Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunol ; 209(7): 1323-1334, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36002235

RESUMEN

Obesity is considered an important comorbidity for a range of noninfectious and infectious disease states including those that originate in the lung, yet the mechanisms that contribute to this susceptibility are not well defined. In this study, we used the diet-induced obesity (DIO) mouse model and two models of acute pulmonary infection, Francisella tularensis subspecies tularensis strain SchuS4 and SARS-CoV-2, to uncover the contribution of obesity in bacterial and viral disease. Whereas DIO mice were more resistant to infection with SchuS4, DIO animals were more susceptible to SARS-CoV-2 infection compared with regular weight mice. In both models, neither survival nor morbidity correlated with differences in pathogen load, overall cellularity, or influx of inflammatory cells in target organs of DIO and regular weight animals. Increased susceptibility was also not associated with exacerbated production of cytokines and chemokines in either model. Rather, we observed pathogen-specific dysregulation of the host lipidome that was associated with vulnerability to infection. Inhibition of specific pathways required for generation of lipid mediators reversed resistance to both bacterial and viral infection. Taken together, our data demonstrate disparity among obese individuals for control of lethal bacterial and viral infection and suggest that dysregulation of the host lipidome contributes to increased susceptibility to viral infection in the obese host.


Asunto(s)
COVID-19 , Francisella tularensis , Tularemia , Virosis , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Lípidos , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , SARS-CoV-2 , Virosis/metabolismo
2.
J Immunol ; 208(5): 1180-1188, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35149529

RESUMEN

Pulmonary infections elicit a combination of tissue-resident and circulating T cell responses. Understanding the contribution of these anatomically distinct cellular pools in protective immune responses is critical for vaccine development. Francisella tularensis is a highly virulent bacterium capable of causing lethal systemic disease following pulmonary infection for which there is no currently licensed vaccine. Although T cells are required for survival of F. tularensis infection, the relative contribution of tissue-resident and circulating T cells is not completely understood, hampering design of effective, long-lasting vaccines directed against this bacterium. We have previously shown that resident T cells were not sufficient to protect against F. tularensis, suggesting circulating cells may serve a critical role in host defense. To elucidate the role of circulating T cells, we used a model of vaccination and challenge of parabiotic mice. Intranasally infected naive mice conjoined to immune animals had increased numbers of circulating memory T cells and similar splenic bacterial burdens as vaccinated-vaccinated pairs. However, bacterial loads in the lungs of naive parabionts were significantly greater than those observed in vaccinated-vaccinated pairs, but despite early control of F. tularensis replication, all naive-vaccinated pairs succumbed to infection. Together, these data define the specific roles of circulating and resident T cells in defense against infection that is initiated in the pulmonary compartment but ultimately causes disseminated disease. These data also provide evidence for employing vaccination strategies that elicit both pools of T cells for immunity against F. tularensis and may be a common theme for other disseminating bacterial infections.


Asunto(s)
Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Francisella tularensis/inmunología , Células T de Memoria/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Carga Bacteriana/inmunología , Femenino , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/metabolismo , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/patología , Ratones , Ratones Endogámicos C57BL , Tularemia/inmunología , Tularemia/patología , Vacunación
3.
Cell Immunol ; 373: 104485, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35149415

RESUMEN

The metabolite itaconate plays a critical role in modulating inflammatory responses among macrophages infected with intracellular pathogens. However, the ability of itaconate to influence developing T cells responses is poorly understood. To determine if itaconate contributes to the quality of T cell mediated immunity against intracellular infection, we used Francisella tularensis as a model of vaccine induced immunity. Following vaccination with F. tularensis live vaccine strain, itaconate deficient mice (ACOD KO) had a prolonged primary infection but were more resistant to secondary infection with virulent F. tularensis relative to wild type controls. Improved resistance to secondary challenge was associated with both increased numbers and effector function of CD4+ and CD8+ T cells in ACOD KO mice. However, additional data suggest that improved T cell responses was not T cell intrinsic. These data underscore the consequences of metabolic perturbations within antigen presenting cells on the development of vaccine-elicited immune responses.


Asunto(s)
Francisella tularensis , Tularemia , Animales , Vacunas Bacterianas , Linfocitos T CD8-positivos , Ratones , Ratones Endogámicos C57BL , Succinatos , Vacunación , Vacunas Atenuadas
4.
J Immunol ; 207(10): 2399-2404, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34607940

RESUMEN

Immunity to pulmonary infection typically requires elicitation of lung-resident T cells that subsequently confer protection against secondary infection. The presence of tissue-resident T cells in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) convalescent patients is unknown. Using a sublethal mouse model of coronavirus disease 2019, we determined if SARS-CoV-2 infection potentiated Ag-specific pulmonary resident CD4+ and CD8+ T cell responses and if these cells mediated protection against secondary infection. S protein-specific T cells were present in resident and circulating populations. However, M and N protein-specific T cells were detected only in the resident T cell pool. Using an adoptive transfer strategy, we found that T cells from SARS-CoV-2 immune animals did not protect naive mice. These data indicate that resident T cells are elicited by SARS-CoV-2 infection but are not sufficient for protective immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , SARS-CoV-2/fisiología , Traslado Adoptivo , Enzima Convertidora de Angiotensina 2/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína de la Espiga del Coronavirus/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T
5.
J Immunol ; 201(4): 1186-1193, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29980611

RESUMEN

The lung is a complex organ with anatomically distinct pools of T cells that play specific roles in combating infection. Our knowledge regarding the generation and/or maintenance of immunity by parenchymal or circulating T cells has been gathered from either persistent (>60 d) or rapidly cleared (<10 d) infections. However, the roles of these distinct T cell pools in infections that are cleared over the course of several weeks are not understood. Clearance of the highly virulent intracellular bacterium Francisella tularensis subspecies tularensis (Ftt) following pulmonary infection of immune animals is a protracted T cell-dependent process requiring ∼30-40 d and serves as a model for infections that are not acutely controlled. Using this model, we found that intranasal vaccination increased the number of tissue-resident CD4+ effector T cells, and subsequent challenge of immune mice with Ftt led to a significant expansion of polyfunctional parenchymal CD4+ effector T cells compared with the circulating pool. Despite the dominant in vivo response by parenchymal CD4+ T cells after vaccination and challenge, circulating CD4+ T cells were superior at controlling intracellular Ftt replication in vitro. Further examination in vivo revealed temporal requirements for resident and circulating T cells during Ftt infection. These requirements were in direct contrast to other pulmonary infections that are cleared rapidly in immune animals. The data in this study provide important insights into the role of specific T cell populations that will be essential for the design of novel effective vaccines against tularemia and potentially other agents of pulmonary infection.


Asunto(s)
Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Francisella tularensis/fisiología , Pulmón/inmunología , Tularemia/inmunología , Animales , Carga Bacteriana , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Vacunación
6.
J Innate Immun ; 10(4): 291-305, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29969788

RESUMEN

Virulent Francisella tularensis subsp. tularensis (Ftt) is a dynamic, intracellular, bacterial pathogen. Its ability to evade and rapidly suppress host inflammatory responses is considered a key element for its profound virulence. We previously established that Ftt lipids play a role in inhibiting inflammation, but we did not determine the lipid species mediating this process. Here, we show that a unique, abundant, phosphatidylethanolamine (PE), present in Francisella, contributes to driving the suppression of inflammatory responses in human and mouse cells. Acyl chain lengths of this PE, C24: 0 and C10: 0, were key to the suppressive capabilities of Francisella PE. Addition of synthetic PE 24: 0-10: 0 resulted in the accumulation of PE in host cells for up to 24 h of incubation, and recapitulated the inhibition of inflammatory responses observed with native Ftt PE. Importantly, this novel PE significantly inhibited inflammatory responses driven by a medically and globally important flavivirus, dengue fever virus. Thus, targeting these lipids and/or the pathways that they manipulate represents a new strategy to combat immunosuppression engendered by Ftt, but they also show promise as a novel therapeutic intervention for significant viral infections.


Asunto(s)
Antiinflamatorios/metabolismo , Células Dendríticas/inmunología , Francisella tularensis/fisiología , Inflamación/inmunología , Macrófagos/inmunología , Fosfatidiletanolaminas/metabolismo , Tularemia/inmunología , Animales , Proteínas Bacterianas/genética , Células Cultivadas , Células Dendríticas/microbiología , Femenino , Humanos , Evasión Inmune , Inflamación/microbiología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Tularemia/microbiología
7.
Front Microbiol ; 9: 607, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29670588

RESUMEN

Francisella tularensis is a highly infectious bacterial pathogen that causes the potentially fatal disease tularemia. The Live Vaccine Strain (LVS) of F. tularensis subsp. holarctica, while no longer licensed as a vaccine, is used as a model organism for identifying correlates of immunity and bacterial factors that mediate a productive immune response against F. tularensis. Recently, it was reported that two biovars of LVS differed in their virulence and vaccine efficacy. Genetic analysis showed that they differ in ferrous iron homeostasis; lower Fe2+ levels contributed to increased resistance to hydrogen peroxide in the vaccine efficacious LVS biovar. This also correlated with resistance to the bactericidal activity of interferon γ-stimulated murine bone marrow-derived macrophages. We have extended these findings further by showing that a mutant lacking bacterioferritin stimulates poor protection against Schu S4 challenge in a mouse model of tularemia. Together these results suggest that the efficacious biovar of LVS stimulates productive immunity by a mechanism that is dependent on its ability to limit the toxic effects of oxidative stress by maintaining optimally low levels of intracellular Fe2+.

8.
Vaccine ; 35(19): 2575-2581, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28372827

RESUMEN

Francisella tularensis subsp. tularensis strain SchuS4 (Ftt) is a highly virulent intracellular bacterium. Inhalation of 10 or fewer organisms results in an acute and potentially lethal disease called pneumonic tularemia. Ftt infections occur naturally in the U.S. and Ftt was developed as a bioweapon. Thus, there is a need for vaccines that protect against this deadly pathogen. Although a live vaccine strain of Francisella tularensis (LVS) exists, LVS fails to generate long-lived protective immunity against modest challenge doses of Ftt. We recently identified an important role for high avidity CD4+ T cells in short-term protection and hypothesized that expanding this pool of cells would improve overall vaccine efficacy with regard to longevity and challenge dose. In support of our hypothesis, application of a prime/boost vaccination strategy increased the pool of high avidity CD4+ T cells which correlated with improved survival following challenge with either increased doses of virulent Ftt or at late time points after vaccination. In summary, we demonstrate that both epitope selection and vaccination strategies that expand antigen-specific T cells correlate with superior immunity to Ftt as measured by survival.


Asunto(s)
Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Francisella tularensis/inmunología , Pulmón/inmunología , Tularemia/inmunología , Animales , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Esquemas de Inmunización , Ratones Endogámicos C57BL , Análisis de Supervivencia , Tularemia/prevención & control , Estados Unidos
9.
J Immunol ; 197(7): 2738-47, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543611

RESUMEN

T cells are the immunological cornerstone in host defense against infections by intracellular bacterial pathogens, such as virulent Francisella tularensis spp. tularensis (Ftt). The general paucity of novel vaccines for Ftt during the past 60 y can, in part, be attributed to the poor understanding of immune parameters required to survive infection. Thus, we developed a strategy utilizing classical immunological tools to elucidate requirements for effective adaptive immune responses directed against Ftt. Following generation of various Francisella strains expressing well-characterized lymphocytic choriomeningitis virus epitopes, we found that survival correlated with persistence of Ag-specific CD4(+) T cells. Function of these cells was confirmed in their ability to more effectively control Ftt replication in vitro. The importance of understanding the Ag-specific response was underscored by our observation that inclusion of an epitope that elicits high-avidity CD4(+) T cells converted a poorly protective vaccine to one that engenders 100% protection. Taken together, these data suggest that improved efficacy of current tularemia vaccine platforms will require targeting appropriate Ag-specific CD4(+) T cell responses and that elucidation of Francisella epitopes that elicit high-avidity CD4(+) T cell responses, specifically in humans, will be required for successful vaccine development.


Asunto(s)
Vacunas Bacterianas/inmunología , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Francisella tularensis/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos
10.
Clin Vaccine Immunol ; 22(1): 119-28, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25410207

RESUMEN

Francisella tularensis is an intracellular, Gram-negative bacterium that causes the fatal disease tularemia. Currently, there are no licensed vaccines for tularemia and the requirements for protection against infection are poorly defined. To identify correlates of vaccine-induced immunity against tularemia, we compared different strains of the live vaccine strain (LVS) for their relative levels of virulence and ability to protect C57BL/6 mice against challenge with virulent F. tularensis strain SchuS4. Successful vaccination, as defined by survival of C57BL/6 mice, was correlated with significantly greater numbers of effector T cells in the spleen and lung. Further, lung cells and splenocytes from fully protected animals were more effective than lung cells and splenocytes from vaccinated but nonimmune animals in limiting intracellular replication of SchuS4 in vitro. Together, our data provide a unique model to compare efficacious vaccines to nonefficacious vaccines, which will enable comprehensive identification of host and bacterial components required for immunization against tularemia.


Asunto(s)
Vacunas Bacterianas/inmunología , Francisella tularensis/inmunología , Linfocitos T/inmunología , Tularemia/inmunología , Tularemia/prevención & control , Animales , Vacunas Bacterianas/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Francisella tularensis/crecimiento & desarrollo , Ratones Endogámicos C57BL , Bazo/inmunología , Análisis de Supervivencia
11.
Front Microbiol ; 5: 438, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25191316

RESUMEN

Activation of the inflammasome is important for the detection and clearance of cytosolic pathogens. In contrast to avirulent Francisella novicida (Fn), infection with virulent Francisella tularensis ssp tularensis does not trigger activation of the host AIM2 inflammasome. Here we show that differential activation of AIM2 following Francisella infection is due to sensitivity of each isolate to reactive oxygen species (ROS). ROS present at the outset of Fn infection contributes to activation of the AIM2 inflammasome, independent of NLRP3 and NADPH oxidase. Rather, mitochondrial ROS (mROS) is critical for Fn stimulation of the inflammasome. This study represents the first demonstration of the importance of mROS in the activation of the AIM2 inflammasome by bacteria. Our results also demonstrate that bacterial resistance to mROS is a mechanism of virulence for early evasion of detection by the host.

12.
J Innate Immun ; 6(6): 793-805, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24902499

RESUMEN

Highly virulent bacterial pathogens have evolved rapid means to suppress host inflammatory responses by unknown mechanisms. Here, we use virulent Francisella tularensis, the cause of lethal tularemia in humans, as a model to elucidate these mechanisms. We show that following infection of murine macrophages F. tularensis rapidly and selectively destabilizes mRNA containing adenylate-uridylate-rich elements that encode for cytokines and chemokines important in controlling bacterial infection. Degradation of host mRNA encoding interleukin (IL)-1ß, IL-6 and CXCL1 did not require viable bacteria or de novo protein synthesis, but did require escape of intracellular organisms from endocytic vesicles into the host cytosol. The specific targeting of host mRNA encoding inflammatory cytokines and chemokines for decay by a bacterial pathogen has not been previously reported. Thus, our findings represent a novel strategy by which a highly virulent pathogen modulates host inflammatory responses critical to the evasion of innate immunity.


Asunto(s)
Citocinas/inmunología , Francisella tularensis/inmunología , Macrófagos/inmunología , Estabilidad del ARN/inmunología , ARN Mensajero/inmunología , Tularemia/inmunología , Animales , Citocinas/genética , Evasión Inmune/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Estabilidad del ARN/genética , ARN Mensajero/genética , Tularemia/genética , Tularemia/patología
13.
Cell Microbiol ; 16(6): 862-77, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24286610

RESUMEN

Autophagy is a key innate immune response to intracellular parasites that promotes their delivery to degradative lysosomes following detection in the cytosol or within damaged vacuoles. Like Listeria and Shigella, which use specific mechanisms to avoid autophagic detection and capture, the bacterial pathogen Francisella tularensis proliferates within the cytosol of macrophages without demonstrable control by autophagy. To examine how Francisella evades autophagy, we screened a library of F. tularensis subsp. tularensis Schu S4 HimarFT transposon mutants in GFP-LC3-expressing murine macrophages by microscopy for clones localized within autophagic vacuoles after phagosomal escape. Eleven clones showed autophagic capture at 6 h post-infection, whose HimarFT insertions clustered to fourgenetic loci involved in lipopolysaccharidic and capsular O-antigen biosynthesis. Consistent with the HimarFT mutants, in-frame deletion mutants of two representative loci, FTT1236 and FTT1448c (manC), lacking both LPS and capsular O-antigen, underwent phagosomal escape but were cleared from the host cytosol. Unlike wild-type Francisella, the O-antigen deletion mutants were ubiquitinated, and recruited the autophagy adaptor p62/SQSTM1 and LC3 prior to cytosolic clearance. Autophagy-deficient macrophages partially supported replication of both mutants, indicating that O-antigen-lacking Francisella are controlled by autophagy. These data demonstrate the intracellular protective role of this bacterial surface polysaccharide against autophagy.


Asunto(s)
Autofagia , Francisella tularensis/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Viabilidad Microbiana , Antígenos O/inmunología , Antígenos O/metabolismo , Animales , Células Cultivadas , Citosol/microbiología , Elementos Transponibles de ADN , Francisella tularensis/fisiología , Interacciones Huésped-Patógeno , Ratones Endogámicos C57BL , Mutagénesis Insercional
14.
PLoS One ; 8(12): e82096, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324751

RESUMEN

Virulent Francisella tularensis ssp tularensis is an intracellular, Gram negative bacterium that causes acute lethal disease following inhalation of fewer than 15 organisms. Pathogenicity of Francisella infections is tied to its unique ability to evade and suppress inflammatory responses in host cells. It has been proposed that induction of alternative activation of infected macrophages is a mechanism by which attenuated Francisella species modulate host responses. In this report we reveal that neither attenuated F. tularensis Live Vaccine Strain (LVS) nor virulent F. tularensis strain SchuS4 induce alternative activation of macrophages in vitro or in vivo. LVS, but not SchuS4, provoked production of arginase1 independent of alternative activation in vitro and in vivo. However, absence of arginase1 did not significantly impact intracellular replication of LVS or SchuS4. Together our data establish that neither induction of alternative activation nor expression of arginase1 are critical features of disease mediated by attenuated or virulent Francisella species.


Asunto(s)
Arginasa/biosíntesis , Francisella tularensis/fisiología , Activación de Macrófagos , Macrófagos/enzimología , Macrófagos/microbiología , Animales , Citocinas/metabolismo , Inducción Enzimática , Francisella tularensis/crecimiento & desarrollo , Francisella tularensis/patogenicidad , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Tularemia/inmunología , Tularemia/microbiología , Tularemia/prevención & control , Vacunas Atenuadas/inmunología
15.
PLoS Pathog ; 9(8): e1003556, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950720

RESUMEN

The intracellular pathogenic bacterium Brucella generates a replicative vacuole (rBCV) derived from the endoplasmic reticulum via subversion of the host cell secretory pathway. rBCV biogenesis requires the expression of the Type IV secretion system (T4SS) VirB, which is thought to translocate effector proteins that modulate membrane trafficking along the endocytic and secretory pathways. To date, only a few T4SS substrates have been identified, whose molecular functions remain unknown. Here, we used an in silico screen to identify putative T4SS effector candidate proteins using criteria such as limited homology in other bacterial genera, the presence of features similar to known VirB T4SS effectors, GC content and presence of eukaryotic-like motifs. Using ß-lactamase and CyaA adenylate cyclase reporter assays, we identified eleven proteins translocated into host cells by Brucella, five in a VirB T4SS-dependent manner, namely BAB1_0678 (BspA), BAB1_0712 (BspB), BAB1_0847 (BspC), BAB1_1671 (BspE) and BAB1_1948 (BspF). A subset of the translocated proteins targeted secretory pathway compartments when ectopically expressed in HeLa cells, and the VirB effectors BspA, BspB and BspF inhibited protein secretion. Brucella infection also impaired host protein secretion in a process requiring BspA, BspB and BspF. Single or combined deletions of bspA, bspB and bspF affected Brucella ability to replicate in macrophages and persist in the liver of infected mice. Taken together, these findings demonstrate that Brucella modulates secretory trafficking via multiple T4SS effector proteins that likely act coordinately to promote Brucella pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/fisiología , Brucella abortus/metabolismo , Brucelosis/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/metabolismo , Animales , Proteínas Bacterianas/genética , Brucella abortus/genética , Brucelosis/patología , Femenino , Células HeLa , Humanos , Hígado/microbiología , Hígado/patología , Macrófagos/patología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Transporte de Proteínas/fisiología , Vacuolas/genética , Vacuolas/metabolismo , Vacuolas/microbiología
16.
PLoS One ; 8(6): e67965, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840797

RESUMEN

Francisella tularensis is a highly infectious bacterium whose virulence relies on its ability to rapidly reach the macrophage cytosol and extensively replicate in this compartment. We previously identified a novel Francisella virulence factor, DipA (FTT0369c), which is required for intramacrophage proliferation and survival, and virulence in mice. DipA is a 353 amino acid protein with a Sec-dependent signal peptide, four Sel1-like repeats (SLR), and a C-terminal coiled-coil (CC) domain. Here, we determined through biochemical and localization studies that DipA is a membrane-associated protein exposed on the surface of the prototypical F. tularensis subsp. tularensis strain SchuS4 during macrophage infection. Deletion and substitution mutagenesis showed that the CC domain, but not the SLR motifs, of DipA is required for surface exposure on SchuS4. Complementation of the dipA mutant with either DipA CC or SLR domain mutants did not restore intracellular growth of Francisella, indicating that proper localization and the SLR domains are required for DipA function. Co-immunoprecipitation studies revealed interactions with the Francisella outer membrane protein FopA, suggesting that DipA is part of a membrane-associated complex. Altogether, our findings indicate that DipA is positioned at the host-pathogen interface to influence the intracellular fate of this pathogen.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Francisella tularensis/crecimiento & desarrollo , Macrófagos/microbiología , Tularemia/microbiología , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Células Cultivadas , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Relación Estructura-Actividad , Tularemia/metabolismo , Tularemia/patología , Factores de Virulencia/genética
17.
J Immunol ; 190(6): 2756-66, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23378429

RESUMEN

B1a cells are an important source of natural Abs, Abs directed against T-independent Ags, and are a primary source of IL-10. Bruton's tyrosine kinase (btk) is a cytoplasmic kinase that is essential for mediating signals from the BCR and is critical for development of B1a cells. Consequentially, animals lacking btk have few B1a cells, minimal Ab responses, and can preferentially generate Th1-type immune responses following infection. B1a cells have been shown to aid in protection against infection with attenuated Francisella tularensis, but their role in infection mediated by fully virulent F. tularensis is not known. Therefore, we used mice with defective btk (CBA/CaHN-Btk(XID)/J [XID mice]) to determine the contribution of B1a cells in defense against the virulent F. tularensis ssp. tularensis strain SchuS4. Surprisingly, XID mice displayed increased resistance to pulmonary infection with F. tularensis. Specifically, XID mice had enhanced clearance of bacteria from the lung and spleen and significantly greater survival of infection compared with wild-type controls. We revealed that resistance to infection in XID mice was associated with decreased numbers of IL-10-producing B1a cells and concomitant increased numbers of IL-12-producing macrophages and IFN-γ-producing NK/NKT cells. Adoptive transfer of wild-type B1a cells into XID mice reversed the control of bacterial replication. Similarly, depletion of NK/NKT cells also increased bacterial burdens in XID mice. Together, our data suggest B cell-NK/NKT cell cross-talk is a critical pivot controlling survival of infection with virulent F. tularensis.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/microbiología , Francisella tularensis/inmunología , Predisposición Genética a la Enfermedad , Células Asesinas Naturales/inmunología , Células T Asesinas Naturales/inmunología , Animales , Anticuerpos Antibacterianos/biosíntesis , Subgrupos de Linfocitos B/trasplante , Comunicación Celular/genética , Comunicación Celular/inmunología , Células Cultivadas , Exudados y Transudados/microbiología , Francisella tularensis/genética , Francisella tularensis/patogenicidad , Células Asesinas Naturales/microbiología , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos CBA , Ratones Mutantes , Células T Asesinas Naturales/microbiología , Células T Asesinas Naturales/patología , Peritoneo/inmunología , Peritoneo/microbiología , Peritoneo/patología , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/fisiología , Análisis de Supervivencia , Células TH1/inmunología , Células TH1/metabolismo , Células TH1/microbiología , Tularemia/inmunología , Tularemia/mortalidad , Tularemia/patología , Virulencia/genética , Virulencia/inmunología
18.
Autophagy ; 8(9): 1342-56, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22863802

RESUMEN

Cytosolic bacterial pathogens must evade intracellular innate immune recognition and clearance systems such as autophagy to ensure their survival and proliferation. The intracellular cycle of the bacterium Francisella tularensis is characterized by rapid phagosomal escape followed by extensive proliferation in the macrophage cytoplasm. Cytosolic replication, but not phagosomal escape, requires the locus FTT0369c, which encodes the dipA gene (deficient in intracellular replication A). Here, we show that a replication-deficient, ∆dipA mutant of the prototypical SchuS4 strain is eventually captured from the cytosol of murine and human macrophages into double-membrane vacuoles displaying the late endosomal marker, LAMP1, and the autophagy-associated protein, LC3, coinciding with a reduction in viable intracellular bacteria. Capture of SchuS4ΔdipA was not dipA-specific as other replication-deficient bacteria, such as chloramphenicol-treated SchuS4 and a purine auxotroph mutant SchuS4ΔpurMCD, were similarly targeted to autophagic vacuoles. Vacuoles containing replication-deficient bacteria were labeled with ubiquitin and the autophagy receptors SQSTM1/p62 and NBR1, and their formation was decreased in macrophages from either ATG5-, LC3B- or SQSTM1-deficient mice, indicating recognition by the ubiquitin-SQSTM1-LC3 pathway. While a fraction of both the wild-type and the replication-impaired strains were ubiquitinated and recruited SQSTM1, only the replication-defective strains progressed to autophagic capture, suggesting that wild-type Francisella interferes with the autophagic cascade. Survival of replication-deficient strains was not restored in autophagy-deficient macrophages, as these bacteria died in the cytosol prior to autophagic capture. Collectively, our results demonstrate that replication-impaired strains of Francisella are cleared by autophagy, while replication-competent bacteria seem to interfere with autophagic recognition, therefore ensuring survival and proliferation.


Asunto(s)
Autofagia , Citosol/microbiología , Replicación del ADN , Francisella tularensis/fisiología , Mutación/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Endocitosis , Endosomas/microbiología , Endosomas/ultraestructura , Francisella tularensis/ultraestructura , Proteínas de Choque Térmico/metabolismo , Humanos , Espacio Intracelular/microbiología , Macrófagos/microbiología , Macrófagos/patología , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana , Fagosomas/microbiología , Fagosomas/ultraestructura , Proteína Sequestosoma-1 , Ubiquitina/metabolismo , Vacuolas/microbiología , Vacuolas/ultraestructura
19.
PLoS One ; 7(5): e37752, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22662210

RESUMEN

Tularemia, caused by the gram-negative bacterium Francisella tularensis, is a severe, sometimes fatal disease. Interest in tularemia has increased over the last decade due to its history as a biological weapon. In particular, development of novel vaccines directed at protecting against pneumonic tularemia has been an important goal. Previous work has demonstrated that, when delivered at very high inoculums, administration of live, highly attenuated strains of virulent F. tularensis can protect against tularemia. However, lower vaccinating inoculums did not offer similar immunity. One concern of using live vaccines is that the host may develop mild tularemia in response to infection and use of high inoculums may contribute to this issue. Thus, generation of a live vaccine that can efficiently protect against tularemia when delivered in low numbers, e.g. <100 organisms, may address this concern. Herein we describe the ability of three defined, attenuated mutants of F. tularensis SchuS4, deleted for FTT0369c, FTT1676, or FTT0369c and FTT1676, respectively, to engender protective immunity against tularemia when delivered at concentrations of approximately 50 or fewer bacteria. Attenuated strains for use as vaccines were selected by their inability to efficiently replicate in macrophages in vitro and impaired replication and dissemination in vivo. Although all strains were defective for replication in vitro within macrophages, protective efficacy of each attenuated mutant was correlated with their ability to modestly replicate and disseminate in the host. Finally, we demonstrate the parenteral vaccination with these strains offered superior protection against pneumonic tularemia than intranasal vaccination. Together our data provides proof of principle that low dose attenuated vaccines may be a viable goal in development of novel vaccines directed against tularemia.


Asunto(s)
Vacunas Bacterianas/administración & dosificación , Francisella tularensis/inmunología , Tularemia/prevención & control , Animales , Vacunas Bacterianas/inmunología , Femenino , Francisella tularensis/genética , Francisella tularensis/patogenicidad , Dosificación Letal Mediana , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Tularemia/mortalidad , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Virulencia/genética
20.
Cell Host Microbe ; 11(1): 33-45, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22264511

RESUMEN

Autophagy is a cellular degradation process that can capture and eliminate intracellular microbes by delivering them to lysosomes for destruction. However, pathogens have evolved mechanisms to subvert this process. The intracellular bacterium Brucella abortus ensures its survival by forming the Brucella-containing vacuole (BCV), which traffics from the endocytic compartment to the endoplasmic reticulum (ER), where the bacterium proliferates. We show that Brucella replication in the ER is followed by BCV conversion into a compartment with autophagic features (aBCV). While Brucella trafficking to the ER was unaffected in autophagy-deficient cells, aBCV formation required the autophagy-initiation proteins ULK1, Beclin 1, and ATG14L and PI3-kinase activity. However, aBCV formation was independent of the autophagy-elongation proteins ATG5, ATG16L1, ATG4B, ATG7, and LC3B. Furthermore, aBCVs were required to complete the intracellular Brucella lifecycle and for cell-to-cell spreading, demonstrating that Brucella selectively co-opts autophagy-initiation complexes to subvert host clearance and promote infection.


Asunto(s)
Autofagia/inmunología , Brucella abortus/inmunología , Brucella abortus/patogenicidad , Evasión Inmune , Vacuolas/microbiología , Animales , Brucella abortus/crecimiento & desarrollo , Línea Celular , Endocitosis , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/microbiología , Humanos , Ratones , Ratones Noqueados , Transporte de Proteínas , Vacuolas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...