Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 583
Filter
1.
J Bacteriol ; 206(9): e0024624, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39194223

ABSTRACT

Francisella spp. are Gram-negative, facultative intracellular pathogens. Francisella tularensis causes the human disease tularemia and is considered a biological threat agent due to its high infectivity and virulence. A central aspect of Francisella virulence is its ability to dampen host immune responses. We previously identified the outer membrane channel (OMC) protein TolC as a critical F. tularensis virulence factor required for suppression of apoptotic and proinflammatory responses during macrophage infection. TolC functions as part of multidrug efflux systems and the type I secretion pathway that exports bacterial effector proteins. In these systems, TolC forms tripartite complexes together with an inner membrane transporter and periplasmic membrane fusion protein (MFP). To advance understanding of TolC function in Francisella, we analyzed OMC and MFP homologs in Francisella novicida, a widely used model species that causes a tularemia-like disease in mice. In agreement with the previous F. tularensis studies, all three OMCs present in F. novicida contributed to multidrug resistance, but only TolC was important for suppressing macrophage cell death. In addition, we identified the EmrA1 MFP as important for resisting antimicrobial compounds and dampening host cell death. In contrast to results obtained with F. tularensis, the cell death triggered during infection with the F. novicida tolC and emrA1 mutants was dominated by pyroptosis rather than apoptosis. These data expand our understanding of TolC function in Francisella and underscore both conserved and differential aspects of F. novicida and F. tularensis. IMPORTANCE: Francisella tularensis is a Gram-negative intracellular bacterial pathogen and causative agent of tularemia. We previously identified the outer membrane channel protein TolC as contributing to antimicrobial resistance and subversion of host responses by F. tularensis. To advance understanding of TolC function in Francisella and to identify components that might work together with TolC, we took advantage of a transposon mutant library in F. novicida, a model species that causes a tularemia-like disease in mice. Our findings identify TolC and the membrane fusion protein EmrA1 as important for both antimicrobial resistance and suppression of macrophage cell death. This study also revealed differences in cell death pathways triggered by F. novicida versus F. tularensis infection that may relate to differences in virulence.


Subject(s)
Bacterial Outer Membrane Proteins , Drug Resistance, Multiple, Bacterial , Francisella , Macrophages , Tularemia , Francisella/genetics , Francisella/pathogenicity , Francisella/metabolism , Animals , Mice , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Macrophages/microbiology , Tularemia/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Cell Death , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , Virulence , Anti-Bacterial Agents/pharmacology , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Francisella tularensis/metabolism
3.
PLoS One ; 19(6): e0305569, 2024.
Article in English | MEDLINE | ID: mdl-38889158

ABSTRACT

Francisella tularensis, the causative agent of tularemia, is divided into three subspecies. Two of these, subspecies holarctica and tularensis, are highly pathogenic to humans and consequently relatively well studied. The third subspecies, mediasiatica, is rarely isolated and remains poorly studied. It is distributed in the sparsely populated regions of Central Asia and Siberia. Curently this subspecies is not known to have been responsible for human infections in spite of its high virulence in laboratory animals. Subspecies mediasiatica is currently divided into three subgroups-MI, present in Central Asia, MII, present in southern Siberia, and MIII represented by a unique strain, 60(B)57, isolated in Uzbekistan in 1960. We describe here the unexpected observation that MIII strain 60(B)57 is avirulent and immunogenic. We observed that infection with this strain protected mice from challenge 21 days later with a virulent subsp. mediasiatica strain. With an increase of this interval, the protection for mice was significantly reduced. In contrast, guinea pigs were protected from challenge with strains of the subspecies holarctica and mediasiatica (but not subsp. tularensis) 90 days after infection with 60(B)57. We performed genome assembly based on whole genome sequencing data obtained using the Nanopore MinION for strain 60(B)57 and two subsp. mediasiatica strains representing the Central Asian MI and Siberian MII phylogenetic subgroups. The prmA gene is truncated due to a nonsense mutation in strain 60(B)57. The deletion of gene prmA has previously been shown to induce a loss of virulence in Francisella novicida the closest model organism suggesting that the observed mutation might the cause of the avirulence of strain 60(B)57.


Subject(s)
Francisella tularensis , Tularemia , Animals , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Mice , Virulence/genetics , Tularemia/microbiology , Guinea Pigs , Mutation , Female , Bacterial Proteins/genetics
4.
PLoS One ; 17(1): e0261938, 2022.
Article in English | MEDLINE | ID: mdl-35077486

ABSTRACT

Francisella tularensis is a highly pathogenic intracellular bacterium that causes the disease tularemia. While its ability to replicate within cells has been studied in much detail, the bacterium also encodes a less characterised type 4 pili (T4P) system. T4Ps are dynamic adhesive organelles identified as major virulence determinants in many human pathogens. In F. tularensis, the T4P is required for adherence to the host cell, as well as for protein secretion. Several components, including pilins, a pili peptidase, a secretin pore and two ATPases, are required to assemble a functional T4P, and these are encoded within distinct clusters on the Francisella chromosome. While some of these components have been functionally characterised, the role of PilO, if any, still is unknown. Here, we examined the role of PilO in the pathogenesis of F. novicida. Our results show that the PilO is essential for pilus assembly on the bacterial surface. In addition, PilO is important for adherence of F. novicida to human monocyte-derived macrophages, secretion of effector proteins and intracellular replication. Importantly, the pilO mutant is attenuated for virulence in BALB/c mice regardless of the route of infection. Following intratracheal and intradermal infection, the mutant caused no histopathology changes, and demonstrated impaired phagosomal escape and replication within lung liver as well as spleen. Thus, PilO is an essential virulence determinant of F. novicida.


Subject(s)
Bacterial Adhesion/genetics , Bacterial Proteins , Fimbriae, Bacterial , Francisella , Tularemia , Virulence Factors , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Francisella/genetics , Francisella/metabolism , Francisella/pathogenicity , Francisella/ultrastructure , Francisella tularensis/genetics , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Francisella tularensis/ultrastructure , Humans , Mice , Mice, Inbred BALB C , Tularemia/genetics , Tularemia/metabolism , Tularemia/pathology , Virulence Factors/genetics , Virulence Factors/metabolism
5.
Comput Biol Chem ; 96: 107601, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34801846

ABSTRACT

Due to the high infectivity and fatal effect on human population, Francisella tularensis (F. tularensis) is classified as a potential biological warfare agent. The interaction between host and pathogen behind the successful establishment of F. tularensis infection within the human host is largely unknown. In our present work, we have studied the molecular level interactions between the host cellular components and F. tularensis genes to understand the interplay between the host and pathogen. Interestingly, we have identified the pathways associated with the pathogen offensive strategies that help in invasion of host defensive systems. The F. tularensis genes purL, katG, proS, rpoB and fusA have displayed high number of interactions with the host genes and thus play a crucial role in vital pathogen pathways. The pathways identified were involved in adaptation to different stress conditions within the host and might be crucial for designing new therapeutic interventions against tularemia.


Subject(s)
Francisella tularensis/genetics , Gene Regulatory Networks , Genes, Microbial/genetics , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Host-Pathogen Interactions , Humans
6.
Front Immunol ; 12: 701341, 2021.
Article in English | MEDLINE | ID: mdl-34777335

ABSTRACT

The essential micronutrient Selenium (Se) is co-translationally incorporated as selenocysteine into proteins. Selenoproteins contain one or more selenocysteines and are vital for optimum immunity. Interestingly, many pathogenic bacteria utilize Se for various biological processes suggesting that Se may play a role in bacterial pathogenesis. A previous study had speculated that Francisella tularensis, a facultative intracellular bacterium and the causative agent of tularemia, sequesters Se by upregulating Se-metabolism genes in type II alveolar epithelial cells. Therefore, we investigated the contribution of host vs. pathogen-associated selenoproteins in bacterial disease using F. tularensis as a model organism. We found that F. tularensis was devoid of any Se utilization traits, neither incorporated elemental Se, nor exhibited Se-dependent growth. However, 100% of Se-deficient mice (0.01 ppm Se), which express low levels of selenoproteins, succumbed to F. tularensis-live vaccine strain pulmonary challenge, whereas 50% of mice on Se-supplemented (0.4 ppm Se) and 25% of mice on Se-adequate (0.1 ppm Se) diet succumbed to infection. Median survival time for Se-deficient mice was 8 days post-infection while Se-supplemented and -adequate mice was 11.5 and >14 days post-infection, respectively. Se-deficient macrophages permitted significantly higher intracellular bacterial replication than Se-supplemented macrophages ex vivo, corroborating in vivo observations. Since Francisella replicates in alveolar macrophages during the acute phase of pneumonic infection, we hypothesized that macrophage-specific host selenoproteins may restrict replication and systemic spread of bacteria. F. tularensis infection led to an increased expression of several macrophage selenoproteins, suggesting their key role in limiting bacterial replication. Upon challenge with F. tularensis, mice lacking selenoproteins in macrophages (TrspM) displayed lower survival and increased bacterial burden in the lung and systemic tissues in comparison to WT littermate controls. Furthermore, macrophages from TrspM mice were unable to restrict bacterial replication ex vivo in comparison to macrophages from littermate controls. We herein describe a novel function of host macrophage-specific selenoproteins in restriction of intracellular bacterial replication. These data suggest that host selenoproteins may be considered as novel targets for modulating immune response to control a bacterial infection.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/metabolism , Selenoproteins/metabolism , Tularemia/etiology , Tularemia/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Mice , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/microbiology , Pneumonia/pathology , Tularemia/mortality , Virulence/genetics , Virulence Factors/genetics
7.
J Bacteriol ; 203(23): e0018521, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34543107

ABSTRACT

Francisella tularensis is a Gram-negative bacterium that causes a fatal human disease known as tularemia. The Centers for Disease Control and Prevention have classified F. tularensis as a category A tier 1 select agent. The virulence mechanisms of Francisella are not entirely understood. Francisella possesses very few transcription regulators, and most of these regulate the expression of genes involved in intracellular survival and virulence. The F. tularensis genome sequence analysis reveals an AraC (FTL_0689) transcriptional regulator homologous to the AraC/XylS family of transcriptional regulators. In Gram-negative bacteria, AraC activates genes required for l-arabinose utilization and catabolism. The role of the FTL_0689 regulator in F. tularensis is not known. In this study, we characterized the role of FTL_0689 in the gene regulation of F. tularensis and investigated its contribution to intracellular survival and virulence. The results demonstrate that FTL_0689 in Francisella is not required for l-arabinose utilization. Instead, FTL_0689 specifically regulates the expression of the oxidative and global stress response, virulence, metabolism, and other key pathways genes required by Francisella when exposed to oxidative stress. The FTL_0689 mutant is attenuated for intramacrophage growth and virulence in mice. Based on the deletion mutant phenotype, FTL_0689 was termed osrR (oxidative stress response regulator). Altogether, this study elucidates the role of the osrR transcriptional regulator in tularemia pathogenesis. IMPORTANCE The virulence mechanisms of category A select agent Francisella tularensis, the causative agent of a fatal human disease known as tularemia, remain largely undefined. The present study investigated the role of a transcriptional regulator and its overall contribution to the oxidative stress resistance of F. tularensis. The results provide an insight into a novel gene regulatory mechanism, especially when Francisella is exposed to oxidative stress conditions. Understanding such Francisella- specific regulatory mechanisms will help identify potential targets for developing effective therapies and vaccines to prevent tularemia.


Subject(s)
AraC Transcription Factor/metabolism , Francisella tularensis/metabolism , Gene Expression Regulation, Bacterial/physiology , Oxidative Stress/physiology , Animals , AraC Transcription Factor/genetics , Down-Regulation , Francisella tularensis/pathogenicity , Gene Deletion , Genetic Complementation Test , Mice , Mice, Inbred C57BL , Tularemia/microbiology , Virulence
8.
Nat Commun ; 12(1): 3696, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140472

ABSTRACT

Extracellular vesicles are thought to facilitate pathogen transmission from arthropods to humans and other animals. Here, we reveal that pathogen spreading from arthropods to the mammalian host is multifaceted. Extracellular vesicles from Ixodes scapularis enable tick feeding and promote infection of the mildly virulent rickettsial agent Anaplasma phagocytophilum through the SNARE proteins Vamp33 and Synaptobrevin 2 and dendritic epidermal T cells. However, extracellular vesicles from the tick Dermacentor andersoni mitigate microbial spreading caused by the lethal pathogen Francisella tularensis. Collectively, we establish that tick extracellular vesicles foster distinct outcomes of bacterial infection and assist in vector feeding by acting on skin immunity. Thus, the biology of arthropods should be taken into consideration when developing strategies to control vector-borne diseases.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/metabolism , Extracellular Vesicles/metabolism , Skin/parasitology , Ticks/metabolism , Ticks/microbiology , Anaplasma phagocytophilum/pathogenicity , Animals , Arthropods/metabolism , Arthropods/microbiology , Arthropods/physiology , Cell Line , Dermacentor/metabolism , Dermacentor/microbiology , Dermacentor/physiology , Extracellular Vesicles/ultrastructure , Francisella tularensis/pathogenicity , Gene Ontology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/parasitology , Intravital Microscopy , Ixodes/metabolism , Ixodes/microbiology , Ixodes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Proteomics , R-SNARE Proteins/metabolism , Skin/immunology , Skin/microbiology , T-Lymphocytes/metabolism , Tandem Mass Spectrometry , Vesicle-Associated Membrane Protein 2/metabolism
9.
Microbiologyopen ; 10(2): e1170, 2021 02.
Article in English | MEDLINE | ID: mdl-33970545

ABSTRACT

Tularemia is a zoonotic disease of global proportions. Francisella tularensis subspecies tularensis (type A) and holarctica (type B) cause disease in healthy humans, with type A infections resulting in higher mortality. Repeated passage of a type B strain in the mid-20th century generated the Live Vaccine Strain (LVS). LVS remains unlicensed, does not protect against high inhalational doses of type A, and its exact mechanisms of attenuation are poorly understood. Recent data suggest that live attenuated vaccines derived from type B may cross-protect against type A. However, there is a dearth of knowledge regarding virulent type B pathogenesis and its capacity to stimulate the host's innate immune response. We therefore sought to increase our understanding of virulent type B in vitro characteristics using strain OR96-0246 as a model. Adding to our knowledge of innate immune kinetics in macrophages following infection with virulent type B, we observed robust replication of strain OR96-0246 in murine and human macrophages, reduced expression of pro-inflammatory cytokine genes from "wild type" type B-infected macrophages compared to LVS, and delayed macrophage cell death suggesting that virulent type B may suppress macrophage activation. One disruption in LVS is in the gene encoding the chloride transporter ClcA. We investigated the role of ClcA in macrophage infection and observed a replication delay in a clcA mutant. Here, we propose its role in acid tolerance. A greater understanding of LVS attenuation may reveal new mechanisms of pathogenesis and inform strategies toward the development of an improved vaccine against tularemia.


Subject(s)
Bacterial Proteins/immunology , Chloride Channels/immunology , Francisella tularensis/immunology , Immunity, Innate , Tularemia/immunology , Tularemia/microbiology , Animals , Bacterial Proteins/genetics , Chloride Channels/genetics , Disease Models, Animal , Francisella tularensis/classification , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Humans , Kinetics , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL
10.
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
11.
Mol Microbiol ; 115(6): 1357-1378, 2021 06.
Article in English | MEDLINE | ID: mdl-33469978

ABSTRACT

Francisella tularensis is a Gram-negative, intracellular bacterium that causes the zoonotic disease tularemia. Intracellular pathogens, including F. tularensis, have evolved mechanisms to survive in the harsh environment of macrophages and neutrophils, where they are exposed to cell envelope-damaging molecules. The bacterial cell wall, primarily composed of peptidoglycan (PG), maintains cell morphology, structure, and membrane integrity. Intracellular Gram-negative bacteria protect themselves from macrophage and neutrophil killing by recycling and repairing damaged PG--a process that involves over 50 different PG synthesis and recycling enzymes. Here, we identified a PG recycling enzyme, L,D-carboxypeptidase A (LdcA), of F. tularensis that is responsible for converting PG tetrapeptide stems to tripeptide stems. Unlike E. coli LdcA and most other orthologs, F. tularensis LdcA does not localize to the cytoplasm and also exhibits L,D-endopeptidase activity, converting PG pentapeptide stems to tripeptide stems. Loss of F. tularensis LdcA led to altered cell morphology and membrane integrity, as well as attenuation in a mouse pulmonary infection model and in primary and immortalized macrophages. Finally, an F. tularensis ldcA mutant protected mice against virulent Type A F. tularensis SchuS4 pulmonary challenge.


Subject(s)
Carboxypeptidases A/metabolism , Cell Wall/metabolism , Francisella tularensis/pathogenicity , Peptidoglycan/metabolism , Tularemia/pathology , Amino Acid Sequence , Animals , Cells, Cultured , Disease Models, Animal , Female , Francisella tularensis/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C3H , Neutrophils/microbiology , Sequence Alignment , Virulence
12.
Sci Rep ; 11(1): 1541, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452341

ABSTRACT

There remains to this day a great gap in understanding as to the role of B cells and their products-antibodies and cytokines-in mediating the protective response to Francisella tularensis, a Gram-negative coccobacillus belonging to the group of facultative intracellular bacterial pathogens. We previously have demonstrated that Francisella interacts directly with peritoneal B-1a cells. Here, we demonstrate that, as early as 12 h postinfection, germ-free mice infected with Francisella tularensis produce infection-induced antibody clones reacting with Francisella tularensis proteins having orthologs or analogs in eukaryotic cells. Production of some individual clones was limited in time and was influenced by virulence of the Francisella strain used. The phylogenetically stabilized defense mechanism can utilize these early infection-induced antibodies both to recognize components of the invading pathogens and to eliminate molecular residues of infection-damaged self cells.


Subject(s)
B-Lymphocytes/metabolism , Tularemia/immunology , Animals , Antibody Formation , B-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Francisella tularensis/pathogenicity , Mice , Mice, Inbred BALB C , Tularemia/microbiology , Virulence
13.
Folia Microbiol (Praha) ; 66(1): 1-14, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32989563

ABSTRACT

Tularemia is a bacterial disease of humans, wild, and domestic animals. Francisella tularensis, which is a Gram-negative coccobacillus-shaped bacterium, is the causative agent of tularemia. Recently, an increase in the number of human tularemia cases has been noticed in several countries around the world. It has been reported mostly from North America, several Scandinavian countries, and certain Asian countries. The disease spreads through vectors such as mosquitoes, horseflies, deer flies, and ticks. Humans can acquire the disease through direct contact of sick animals, consumption of infected animals, drinking or direct contact of contaminated water, and inhalation of bacteria-loaded aerosols. Low infectious dose, aerosol route of infection, and its ability to induce fatal disease make it a potential agent of biological warfare. Tularemia leads to several clinical forms, such as glandular, ulceroglandular, oculoglandular, oropharyngeal, respiratory, and typhoidal forms. The disease is diagnosed through the use of culture, serology, or molecular methods. Quinolones, tetracyclines, or aminoglycosides are frequently used in the treatment of tularemia. No licensed vaccine is available in the prophylaxis of tularemia and this is need of the time and high-priority research area. This review mostly focuses on general features, importance, current status, and preventive measures of this disease.


Subject(s)
Communicable Diseases, Emerging/microbiology , Francisella tularensis/pathogenicity , Tularemia/microbiology , Animals , Anti-Bacterial Agents/therapeutic use , Biological Warfare Agents , Communicable Diseases, Emerging/drug therapy , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/prevention & control , Disease Transmission, Infectious/prevention & control , Francisella tularensis/isolation & purification , Humans , Tick-Borne Diseases/drug therapy , Tick-Borne Diseases/epidemiology , Tick-Borne Diseases/microbiology , Tick-Borne Diseases/prevention & control , Tularemia/drug therapy , Tularemia/epidemiology , Tularemia/prevention & control
15.
Mol Cell ; 81(1): 139-152.e10, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33217319

ABSTRACT

The bacterium Francisella tularensis (Ft) is one of the most infectious agents known. Ft virulence is controlled by a unique combination of transcription regulators: the MglA-SspA heterodimer, PigR, and the stress signal, ppGpp. MglA-SspA assembles with the σ70-associated RNAP holoenzyme (RNAPσ70), forming a virulence-specialized polymerase. These factors activate Francisella pathogenicity island (FPI) gene expression, which is required for virulence, but the mechanism is unknown. Here we report FtRNAPσ70-promoter-DNA, FtRNAPσ70-(MglA-SspA)-promoter DNA, and FtRNAPσ70-(MglA-SspA)-ppGpp-PigR-promoter DNA cryo-EM structures. Structural and genetic analyses show MglA-SspA facilitates σ70 binding to DNA to regulate virulence and virulence-enhancing genes. Our Escherichia coli RNAPσ70-homodimeric EcSspA structure suggests this is a general SspA-transcription regulation mechanism. Strikingly, our FtRNAPσ70-(MglA-SspA)-ppGpp-PigR-DNA structure reveals ppGpp binding to MglA-SspA tethers PigR to promoters. PigR in turn recruits FtRNAP αCTDs to DNA UP elements. Thus, these studies unveil a unique mechanism for Ft pathogenesis involving a virulence-specialized RNAP that employs two (MglA-SspA)-based strategies to activate virulence genes.


Subject(s)
DNA-Directed RNA Polymerases , Francisella tularensis , Promoter Regions, Genetic , Sigma Factor , Virulence Factors , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Francisella tularensis/genetics , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Sigma Factor/genetics , Sigma Factor/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
16.
Front Immunol ; 11: 1773, 2020.
Article in English | MEDLINE | ID: mdl-32849637

ABSTRACT

Mucosa-associated invariant T (MAIT) cells are a unique T cell subset that contributes to protective immunity against microbial pathogens, but little is known about the role of chemokines in recruiting MAIT cells to the site of infection. Pulmonary infection with Francisella tularensis live vaccine strain (LVS) stimulates the accrual of large numbers of MAIT cells in the lungs of mice. Using this infection model, we find that MAIT cells are predominantly CXCR6+ but do not require CXCR6 for accumulation in the lungs. However, CXCR6 does contribute to long-term retention of MAIT cells in the airway lumen after clearance of the infection. We also find that MAIT cells are not recruited from secondary lymphoid organs and largely proliferate in situ in the lungs after infection. Nevertheless, the only known ligand for CXCR6, CXCL16, is sufficient to drive MAIT cell accumulation in the lungs in the absence of infection when administered in combination with the MAIT cell antigen 5-OP-RU. Overall, this new data advances the understanding of mechanisms that facilitate MAIT cell accumulation and retention in the lungs.


Subject(s)
Chemokine CXCL16/administration & dosage , Chemotaxis, Leukocyte/drug effects , Francisella tularensis/pathogenicity , Lung/drug effects , Mucosal-Associated Invariant T Cells/drug effects , Pneumonia, Bacterial/metabolism , Receptors, CXCR6/metabolism , Administration, Intranasal , Animals , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL16/metabolism , Coculture Techniques , Disease Models, Animal , Francisella tularensis/immunology , Host-Pathogen Interactions , Lung/immunology , Lung/metabolism , Lung/microbiology , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Mucosal-Associated Invariant T Cells/microbiology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Receptors, CXCR6/deficiency , Receptors, CXCR6/genetics , Ribitol/administration & dosage , Ribitol/analogs & derivatives , Signal Transduction , Uracil/administration & dosage , Uracil/analogs & derivatives
17.
Int J Mol Sci ; 21(15)2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32752076

ABSTRACT

Responsible for tularemia, Francisella tularensis bacteria are highly infectious Gram-negative, category A bioterrorism agents. The molecular mechanisms for their virulence and resistance to antibiotics remain largely unknown. FupA (Fer Utilization Protein), a protein mediating high-affinity transport of ferrous iron across the outer membrane, is associated with both. Recent studies demonstrated that fupA deletion contributed to lower F. tularensis susceptibility towards fluoroquinolones, by increasing the production of outer membrane vesicles. Although the paralogous FupB protein lacks such activity, iron transport capacity and a role in membrane stability were reported for the FupA/B chimera, a protein found in some F. tularensis strains, including the live vaccine strain (LVS). To investigate the mode of action of these proteins, we purified recombinant FupA, FupB and FupA/B proteins expressed in Escherichia coli and incorporated them into mixed lipid bilayers. We examined the porin-forming activity of the FupA/B proteoliposomes using a fluorescent 8-aminonaphthalene-1,3,6-trisulfonic acid, disodium salt (ANTS) probe. Using electrophysiology on tethered bilayer lipid membranes, we confirmed that the FupA/B fusion protein exhibits pore-forming activity with large ionic conductance, a property shared with both FupA and FupB. This demonstration opens up new avenues for identifying functional genes, and novel therapeutic strategies against F. tularensis infections.


Subject(s)
Francisella tularensis/genetics , Iron/metabolism , Porins/genetics , Tularemia/genetics , Bacterial Proteins/genetics , Bacterial Vaccines , Biological Transport/genetics , Biological Transport/immunology , Biological Warfare Agents , Escherichia coli/genetics , Fluoroquinolones/adverse effects , Fluoroquinolones/therapeutic use , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Humans , Porins/metabolism , Tularemia/drug therapy , Tularemia/microbiology
18.
FEBS Lett ; 594(17): 2782-2799, 2020 09.
Article in English | MEDLINE | ID: mdl-32484234

ABSTRACT

Intracellular pathogens affect diverse host cellular defence and metabolic pathways. Here, we used infection with Francisella tularensis to identify SON DNA-binding protein as a central determinant of macrophage activities. RNAi knockdown of SON increases survival of human macrophages following F. tularensis infection or inflammasome stimulation. SON is required for macrophage autophagy, interferon response factor 3 expression, type I interferon response and inflammasome-associated readouts. SON knockdown has gene- and stimulus-specific effects on inflammatory gene expression. SON is required for accurate splicing and expression of GBF1, a key mediator of cis-Golgi structure and function. Chemical GBF1 inhibition has similar effects to SON knockdown, suggesting that SON controls macrophage functions at least in part by controlling Golgi-associated processes.


Subject(s)
Autophagy/genetics , DNA-Binding Proteins/genetics , Francisella tularensis/pathogenicity , Golgi Apparatus/immunology , Guanine Nucleotide Exchange Factors/genetics , Host-Pathogen Interactions/genetics , Macrophages/immunology , Minor Histocompatibility Antigens/genetics , Autophagy/drug effects , Cell Death , Cell Differentiation/drug effects , Cell Line , Cell Survival , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Francisella tularensis/genetics , Francisella tularensis/immunology , Gene Expression Profiling , Gene Expression Regulation , Golgi Apparatus/metabolism , Golgi Apparatus/microbiology , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/immunology , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Macrophages/metabolism , Macrophages/microbiology , Minor Histocompatibility Antigens/immunology , Pyridines/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology
19.
BMC Infect Dis ; 20(1): 327, 2020 May 07.
Article in English | MEDLINE | ID: mdl-32380974

ABSTRACT

BACKGROUND: Tularemia, a re-emerging, potential life threatening infectious disease, can present itself with nonspecific clinical symptoms including fever, chills and malaise. Taking a detailed history of exposure and a highly raised index of clinical suspicion are necessary to take the appropriate diagnostic and therapeutic steps in this setting. Here, a case report of typhoid tularaemia is presented. CASE PRESENTATION: A 53-year old male forester and farmer with protracted fever, abdominal pain, diarrhoea and loss of weight, who experienced productive cough and a pulmonary infiltrate later in the course of disease, was admitted for further investigation. Tularaemia was suspected only owing to history and confirmed by serologic testing more than three weeks after the beginning of the symptoms. The initial antibiotic therapy with ceftriaxone/doxycycline was switched to ciprofloxacin, resulting in the resolution of fever and symptoms. CONCLUSION: Tularaemia has to be considered as a differential diagnosis in febrile patients, even more in cases with protracted fever. Since tularaemia is expanding geographically, involving more animal hosts and causing larger outbreaks, clinicians have to be aware of this potentially fatal disease.


Subject(s)
Fever/microbiology , Tularemia/diagnosis , Tularemia/etiology , Abdominal Pain/diagnosis , Abdominal Pain/drug therapy , Anti-Bacterial Agents/therapeutic use , Body Weight , Ceftriaxone/therapeutic use , Ciprofloxacin/therapeutic use , Cough/drug therapy , Cough/microbiology , Diagnosis, Differential , Diarrhea/drug therapy , Diarrhea/microbiology , Doxycycline/therapeutic use , Farmers , Francisella tularensis/pathogenicity , Humans , Male , Middle Aged , Serologic Tests , Tularemia/drug therapy
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL