Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Front Immunol ; 12: 701341, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777335

RESUMO

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.


Assuntos
Francisella tularensis/imunologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Selenoproteínas/metabolismo , Tularemia/etiologia , Tularemia/metabolismo , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Camundongos , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/microbiologia , Pneumonia/patologia , Tularemia/mortalidade , Virulência/genética , Fatores de Virulência/genética
2.
Microbiologyopen ; 10(2): e1170, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33970545

RESUMO

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.


Assuntos
Proteínas de Bactérias/imunologia , Canais de Cloreto/imunologia , Francisella tularensis/imunologia , Imunidade Inata , Tularemia/imunologia , Tularemia/microbiologia , Animais , Proteínas de Bactérias/genética , Canais de Cloreto/genética , Modelos Animais de Doenças , Francisella tularensis/classificação , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Humanos , Cinética , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
3.
Mol Microbiol ; 115(6): 1357-1378, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33469978

RESUMO

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.


Assuntos
Carboxipeptidases A/metabolismo , Parede Celular/metabolismo , Francisella tularensis/patogenicidade , Peptidoglicano/metabolismo , Tularemia/patologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Francisella tularensis/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C3H , Neutrófilos/microbiologia , Alinhamento de Sequência , Virulência
4.
FEBS Lett ; 594(17): 2782-2799, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32484234

RESUMO

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.


Assuntos
Autofagia/genética , Proteínas de Ligação a DNA/genética , Francisella tularensis/patogenicidade , Complexo de Golgi/imunologia , Fatores de Troca do Nucleotídeo Guanina/genética , Interações Hospedeiro-Patógeno/genética , Macrófagos/imunologia , Antígenos de Histocompatibilidade Menor/genética , Autofagia/efeitos dos fármacos , Morte Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/imunologia , Francisella tularensis/genética , Francisella tularensis/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/microbiologia , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Antígenos de Histocompatibilidade Menor/imunologia , Piridinas/farmacologia , Quinolinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células THP-1 , Acetato de Tetradecanoilforbol/farmacologia
5.
Microb Pathog ; 137: 103742, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31513897

RESUMO

Low molecular mass penicillin binding proteins (LMM PBP) are bacterial enzymes involved in the final steps of peptidoglycan biosynthesis. In Escherichia coli, most LMM PBP exhibit dd-carboxypeptidase activity, are not essential for growth in routine laboratory media, and contributions to virulent phenotypes remain largely unknown. The Francisella tularensis Schu S4 genome harbors the dacD gene (FTT_1029), which encodes a LMM PBP with homology to PBP6b of E. coli. Disruption of this locus in the fully virulent Schu S4 strain resulted in a mutant that could not grow in Chamberlain's Defined Medium and exhibited severe morphological defects. Further characterization studies demonstrated that the growth defects of the dacD mutant were pH-dependent, and could be partially restored by growth at neutral pH or fully restored by genetic complementation. Infection of murine macrophage-like cells showed that the Schu S4 dacD mutant is capable of intracellular replication. However, this mutant was attenuated in BALB/c mice following intranasal challenge (LD50 = 603 CFU) as compared to mice challenged with the parent (LD50 = 1 CFU) or complemented strain (LD50 = 1 CFU). Additionally, mice that survived infection with the dacD mutant showed significant protection against subsequent challenge with the parent strain. Collectively, these results indicate that the DacD protein of F. tularensis is essential for growth in low pH environments and virulence in vivo. These results also suggest that a PBP mutant could serve as the basis of a novel, live attenuated vaccine strain.


Assuntos
Francisella tularensis/enzimologia , Francisella tularensis/patogenicidade , D-Ala-D-Ala Carboxipeptidase Tipo Serina/metabolismo , Tularemia/imunologia , Animais , Proteínas de Bactérias/genética , Vacinas Bacterianas/imunologia , Linhagem Celular , Modelos Animais de Doenças , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Francisella tularensis/genética , Pulmão/microbiologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Proteínas de Ligação às Penicilinas , D-Ala-D-Ala Carboxipeptidase Tipo Serina/genética , Tularemia/microbiologia , Vacinas Atenuadas/imunologia , Virulência , Fatores de Virulência/genética
6.
Artigo em Inglês | MEDLINE | ID: mdl-31334134

RESUMO

Primary interaction of an intracellular bacterium with its host cell is initiated by activation of multiple signaling pathways in response to bacterium recognition itself or as cellular responses to stress induced by the bacterium. The leading molecules in these processes are cell surface membrane receptors as well as cytosolic pattern recognition receptors recognizing pathogen-associated molecular patterns or damage-associated molecular patterns induced by the invading bacterium. In this review, we demonstrate possible sequences of events leading to recognition of Francisella tularensis, present findings on known mechanisms for manipulating cell responses to protect Francisella from being killed, and discuss newly published data from the perspective of early stages of host-pathogen interaction.


Assuntos
Francisella tularensis/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Moléculas com Motivos Associados a Patógenos/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Tularemia/imunologia , Alarminas/genética , Alarminas/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Fagocitose/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores de Reconhecimento de Padrão/genética , Transdução de Sinais , Tularemia/genética , Tularemia/microbiologia
7.
Nat Commun ; 10(1): 2121, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31073183

RESUMO

Pulmonary immune control is crucial for protection against pathogens. Here we identify a pathway that promotes host responses during pulmonary bacterial infection; the expression of CD200 receptor (CD200R), which is known to dampen pulmonary immune responses, promotes effective clearance of the lethal intracellular bacterium Francisella tularensis. We show that depletion of CD200R in mice increases in vitro and in vivo infectious burden. In vivo, CD200R deficiency leads to enhanced bacterial burden in neutrophils, suggesting CD200R normally limits the neutrophil niche for infection. Indeed, depletion of this neutrophil niche in CD200R-/- mice restores F. tularensis infection to levels seen in wild-type mice. Mechanistically, CD200R-deficient neutrophils display significantly reduced reactive oxygen species production (ROS), suggesting that CD200R-mediated ROS production in neutrophils is necessary for limiting F. tularensis colonisation and proliferation. Overall, our data show that CD200R promotes the antimicrobial properties of neutrophils and may represent a novel antibacterial therapeutic target.


Assuntos
Francisella tularensis/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Glicoproteínas de Membrana/imunologia , Neutrófilos/imunologia , Tularemia/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Cultura Primária de Células , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Tularemia/microbiologia
8.
PLoS One ; 14(3): e0213699, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30870480

RESUMO

Francisella tularensis is a Gram-negative, facultative intracellular pathogen and the causative agent of a lethal human disease known as tularemia. Due to its extremely high virulence and potential to be used as a bioterror agent, F. tularensis is classified by the CDC as a Category A Select Agent. As an intracellular pathogen, F. tularensis during its intracellular residence encounters a number of oxidative and nitrosative stresses. The roles of the primary antioxidant enzymes SodB, SodC and KatG in oxidative stress resistance and virulence of F. tularensis live vaccine strain (LVS) have been characterized in previous studies. However, very fragmentary information is available regarding the role of peroxiredoxin of the AhpC/TSA family (annotated as AhpC) of F. tularensis SchuS4; whereas the role of AhpC of F. tularensis LVS in tularemia pathogenesis is not known. This study was undertaken to exhaustively investigate the role of AhpC in oxidative stress resistance of F. tularensis LVS and SchuS4. We report that AhpC of F. tularensis LVS confers resistance against a wide range of reactive oxygen and nitrogen species, and serves as a virulence factor. In highly virulent F. tularensis SchuS4 strain, AhpC serves as a key antioxidant enzyme and contributes to its robust oxidative and nitrosative stress resistance, and intramacrophage survival. We also demonstrate that there is functional redundancy among primary antioxidant enzymes AhpC, SodC, and KatG of F. tularensis SchuS4. Collectively, this study highlights the differences in antioxidant defense mechanisms of F. tularensis LVS and SchuS4.


Assuntos
Antioxidantes/fisiologia , Francisella tularensis/enzimologia , Estresse Oxidativo , Peroxirredoxinas/fisiologia , Tularemia/microbiologia , Animais , Proteínas de Bactérias/fisiologia , Vacinas Bacterianas/imunologia , Francisella tularensis/patogenicidade , Teste de Complementação Genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Superóxido Dismutase/fisiologia , Tularemia/imunologia , Vacinas Atenuadas/imunologia , Virulência
9.
Infect Immun ; 87(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30670554

RESUMO

Francisella tularensis is a Gram-negative, facultative intracellular pathogen and the causative agent of tularemia. Previous studies with the attenuated live vaccine strain (LVS) identified a role for the outer membrane protein TolC in modulation of host cell responses during infection and virulence in the mouse model of tularemia. TolC is an integral part of efflux pumps that export small molecules and type I secretion systems that export a range of bacterial virulence factors. In this study, we analyzed TolC and its two orthologs, FtlC and SilC, present in the fully virulent F. tularensis Schu S4 strain for their contributions to multidrug efflux, suppression of innate immune responses, and virulence. We found that each TolC ortholog participated in multidrug efflux, with overlapping substrate specificities for TolC and FtlC and a distinct substrate profile for SilC. In contrast to their shared roles in drug efflux, only TolC functioned in the modulation of macrophage apoptotic and proinflammatory responses to Schu S4 infection, consistent with a role in virulence factor delivery to host cells. In agreement with previous results with the LVS, the Schu S4 ΔtolC mutant was highly attenuated for virulence in mice by both the intranasal and intradermal routes of infection. Unexpectedly, FtlC was also critical for Schu S4 virulence, but only by the intradermal route. Our data demonstrate a conserved and critical role for TolC in modulation of host immune responses and Francisella virulence and also highlight strain- and route-dependent differences in the pathogenesis of tularemia.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Farmacorresistência Bacteriana Múltipla , Francisella tularensis/efeitos dos fármacos , Francisella tularensis/patogenicidade , Tularemia/microbiologia , Animais , Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/genética , Modelos Animais de Doenças , Feminino , Francisella tularensis/genética , Francisella tularensis/metabolismo , Deleção de Genes , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C3H , Tularemia/genética , Tularemia/imunologia , Virulência , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
10.
Microbiologyopen ; 8(4): e00684, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29978561

RESUMO

Francisella tularensis is the causative agent of the infectious disease tularemia and is designated a category A bioterrorism agent. The type VI secretion system encoded by the Francisella pathogenicity island (FPI) is necessary for intracellular growth; however, the functions of FPI proteins are largely unknown. In this study, we found that the FPI protein intracellular growth locus E (IglE) showed a unique localization pattern compared to other FPI proteins. Deleting iglE from Francisella tularensis subsp. novicida (F. novicida) decreased intracellular growth. Immunoprecipitation and pull-down assays revealed that IglE was associated with ß-tubulin. Additionally, GFP-fused IglE colocalized with microtubule organizing centers (MTOCs) in 293T cells. The iglE deletion mutant was transferred with dynein toward MTOCs and packed into lysosome-localizing areas. Conversely, the wild-type F. novicida exhibited intracellular growth distant from MTOCs. In addition, IglE expressed in 293T cells colocalized with dynein. These results suggest that IglE helps to prevent dynein- and MTOC-mediated intracellular trafficking in host cells to inhibit the transport of F. novicida toward lysosomes.


Assuntos
Proteínas de Bactérias/metabolismo , Francisella tularensis/patogenicidade , Ilhas Genômicas , Centro Organizador dos Microtúbulos/microbiologia , Tularemia/microbiologia , Proteínas de Bactérias/genética , Linhagem Celular , Dineínas/genética , Dineínas/metabolismo , Francisella tularensis/genética , Francisella tularensis/metabolismo , Humanos , Lisossomos/metabolismo , Lisossomos/microbiologia , Transporte Proteico , Tularemia/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
11.
Sci Rep ; 8(1): 11324, 2018 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-30054549

RESUMO

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.


Assuntos
Endopeptidase Clp/genética , Francisella tularensis/genética , Tularemia/genética , Sistemas de Secreção Tipo VI/deficiência , Animais , Vacinas Bacterianas/genética , Vacinas Bacterianas/imunologia , Citoplasma/genética , Citoplasma/microbiologia , Modelos Animais de Doenças , Francisella tularensis/classificação , Francisella tularensis/patogenicidade , Humanos , Macrófagos/microbiologia , Camundongos , Especificidade da Espécie , Tularemia/microbiologia , Sistemas de Secreção Tipo VI/genética
12.
Infect Immun ; 86(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29760217

RESUMO

Francisella tularensis subsp. tularensis is a highly pathogenic intracellular bacterium that suppresses host inflammation by impairing the metabolic shift from oxidative phosphorylation to glycolysis. Decreased mitochondrial metabolism is central to initiating a metabolic shift to glycolysis and regulating inflammation, but F. tularensis subsp. tularensis manipulation of host mitochondrial function has not been explored. We demonstrate, using extracellular flux analysis, that F. tularensis subsp. tularensis infection initially improves host macrophage mitochondrial bioenergetics in a capsule-dependent manner. Enhancement of mitochondrial function by F. tularensis subsp. tularensis allowed for modest replication and inhibition of apoptosis early after infection. However, using live cell imaging, we found that F. tularensis subsp. tularensis facilitated the loss of mitochondrial function at later time points during infection in a capsule-independent fashion. This loss of function was paired with oncosis and rapid bacterial replication. Inhibition of oncosis reduced intracellular bacterial numbers, underscoring the requirement for this process during F. tularensis subsp. tularensis infection. These findings establish that temporal mitochondrial manipulation by F. tularensis subsp. tularensis is critical for maintenance of a noninflammatory environment and subsequently aids in optimal replication and dissemination of this pathogenic organism.


Assuntos
Cápsulas Bacterianas/metabolismo , Morte Celular , Metabolismo Energético , Francisella tularensis/patogenicidade , Interações Hospedeiro-Patógeno , Mitocôndrias/metabolismo , Mitocôndrias/microbiologia , Animais , Carga Bacteriana , Células Cultivadas , Citoplasma/microbiologia , Feminino , Francisella tularensis/crescimento & desenvolvimento , Evasão da Resposta Imune , Inflamação/patologia , Microscopia Intravital , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos Endogâmicos C57BL
13.
PLoS One ; 13(5): e0198140, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29799870

RESUMO

There are no defined correlates of protection for any intracellular pathogen, including the bacterium Francisella tularensis, which causes tularemia. Evaluating vaccine efficacy against sporadic diseases like tularemia using field trials is problematic, and therefore alternative strategies to test vaccine candidates like the Francisella Live Vaccine Strain (LVS), such as testing in animals and applying correlate measurements, are needed. Recently, we described a promising correlate strategy that predicted the degree of vaccine-induced protection in mice given parenteral challenges, primarily when using an attenuated Francisella strain. Here, we demonstrate that using peripheral blood lymphocytes (PBLs) in this approach predicts LVS-mediated protection against respiratory challenge of Fischer 344 rats with fully virulent F. tularensis, with exceptional sensitivity and specificity. Rats were vaccinated with a panel of LVS-derived vaccines and subsequently given lethal respiratory challenges with Type A F. tularensis. In parallel, PBLs from vaccinated rats were evaluated for their functional ability to control intramacrophage Francisella growth in in vitro co-culture assays. PBLs recovered from co-cultures were also evaluated for relative gene expression using a large panel of genes identified in murine studies. In vitro control of LVS intramacrophage replication reflected the hierarchy of protection. Further, despite variability between individuals, 22 genes were significantly more up-regulated in PBLs from rats vaccinated with LVS compared to those from rats vaccinated with the variant LVS-R or heat-killed LVS, which were poorly protective. These genes included IFN-γ, IL-21, NOS2, LTA, T-bet, IL-12rß2, and CCL5. Most importantly, combining quantifications of intramacrophage growth control with 5-7 gene expression levels using multivariate analyses discriminated protected from non-protected individuals with greater than 95% sensitivity and specificity. The results therefore support translation of this approach to non-human primates and people to evaluate new vaccines against Francisella and other intracellular pathogens.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Imunização , Sistema Respiratório/microbiologia , Animais , Feminino , Regulação da Expressão Gênica/imunologia , Imunidade Humoral/imunologia , Macrófagos/microbiologia , Análise Multivariada , Ratos , Linfócitos T/imunologia , Virulência
14.
Artigo em Inglês | MEDLINE | ID: mdl-29740542

RESUMO

Francisella tularensisis subsp. tularensis is an intracellular bacterial pathogen and the causative agent of the life-threatening zoonotic disease tularemia. The Francisella Pathogenicity Island encodes a large secretion apparatus, known as a Type VI Secretion System (T6SS), which is essential for Francisella to escape from its phagosome and multiply within host macrophages and to cause disease in animals. The T6SS, found in one-quarter of Gram-negative bacteria including many highly pathogenic ones, is a recently discovered secretion system that is not yet fully understood. Nevertheless, there have been remarkable advances in our understanding of the structure, composition, and function of T6SSs of several bacteria in the past few years. The system operates like an inside-out headless contractile phage that is anchored to the bacterial membrane via a baseplate and membrane complex. The system injects effector molecules across the inner and outer bacterial membrane and into host prokaryotic or eukaryotic targets to kill, intoxicate, or in the case of Francisella, hijack the target cell. Recent advances include an atomic model of the contractile sheath, insights into the mechanics of sheath contraction, the composition of the baseplate and membrane complex, the process of assembly of the apparatus, and identification of numerous effector molecules and activities. While Francisella T6SS appears to be an outlier among T6SSs, with limited or no sequence homology with other systems, its structure and organization are strikingly similar to other systems. Nevertheless, we have only scratched the surface in uncovering the mysteries of the Francisella T6SS, and there are numerous questions that remain to be answered.


Assuntos
Francisella tularensis/patogenicidade , Tularemia/patologia , Sistemas de Secreção Tipo VI/metabolismo , Animais , Francisella tularensis/genética , Ilhas Genômicas/genética , Humanos , Macrófagos/microbiologia , Transdução de Sinais/fisiologia , Tularemia/microbiologia , Sistemas de Secreção Tipo VI/genética
15.
J Bacteriol ; 200(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29378894

RESUMO

Francisella tularensis, the causative agent of tularemia, lacks typical bacterial virulence factors and toxins but still exhibits extreme virulence. The bacterial multidrug efflux systems consist of an inner membrane, a transmembrane membrane fusion protein, and an outer membrane (OM) component that form a contiguous channel for the secretion of a multitude of bacterial products. Francisella contains three orthologs of the OM proteins; two of these, termed TolC and FtlC, are important for tularemia pathogenesis. The third OM protein, SilC, is homologous to the silver cation efflux protein of other bacterial pathogens. The silC gene (FTL_0686) is located on an operon encoding an Emr-type multidrug efflux pump of F. tularensis The role of SilC in tularemia pathogenesis is not known. In this study, we investigated the role of SilC in secretion and virulence of F. tularensis by generating a silC gene deletion (ΔsilC) mutant and its transcomplemented strain. Our results demonstrate that the ΔsilC mutant exhibits increased sensitivity to antibiotics, oxidants, silver, diminished intramacrophage growth, and attenuated virulence in mice compared to wild-type F. tularensis However, the secretion of antioxidant enzymes of F. tularensis is not impaired in the ΔsilC mutant. The virulence of the ΔsilC mutant is restored in NADPH oxidase-deficient mice, indicating that SilC resists oxidative stress in vivo Collectively, this study demonstrates that the OM component SilC serves a specialized role in virulence of F. tularensis by conferring resistance against oxidative stress and silver.IMPORTANCEFrancisella tularensis, the causative agent of a fatal human disease known as tularemia, is a category A select agent and a potential bioterror agent. The virulence mechanisms of Francisella are not completely understood. This study investigated the role of a unique outer membrane protein, SilC, of a multidrug efflux pump in the virulence of F. tularensis This is the first report demonstrating that the OM component SilC plays an important role in efflux of silver and contributes to the virulence of F. tularensis primarily by providing resistance against oxidative stress. Characterization of these unique virulence mechanisms will provide an understanding of the pathogenesis of tularemia and identification of potential targets for the development of effective therapeutics and prophylactics for protection from this lethal disease.


Assuntos
Proteínas de Bactérias/metabolismo , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidade , Proteínas de Membrana Transportadoras/metabolismo , Estresse Oxidativo , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Francisella tularensis/genética , Deleção de Genes , Macrófagos/microbiologia , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , NADPH Oxidases/genética , Células RAW 264.7 , Prata/farmacologia , Superóxido Dismutase/genética , Virulência
16.
J Bacteriol ; 200(4)2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29109188

RESUMO

Zinc is an essential nutrient for bacterial growth. Because host cells can restrict pathogen access to zinc as an antimicrobial defense mechanism, intracellular pathogens such as Francisella must sense their environment and acquire zinc in response. In many bacteria, the conserved transcription factor Zur is a key regulator of zinc acquisition. To identify mechanisms of zinc uptake in Francisella novicida U112, transcriptome sequencing of wild-type and putative zur mutant bacteria was performed. Only three genes were confirmed as directly regulated by Zur and zinc limitation by quantitative reverse transcription-PCR. One of these genes, FTN_0879, is predicted to encode a protein with similarity to the zupT family of zinc transporters, which are not typically regulated by Zur. While a putative znuACB operon encoding a high-affinity zinc transporter was identified in U112, expression of this operon was not controlled by Zur or zinc concentration. Disruption of zupT but not znuA in U112 impaired growth under zinc limitation, suggesting that ZupT is the primary mechanism of zinc acquisition under these conditions. In the virulent Francisella tularensis subsp. tularensis Schu S4 strain, zupT is a pseudogene, and attempts to delete znuA were unsuccessful, suggesting that it is essential in this strain. A reverse TetR repression system was used to knock down the expression of znuA in Schu S4, revealing that znuA is required for growth under zinc limitation and contributes to intracellular growth within macrophages. Overall, this work identifies genes necessary for adaptation to zinc limitation and highlights nutritional differences between environmental and virulent Francisella strains.IMPORTANCEFrancisella tularensis is a tier 1 select agent with a high potential for lethality and no approved vaccine. A better understanding of Francisella virulence factors is required for the development of therapeutics. While acquisition of zinc has been shown to be required for the virulence of numerous intracellular pathogens, zinc uptake has not been characterized in Francisella This work characterizes the Zur regulon in F. novicida and identifies two transporters that contribute to bacterial growth under zinc limitation. In addition, these data identify differences in mechanisms of zinc uptake and tolerance to zinc limitation between F. tularensis and F. novicida, highlighting the role of znuA in the growth of Schu S4 under zinc limitation.


Assuntos
Microbiologia Ambiental , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidade , Regulação Bacteriana da Expressão Gênica , Zinco/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Francisella tularensis/efeitos dos fármacos , Francisella tularensis/genética , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/genética , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Proteínas de Membrana Transportadoras/genética , Mutação , Óperon , Regulon/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tularemia/microbiologia , Virulência/genética , Zinco/farmacologia
17.
J Bacteriol ; 200(3)2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158241

RESUMO

The alarmone ppGpp is a critical regulator of virulence gene expression in Francisella tularensis In this intracellular pathogen, ppGpp is thought to work in concert with the putative DNA-binding protein PigR and the SspA protein family members MglA and SspA to control a common set of genes. MglA and SspA form a complex that interacts with RNA polymerase (RNAP), and PigR functions by interacting with the RNAP-associated MglA-SspA complex. Prior work suggested that ppGpp indirectly exerts its regulatory effects in F. tularensis by promoting the accumulation of polyphosphate in the cell, which in turn was required for formation of the MglA-SspA complex. Here we show that in Escherichia coli, neither polyphosphate nor ppGpp is required for formation of the MglA-SspA complex but that ppGpp promotes the interaction between PigR and the MglA-SspA complex. Moreover, we show that polyphosphate kinase, the enzyme responsible for the synthesis of polyphosphate, antagonizes virulence gene expression in F. tularensis, a finding that is inconsistent with the notion that polyphosphate accumulation promotes virulence gene expression in this organism. Our findings identify polyphosphate kinase as a novel negative regulator of virulence gene expression in F. tularensis and support a model in which ppGpp exerts its positive regulatory effects by promoting the interaction between PigR and the MglA-SspA complex.IMPORTANCE In Francisella tularensis, MglA and SspA form a complex that associates with RNA polymerase to positively control the expression of key virulence genes. The MglA-SspA complex works together with the putative DNA-binding protein PigR and the alarmone ppGpp. PigR functions by interacting directly with the MglA-SspA complex, but how ppGpp exerts its effects was unclear. Prior work indicated that ppGpp acts by promoting the accumulation of polyphosphate, which is required for MglA and SspA to interact. Here we show that formation of the MglA-SspA complex does not require polyphosphate. Furthermore, we find that polyphosphate antagonizes the expression of virulence genes in F. tularensis Thus, ppGpp does not promote virulence gene expression in this organism through an effect on polyphosphate.


Assuntos
Francisella tularensis/genética , Francisella tularensis/patogenicidade , Regulação Bacteriana da Expressão Gênica , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Escherichia coli/genética , Francisella tularensis/enzimologia , Ilhas Genômicas , Macrófagos/microbiologia , Camundongos , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Polifosfatos/metabolismo , Ligação Proteica , Fatores de Transcrição/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Virulência/genética
18.
mBio ; 8(5)2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-29018123

RESUMO

Francisella tularensis is the causative agent of tularemia and a potential bioterrorism agent. In the present study, we isolated, identified, and quantified the proteins present in the membranes of the virulent type A strain, Schu S4, and the attenuated type B strain, LVS (live vaccine strain). Spectral counting of mass spectrometric data showed enrichment for membrane proteins in both strains. Mice vaccinated with whole LVS membranes encapsulated in poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing the adjuvant polyinosinic-polycytidylic acid [poly(I·C)] showed significant protection against a challenge with LVS compared to the results seen with naive mice or mice vaccinated with either membranes or poly(I·C) alone. The PLGA-encapsulated Schu S4 membranes with poly(I·C) alone did not significantly protect mice from a lethal intraperitoneal challenge with Schu S4; however, this vaccination strategy provided protection from LVS challenge. Mice that received the encapsulated Schu S4 membranes followed by a booster of LVS bacteria showed significant protection with respect to a lethal Schu S4 challenge compared to control mice. Western blot analyses of the sera from the Schu S4-vaccinated mice that received an LVS booster showed four immunoreactive bands. One of these bands from the corresponding one-dimensional (1D) SDS-PAGE experiment represented capsule. The remaining bands were excised, digested with trypsin, and analyzed using mass spectrometry. The most abundant proteins present in these immunoreactive samples were an outer membrane OmpA-like protein, FopA; the type IV pilus fiber building block protein; a hypothetical membrane protein; and lipoproteins LpnA and Lpp3. These proteins should serve as potential targets for future recombinant protein vaccination studies.IMPORTANCE The low infectious dose, the high potential mortality/morbidity rates, and the ability to be disseminated as an aerosol make Francisella tularensis a potential agent for bioterrorism. These characteristics led the Centers for Disease Control (CDC) to classify F. tularensis as a Tier 1 pathogen. Currently, there is no vaccine approved for general use in the United States.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Proteínas de Membrana/imunologia , Tularemia/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia , Adjuvantes Imunológicos , Animais , Modelos Animais de Doenças , Francisella tularensis/química , Francisella tularensis/patogenicidade , Ácido Láctico , Macrófagos/imunologia , Macrófagos/microbiologia , Espectrometria de Massas , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas , Poli I-C/imunologia , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Proteômica , Tularemia/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/genética
19.
Microbiology (Reading) ; 163(11): 1664-1679, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29034854

RESUMO

During conditions of nutrient limitation bacteria undergo a series of global gene expression changes to survive conditions of amino acid and fatty acid starvation. Rapid reallocation of cellular resources is brought about by gene expression changes coordinated by the signalling nucleotides' guanosine tetraphosphate or pentaphosphate, collectively termed (p)ppGpp and is known as the stringent response. The stringent response has been implicated in bacterial virulence, with elevated (p)ppGpp levels being associated with increased virulence gene expression. This has been observed in the highly pathogenic Francisella tularensis sub spp. tularensis SCHU S4, the causative agent of tularaemia. Here, we aimed to artificially induce the stringent response by culturing F. tularensis in the presence of the amino acid analogue l-serine hydroxamate. Serine hydroxamate competitively inhibits tRNAser aminoacylation, causing an accumulation of uncharged tRNA. The uncharged tRNA enters the A site on the translating bacterial ribosome and causes ribosome stalling, in turn stimulating the production of (p)ppGpp and activation of the stringent response. Using the essential virulence gene iglC, which is encoded on the Francisella pathogenicity island (FPI) as a marker of active stringent response, we optimized the culture conditions required for the investigation of virulence gene expression under conditions of nutrient limitation. We subsequently used whole genome RNA-seq to show how F. tularensis alters gene expression on a global scale during active stringent response. Key findings included up-regulation of genes involved in virulence, stress responses and metabolism, and down-regulation of genes involved in metabolite transport and cell division. F. tularensis is a highly virulent intracellular pathogen capable of causing debilitating or fatal disease at extremely low infectious doses. However, virulence mechanisms are still poorly understood. The stringent response is widely recognized as a diverse and complex bacterial stress response implicated in virulence. This work describes the global gene expression profile of F. tularensis SCHU S4 under active stringent response for the first time. Herein we provide evidence for an association of active stringent response with FPI virulence gene expression. Our results further the understanding of the molecular basis of virulence and regulation thereof in F. tularensis. These results also support research into genes involved in (p)ppGpp production and polyphosphate biosynthesis and their applicability as targets for novel antimicrobials.


Assuntos
Adaptação Biológica/fisiologia , Francisella tularensis/fisiologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Ilhas Genômicas/genética , Transcriptoma/fisiologia , Virulência/fisiologia , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Regulação Bacteriana da Expressão Gênica/genética , Genes Bacterianos/genética , Genes Reguladores/genética , Genes Reguladores/fisiologia , Ilhas Genômicas/fisiologia , Sequenciamento de Nucleotídeos em Larga Escala , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/fisiologia , Estresse Oxidativo/genética , Estresse Oxidativo/fisiologia , Proteoma/fisiologia , Análise de Sequência de RNA , Serina/análogos & derivados , Serina/toxicidade , Estresse Fisiológico , Ativação Transcricional/genética , Ativação Transcricional/fisiologia , Transcriptoma/genética , Virulência/genética
20.
Nat Commun ; 8(1): 853, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29021545

RESUMO

The enzyme fructose-bisphosphate aldolase occupies a central position in glycolysis and gluconeogenesis pathways. Beyond its housekeeping role in metabolism, fructose-bisphosphate aldolase has been involved in additional functions and is considered as a potential target for drug development against pathogenic bacteria. Here, we address the role of fructose-bisphosphate aldolase in the bacterial pathogen Francisella novicida. We demonstrate that fructose-bisphosphate aldolase is important for bacterial multiplication in macrophages in the presence of gluconeogenic substrates. In addition, we unravel a direct role of this metabolic enzyme in transcription regulation of genes katG and rpoA, encoding catalase and an RNA polymerase subunit, respectively. We propose a model in which fructose-bisphosphate aldolase participates in the control of host redox homeostasis and the inflammatory immune response.The enzyme fructose-bisphosphate aldolase (FBA) plays central roles in glycolysis and gluconeogenesis. Here, Ziveri et al. show that FBA of the pathogen Francisella novicida acts, in addition, as a transcriptional regulator and is important for bacterial multiplication in macrophages.


Assuntos
Francisella tularensis/enzimologia , Frutose-Bifosfato Aldolase/metabolismo , Regulação Bacteriana da Expressão Gênica , Animais , Feminino , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Frutose-Bifosfato Aldolase/genética , Gluconeogênese , Glucose/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Metabolômica , Camundongos Endogâmicos BALB C , Estresse Oxidativo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA