Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Infect Immun ; 91(4): e0006423, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36916918

ABSTRACT

The mesenteric lymph nodes (MLN) function as a barrier to systemic spread for both commensal and pathogenic bacteria in the gut. Listeria monocytogenes, a facultative intracellular foodborne pathogen, readily overcomes this barrier and spreads into the bloodstream, causing life-threatening systemic infections. We show here that intracellular replication protected L. monocytogenes from clearance by monocytes and neutrophils and promoted colonization of the small intestine-draining MLN (sMLN) but was not required for dissemination to the colon-draining MLN (cMLN). Intestinal tissue had enough free lipoate to support LplA2-dependent extracellular growth of L. monocytogenes, but exogenous lipoate in the MLN was severely limited, and so the bacteria could replicate only inside cells, where they used LplA1 to scavenge lipoate from host peptides. When foodborne infection was manipulated to allow ΔlplA1 L. monocytogenes to colonize the MLN to the same extent as wild-type bacteria, the mutant was still never recovered in the spleen or liver of any animal. We found that intracellular replication in the MLN promoted actin-based motility and cell-to-cell spread of L. monocytogenes and that rapid efficient exit from the MLN was actA dependent. We conclude that intracellular replication of L. monocytogenes in intestinal tissues is not essential and serves primarily to amplify bacterial burdens above a critical threshold needed to efficiently colonize the cMLN. In contrast, intracellular replication in the MLN is absolutely required for further systemic spread and serves primarily to promote ActA-mediated cell-to-cell spread.


Subject(s)
Listeria monocytogenes , Listeriosis , Animals , Listeriosis/microbiology , Bacterial Proteins/genetics , Liver/pathology , Lymph Nodes/microbiology
2.
Infect Immun ; 91(9): e0025123, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37594272

ABSTRACT

Apolipoprotein E (ApoE) is a lipid transport protein that is hypothesized to suppress proinflammatory cytokine production, particularly after stimulation with Toll-like receptor (TLR) ligands such as lipopolysaccharide (LPS). Studies using transgenic ApoE human replacement mice (APOE) expressing one of three different allelic variants suggest that there is a hierarchy in terms of responsiveness to proinflammatory stimuli such as APOE4/E4 > APOE3/E3 > APOE2/E2. In this study, we test the hypothesis that APOE genotype can also predict susceptibility to infection with the facultative intracellular gram-positive bacterium Listeria monocytogenes. We found that bone-marrow-derived macrophages isolated from aged APOE4/E4 mice expressed elevated levels of nitric oxide synthase 2 and were highly resistant to in vitro infection with L. monocytogenes compared to APOE3/E3 and APOE2/E2 mice. However, we did not find statistically significant differences in cytokine or chemokine output from either macrophages or whole splenocytes isolated from APOE2/E2, APOE3/E3, or APOE4/E4 mice following L. monocytogenes infection. In vivo, overall susceptibility to foodborne listeriosis also did not differ by APOE genotype in either young (2 mo old) or aged (15 mo old) C57BL/6 mice. However, we observed a sex-dependent susceptibility to infection in aged APOE2/E2 male mice and a sex-dependent resistance to infection in aged APOE4/E4 male mice that was not present in female mice. Thus, these results suggest that APOE genotype does not play an important role in innate resistance to infection with L. monocytogenes but may be linked to sex-dependent changes that occur during immune senescence.


Subject(s)
Listeria monocytogenes , Listeriosis , Animals , Female , Humans , Male , Mice , Apolipoprotein E2 , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins E/genetics , Cytokines , Genotype , Mice, Inbred C57BL , Mice, Transgenic
3.
Proc Natl Acad Sci U S A ; 116(52): 26892-26899, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31818955

ABSTRACT

Mineral-respiring bacteria use a process called extracellular electron transfer to route their respiratory electron transport chain to insoluble electron acceptors on the exterior of the cell. We recently characterized a flavin-based extracellular electron transfer system that is present in the foodborne pathogen Listeria monocytogenes, as well as many other Gram-positive bacteria, and which highlights a more generalized role for extracellular electron transfer in microbial metabolism. Here we identify a family of putative extracellular reductases that possess a conserved posttranslational flavinylation modification. Phylogenetic analyses suggest that divergent flavinylated extracellular reductase subfamilies possess distinct and often unidentified substrate specificities. We show that flavinylation of a member of the fumarate reductase subfamily allows this enzyme to receive electrons from the extracellular electron transfer system and support L. monocytogenes growth. We demonstrate that this represents a generalizable mechanism by finding that a L. monocytogenes strain engineered to express a flavinylated extracellular urocanate reductase uses urocanate by a related mechanism and to a similar effect. These studies thus identify an enzyme family that exploits a modular flavin-based electron transfer strategy to reduce distinct extracellular substrates and support a multifunctional view of the role of extracellular electron transfer activities in microbial physiology.

4.
J Immunol ; 202(12): 3474-3482, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31061007

ABSTRACT

PGE2 is a lipid-signaling molecule with complex roles in both homeostasis and inflammation. Depending on the cellular context, PGE2 may also suppress certain immune responses. In this study, we tested whether PGE2 could inhibit bacterial killing by polymorphonuclear neutrophils (PMN) using a mouse model of foodborne listeriosis. We found that PGE2 pretreatment decreased the ability of PMN harvested from the bone marrow of either BALB/cByJ or C57BL/6J mice to kill Listeria monocytogenes in vitro. PGE2 treatment slowed the migration of PMN toward the chemoattractant leukotriene B4, decreased uptake of L. monocytogenes by PMN, and inhibited the respiratory burst of PMN compared with vehicle-treated cells. When immune cells were isolated from the livers of infected mice and tested directly ex vivo for the presence of PGE2, BALB/cByJ cells produced significantly more than C57BL/6J cells. Together, these data suggest that robust PGE2 production can suppress PMN effector functions, leading to decreased bacterial killing, which may contribute to the innate susceptibility of BALB/cByJ mice to infection with the facultative intracellular bacterial pathogen L. monocytogenes.


Subject(s)
Dinoprostone/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Monocytes/immunology , Neutrophils/immunology , Animals , Apoptosis , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2/genetics , Respiratory Burst
5.
J Immunol ; 198(7): 2796-2804, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28213502

ABSTRACT

After foodborne transmission of the facultative intracellular bacterial pathogen Listeria monocytogenes, most of the bacterial burden in the gut is extracellular. However, we previously demonstrated that intracellular replication in an as yet unidentified cell type was essential for dissemination and systemic spread of L. monocytogenes In this article, we show that the vast majority of cell-associated L. monocytogenes in the gut were adhered to Ly6Chi monocytes, a cell type that inefficiently internalized L. monocytogenes With bone marrow-derived in vitro cultures, high multiplicity of infection or the use of opsonized bacteria enhanced uptake of L. monocytogenes in CD64- monocytes, but very few bacteria reached the cell cytosol. Surprisingly, monocytes that had upregulated CD64 expression in transition toward becoming macrophages fully supported intracellular growth of L. monocytogenes In contrast, inflammatory monocytes that had increased CD64 expression in the bone marrow of BALB/c/By/J mice prior to L. monocytogenes exposure in the gut did not support L. monocytogenes growth. Thus, contrary to the perception that L. monocytogenes can infect virtually all cell types, neither naive nor inflammatory Ly6Chi monocytes served as a productive intracellular growth niche for L. monocytogenes. These results have broad implications for innate immune recognition of L. monocytogenes in the gut and highlight the need for additional studies on the interaction of extracellular, adherent L. monocytogenes with the unique subsets of myeloid-derived inflammatory cells that infiltrate sites of infection.


Subject(s)
Intestines/immunology , Listeriosis/immunology , Monocytes/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Intestines/microbiology , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Mice , Mice, Inbred BALB C , Phagocytosis , Virulence
6.
J Immunol ; 199(11): 3789-3797, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29055001

ABSTRACT

Recent fate-mapping studies and gene-expression profiles suggest that commonly used protocols to generate bone marrow-derived cultured dendritic cells yield a heterogeneous mixture, including some CD11chi cells that may not have a bona fide counterpart in vivo. In this study, we provide further evidence of the discordance between ex vivo-isolated and in vitro-cultured CD11c+ cells by analyzing an additional phenotype, the ability to support cytosolic growth of the facultative intracellular bacterial pathogen Listeria monocytogenes Two days after foodborne infection of mice with GFP-expressing L. monocytogenes, a small percentage of CD103neg and CD103+ conventional dendritic cells (cDC) in the intestinal lamina propria and mesenteric lymph nodes were GFP+ However, in vitro infection of the same subsets of cells harvested from naive mice resulted in inefficient invasion by the bacteria (<0.1% of the inoculum). The few intracellular bacteria detected survived for only a few hours. In contrast, cultured CD103negCD11c+ cells induced by GM-CSF readily supported exponential growth of L. monocytogenes Flt3 ligand-induced cultures yielded CD103+CD11c+ cells that more closely resembled cDC, with only a modest level of L. monocytogenes replication. For both culture protocols, the longer the cells were maintained in vitro, the more readily they supported intracellular growth. The results of this study suggest that cDC are not a niche for intracellular growth of L. monocytogenes during intestinal infection of mice.


Subject(s)
Bone Marrow/immunology , Dendritic Cells/immunology , Gastrointestinal Tract/immunology , Listeria monocytogenes/physiology , Listeriosis/immunology , Animals , Antigens, CD/metabolism , Bone Marrow/microbiology , CD11 Antigens/metabolism , Cell Growth Processes , Cells, Cultured , DNA Replication , DNA, Bacterial/genetics , Dendritic Cells/microbiology , Female , Gastrointestinal Tract/microbiology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunophenotyping , Integrin alpha Chains/metabolism , Mice , Mice, Inbred BALB C , fms-Like Tyrosine Kinase 3/metabolism
7.
Infect Immun ; 86(4)2018 04.
Article in English | MEDLINE | ID: mdl-29426040

ABSTRACT

Inbred mouse strains differ in their susceptibility to infection with the facultative intracellular bacterium Listeria monocytogenes, largely due to delayed or deficient innate immune responses. Previous antibody depletion studies suggested that neutrophils (polymorphonuclear leukocytes [PMN]) were particularly important for clearance in the liver, but the ability of PMN from susceptible and resistant mice to directly kill L. monocytogenes has not been examined. In this study, we showed that PMN infiltrated the livers of BALB/c/By/J (BALB/c) and C57BL/6 (B6) mice in similar numbers and that both cell types readily migrated toward leukotriene B4 in an in vitro chemotaxis assay. However, CFU burdens in the liver were significantly higher in BALB/c mice than in other strains, suggesting that PMN in the BALB/c liver might not be able to clear L. monocytogenes as efficiently as B6 PMN. Unprimed PMN harvested from either BALB/c or B6 bone marrow killed L. monocytogenes directly ex vivo, and pretreatment with autologous serum significantly enhanced killing efficiency for both. L. monocytogenes were internalized within 10 min and rapidly triggered intracellular production of reactive oxygen species in a dose-dependent manner. However, PMN from gp91phox-deficient mice also readily killed L. monocytogenes, which suggested that nonoxidative killing mechanisms may be sufficient for bacterial clearance. Together, these results indicate that there is not an intrinsic defect in the ability of PMN from susceptible BALB/c mice to kill L. monocytogenes and further suggest that if PMN function is impaired in BALB/c mice, it is likely due to locally produced modulating factors present in the liver during infection.


Subject(s)
Disease Resistance/immunology , Disease Susceptibility/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis/immunology , Animals , Biomarkers , Immunity, Innate , Listeriosis/metabolism , Liver/immunology , Liver/metabolism , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Viability/immunology , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Respiratory Burst/immunology , Species Specificity
8.
J Immunol ; 196(7): 3109-16, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26895837

ABSTRACT

Type I IFN (IFN-α/ß) is thought to enhance growth of the foodborne intracellular pathogen Listeria monocytogenes by promoting mechanisms that dampen innate immunity to infection. However, the type I IFN response has been studied primarily using methods that bypass the stomach and, therefore, fail to replicate the natural course of L. monocytogenes infection. In this study, we compared i.v. and foodborne transmission of L. monocytogenes in mice lacking the common type I IFN receptor (IFNAR1(-/-)). Contrary to what was observed using i.v. infection, IFNAR1(-/-) and wild-type mice had similar bacterial burdens in the liver and spleen following foodborne infection. Splenocytes from wild-type mice infected i.v. produced significantly more IFN-ß than did those infected by the foodborne route. Consequently, the immunosuppressive effects of type I IFN signaling, which included T cell death, increased IL-10 secretion, and repression of neutrophil recruitment to the spleen, were all observed following i.v. but not foodborne transmission of L. monocytogenes. Type I IFN was also previously shown to cause a loss of responsiveness to IFN-γ through downregulation of the IFN-γ receptor α-chain on macrophages and dendritic cells. However, we detected a decrease in surface expression of IFN-γ receptor α-chain even in the absence of IFN-α/ß signaling, suggesting that in vivo, this infection-induced phenotype is not type I IFN-dependent. These results highlight the importance of using the natural route of infection for studies of host-pathogen interactions and suggest that the detrimental effects of IFN-α/ß signaling on the innate immune response to L. monocytogenes may be an artifact of the i.v. infection model.


Subject(s)
Disease Susceptibility , Interferon Type I/genetics , Listeria monocytogenes/immunology , Listeriosis/genetics , Listeriosis/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Gene Expression , Interferon Type I/metabolism , Interferon-beta/biosynthesis , Listeriosis/metabolism , Lymphocyte Depletion , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Signal Transduction , Spleen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
PLoS Pathog ; 10(8): e1004301, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25101646

ABSTRACT

We characterized key components and major targets of the c-di-GMP signaling pathways in the foodborne pathogen Listeria monocytogenes, identified a new c-di-GMP-inducible exopolysaccharide responsible for motility inhibition, cell aggregation, and enhanced tolerance to disinfectants and desiccation, and provided first insights into the role of c-di-GMP signaling in listerial virulence. Genome-wide genetic and biochemical analyses of c-di-GMP signaling pathways revealed that L. monocytogenes has three GGDEF domain proteins, DgcA (Lmo1911), DgcB (Lmo1912) and DgcC (Lmo2174), that possess diguanylate cyclase activity, and three EAL domain proteins, PdeB (Lmo0131), PdeC (Lmo1914) and PdeD (Lmo0111), that possess c-di-GMP phosphodiesterase activity. Deletion of all phosphodiesterase genes (ΔpdeB/C/D) or expression of a heterologous diguanylate cyclase stimulated production of a previously unknown exopolysaccharide. The synthesis of this exopolysaccharide was attributed to the pssA-E (lmo0527-0531) gene cluster. The last gene of the cluster encodes the fourth listerial GGDEF domain protein, PssE, that functions as an I-site c-di-GMP receptor essential for exopolysaccharide synthesis. The c-di-GMP-inducible exopolysaccharide causes cell aggregation in minimal medium and impairs bacterial migration in semi-solid agar, however, it does not promote biofilm formation on abiotic surfaces. The exopolysaccharide also greatly enhances bacterial tolerance to commonly used disinfectants as well as desiccation, which may contribute to survival of L. monocytogenes on contaminated food products and in food-processing facilities. The exopolysaccharide and another, as yet unknown c-di-GMP-dependent target, drastically decrease listerial invasiveness in enterocytes in vitro, and lower pathogen load in the liver and gallbladder of mice infected via an oral route, which suggests that elevated c-di-GMP levels play an overall negative role in listerial virulence.


Subject(s)
Bacterial Proteins/metabolism , Cyclic GMP/analogs & derivatives , Gene Expression Regulation, Bacterial/physiology , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Animals , Bacterial Proteins/genetics , Chromatography, High Pressure Liquid , Cyclic GMP/metabolism , Disease Models, Animal , Escherichia coli Proteins/metabolism , Female , Listeria monocytogenes/genetics , Listeriosis/genetics , Mice , Mice, Inbred BALB C , Phosphorus-Oxygen Lyases/metabolism , Signal Transduction/physiology , Virulence/physiology
10.
Infect Immun ; 83(8): 3146-56, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26015479

ABSTRACT

Listeria monocytogenes is a highly adaptive bacterium that replicates as a free-living saprophyte in the environment as well as a facultative intracellular pathogen that causes invasive foodborne infections. The intracellular life cycle of L. monocytogenes is considered to be its primary virulence determinant during mammalian infection; however, the proportion of L. monocytogenes that is intracellular in vivo has not been studied extensively. In this report, we demonstrate that the majority of wild-type (strain EGDe) and mouse-adapted (InlA(m)-expressing) L. monocytogenes recovered from the mesenteric lymph nodes (MLN) was extracellular within the first few days after foodborne infection. In addition, significantly lower burdens of L. monocytogenes were recovered from the colon, spleen, and liver of gentamicin-treated mice than of control mice. This led us to investigate whether intracellular replication of L. monocytogenes was essential during the intestinal phase of infection. We found that lipoate protein ligase-deficient L. monocytogenes (ΔlplA1) mutants, which display impaired intracellular growth, were able to colonize the colon but did not persist efficiently and had a significant defect in spreading to the MLN, spleen, and liver. Together, these data indicate that the majority of the L. monocytogenes burden in the gastrointestinal tract is extracellular, but the small proportion of intracellular L. monocytogenes is essential for dissemination to the MLN and systemic organs.


Subject(s)
Foodborne Diseases/microbiology , Intestines/microbiology , Listeria monocytogenes/growth & development , Listeriosis/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Liver/microbiology , Lymph Nodes/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/microbiology
11.
PLoS Pathog ; 8(11): e1003015, 2012.
Article in English | MEDLINE | ID: mdl-23166492

ABSTRACT

Intestinal Listeria monocytogenes infection is not efficient in mice and this has been attributed to a low affinity interaction between the bacterial surface protein InlA and E-cadherin on murine intestinal epithelial cells. Previous studies using either transgenic mice expressing human E-cadherin or mouse-adapted L. monocytogenes expressing a modified InlA protein (InlA(m)) with high affinity for murine E-cadherin showed increased efficiency of intragastric infection. However, the large inocula used in these studies disseminated to the spleen and liver rapidly, resulting in a lethal systemic infection that made it difficult to define the natural course of intestinal infection. We describe here a novel mouse model of oral listeriosis that closely mimics all phases of human disease: (1) ingestion of contaminated food, (2) a distinct period of time during which L. monocytogenes colonize only the intestines, (3) varying degrees of systemic spread in susceptible vs. resistant mice, and (4) late stage spread to the brain. Using this natural feeding model, we showed that the type of food, the time of day when feeding occurred, and mouse gender each affected susceptibility to L. monocytogenes infection. Co-infection studies using L. monocytogenes strains that expressed either a high affinity ligand for E-cadherin (InlA(m)), a low affinity ligand (wild type InlA from Lm EGDe), or no InlA (ΔinlA) showed that InlA was not required to establish intestinal infection in mice. However, expression of InlA(m) significantly increased bacterial persistence in the underlying lamina propria and greatly enhanced dissemination to the mesenteric lymph nodes. Thus, these studies revealed a previously uncharacterized role for InlA in facilitating systemic spread via the lymphatic system after invasion of the gut mucosa.


Subject(s)
Bacterial Proteins/immunology , Bacterial Translocation/immunology , Foodborne Diseases/immunology , Intestinal Diseases/immunology , Listeria monocytogenes/physiology , Listeriosis/immunology , Lymph Nodes/immunology , Mesentery/immunology , Animals , Bacterial Proteins/genetics , Cadherins/genetics , Cadherins/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Foodborne Diseases/genetics , Foodborne Diseases/microbiology , Foodborne Diseases/pathology , Humans , Intestinal Diseases/genetics , Intestinal Diseases/microbiology , Intestinal Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Listeriosis/genetics , Listeriosis/pathology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Mesentery/microbiology , Mesentery/physiology , Mice , Mice, Inbred BALB C
12.
J Leukoc Biol ; 116(1): 132-145, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38416405

ABSTRACT

Lymph node stromal cells (LNSCs) are an often overlooked component of the immune system but play a crucial role in maintaining tissue homeostasis and orchestrating immune responses. Our understanding of the functions these cells serve in the context of bacterial infections remains limited. We previously showed that Listeria monocytogenes, a facultative intracellular foodborne bacterial pathogen, must replicate within an as-yet-unidentified cell type in the mesenteric lymph node (MLN) to spread systemically. Here, we show that L. monocytogenes could invade, escape from the vacuole, replicate exponentially, and induce a type I interferon response in the cytosol of 2 LNSC populations infected in vitro, fibroblastic reticular cells (FRCs) and blood endothelial cells (BECs). Infected FRCs and BECs also produced a significant chemokine and proinflammatory cytokine response after in vitro infection. Flow cytometric analysis confirmed that GFP+  L. monocytogenes were associated with a small percentage of MLN stromal cells in vivo following foodborne infection of mice. Using fluorescent microscopy, we showed that these cell-associated bacteria were intracellular L. monocytogenes and that the number of infected FRCs and BECs changed over the course of a 3-day infection in mice. Ex vivo culturing of these infected LNSC populations revealed viable, replicating bacteria that grew on agar plates. These results highlight the unexplored potential of FRCs and BECs to serve as suitable growth niches for L. monocytogenes during foodborne infection and to contribute to the proinflammatory environment within the MLN that promotes clearance of listeriosis.


Subject(s)
Listeria monocytogenes , Listeriosis , Lymph Nodes , Stromal Cells , Animals , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/pathology , Lymph Nodes/microbiology , Lymph Nodes/immunology , Lymph Nodes/pathology , Stromal Cells/microbiology , Stromal Cells/metabolism , Mice , Mice, Inbred C57BL , Cytokines/metabolism , Endothelial Cells/microbiology , Endothelial Cells/metabolism , Disease Susceptibility , Female
13.
J Immunol ; 186(3): 1703-12, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21191063

ABSTRACT

A subset of CD44(hi)CD8(+) T cells isolated from C57BL/6/J (B6) mice, but not BALB/c/By/J (BALB/c) mice, rapidly secrete IFN-γ within 16 h of infection with Listeria monocytogenes. This Ag-independent response requires the presence of both IL-12 and IL-18. Previous studies showed that dendritic cells from B6 mice produced more Th1-type cytokines such as IL-12 than did those from BALB/c mice in response to L. monocytogenes infection. In this report, we demonstrate that the microenvironment in L. monocytogenes-infected BALB/c mice is sufficient to induce responsive B6 CD8(+) T cells to rapidly secrete IFN-γ. Furthermore, BALB/c CD8(+) T cells did not rapidly secrete IFN-γ even when they were exposed to high concentrations of IL-12 plus IL-18 in vitro. In the presence of IL-12 and IL-18, B6 CD44(hi)CD8(+) T cells upregulated expression of the receptor subunits for these cytokines more rapidly than did BALB/c T cells. In comparing particular subsets of memory phenotype CD8(+) T cells, we found that virtual memory cells, rather than true Ag-experienced cells, had the greatest level of impairment in BALB/c mice. These data suggest that the degree of cytokine-driven bystander activation of CD8(+) T cells that occurs during infection depends on both APCs and T cell-intrinsic properties that can vary among mouse strains.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Interferon-gamma/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Homeostasis/genetics , Homeostasis/immunology , Immunologic Memory/genetics , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-15/metabolism , Interleukin-18/metabolism , Listeriosis/immunology , Listeriosis/metabolism , Listeriosis/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Time Factors
14.
J Exp Med ; 203(2): 383-91, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16461341

ABSTRACT

Studies using major histocompatibility complex (MHC)-Ia-deficient mice have shown that MHC-Ib-restricted CD8+ T cells can clear infections caused by intracellular pathogens such as Listeria monocytogenes. M3-restricted CD8+ T cells, which recognize short hydrophobic N-formylated peptides, appear to comprise a substantial portion of the MHC-Ib-restricted T cell response in the mouse model of L. monocytogenes infection. In this study, we isolated formyltransferase (fmt) mutant strains of L. monocytogenes that lacked the ability to add formyl groups to nascent polypeptides. These fmt mutant Listeria strains did not produce antigens that could be recognized by M3-restricted T cells. We showed that immunization of MHC-Ia-deficient mice with fmt mutant Listeria resulted in stimulation of a protective memory response that cleared subsequent challenge with wild-type L. monocytogenes, despite the fact that M3-restricted CD8+ T cells did not proliferate in these mice. These data suggest that M3-restricted T cells are not required for protection against L. monocytogenes and underscore the importance of searching for new antigen-presenting molecules among the large MHC-Ib family of proteins.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Histocompatibility Antigens Class I/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Histocompatibility Antigens Class I/physiology , Hydroxymethyl and Formyl Transferases/genetics , Listeria monocytogenes/enzymology , Listeria monocytogenes/genetics , Listeriosis/microbiology , Liver/immunology , Liver/microbiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutation , Peptides/metabolism , Spleen/immunology , Spleen/microbiology
15.
Infect Immun ; 79(1): 177-84, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20956571

ABSTRACT

Heat shock factor 1 (HSF1) is a stress-induced transcription factor that promotes expression of genes that protect mammalian cells from the lethal effects of severely elevated temperatures (>42°C). However, we recently showed that HSF1 is activated at a lower temperature (39.5°C) in T cells, suggesting that HSF1 may be important for preserving T cell function during pathogen-induced fever responses. To test this, we examined the role of HSF1 in clearance of Listeria monocytogenes, an intracellular bacterial pathogen that elicits a strong CD8(+) T cell response in mice. Using temperature transponder microchips, we showed that the core body temperature increased approximately 2°C in L. monocytogenes-infected mice and that the fever response was maintained for at least 24 h. HSF1-deficient mice cleared a low-dose infection with slightly slower kinetics than did HSF1(+/+) littermate controls but were significantly more susceptible to challenges with higher doses of bacteria. Surprisingly, HSF1-deficient mice did not show a defect in CD8(+) T cell responses following sublethal infection. However, when HSF1-deficient mice were challenged with high doses of L. monocytogenes, increased levels of serum tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ) compared to those of littermate control mice were observed, and rapid death of the animals occurred within 48 to 60 h of infection. Neutralization of TNF-α enhanced the survival of HSF1-deficient mice. These results suggest that HSF1 is needed to prevent the overproduction of proinflammatory cytokines and subsequent death due to septic shock that can result following high-dose challenge with bacterial pathogens.


Subject(s)
DNA-Binding Proteins/metabolism , Fever/metabolism , Listeria monocytogenes , Listeriosis/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , CD8-Positive T-Lymphocytes/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Genotype , Heat Shock Transcription Factors , Interferon-gamma/blood , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
16.
Cell Immunol ; 272(1): 79-86, 2011.
Article in English | MEDLINE | ID: mdl-21978649

ABSTRACT

A subset of CD44(hi)CD8+ T cells in some, but not all mice, can be induced to rapidly secrete IFNγ during infection with Listeria monocytogenes. This response is dependent on the presence of both IL-12 and IL-18 and does not require engagement of the T cell receptor. In this study, we demonstrate that human CD8+ T cells also vary widely in their ability to secrete IFNγ within 15h of either Listeria infection or cytokine stimulation. The magnitude of the rapid IFNγ response correlated more closely with the intrinsic responsiveness of the T cells to cytokine stimulation rather than the amount of IL-12 produced. CD8+ T cells from 2 out of 16 blood donors (12.5%) failed to generate a significant IFNγ response. These results demonstrate that bystander activation of CD8+ T cells varies among individuals and validate further study of the differential responses observed using BALB/c vs. C57BL/6 mice.


Subject(s)
CD8-Positive T-Lymphocytes , Immunity, Innate , Interferon-gamma/biosynthesis , Interleukin-12/pharmacology , Interleukin-18/pharmacology , Listeriosis/immunology , Adult , Animals , Bystander Effect/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunologic Memory/drug effects , Immunologic Memory/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-18/immunology , Listeria monocytogenes/drug effects , Listeria monocytogenes/immunology , Listeriosis/metabolism , Listeriosis/microbiology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
17.
Microbiol Resour Announc ; 10(18)2021 May 06.
Article in English | MEDLINE | ID: mdl-33958419

ABSTRACT

We report the whole-genome sequence of Listeria monocytogenes UKVDL9 and an edited draft genome sequence of L. monocytogenes 2010L-2198. Both are neurotropic lineage III strains; UKVDL9 was isolated from a sheep brain, and 2010L-2198 was isolated from a human subject with rhombencephalitis.

18.
Curr Protoc Immunol ; 130(1): e102, 2020 09.
Article in English | MEDLINE | ID: mdl-32710703

ABSTRACT

Listeria monocytogenes is a foodborne pathogen that causes serious, often deadly, systemic disease in susceptible individuals such as neonates and the elderly. These facultative intracellular bacteria have been an invaluable tool in immunology research for more than three decades. Intravenous (i.v.) injection is the most commonly used transmission route in mice, but oral models of infection have also been developed in recent years, and these may be more appropriate for many studies. This article includes detailed instructions for use of either foodborne or i.v. inoculation of mice and discusses the rationale for choosing either model. Additionally, a protocol is provided for enrichment of neutrophils and monocytes from the infected liver in a manner that allows for determination of bacterial burden while still providing sufficient cells for use in flow cytometric analysis or in vitro assays. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Foodborne L. monocytogenes infection Support Protocol 1: Preparing L. monocytogenes for foodborne infection Basic Protocol 2: Intravenous L. monocytogenes infection Support Protocol 2: Preparing L. monocytogenes for intravenous infection Basic Protocol 3: Enrichment of non-parenchymal cells from the infected liver.


Subject(s)
Listeria monocytogenes/physiology , Listeriosis/etiology , Listeriosis/pathology , Liver/microbiology , Liver/pathology , Monocytes/pathology , Neutrophils/pathology , Animals , Biomarkers , Biopsy , Disease Models, Animal , Disease Susceptibility , Female , Foodborne Diseases/microbiology , Humans , Immunophenotyping , Listeriosis/metabolism , Listeriosis/transmission , Mice , Monocytes/immunology , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Species Specificity
19.
mSphere ; 5(5)2020 09 16.
Article in English | MEDLINE | ID: mdl-32938704

ABSTRACT

Listeria monocytogenes is thought to colonize the brain using one of three mechanisms: direct invasion of the blood-brain barrier, transportation across the barrier by infected monocytes, and axonal migration to the brain stem. The first two pathways seem to occur following unrestricted bacterial growth in the blood and thus have been linked to immunocompromise. In contrast, cell-to-cell spread within nerves is thought to be mediated by a particular subset of neurotropic L. monocytogenes strains. In this study, we used a mouse model of foodborne transmission to evaluate the neurotropism of several L. monocytogenes isolates. Two strains preferentially colonized the brain stems of BALB/cByJ mice 5 days postinfection and were not detectable in blood at that time point. In contrast, infection with other strains resulted in robust systemic infection of the viscera but no dissemination to the brain. Both neurotropic strains (L2010-2198, a human rhombencephalitis isolate, and UKVDL9, a sheep brain isolate) typed as phylogenetic lineage III, the least characterized group of L. monocytogenes Neither of these strains encodes InlF, an internalin-like protein that was recently shown to promote invasion of the blood-brain barrier. Acute neurologic deficits were observed in mice infected with the neurotropic strains, and milder symptoms persisted for up to 16 days in some animals. These results demonstrate that neurotropic L. monocytogenes strains are not restricted to any one particular lineage and suggest that the foodborne mouse model of listeriosis can be used to investigate the pathogenic mechanisms that allow L. monocytogenes to invade the brain stem.IMPORTANCE Progress in understanding the two naturally occurring central nervous system (CNS) manifestations of listeriosis (meningitis/meningoencephalitis and rhombencephalitis) has been limited by the lack of small animal models that can readily distinguish between these distinct infections. We report here that certain neurotropic strains of Listeria monocytogenes can spread to the brains of young otherwise healthy mice and cause neurological deficits without causing a fatal bacteremia. The novel strains described here fall within phylogenetic lineage III, a small collection of L. monocytogenes isolates that have not been well characterized to date. The animal model reported here mimics many features of human rhombencephalitis and will be useful for studying the mechanisms that allow L. monocytogenes to disseminate to the brain stem following natural foodborne transmission.


Subject(s)
Brain/microbiology , Listeria monocytogenes/pathogenicity , Listeriosis/blood , Viral Tropism , Animals , Brain/pathology , Central Nervous System/microbiology , Disease Models, Animal , Female , Humans , Infectious Encephalitis/microbiology , Listeria monocytogenes/isolation & purification , Listeriosis/microbiology , Listeriosis/transmission , Mice , Mice, Inbred BALB C , Phylogeny , Sheep , Virulence
20.
Infect Immun ; 77(4): 1492-501, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179413

ABSTRACT

A subset of CD8+ T cells can rapidly secrete gamma interferon (IFN-gamma) in an antigen-independent and interleukin-12 (IL-12)- and IL-18-dependent manner within 16 h of infection with the intracellular bacterial pathogen Listeria monocytogenes. This rapid IFN-gamma response is robust enough to be detected directly ex vivo and is not observed following infection with intracellular bacterial pathogens that remain sequestered within host cell vacuoles. We demonstrate here that three distinct pathways can lead to rapid secretion of IFN-gamma by CD8+ T cells during L. monocytogenes infection: (i) a direct cytokine-inducing activity encoded by the cholesterol-dependent cytolysin (CDC) listeriolysin O (LLO) acts within the infected cell, (ii) the pore-forming activity of LLO promotes cytosolic localization of bacterial products that trigger cytosol-specific signaling pathways, and (iii) the sustained presence of high concentrations of bacterial products can exogenously trigger cytokine production. Although it has been suggested that CDC protein toxins may act as Toll-like receptor 4 (TLR4) agonists to trigger proinflammatory cytokine secretion, we show in this report that TLR4 signaling is not required to induce a maximal rapid IFN-gamma response by CD8+ T cells. The results presented here indicate that multiple mechanisms contribute to the induction of rapid IFN-gamma secretion by CD8+ T cells during Listeria infection and that care must be taken when interpreting the results of in vitro assays, since the contribution of each pathway can vary depending on how the assay is performed.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/biosynthesis , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Time Factors , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL