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










Publication year range
1.
J Immunol ; 209(7): 1323-1334, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36002235

ABSTRACT

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.


Subject(s)
COVID-19 , Francisella tularensis , Tularemia , Virus Diseases , Animals , Chemokines/metabolism , Cytokines/metabolism , Lipids , Lung/microbiology , Mice , Mice, Inbred C57BL , Obesity/metabolism , SARS-CoV-2 , Virus Diseases/metabolism
2.
J Immunol ; 208(5): 1180-1188, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35149529

ABSTRACT

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.


Subject(s)
Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Francisella tularensis/immunology , Memory T Cells/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Load/immunology , Female , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred C57BL , Tularemia/immunology , Tularemia/pathology , Vaccination
3.
Cell Immunol ; 373: 104485, 2022 03.
Article in English | MEDLINE | ID: mdl-35149415

ABSTRACT

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.


Subject(s)
Francisella tularensis , Tularemia , Animals , Bacterial Vaccines , CD8-Positive T-Lymphocytes , Mice , Mice, Inbred C57BL , Succinates , Vaccination , Vaccines, Attenuated
4.
J Immunol ; 207(10): 2399-2404, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34607940

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lung/immunology , SARS-CoV-2/physiology , Adoptive Transfer , Angiotensin-Converting Enzyme 2/genetics , Animals , Cells, Cultured , Disease Models, Animal , Disease Resistance , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spike Glycoprotein, Coronavirus/immunology , T-Cell Antigen Receptor Specificity
5.
iScience ; 24(9): 103025, 2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34522865

ABSTRACT

Resolution of infection results in development of trained innate immunity which is typically beneficial for defense against unrelated secondary infection. Epigenetic changes including modification of histones via binding of various polar metabolites underlie the establishment of trained innate immunity. Therefore, host metabolism and this response are intimately linked. However, little is known regarding the influence of lipids on the development and function of trained immunity. Utilizing two models of pulmonary bacterial infection combined with multi-omic approaches, we identified persistent, pathogen-specific changes to the lung lipidome that correlated with differences in the trained immune response against a third unrelated pathogen. Further, we establish the specific cellular populations in the lung that contribute to this altered lipidome. Together these results expand our understanding of the pulmonary trained innate immune response and the contributions of host lipids in informing that response.

6.
Infect Immun ; 88(2)2020 01 22.
Article in English | MEDLINE | ID: mdl-31740527

ABSTRACT

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.


Subject(s)
Francisella tularensis/drug effects , Interferon-gamma/pharmacology , Mitochondria/drug effects , Animals , Cell Membrane/metabolism , Cell Membrane/microbiology , Cytosol/metabolism , Cytosol/microbiology , Humans , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/microbiology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Succinates/pharmacology , Tularemia/drug therapy , Tularemia/metabolism , Tularemia/microbiology
7.
J Immunol ; 201(4): 1186-1193, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29980611

ABSTRACT

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.


Subject(s)
Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Francisella tularensis/physiology , Lung/immunology , Tularemia/immunology , Animals , Bacterial Load , Cell Proliferation , Disease Models, Animal , Female , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Vaccination
8.
J Innate Immun ; 10(4): 291-305, 2018.
Article in English | MEDLINE | ID: mdl-29969788

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/metabolism , Dendritic Cells/immunology , Francisella tularensis/physiology , Inflammation/immunology , Macrophages/immunology , Phosphatidylethanolamines/metabolism , Tularemia/immunology , Animals , Bacterial Proteins/genetics , Cells, Cultured , Dendritic Cells/microbiology , Female , Humans , Immune Evasion , Inflammation/microbiology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutation/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Tularemia/microbiology
9.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29760217

ABSTRACT

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.


Subject(s)
Bacterial Capsules/metabolism , Cell Death , Energy Metabolism , Francisella tularensis/pathogenicity , Host-Pathogen Interactions , Mitochondria/metabolism , Mitochondria/microbiology , Animals , Bacterial Load , Cells, Cultured , Cytoplasm/microbiology , Female , Francisella tularensis/growth & development , Immune Evasion , Inflammation/pathology , Intravital Microscopy , Macrophages/microbiology , Macrophages/physiology , Mice, Inbred C57BL
10.
Front Microbiol ; 9: 607, 2018.
Article in English | MEDLINE | ID: mdl-29670588

ABSTRACT

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+.

11.
Vaccine ; 35(19): 2575-2581, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28372827

ABSTRACT

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.


Subject(s)
Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Francisella tularensis/immunology , Lung/immunology , Tularemia/immunology , Animals , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Immunization Schedule , Mice, Inbred C57BL , Survival Analysis , Tularemia/prevention & control , United States
12.
J Immunol ; 197(7): 2738-47, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27543611

ABSTRACT

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.


Subject(s)
Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Francisella tularensis/immunology , Animals , Female , Mice , Mice, Inbred Strains
13.
Clin Vaccine Immunol ; 22(1): 119-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25410207

ABSTRACT

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.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , T-Lymphocytes/immunology , Tularemia/immunology , Tularemia/prevention & control , Animals , Bacterial Vaccines/administration & dosage , Disease Models, Animal , Female , Francisella tularensis/growth & development , Mice, Inbred C57BL , Spleen/immunology , Survival Analysis
14.
Front Microbiol ; 5: 438, 2014.
Article in English | MEDLINE | ID: mdl-25191316

ABSTRACT

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.

15.
J Innate Immun ; 6(6): 793-805, 2014.
Article in English | MEDLINE | ID: mdl-24902499

ABSTRACT

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.


Subject(s)
Cytokines/immunology , Francisella tularensis/immunology , Macrophages/immunology , RNA Stability/immunology , RNA, Messenger/immunology , Tularemia/immunology , Animals , Cytokines/genetics , Immune Evasion/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , RNA Stability/genetics , RNA, Messenger/genetics , Tularemia/genetics , Tularemia/pathology
16.
Cell Microbiol ; 16(6): 862-77, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24286610

ABSTRACT

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.


Subject(s)
Autophagy , Francisella tularensis/immunology , Macrophages/immunology , Macrophages/microbiology , Microbial Viability , O Antigens/immunology , O Antigens/metabolism , Animals , Cells, Cultured , Cytosol/microbiology , DNA Transposable Elements , Francisella tularensis/physiology , Host-Pathogen Interactions , Mice, Inbred C57BL , Mutagenesis, Insertional
17.
PLoS One ; 8(12): e82096, 2013.
Article in English | MEDLINE | ID: mdl-24324751

ABSTRACT

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.


Subject(s)
Arginase/biosynthesis , Francisella tularensis/physiology , Macrophage Activation , Macrophages/enzymology , Macrophages/microbiology , Animals , Cytokines/metabolism , Enzyme Induction , Francisella tularensis/growth & development , Francisella tularensis/pathogenicity , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Tularemia/immunology , Tularemia/microbiology , Tularemia/prevention & control , Vaccines, Attenuated/immunology
18.
PLoS Pathog ; 9(8): e1003556, 2013.
Article in English | MEDLINE | ID: mdl-23950720

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/physiology , Brucella abortus/metabolism , Brucellosis/metabolism , Liver/metabolism , Macrophages/metabolism , Macrophages/microbiology , Membrane Proteins/metabolism , Animals , Bacterial Proteins/genetics , Brucella abortus/genetics , Brucellosis/pathology , Female , HeLa Cells , Humans , Liver/microbiology , Liver/pathology , Macrophages/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Protein Transport/physiology , Vacuoles/genetics , Vacuoles/metabolism , Vacuoles/microbiology
19.
PLoS One ; 8(6): e67965, 2013.
Article in English | MEDLINE | ID: mdl-23840797

ABSTRACT

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.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Francisella tularensis/growth & development , Macrophages/microbiology , Tularemia/microbiology , Virulence Factors/chemistry , Virulence Factors/metabolism , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/genetics , Cells, Cultured , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Structure-Activity Relationship , Tularemia/metabolism , Tularemia/pathology , Virulence Factors/genetics
20.
J Immunol ; 190(6): 2756-66, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23378429

ABSTRACT

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.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Francisella tularensis/immunology , Genetic Predisposition to Disease , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , Animals , Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/transplantation , Cell Communication/genetics , Cell Communication/immunology , Cells, Cultured , Exudates and Transudates/microbiology , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Natural Killer T-Cells/microbiology , Natural Killer T-Cells/pathology , Peritoneum/immunology , Peritoneum/microbiology , Peritoneum/pathology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Survival Analysis , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/microbiology , Tularemia/immunology , Tularemia/mortality , Tularemia/pathology , Virulence/genetics , Virulence/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...