Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
PLoS One ; 18(10): e0293450, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37883420

RESUMEN

Route of exposure to pathogens can inform divergent disease pathogenesis and mortality rates. However, the features that contribute to these differences are not well established. Host metabolism has emerged as a critical element governing susceptibility and the metabolism of tissue exposure sites are unique. Therefore, specific metabolic niches may contribute to the course and outcome of infection depending on route of infection. In the current study, we utilized a combination of imaging and systems metabolomics to map the spatiotemporal dynamics of the host response to intranasal (i.n.) or intradermal (i.d.) infection of mice using the bacterium Francisella tularensis subsp tularensis (FTT). FTT causes lethal disease through these infection routes with similar inoculation doses and replication kinetics, which allowed for isolation of host outcomes independent of bacterial burden. We observed metabolic modifications that were both route dependent and independent. Specifically, i.d. infection resulted in early metabolic reprogramming at the site of infection and draining lymph nodes, whereas the lungs and associated draining lymph nodes were refractory to metabolic reprogramming following i.n. infection. Irrespective of exposure route, FTT promoted metabolic changes in systemic organs prior to colonization, and caused massive dysregulation of host metabolism in these tissues prior to onset of morbidity. Preconditioning infection sites towards a more glycolytic and pro-inflammatory state prior to infection exacerbated FTT replication within the lungs but not intradermal tissue. This enhancement of replication in the lungs was associated with the ability of FTT to limit redox imbalance and alter the pentose phosphate pathway. Together, these studies identify central metabolic features of the lung and dermal compartments that contribute to disease progression and identify potential tissue specific targets that may be exploited for novel therapeutic approaches.


Asunto(s)
Francisella tularensis , Tularemia , Ratones , Animales , Tularemia/metabolismo , Ratones Endogámicos C57BL , Inflamación , Pulmón/metabolismo
2.
mBio ; 14(4): e0113623, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37404047

RESUMEN

Francisella tularensis is a zoonotic pathogen and the causative agent of tularemia. F. tularensis replicates to high levels within the cytosol of macrophages and other host cells while subverting the host response to infection. Critical to the success of F. tularensis is its ability to delay macrophage apoptosis to maintain its intracellular replicative niche. However, the host-signaling pathway(s) modulated by F. tularensis to delay apoptosis are poorly characterized. The outer membrane channel protein TolC is required for F. tularensis virulence and its ability to suppress apoptosis and cytokine expression during infection of macrophages. We took advantage of the F. tularensis ∆tolC mutant phenotype to identify host pathways that are important for activating macrophage apoptosis and that are disrupted by the bacteria. Comparison of macrophages infected with wild-type or ∆tolC F. tularensis revealed that the bacteria interfere with TLR2-MYD88-p38 signaling at early times post infection to delay apoptosis, dampen innate host responses, and preserve the intracellular replicative niche. Experiments using the mouse pneumonic tularemia model confirmed the in vivo relevance of these findings, revealing contributions of TLR2 and MYD88 signaling to the protective host response to F. tularensis, which is modulated by the bacteria to promote virulence. IMPORTANCE Francisella tularensis is a Gram-negative intracellular bacterial pathogen and the causative agent of the zoonotic disease tularemia. F. tularensis, like other intracellular pathogens, modulates host-programmed cell death pathways to ensure its replication and survival. We previously identified the outer membrane channel protein TolC as required for the ability of F. tularensis to delay host cell death. However, the mechanism by which F. tularensis delays cell death pathways during intracellular replication is unclear despite being critical to pathogenesis. In the present study, we address this gap in knowledge by taking advantage of ∆tolC mutants of F. tularensis to uncover signaling pathways governing host apoptotic responses to F. tularensis and which are modulated by the bacteria during infection to promote virulence. These findings reveal mechanisms by which intracellular pathogens subvert host responses and enhance our understanding of the pathogenesis of tularemia.


Asunto(s)
Francisella tularensis , Tularemia , Ratones , Animales , Francisella tularensis/metabolismo , Tularemia/metabolismo , Virulencia , Receptor Toll-Like 2/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Macrófagos/microbiología , Transducción de Señal , Apoptosis , Canales Iónicos/metabolismo
3.
Microbiol Spectr ; 10(5): e0241522, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36190401

RESUMEN

The life-threatening disease tularemia is caused by Francisella tularensis, an intracellular Gram-negative bacterial pathogen. Due to the high mortality rates of the disease, as well as the low respiratory infectious dose, F. tularensis is categorized as a Tier 1 bioterror agent. The identification and isolation from clinical blood cultures of F. tularensis are complicated by its slow growth. Iron was shown to be one of the limiting nutrients required for F. tularensis metabolism and growth. Bacterial growth was shown to be restricted or enhanced in the absence or addition of iron. In this study, we tested the beneficial effect of enhanced iron concentrations on expediting F. tularensis blood culture diagnostics. Accordingly, bacterial growth rates in blood cultures with or without Fe2+ supplementation were evaluated. Growth quantification by direct CFU counts demonstrated significant improvement of growth rates of up to 6 orders of magnitude in Fe2+-supplemented media compared to the corresponding nonmodified cultures. Fe2+ supplementation significantly shortened incubation periods for successful diagnosis and isolation of F. tularensis by up to 92 h. This was achieved in a variety of blood culture types in spite of a low initial bacterial inoculum representative of low levels of bacteremia. These improvements were demonstrated with culture of either Francisella tularensis subsp. tularensis or subsp. holarctica in all examined commercial blood culture types routinely used in a clinical setup. Finally, essential downstream identification assays, such as matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF-MS), immunofluorescence, or antibiotic susceptibility tests, were not affected in the presence of Fe2+. To conclude, supplementing blood cultures with Fe2+ enables a significant shortening of incubation times for F. tularensis diagnosis, without affecting subsequent identification or isolation assays. IMPORTANCE In this study, we evaluated bacterial growth rates of Francisella tularensis strains in iron (Fe)-enriched blood cultures as a means of improving and accelerating bacterial growth. The shortening of the culturing time should facilitate rapid pathogen detection and isolation, positively impacting clinical diagnosis and enabling prompt onset of efficient therapy.


Asunto(s)
Francisella tularensis , Tularemia , Humanos , Francisella tularensis/metabolismo , Cultivo de Sangre , Tularemia/diagnóstico , Tularemia/metabolismo , Tularemia/microbiología , Hierro/metabolismo , Antibacterianos/farmacología
4.
PLoS One ; 17(1): e0261938, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35077486

RESUMEN

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.


Asunto(s)
Adhesión Bacteriana/genética , Proteínas Bacterianas , Fimbrias Bacterianas , Francisella , Tularemia , Factores de Virulencia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/ultraestructura , Francisella/genética , Francisella/metabolismo , Francisella/patogenicidad , Francisella/ultraestructura , Francisella tularensis/genética , Francisella tularensis/metabolismo , Francisella tularensis/patogenicidad , Francisella tularensis/ultraestructura , Humanos , Ratones , Ratones Endogámicos BALB C , Tularemia/genética , Tularemia/metabolismo , Tularemia/patología , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
Front Immunol ; 12: 701341, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777335

RESUMEN

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.


Asunto(s)
Francisella tularensis/inmunología , Interacciones Huésped-Patógeno/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Selenoproteínas/metabolismo , Tularemia/etiología , Tularemia/metabolismo , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Francisella tularensis/genética , Francisella tularensis/patogenicidad , Ratones , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/microbiología , Neumonía/patología , Tularemia/mortalidad , Virulencia/genética , Factores de Virulencia/genética
6.
Sci Rep ; 10(1): 11746, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32678173

RESUMEN

Category A and B biothreat agents are deemed to be of great concern by the US Centers for Disease Control and Prevention (CDC) and include the bacteria Francisella tularensis, Yersinia pestis, Burkholderia mallei, and Brucella species. Underscored by the impact of the 2020 SARS-CoV-2 outbreak, 2016 Zika pandemic, 2014 Ebola outbreak, 2001 anthrax letter attacks, and 1984 Rajneeshee Salmonella attacks, the threat of future epidemics/pandemics and/or terrorist/criminal use of pathogenic organisms warrants continued exploration and development of both classic and alternative methods of detecting biothreat agents. Volatile organic compounds (VOCs) comprise a large and highly diverse group of carbon-based molecules, generally related by their volatility at ambient temperature. Recently, the diagnostic potential of VOCs has been realized, as correlations between the microbial VOC metabolome and specific bacterial pathogens have been identified. Herein, we describe the use of microbial VOC profiles as fingerprints for the identification of biothreat-relevant microbes, and for differentiating between a kanamycin susceptible and resistant strain. Additionally, we demonstrate microbial VOC profiling using a rapid-throughput VOC metabolomics method we refer to as 'simultaneous multifiber headspace solid-phase microextraction' (simulti-hSPME). Finally, through VOC analysis, we illustrate a rapid non-invasive approach to the diagnosis of BALB/c mice infected with either F. tularensis SCHU S4 or Y. pestis CO92.


Asunto(s)
Metabolómica/métodos , Tularemia/metabolismo , Compuestos Orgánicos Volátiles/metabolismo , Animales , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Brotes de Enfermedades , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Femenino , Francisella tularensis/efectos de los fármacos , Francisella tularensis/aislamiento & purificación , Francisella tularensis/metabolismo , Kanamicina/farmacología , Ratones , Ratones Endogámicos BALB C , Pandemias , Neumonía Viral/epidemiología , Neumonía Viral/metabolismo , Neumonía Viral/virología , SARS-CoV-2 , Microextracción en Fase Sólida , Tularemia/microbiología , Tularemia/patología , Tularemia/veterinaria , Compuestos Orgánicos Volátiles/análisis , Compuestos Orgánicos Volátiles/aislamiento & purificación , Yersinia pestis/efectos de los fármacos , Yersinia pestis/aislamiento & purificación , Yersinia pestis/metabolismo
7.
PLoS Pathog ; 16(6): e1008566, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32492066

RESUMEN

Host-derived glutathione (GSH) is an essential source of cysteine for the intracellular pathogen Francisella tularensis. In a comprehensive transposon insertion sequencing screen, we identified several F. tularensis genes that play central and previously unappreciated roles in the utilization of GSH during the growth of the bacterium in macrophages. We show that one of these, a gene we named dptA, encodes a proton-dependent oligopeptide transporter that enables growth of the organism on the dipeptide Cys-Gly, a key breakdown product of GSH generated by the enzyme γ-glutamyltranspeptidase (GGT). Although GGT was thought to be the principal enzyme involved in GSH breakdown in F. tularensis, our screen identified a second enzyme, referred to as ChaC, that is also involved in the utilization of exogenous GSH. However, unlike GGT and DptA, we show that the importance of ChaC in supporting intramacrophage growth extends beyond cysteine acquisition. Taken together, our findings provide a compendium of F. tularensis genes required for intracellular growth and identify new players in the metabolism of GSH that could be attractive targets for therapeutic intervention.


Asunto(s)
Proteínas Bacterianas , Francisella tularensis/fisiología , Glutatión , Interacciones Huésped-Patógeno/fisiología , Macrófagos , Transglutaminasas , Tularemia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Dipéptidos/genética , Dipéptidos/metabolismo , Femenino , Glutatión/genética , Glutatión/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Ratones , Transglutaminasas/genética , Transglutaminasas/metabolismo , Tularemia/genética , Tularemia/metabolismo
8.
Infect Immun ; 88(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31740527

RESUMEN

The mechanisms by which interferon gamma (IFN-γ) controls the replication of cytosolic pathogens independent of responses, such as the generation of reactive oxygen species/reactive nitrogen species (ROS/RNS), have not been fully elucidated. In the current study, we developed a model using Francisella tularensis, the causative agent of tularemia, in which pathways triggered by IFN-γ commonly associated with bacterial control were not required. Using this model, we demonstrated that IFN-γ-mediated production of itaconate and its ability to impair host mitochondrial function, independent of activity on the pathogen, were central for the restriction of bacterial replication in vitro and in vivo We then demonstrate that IFN-γ-driven itaconate production was dispensable, as directly targeting complex II using cell membrane-permeable metabolites also controlled infection. Together, these findings show that while reprogramming of mitochondrial metabolism is a key factor in IFN-γ control of intracellular bacteria, the development of antimicrobial strategies based on targeting host mitochondrial metabolism independent of this cytokine may be an effective therapeutic approach.


Asunto(s)
Francisella tularensis/efectos de los fármacos , Interferón gamma/farmacología , Mitocondrias/efectos de los fármacos , Animales , Membrana Celular/metabolismo , Membrana Celular/microbiología , Citosol/metabolismo , Citosol/microbiología , Humanos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/microbiología , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Succinatos/farmacología , Tularemia/tratamiento farmacológico , Tularemia/metabolismo , Tularemia/microbiología
9.
Methods Mol Biol ; 2024: 59-78, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31364042

RESUMEN

The study of the humoral immune response to infectious and chronic diseases is important for understanding the disease progression, identification of protective antigens, vaccine development, and discovery of biomarkers for early diagnosis. Proteomic approaches, including serological proteome analysis (SERPA), have been used to identify the repertoire of immunoreactive proteins in various diseases. In this chapter, we provide an outline of the SERPA approach, using the analysis of sera from mice vaccinated with a live attenuated tularemia vaccine as an example.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Proteómica/métodos , Animales , Biomarcadores/metabolismo , Western Blotting , Ratones , Tularemia/inmunología , Tularemia/metabolismo
10.
Microbiologyopen ; 8(4): e00684, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29978561

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Francisella tularensis/patogenicidad , Islas Genómicas , Centro Organizador de los Microtúbulos/microbiología , Tularemia/microbiología , Proteínas Bacterianas/genética , Línea Celular , Dineínas/genética , Dineínas/metabolismo , Francisella tularensis/genética , Francisella tularensis/metabolismo , Humanos , Lisosomas/metabolismo , Lisosomas/microbiología , Transporte de Proteínas , Tularemia/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
11.
mBio ; 9(6)2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30459188

RESUMEN

Francisella tularensis is a Gram-negative, facultative, intracellular bacterial pathogen and one of the most virulent organisms known. A hallmark of F. tularensis pathogenesis is the bacterium's ability to replicate to high densities within the cytoplasm of infected cells in over 250 known host species, including humans. This demonstrates that F. tularensis is adept at modulating its metabolism to fluctuating concentrations of host-derived nutrients. The precise metabolic pathways and nutrients utilized by F. tularensis during intracellular growth, however, are poorly understood. Here, we use systematic mutational analysis to identify the carbon catabolic pathways and host-derived nutrients required for F. tularensis intracellular replication. We demonstrate that the glycolytic enzyme phosphofructokinase (PfkA), and thus glycolysis, is dispensable for F. tularensis SchuS4 virulence, and we highlight the importance of the gluconeogenic enzyme fructose 1,6-bisphosphatase (GlpX). We found that the specific gluconeogenic enzymes that function upstream of GlpX varied based on infection model, indicating that F. tularensis alters its metabolic flux according to the nutrients available within its replicative niche. Despite this flexibility, we found that glutamate dehydrogenase (GdhA) and glycerol 3-phosphate (G3P) dehydrogenase (GlpA) are essential for F. tularensis intracellular replication in all infection models tested. Finally, we demonstrate that host cell lipolysis is required for F. tularensis intracellular proliferation, suggesting that host triglyceride stores represent a primary source of glycerol during intracellular replication. Altogether, the data presented here reveal common nutritional requirements for a bacterium that exhibits characteristic metabolic flexibility during infection.IMPORTANCE The widespread onset of antibiotic resistance prioritizes the need for novel antimicrobial strategies to prevent the spread of disease. With its low infectious dose, broad host range, and high rate of mortality, F. tularensis poses a severe risk to public health and is considered a potential agent for bioterrorism. F. tularensis reaches extreme densities within the host cell cytosol, often replicating 1,000-fold in a single cell within 24 hours. This remarkable rate of growth demonstrates that F. tularensis is adept at harvesting and utilizing host cell nutrients. However, like most intracellular pathogens, the types of nutrients utilized by F. tularensis and how they are acquired is not fully understood. Identifying the essential pathways for F. tularensis replication may reveal new therapeutic strategies for targeting this highly infectious pathogen and may provide insight for improved targeting of intracellular pathogens in general.


Asunto(s)
Carbono/metabolismo , Citoplasma/microbiología , Francisella tularensis/crecimiento & desarrollo , Redes y Vías Metabólicas , Animales , Replicación del ADN , Femenino , Francisella tularensis/metabolismo , Fructosa-Bifosfatasa/metabolismo , Gluconeogénesis , Glucólisis , Macrófagos/microbiología , Macrófagos/fisiología , Análisis de Flujos Metabólicos , Ratones , Ratones Endogámicos C57BL , Fosfofructoquinasas/metabolismo , Tularemia/metabolismo , Virulencia
12.
Cell Microbiol ; 20(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29063667

RESUMEN

Francisella tularensis infects several cell types including neutrophils, and aberrant neutrophil accumulation contributes to tissue destruction during tularaemia. We demonstrated previously that F. tularensis strains Schu S4 and live vaccine strain markedly delay human neutrophil apoptosis and thereby prolong cell lifespan, but the bacterial factors that mediate this aspect of virulence are undefined. Herein, we demonstrate that bacterial conditioned medium (CM) can delay apoptosis in the absence of direct infection. Biochemical analyses show that CM contained F. tularensis surface factors as well as outer membrane components. Our previous studies excluded roles for lipopolysaccharide and capsule in apoptosis inhibition, and current studies of [14 C] acetate-labelled bacteria argue against a role for other bacterial lipids in this process. At the same time, studies of isogenic mutants indicate that TolC and virulence factors whose expression requires FevR or MglA were also dispensable, demonstrating that apoptosis inhibition does not require Type I or Type VI secretion. Instead, we identified bacterial lipoproteins (BLPs) as active factors in CM. Additional studies of isolated BLPs demonstrated dose-dependent neutrophil apoptosis inhibition via a TLR2-dependent mechanism that is significantly influenced by a common polymorphism, rs5743618, in human TLR1. These data provide fundamental new insight into pathogen manipulation of neutrophil lifespan and BLP function.


Asunto(s)
Apoptosis/fisiología , Proteínas Bacterianas/metabolismo , Francisella tularensis/metabolismo , Lipoproteínas/metabolismo , Neutrófilos/fisiología , Polimorfismo de Nucleótido Simple/genética , Receptor Toll-Like 1/genética , Francisella tularensis/genética , Humanos , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/fisiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Tularemia/metabolismo , Tularemia/microbiología , Virulencia/genética , Factores de Virulencia/metabolismo
13.
Mol Cell Proteomics ; 17(1): 81-94, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046388

RESUMEN

Dendritic cells (DCs) infected by Francisella tularensis are poorly activated and do not undergo classical maturation process. Although reasons of such unresponsiveness are not fully understood, their impact on the priming of immunity is well appreciated. Previous attempts to explain the behavior of Francisella-infected DCs were hypothesis-driven and focused on events at later stages of infection. Here, we took an alternative unbiased approach by applying methods of global phosphoproteomics to analyze the dynamics of cell signaling in primary DCs during the first hour of infection by Francisella tularensis Presented results show that the early response of DCs to Francisella occurs in phases and that ERK and p38 signaling modules induced at the later stage are differentially regulated by virulent and attenuated ΔdsbA strain. These findings imply that the temporal orchestration of host proinflammatory pathways represents the integral part of Francisella life-cycle inside hijacked DCs.


Asunto(s)
Células Dendríticas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Francisella tularensis , Tularemia/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Células Dendríticas/microbiología , Femenino , Ratones Endogámicos C57BL , Fosforilación
14.
J Leukoc Biol ; 102(6): 1441-1450, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28951422

RESUMEN

Francisella tularensis is a highly infectious intracellular bacterium that causes the potentially fatal disease tularemia. We used mice with conditional MyD88 deficiencies to investigate cellular and molecular mechanisms by which MyD88 restricts type A F. tularensis infection. F. tularensis-induced weight loss was predominately dependent on MyD88 signaling in nonhematopoietic cells. In contrast, MyD88 signaling in hematopoietic cells, but not in myeloid and dendritic cells, was essential for control of F. tularensis infection in tissue. Myeloid and dendritic cell MyD88 deficiency also did not markedly impair cytokine production during infection. Although the production of IL-12 or -18 was not significantly reduced in hematopoietic MyD88-deficient mice, IFN-γ production was abolished in these animals. In addition, neutralization studies revealed that control of F. tularensis infection mediated by hematopoietic MyD88 was entirely dependent on IFN-γ. Although IL-18 production was not significantly affected by MyD88 deficiency, IL-18 was essential for IFN-γ production and restricted bacterial replication in an IFN-γ-dependent manner. Caspase-1 was also found to be partially necessary for the production of IL-18 and IFN-γ and for control of F. tularensis replication. Our collective data show that the response of leukocytes to caspase-1-dependent IL-18 via MyD88 is critical, whereas MyD88 signaling in myeloid and dendritic cells is dispensable for IFN-γ-dependent control of type A F. tularensis infection.


Asunto(s)
Francisella tularensis/fisiología , Hematopoyesis , Interferón gamma/metabolismo , Interleucina-18/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Tularemia/metabolismo , Tularemia/microbiología , Animales , Caspasa 1/metabolismo , Células Dendríticas/metabolismo , Francisella tularensis/patogenicidad , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Transducción de Señal , Tularemia/patología , Virulencia
15.
Artículo en Inglés | MEDLINE | ID: mdl-28680859

RESUMEN

Francisella tularensis is an intracellular pathogen for many animals causing the infectious disease, tularemia. Whereas F. tularensis subsp. holarctica is highly pathogenic for humans, F. novicida is almost avirulent for humans, but virulent for mice. In order to compare metabolic fluxes between these strains, we performed 13C-labeling experiments with F. tularensis subsp. holarctica wild type (beaver isolate), F. tularensis subsp. holarctica strain LVS, or F. novicida strain U112 in complex media containing either [U-13C6]glucose, [1,2-13C2]glucose, [U-13C3]serine, or [U-13C3]glycerol. GC/MS-based isotopolog profiling of amino acids, polysaccharide-derived glucose, free fructose, amino sugars derived from the cell wall, fatty acids, 3-hydroxybutyrate, lactate, succinate and malate revealed uptake and metabolic usage of all tracers under the experimental conditions with glucose being the major carbon source for all strains under study. The labeling patterns of the F. tularensis subsp. holarctica wild type were highly similar to those of the LVS strain, but showed remarkable differences to the labeling profiles of the metabolites from the F. novicida strain. Glucose was directly used for polysaccharide and cell wall biosynthesis with higher rates in F. tularensis subsp. holarctica or metabolized, with higher rates in F. novicida, via glycolysis and the non-oxidative pentose phosphate pathway (PPP). Catabolic turnover of glucose via gluconeogenesis was also observed. In all strains, Ala was mainly synthesized from pyruvate, although no pathway from pyruvate to Ala is annotated in the genomes of F. tularensis and F. novicida. Glycerol efficiently served as a gluconeogenetic substrate in F. novicida, but only less in the F. tularensis subsp. holarctica strains. In any of the studied strains, serine did not serve as a major substrate and was not significantly used for gluconeogenesis under the experimental conditions. Rather, it was only utilized, at low rates, in downstream metabolic processes, e.g., via acetyl-CoA in the citrate cycle and for fatty acid biosynthesis, especially in the F. tularensis subsp. holarctica strains. In summary, the data reflect differential metabolite fluxes in F. tularensis subsp. holarctica and F. novicida suggesting that the different utilization of substrates could be related to host specificity and virulence of Francisella.


Asunto(s)
Francisella tularensis/metabolismo , Francisella/metabolismo , Redes y Vías Metabólicas , Aminoácidos/metabolismo , Pared Celular/química , Medios de Cultivo/química , Francisella/crecimiento & desarrollo , Francisella/patogenicidad , Francisella tularensis/crecimiento & desarrollo , Francisella tularensis/patogenicidad , Glucosa/metabolismo , Glicerol/metabolismo , Polisacáridos/metabolismo , Serina/metabolismo , Coloración y Etiquetado , Tularemia/metabolismo , Tularemia/microbiología , Virulencia
16.
Artículo en Inglés | MEDLINE | ID: mdl-28401066

RESUMEN

Francisella tularensis is a highly infectious Gram-negative bacterium and the causative agent of the zoonotic disease tularemia. This bacterial pathogen can infect a broad variety of animal species and can be transmitted to humans in numerous ways with various clinical outcomes. Although, Francisella possesses the capacity to infect numerous mammalian cell types, the macrophage constitutes the main intracellular niche, used for in vivo bacterial dissemination. To survive and multiply within infected macrophages, Francisella must imperatively escape from the phagosomal compartment. In the cytosol, the bacterium needs to control the host innate immune response and adapt its metabolism to this nutrient-restricted niche. Our laboratory has shown that intracellular Francisella mainly relied on host amino acid as major gluconeogenic substrates and provided evidence that the host metabolism was also modified upon Francisella infection. We will review here our current understanding of how Francisella copes with the available nutrient sources provided by the host cell during the course of infection.


Asunto(s)
Adaptación Fisiológica , Francisella/metabolismo , Francisella/patogenicidad , Interacciones Huésped-Patógeno , Tularemia/metabolismo , Adaptación Fisiológica/genética , Aminoácidos/metabolismo , Animales , Metabolismo de los Hidratos de Carbono , Citosol/metabolismo , Citosol/microbiología , Francisella/genética , Glucólisis , Inmunidad Innata , Macrófagos/metabolismo , Macrófagos/microbiología , Fagosomas/metabolismo , Fagosomas/microbiología , Tularemia/inmunología , Tularemia/microbiología , Factores de Virulencia/metabolismo , Zoonosis/microbiología
17.
J Am Heart Assoc ; 5(9)2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27620887

RESUMEN

BACKGROUND: Sepsis patients with cardiac dysfunction have significantly higher mortality. Although several pathways are associated with myocardial damage in sepsis, the precise cause(s) remains unclear and treatment options are limited. This study was designed to develop a new model to investigate the early events of cardiac damage during sepsis progression. METHODS AND RESULTS: Francisella tularensis subspecies novicida (Ft.n) is a Gram-negative intracellular pathogen causing severe sepsis syndrome in mice. BALB/c mice (N=12) were sham treated or infected with Ft.n through the intranasal route. Serial electrocardiograms were recorded at multiple time points until 96 hours. Hearts were then harvested for histology and gene expression studies. Similar to septic patients, we illustrate both cardiac electrical and structural phenotypes in our murine Ft.n infection model, including prominent R' wave formation, prolonged QRS intervals, and significant left ventricular dysfunction. Notably, in infected animals, we detected numerous microlesions in the myocardium, previously observed following nosocomial Streptococcus infection and in sepsis patients. We show that Ft.n-mediated microlesions are attributed to cardiomyocyte apoptosis, increased immune cell infiltration, and expression of inflammatory mediators (tumor necrosis factor, interleukin [IL]-1ß, IL-8, and superoxide dismutase 2). Finally, we identify increased expression of microRNA-155 and rapid degradation of heat shock factor 1 following cardiac Ft.n infection as a primary cause of myocardial inflammation and apoptosis. CONCLUSIONS: We have developed and characterized an Ft.n infection model to understand the pathogenesis of cardiac dysregulation in sepsis. Our findings illustrate novel in vivo phenotypes underlying cardiac dysfunction during Ft.n infection with significant translational impact on our understanding of sepsis pathophysiology.


Asunto(s)
Corazón/fisiopatología , Miocardio/patología , Sepsis/fisiopatología , Tularemia/fisiopatología , Animales , Apoptosis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Electrocardiografía , Factores de Transcripción del Choque Térmico/metabolismo , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Ratones , MicroARNs/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/patología , Sepsis/metabolismo , Sepsis/patología , Superóxido Dismutasa/metabolismo , Tularemia/metabolismo , Tularemia/patología , Factor de Necrosis Tumoral alfa/metabolismo
18.
PLoS One ; 11(7): e0159740, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27448164

RESUMEN

Pullulanase, an enzyme that catalyzes the hydrolysis of polysaccharides, has been identified in a broad range of organisms, including bacteria, yeasts, fungi, and animals. The pullulanase (pulB; FTT_0412c) of F. tularensis subspecies tularensis Schu S4 is considered to be a homologue of the type I pullulanase (pulA) of the other Francisella subspecies. The significance of Francisella pullulanase has been obscure until now. In the present study, we characterized a recombinant PulB of F. tularensis SCHU P9, which was expressed as a his-tagged protein in Escherichia coli. The recombinant PulB was confirmed to be a type I pullulanase by its enzymatic activity in vitro. A pulB gene knockout mutant of F. tularensis SCHU P9 (ΔpulB) was constructed using the TargeTron Knockout system and plasmid pKEK1140 to clarify the function of PulB during the growth of F. tularensis in macrophages. The intracellular growth of the ΔpulB mutant in murine macrophage J774.1 cells was significantly reduced compared with that of the parental strain SCHU P9. Expression of PulB in ΔpulB, using an expression plasmid, resulted in the complementation of the reduced growth in macrophages, suggesting that PulB is necessary for the efficient growth of F. tularensis in macrophages. To assess the role of PulB in virulence, the knockout and parent bacterial strains were used to infect C57BL/6J mice. Histopathological analyses showed that tissues from ΔpulB-infected mice showed milder lesions compared to those from SCHU P9-infected mice. However, all mice infected with SCHU P9 and ΔpulB showed the similar levels of bacterial loads in their tissues. The results suggest that PulB plays a significant role in bacterial growth within murine macrophage but does not contribute to bacterial virulence in vivo.


Asunto(s)
Francisella tularensis/enzimología , Francisella tularensis/crecimiento & desarrollo , Glicósido Hidrolasas/metabolismo , Tularemia/microbiología , Animales , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Francisella tularensis/genética , Francisella tularensis/patogenicidad , Glicósido Hidrolasas/genética , Concentración de Iones de Hidrógeno , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Mutación , Temperatura , Tularemia/inmunología , Tularemia/metabolismo , Tularemia/patología , Virulencia
19.
J Biol Chem ; 291(8): 3871-81, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26679996

RESUMEN

As an innate defense mechanism, macrophages produce reactive oxygen species that weaken pathogens and serve as secondary messengers involved in immune function. The Gram-negative bacterium Francisella tularensis utilizes its antioxidant armature to limit the host immune response, but the mechanism behind this suppression is not defined. Here we establish that F. tularensis limits Ca(2+) entry in macrophages, thereby limiting actin reorganization and IL-6 production in a redox-dependent fashion. Wild type (live vaccine strain) or catalase-deficient F. tularensis (ΔkatG) show distinct profiles in their H2O2 scavenging rates, 1 and 0.015 pm/s, respectively. Murine alveolar macrophages infected with ΔkatG display abnormally high basal intracellular Ca(2+) concentration that did not increase further in response to H2O2. Additionally, ΔkatG-infected macrophages displayed limited Ca(2+) influx in response to ionomycin, as a result of ionophore H2O2 sensitivity. Exogenously added H2O2 or H2O2 generated by ΔkatG likely oxidizes ionomycin and alters its ability to transport Ca(2+). Basal increases in cytosolic Ca(2+) and insensitivity to H2O2-mediated Ca(2+) entry in ΔkatG-infected cells are reversed by the Ca(2+) channel inhibitors 2-aminoethyl diphenylborinate and SKF-96365. 2-Aminoethyl diphenylborinate but not SKF-96365 abrogated ΔkatG-dependent increases in macrophage actin remodeling and IL-6 secretion, suggesting a role for H2O2-mediated Ca(2+) entry through the transient receptor potential melastatin 2 (TRPM2) channel in macrophages. Indeed, increases in basal Ca(2+), actin polymerization, and IL-6 production are reversed in TRPM2-null macrophages infected with ΔkatG. Together, our findings provide compelling evidence that F. tularensis catalase restricts reactive oxygen species to temper macrophage TRPM2-mediated Ca(2+) signaling and limit host immune function.


Asunto(s)
Proteínas Bacterianas/inmunología , Catalasa/inmunología , Francisella tularensis/inmunología , Inmunidad Innata , Macrófagos/inmunología , Canales Catiónicos TRPM/inmunología , Tularemia/inmunología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Calcio/inmunología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Catalasa/genética , Catalasa/metabolismo , Femenino , Francisella tularensis/enzimología , Francisella tularensis/genética , Eliminación de Gen , Peróxido de Hidrógeno/inmunología , Peróxido de Hidrógeno/metabolismo , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-6/metabolismo , Ionomicina/farmacología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Noqueados , Oxidación-Reducción/efectos de los fármacos , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo , Tularemia/genética , Tularemia/metabolismo
20.
Mol Gen Mikrobiol Virusol ; 33(3): 33-9, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26665740

RESUMEN

The live vaccine based on the Francisella tularensis subsp. holarctica vaccine strain 15 NIIEG line is used in Russia against tularemia. This vaccine is highly effective, but fairly unstable. Therefore, development of stable live tularemia vaccine with minimal side effect is rather urgent. The method of allel removal in the F. tularensis vaccine strain was used to remove one copy of the iglC gene, which is required to provide intracellular production of the vaccine strain, as well as removal of the recA gene. The latter is crucial for homological recombination. pGM5 suicide vector based on pHV33 bireplicon plasmid was constructed to provide replacement of intact F. tularensis chromosome segments by modified segments. Modified chromosome segments contain F. Tularensis DNA fragment without iglC structural gene segment 545 p. b. (in pGMΔiglC plasmid), as well as DNA fragment containing no recA structural gene segment 1060 p.b. (pGMΔrecA plasmid). The constructed 15/23-1ΔrecA mutant, in contrast to the vaccine strain 15, was capable of reproducing in the macrophage-like cells J774A.1 line, whereas the efficiency of the reproduction was 8-10 times less. BALB/c mouse responded to immunization by the 15/23-1ΔrecA strain by smaller weight decrease (-2%) as compared to the strain 15 (-14%). Bacteria of the 15/23-1ΔrecA strain were virtually incapable of germinating from the BALB/c murine spleen 14 days after invasion, whereas bacteria of the strain 15 were found in the murine organs even after 21 days. The F. tularensis 15/23-1ΔrecA strain having smaller reaction ability can be used as a basis for construction of stable live safe tularemia vaccine.


Asunto(s)
Proteínas Bacterianas , Genes Bacterianos/inmunología , Vectores Genéticos , Rec A Recombinasas , Tularemia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/metabolismo , Línea Celular , Francisella tularensis/genética , Francisella tularensis/inmunología , Francisella tularensis/metabolismo , Ratones , Rec A Recombinasas/genética , Rec A Recombinasas/inmunología , Rec A Recombinasas/metabolismo , Tularemia/genética , Tularemia/inmunología , Tularemia/metabolismo , Tularemia/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...