immunity is critical for protection, but mechanisms of protection against highly virulent variants, such as the prototypic strain F. tularensis strain SCHU S4, are poorly understood. To this end, we established a co-culture system, based on splenocytes from naïve, or immunized mice and in vitro infected bone marrow-derived macrophages that allowed assessment of mechanisms controlling infection with F. tularensis. We utilized the system to understand why the clpB gene deletion mutant, ΔclpB, of SCHU S4 shows superior efficacy as a vaccine in the mouse model as compared to the existing human vaccine, the live vaccine strain (LVS). Compared to naïve splenocytes, ΔclpB-, or LVS-immune splenocytes conferred very significant control of a SCHU S4 infection and the ΔclpB-immune splenocytes were superior to the LVS-immune splenocytes. Cultures with the ΔclpB-immune splenocytes also contained higher levels of IFN-γ, IL-17, and GM-CSF and nitric oxide, and T cells expressing combinations of IFN-γ, TNF-α, and IL-17, than did cultures with LVS-immune splenocytes. There was strong inverse correlation between bacterial replication and levels of nitrite, an end product of nitric oxide, and essentially no control was observed when BMDM from iNOS-/- mice were infected. Collectively, the co-culture model identified a critical role of nitric oxide for protection against a highly virulent strain of F. tularensis.


Subject(s)
Coculture Techniques/methods , Francisella tularensis/immunology , Nitric Oxide/analysis , Tularemia/prevention & control , Vaccination , Vaccines/immunology , Animals , Bacterial Proteins/genetics , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Cytokines/metabolism , DNA, Bacterial , DNA, Recombinant , Disease Models, Animal , Endopeptidase Clp , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Heat-Shock Proteins/genetics , Humans , Immunity, Cellular/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Macrophages , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase Type II , Nitrites/analysis , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/prevention & control , Sequence Deletion , T-Lymphocytes/immunology , Tularemia/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccines, Attenuated/immunology
12.
J Antimicrob Chemother ; 71(10): 2815-23, 2016 10.
Article in English | MEDLINE | ID: mdl-27334667

ABSTRACT

OBJECTIVES: We analysed diverse strains of Francisella tularensis subsp. holarctica to assess if its division into biovars I and II is associated with specific mutations previously linked to erythromycin resistance and to determine the distribution of this resistance trait across this subspecies. METHODS: Three-hundred and fourteen F. tularensis subsp. holarctica strains were tested for erythromycin susceptibility and whole-genome sequences for these strains were examined for SNPs in genes previously associated with erythromycin resistance. Each strain was assigned to a global phylogenetic framework using genome-wide canonical SNPs. The contribution of a specific SNP to erythromycin resistance was examined using allelic exchange. The geographical distribution of erythromycin-resistant F. tularensis strains was further investigated by literature search. RESULTS: There was a perfect correlation between biovar II strains (erythromycin resistance) and the phylogenetic group B.12. Only B.12 strains had an A → C SNP at position 2059 in the three copies of the rrl gene. Introducing 2059C into an rrl gene of an erythromycin-susceptible F. tularensis strain resulted in resistance. An additional 1144 erythromycin-resistant strains were identified from the scientific literature, all of them from Eurasia. CONCLUSIONS: Erythromycin resistance in F. tularensis is caused by an A2059C rrl gene mutation, which exhibits a strictly clonal inheritance pattern found only in phylogenetic group B.12. This group is an extremely successful clone, representing the most common type of F. tularensis throughout Eurasia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Erythromycin/pharmacology , Francisella tularensis/drug effects , Francisella tularensis/genetics , Polymorphism, Single Nucleotide , Drug Resistance, Bacterial/genetics , Genes, Bacterial , Genome, Bacterial , Mutation , Phenotype , Phylogeny , RNA, Ribosomal, 23S/genetics
13.
PLoS One ; 10(3): e0119143, 2015.
Article in English | MEDLINE | ID: mdl-25756756

ABSTRACT

Francisella tularensis is a highly virulent facultative intracellular pathogen causing the severe disease tularemia in mammals. As for other bacteria, iron is essential for its growth but very few mechanisms for iron acquisition have been identified. Here, we analyzed if and how F. tularensis can utilize heme, a major source of iron in vivo. This is by no means obvious since the bacterium lacks components of traditional heme-uptake systems. We show that SCHU S4, the prototypic strain of subspecies tularensis, grew in vitro with heme as the sole iron source. By screening a SCHU S4 transposon insertion library, 16 genes were identified as important to efficiently utilize heme, two of which were required to avoid heme toxicity. None of the identified genes appeared to encode components of a potential heme-uptake apparatus. Analysis of SCHU S4 deletion mutants revealed that each of the components FeoB, the siderophore system, and FupA, contributed to the heme-dependent growth. In the case of the former two systems, iron acquisition was impaired, whereas the absence of FupA did not affect iron uptake but led to abnormally high binding of iron to macromolecules. Overall, the present study demonstrates that heme supports growth of F. tularensis and that the requirements for the utilization are highly complex and to some extent novel.


Subject(s)
Francisella tularensis/metabolism , Heme/metabolism , Metabolic Networks and Pathways/genetics , Ferrous Compounds/metabolism , Francisella tularensis/genetics , Francisella tularensis/growth & development , Gene Expression Regulation, Bacterial , Genes, Bacterial
14.
Infect Immun ; 82(9): 3622-35, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24935978

ABSTRACT

Previously, we identified a spontaneous, essentially avirulent mutant, FSC043, of the highly virulent strain SCHU S4 of Francisella tularensis subsp. tularensis. We have now characterized the phenotype of the mutant and the mechanisms of its attenuation in more detail. Genetic and proteomic analyses revealed that the pdpE gene and most of the pdpC gene were very markedly downregulated and, as previously demonstrated, that the strain expressed partially deleted and fused fupA and fupB genes. FSC043 showed minimal intracellular replication and induced no cell cytotoxicity. The mutant showed delayed phagosomal escape; at 18 h, colocalization with LAMP-1 was 80%, indicating phagosomal localization, whereas the corresponding percentages for SCHU S4 and the ΔfupA mutant were <10%. However, a small subset of the FSC043-infected cells contained up to 100 bacteria with LAMP-1 colocalization of around 30%. The unusual intracellular phenotype was similar to that of the ΔpdpC and ΔpdpC ΔpdpE mutants. Complementation of FSC043 with the intact fupA and fupB genes did not affect the phenotype, whereas complementation with the pdpC and pdpE genes restored intracellular replication and led to marked virulence. Even higher virulence was observed after complementation with both double-gene constructs. After immunization with the FSC043 strain, moderate protection against respiratory challenge with the SCHU S4 strain was observed. In summary, FSC043 showed a highly unusual intracellular phenotype, and based on our findings, we hypothesize that the mutation in the pdpC gene makes an essential contribution to the phenotype.


Subject(s)
Bacterial Proteins/genetics , Francisella tularensis/genetics , Tularemia/genetics , Animals , Cell Line , Down-Regulation/genetics , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutation/genetics , Phagosomes/genetics , Phenotype , Proteomics/methods , Tularemia/microbiology , Virulence/genetics
15.
PLoS One ; 8(11): e78671, 2013.
Article in English | MEDLINE | ID: mdl-24236032

ABSTRACT

Francisella tularensis subsp. tularensis is a highly virulent pathogen for humans especially if inhaled. Consequently, it is considered to be a potential biothreat agent. An experimental vaccine, F. tularensis live vaccine strain, derived from the less virulent subsp. holarctica, was developed more than 50 years ago, but remains unlicensed. Previously, we developed a novel live vaccine strain, by deleting the chaperonin clpB gene from F. tularensis subsp. tularensis strain, SCHU S4. SCHU S4ΔclpB was less virulent for mice than LVS and a more effective vaccine against respiratory challenge with wild type SCHU S4. In the current study, we were interested to determine whether a similar mutant on the less virulent subsp. holarctica background would also outperform LVS in terms of safety and efficacy. To this end, clpB was deleted from clinical holarctica strain, FSC200. FSC200ΔclpB had a significantly higher intranasal LD50 than LVS for BALB/c mice, but replicated to higher numbers at foci of infection after dermal inoculation. Moreover, FSC200ΔclpB killed SCID mice more rapidly than LVS. However, dermal vaccination of BALB/c mice with the former versus the latter induced greater protection against respiratory challenge with SCHU S4. This increased efficacy was associated with enhanced production of pulmonary IL-17 after SCHU S4 challenge.


Subject(s)
Bacterial Vaccines/genetics , Francisella tularensis/genetics , Respiratory Tract Infections/prevention & control , Tularemia/prevention & control , Vaccination , Animals , Bacterial Proteins/genetics , Bacterial Vaccines/immunology , Cytokines/metabolism , Francisella tularensis/immunology , Gene Deletion , Heat-Shock Proteins/genetics , Humans , Kinetics , Lung/metabolism , Lung/microbiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Tularemia/immunology , Tularemia/microbiology , Vaccine Potency , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
16.
BMC Microbiol ; 13: 20, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23356941

ABSTRACT

BACKGROUND: A prerequisite for the virulence of the facultative intracellular bacterium Francisella tularensis is effective intramacrophage proliferation, which is preceded by phagosomal escape into the cytosol, and ultimately leads to host cell death. Many components essential for the intracellular life cycle are encoded by a gene cluster, the Francisella pathogenicity island (FPI), constituting a type VI secretion system. RESULTS: We characterized the FPI mutant ΔpdpC of the live vaccine strain (LVS) of F. tularensis and found that it exhibited lack of intracellular replication, incomplete phagosomal escape, and marked attenuation in the mouse model, however, unlike a phagosomally contained FPI mutant, it triggered secretion of IL-1ß, albeit lower than LVS, and markedly induced LDH release. CONCLUSIONS: The phenotype of the ΔpdpC mutant appears to be unique compared to previously described F. tularensis FPI mutants.


Subject(s)
Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Macrophages/microbiology , Virulence Factors/deficiency , Animals , Bacterial Vaccines/genetics , Disease Models, Animal , Female , Francisella tularensis/growth & development , Mice , Mice, Inbred C57BL , Phenotype , Tularemia/microbiology , Tularemia/pathology , Vaccines, Attenuated/genetics , Virulence Factors/genetics
17.
J Med Microbiol ; 60(Pt 11): 1570-1583, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21700740

ABSTRACT

Francisella tularensis is a highly virulent intracellular bacterium capable of rapid multiplication in phagocytic cells. Previous studies have revealed that activation of F. tularensis-infected macrophages leads to control of infection and reactive nitrogen and oxygen species make important contributions to the bacterial killing. We investigated the effects of adding S-nitroso-acetyl-penicillamine (SNAP), which generates nitric oxide, or 3-morpholinosydnonimine hydrochloride, which indirectly leads to formation of peroxynitrite, to J774 murine macrophage-like cell cultures infected with F. tularensis LVS. Addition of SNAP led to significantly increased colocalization between LAMP-1 and bacteria, indicating containment of F. tularensis in the phagosome within 2 h, although no killing occurred within 4 h. A specific inhibitory effect on bacterial transcription was observed since the gene encoding the global regulator MglA was inhibited 50-100-fold. F. tularensis-infected J774 cells were incapable of secreting TNF-α in response to Escherichia coli LPS but addition of SNAP almost completely reversed the suppression. Similarly, infection with an MglA mutant did not inhibit LPS-induced TNF-α secretion of J774 cells. Strong staining of nitrotyrosine was observed in SNAP-treated bacteria, and MS identified nitration of two ribosomal 50S proteins, a CBS domain pair protein and bacterioferritin. The results demonstrated that addition of SNAP initially did not affect the viability of intracellular F. tularensis LVS but led to containment of the bacteria in the phagosome. Moreover, the treatment resulted in modification by nitration of several F. tularensis proteins.


Subject(s)
Francisella tularensis/immunology , Macrophages/immunology , Molsidomine/analogs & derivatives , Nitric Oxide Donors/pharmacology , S-Nitroso-N-Acetylpenicillamine/pharmacology , Tularemia/immunology , Animals , Asialoglycoproteins/genetics , Asialoglycoproteins/immunology , Bacterial Proteins/immunology , Blotting, Western , Cell Line , Cell Survival/immunology , Francisella tularensis/genetics , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Macrophages/microbiology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Molsidomine/pharmacology , Peroxynitrous Acid/immunology , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tularemia/microbiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
18.
BMC Microbiol ; 10: 227, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20796283

ABSTRACT

BACKGROUND: All four Francisella tularensis subspecies possess gene clusters with potential to express type IV pili (Tfp). These clusters include putative pilin genes, as well as pilB, pilC and pilQ, required for secretion and assembly of Tfp. A hallmark of Tfp is the ability to retract the pilus upon surface contact, a property mediated by the ATPase PilT. Interestingly, out of the two major human pathogenic subspecies only the highly virulent type A strains have a functional pilT gene. RESULTS: In a previous study, we were able to show that one pilin gene, pilA, was essential for virulence of a type B strain in a mouse infection model. In this work we have examined the role of several Tfp genes in the virulence of the pathogenic type A strain SCHU S4. pilA, pilC, pilQ, and pilT were mutated by in-frame deletion mutagenesis. Interestingly, when mice were infected with a mixture of each mutant strain and the wild-type strain, the pilA, pilC and pilQ mutants were out-competed, while the pilT mutant was equally competitive as the wild-type. CONCLUSIONS: This suggests that expression and surface localisation of PilA contribute to virulence in the highly virulent type A strain, while PilT was dispensable for virulence in the mouse infection model.


Subject(s)
Fimbriae Proteins/metabolism , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Virulence Factors/metabolism , Animals , Female , Fimbriae Proteins/genetics , Francisella tularensis/genetics , Humans , Mice , Mice, Inbred C57BL , Mutation , Random Allocation , Virulence , Virulence Factors/genetics
19.
Vaccine ; 28(7): 1824-31, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20018266

ABSTRACT

Francisella tularensis subspecies tularensis is a highly virulent facultative intracellular pathogen of humans and a potential biological weapon. A live vaccine strain, F. tularensis LVS, was developed more than 50 years ago by pragmatic attenuation of a strain of the less virulent holarctica subspecies. LVS was demonstrated to be highly effective in human volunteers who were exposed to intradermal challenge with fully virulent subsp. tularensis, but was less effective against aerosol exposure. LVS faces regulatory hurdles that to date have prevented its licensure for general use. Therefore, a better defined and more effective vaccine is being sought. To this end we have created gene deletion mutants in the virulent subsp. tularensis strain and tested them for their ability to elicit a protective immune response against systemic or aerosol challenge with the highly virulent wild-type subsp. tularensis strain, SCHU S4. Both oral and intradermal (ID) primary vaccination routes were assessed in BALB/c and C3H/HeN mice as was oral boosting. One SCHU S4 mutant missing the heat shock gene, clpB, was significantly more attenuated than LVS whereas a double deletion mutant missing genes FTT0918 and capB was as attenuated as LVS. In general mice immunized with SCHU S4DeltaclpB were significantly better protected against aerosol challenge than mice immunized with LVS. A single ID immunization of BALB/c mice with SCHU S4DeltaclpB was at least as effective as any other regimen examined. Mice immunized with SCHU S4Delta0918DeltacapB were generally protected to a similar degree as mice immunized with LVS. A preliminary examination of immune responses to vaccination with LVS, SCHU S4DeltaclpB, or SCHU S4Delta0918DeltacapB provided no obvious correlate to their relative efficacies.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/pathogenicity , Tularemia/prevention & control , Administration, Oral , Aerosols , Animals , Bacterial Vaccines/genetics , Female , Francisella tularensis/genetics , Francisella tularensis/immunology , Immunization, Secondary , Injections, Intradermal , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Sequence Deletion , Tularemia/immunology , Tularemia/pathology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
20.
Infect Immun ; 77(8): 3424-31, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19506014

ABSTRACT

A disadvantage of several old vaccines is that the genetic events resulting in the attenuation are often largely unknown and reversion to virulence cannot be excluded. In the 1950s, a live vaccine strain, LVS, was developed from a type B strain of Francisella tularensis, the causative agent of tularemia. LVS, which is highly attenuated for humans but still virulent for mice by some infection routes, has been extensively studied and found to protect staff from laboratory-acquired tularemia. The efforts to improve biopreparedness have identified a demand for a vaccine against tularemia. Recently the rapid progress in genomics of different Francisella strains has led to identification of several regions of differences (RDs). Two genes carried within RDs, pilA, encoding a putative type IV pilin, and FTT0918, encoding an outer membrane protein, have been linked to virulence. Interestingly, LVS has lost these two genes via direct repeat-mediated deletions. Here we show that reintroduction of the two deleted regions restores virulence of LVS in a mouse infection model to a level indistinguishable from that of virulent type B strains. The identification of the two attenuating deletion events could facilitate the licensing of LVS for use in humans.


Subject(s)
Bacterial Vaccines/genetics , Francisella tularensis/pathogenicity , Genetic Complementation Test , Tularemia/microbiology , Virulence Factors/physiology , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/physiology , Female , Fimbriae Proteins/genetics , Fimbriae Proteins/physiology , Francisella tularensis/genetics , Mice , Mice, Inbred C57BL , Survival Analysis , Virulence , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...