Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 16(3): e0249142, 2021.
Article in English | MEDLINE | ID: mdl-33760886

ABSTRACT

CCR2 is the major chemokine receptor that regulates appropriate trafficking of inflammatory monocytes, but the role of this chemokine receptor and its ligands during primary and secondary infection with intracellular infections remains incompletely understood. Here we used murine infection with the Live Vaccine Strain (LVS) of Francisella tularensis to evaluate the role of CCR2 during primary and secondary parenteral responses to this prototype intracellular bacterium. We find that mice deficient in CCR2 are highly compromised in their ability to survive intradermal infection with LVS, indicating the importance of this receptor during primary parenteral responses. Interestingly, this defect could not be readily attributed to the activities of the known murine CCR2 ligands MCP-1/CCL2, MCP-3/CCL7, or MCP-5/CCL12. Nonetheless, CCR2 knockout mice vaccinated by infection with low doses of LVS generated optimal T cell responses that controlled the intramacrophage replication of Francisella, and LVS-immune CCR2 knockout mice survived maximal lethal Francisella challenge. Thus, fully protective adaptive immune memory responses to this intracellular bacterium can be readily generated in the absence of CCR2.


Subject(s)
Francisella tularensis/physiology , Receptors, CCR2/genetics , Tularemia/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL7/deficiency , Chemokine CCL7/genetics , Chemokine CCL7/immunology , Disease Models, Animal , Disease Susceptibility , Francisella tularensis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Receptors, CCR2/deficiency , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tularemia/mortality , Tularemia/pathology , Tularemia/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
2.
J Exp Med ; 213(12): 2793-2809, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27799620

ABSTRACT

Mucosa-associated invariant T (MAIT) cells are a unique innate T cell subset that is necessary for rapid recruitment of activated CD4+ T cells to the lungs after pulmonary F. tularensis LVS infection. Here, we investigated the mechanisms behind this effect. We provide evidence to show that MAIT cells promote early differentiation of CCR2-dependent monocytes into monocyte-derived DCs (Mo-DCs) in the lungs after F. tularensis LVS pulmonary infection. Adoptive transfer of Mo-DCs to MAIT cell-deficient mice (MR1-/- mice) rescued their defect in the recruitment of activated CD4+ T cells to the lungs. We further demonstrate that MAIT cell-dependent GM-CSF production stimulated monocyte differentiation in vitro, and that in vivo production of GM-CSF was delayed in the lungs of MR1-/- mice. Finally, GM-CSF-deficient mice exhibited a defect in monocyte differentiation into Mo-DCs that was phenotypically similar to MR1-/- mice. Overall, our data demonstrate that MAIT cells promote early pulmonary GM-CSF production, which drives the differentiation of inflammatory monocytes into Mo-DCs. Further, this delayed differentiation of Mo-DCs in MR1-/- mice was responsible for the delayed recruitment of activated CD4+ T cells to the lungs. These findings establish a novel mechanism by which MAIT cells function to promote both innate and adaptive immune responses.


Subject(s)
Cell Differentiation , Dendritic Cells/immunology , Intracellular Space/microbiology , Lung Diseases/immunology , Lung Diseases/microbiology , Lung/pathology , Monocytes/pathology , Mucosal-Associated Invariant T Cells/immunology , Adoptive Transfer , Animals , Bone Marrow/pathology , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Cytokines/pharmacology , Female , Francisella tularensis/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Histocompatibility Antigens Class I/immunology , Lung/microbiology , Lung Diseases/pathology , Male , Mice, Inbred C57BL , Mucous Membrane/microbiology , Mucous Membrane/pathology , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/pathology , Receptors, CCR2/metabolism , Recombinant Proteins/pharmacology , Tularemia/immunology , Tularemia/microbiology , Tularemia/pathology , Vaccines/immunology
3.
PLoS One ; 10(9): e0138565, 2015.
Article in English | MEDLINE | ID: mdl-26379269

ABSTRACT

The virulence of F. tularensis is often associated with its ability to grow in macrophages, although recent studies show that Francisella proliferates in multiple host cell types, including pulmonary epithelial cells. Thus far little is known about the requirements for killing of F. tularensis in the non-macrophage host cell types that support replication of this organism. Here we sought to address this question through the use of a murine lung epithelial cell line (TC-1 cells). Our data show that combinations of the cytokines IFN-γ, TNF, and IL-17A activated murine pulmonary epithelial cells to inhibit the intracellular growth of the F. tularensis Live Vaccine Strain (LVS) and the highly virulent F. tularensis Schu S4 strain. Although paired combinations of IFN-γ, TNF, and IL-17A all significantly controlled LVS growth, simultaneous treatment with all three cytokines had the greatest effect on LVS growth inhibition. In contrast, Schu S4 was more resistant to cytokine-induced growth effects, exhibiting significant growth inhibition only in response to all three cytokines. Since one of the main antimicrobial mechanisms of activated macrophages is the release of reactive nitrogen intermediates (RNI) via the activity of iNOS, we investigated the role of RNI and iNOS in Francisella growth control by pulmonary epithelial cells. NOS2 gene expression was significantly up-regulated in infected, cytokine-treated pulmonary epithelial cells in a manner that correlated with LVS and Schu S4 growth control. Treatment of LVS-infected cells with an iNOS inhibitor significantly reversed LVS killing in cytokine-treated cultures. Further, we found that mouse pulmonary epithelial cells produced iNOS during in vivo respiratory LVS infection. Overall, these data demonstrate that lung epithelial cells produce iNOS both in vitro and in vivo, and can inhibit Francisella intracellular growth via reactive nitrogen intermediates.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/microbiology , Francisella tularensis/growth & development , Lung/metabolism , Lung/microbiology , Animals , Cell Line , Interferon-gamma/metabolism , Interleukin-17/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Reactive Nitrogen Species/metabolism , Tumor Necrosis Factors/metabolism , Virulence/physiology
4.
J Immunol ; 184(10): 5791-801, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20393138

ABSTRACT

For several intracellular infections, pulmonary vaccination provides measurably better protection against pulmonary challenge. The unique factors that contribute to pulmonary immune responses are not well characterized. In this study, we show that CD4(-)CD8(-) double negative (DN) T cells are a major responding T cell subset in the lungs of mice during pulmonary Francisella tularensis live vaccine strain (LVS) infection. DN T cells were a minor (<2%) subset in spleens and lungs of mice during sublethal intradermal infection with LVS. In contrast, they were a major responding T cell subset in lungs during pulmonary LVS infection, producing large quantities of IFN-gamma and IL-17A. The numbers of IL-17A(+) DN T cells in the lungs exceeded that of CD4(+) and CD8(+) T cells on day 7 postinfection; by day 14 postinfection, all three IL-17A-producing T cell subsets were present in equivalent numbers. CD4(+), CD8(+), and DN T cell production of IL-17A was not observed in the spleens of pulmonary-infected mice or the lungs and spleens of intradermally infected mice. Correspondingly, IL-17A knockout mice were more susceptible to respiratory than intradermal LVS infection, with delayed clearance 1-3 wk postinfection. Finally, in vitro treatment of LVS-infected macrophages and alveolar type II epithelial cells with IFN-gamma and IL-17A affected significantly greater LVS growth control than treatment with either cytokine alone. The data presented in this study demonstrate that DN cells contribute to production of IL-17A and IFN-gamma in the lungs during inhalational Francisella infection and that these cytokines additively activate host cells to control LVS intracellular growth.


Subject(s)
Bacterial Vaccines/immunology , CD4 Antigens , CD8 Antigens , Francisella tularensis/immunology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Pneumonia, Bacterial/immunology , T-Lymphocyte Subsets/immunology , Animals , Bacterial Vaccines/administration & dosage , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Francisella tularensis/growth & development , Interleukin-17/deficiency , Interleukin-17/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/microbiology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , T-Lymphocyte Subsets/microbiology , Tularemia/immunology , Tularemia/microbiology
5.
Microbes Infect ; 12(1): 28-36, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19781659

ABSTRACT

Although survival of primary infection with the live vaccine strain (LVS) of Francisella tularensis depends on interferon gamma (IFN-gamma), the relative importance of IFN-gamma to secondary protective immunity in vivo has not been clearly established. Here we examine the role of IFN-gamma in T cell priming and expression of vaccine-induced protection against lethal intraperitoneal challenge of mice. Large amounts of IFN-gamma were detected between days 3 and 7 in the sera of LVS-immunized mice, while relatively small amounts were found transiently after secondary LVS challenge. Consistent with the production of this cytokine, mice lacking IFN-gamma (gamma interferon knockout, GKO, mice) could not be successfully vaccinated with LVS or an attenuated mglA mutant of F. novicida to withstand secondary Francisella LVS challenge. Further, splenocytes from such primed mice did not adoptively transfer protection to naive GKO recipient mice in vivo, nor control the intramacrophage growth of LVS in vitro. Finally, LVS-immune WT mice depleted of IFN-gamma prior to intraperitoneal challenge survived only the lowest doses of challenge. Thus successful priming of protective LVS-immune T cells, as well as complete expression of protection against Francisella during secondary challenge, depends heavily on IFN-gamma.


Subject(s)
Francisella tularensis/immunology , Interferon-gamma/immunology , Tularemia/immunology , Tularemia/mortality , Adoptive Transfer , Animals , Bacterial Vaccines/immunology , Interferon-gamma/blood , Interferon-gamma/deficiency , Lethal Dose 50 , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Survival Analysis , Vaccination/methods
6.
Infect Immun ; 77(5): 2010-21, 2009 May.
Article in English | MEDLINE | ID: mdl-19237526

ABSTRACT

Parenteral and respiratory vaccinations with the intracellular bacterium Francisella tularensis have been studied using the live vaccine strain (LVS) in a mouse model, and spleen cells from immune mice are often used for immunological studies. However, mechanisms of host immunological responses may be different in nonlymphoid organs that are important sites of infection, such as lung and liver. Using parenteral (intradermal) or respiratory (cloud aerosol) vaccination, here we examine the functions of resulting LVS-immune liver or lung cells, respectively. Surprisingly, LVS was considerably more virulent when administered by cloud aerosol than by intranasal instillation, suggesting method-dependent differences in initial localization and/or dissemination patterns. Only low doses were sublethal, and resolution of sublethal cloud aerosol infection was dependent on gamma interferon (IFN-gamma), tumor necrosis factor alpha, and inducible nitric oxide synthase. Nonetheless, survival of cloud aerosol or parenteral infection resulted in the development of a protective immune response against lethal LVS intraperitoneal or aerosol challenge, reflecting development of systemic secondary immunity in both cases. Such immunity was further detected by directly examining the functions of LVS-immune lung or liver lymphocytes in vitro. Lung lymphocytes primed by respiratory infection, as well as liver lymphocytes primed by parenteral infection, clearly controlled in vitro intracellular bacterial growth primarily via mechanisms that were not dependent on IFN-gamma activity. Thus, our results indicate functional similarities between immune T cells residing in spleens, livers, and lungs of LVS-immune mice.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Liver/immunology , Lung/immunology , T-Lymphocytes/immunology , Tularemia/prevention & control , Animals , Colony Count, Microbial , Female , Francisella tularensis/growth & development , Interferon-gamma/deficiency , Interferon-gamma/immunology , Liver/microbiology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/microbiology , Survival Analysis
7.
Microbes Infect ; 8(3): 779-90, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16513388

ABSTRACT

The means by which Francisella tularensis, the causative agent of tularemia, are recognized by mammalian immune systems are poorly understood. Here we wished to explore the contribution of the MyD88/Toll-like receptor signaling pathway in initiating murine responses to F. tularensis Live Vaccine Strain (LVS). MyD88 knockout (KO) mice, but not TLR2-, TLR4- or TLR9-deficient mice, rapidly succumbed following in vivo bacterial infection via the intradermal route even with a very low dose of LVS (5 x 10(1)) that was 100,000-fold less than the LD(50) of normal wild-type (WT) mice. By day 5 after LVS infection, bacterial organ burdens were 5-6 logs higher in MyD88 knockout mice; further, unlike infected WT mice, levels of interferon-gamma in the sera of LVS-infected MyD88 KO were undetectable. An in vitro culture system was used to assess the ability of bone marrow macrophages derived from either KO or WT mice to support bacterial growth, or to control intracellular bacterial replication when co-cultured with immune lymphocytes. In this assay, bacterial replication was similar in macrophages derived from either WT or any of the TLR KO mice. Bacterial growth was controlled in co-cultures containing macrophages from MyD88 KO mice or TLR KO mice as well as in co-cultures containing immune WT splenic lymphocytes and WT macrophages. Further, MyD88-deficient LVS-immune splenocytes controlled intracellular growth comparably to those from normal mice. Thus MyD88 is essential for innate host resistance to LVS infection, but is not required for macrophage control of intracellular bacterial growth.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Francisella tularensis/metabolism , Macrophages/microbiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Interleukin-1/metabolism , Interleukin-18/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88 , Signal Transduction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
8.
J Bacteriol ; 186(19): 6430-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15375123

ABSTRACT

Francisella tularensis is a gram-negative, facultative intracellular pathogen that causes the highly infectious zoonotic disease tularemia. We have discovered a ca. 30-kb pathogenicity island of F. tularensis (FPI) that includes four large open reading frames (ORFs) of 2.5 to 3.9 kb and 13 ORFs of 1.5 kb or smaller. Previously, two small genes located near the center of the FPI were shown to be needed for intramacrophage growth. In this work we show that two of the large ORFs, located toward the ends of the FPI, are needed for virulence. Although most genes in the FPI encode proteins with amino acid sequences that are highly conserved between high- and low-virulence strains, one of the FPI genes is present in highly virulent type A F. tularensis, absent in moderately virulent type B F. tularensis, and altered in F. tularensis subsp. novicida, which is highly virulent for mice but avirulent for humans. The G+C content of a 17.7-kb stretch of the FPI is 26.6%, which is 6.6% below the average G+C content of the F. tularensis genome. This extremely low G+C content suggests that the DNA was imported from a microbe with a very low G+C-containing chromosome.


Subject(s)
Francisella tularensis/genetics , Genomic Islands/physiology , Macrophages/microbiology , Animals , Base Composition , Base Sequence , Francisella tularensis/growth & development , Francisella tularensis/pathogenicity , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Polymerase Chain Reaction , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...