Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2345, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38528069

ABSTRACT

Loss-of-function mutations have provided crucial insights into the immunoregulatory actions of Foxp3+ regulatory T cells (Tregs). By contrast, we know very little about the consequences of defects that amplify aspects of Treg function or differentiation. Here we show that mice heterozygous for an Ikbkb gain-of-function mutation develop psoriasis. Doubling the gene dose (IkbkbGoF/GoF) results in dactylitis, spondylitis, and characteristic nail changes, which are features of psoriatic arthritis. IkbkbGoF mice exhibit a selective expansion of Foxp3 + CD25+ Tregs of which a subset express IL-17. These modified Tregs are enriched in both inflamed tissues, blood and spleen, and their transfer is sufficient to induce disease without conventional T cells. Single-cell transcriptional and phenotyping analyses of isolated Tregs reveal expansion of non-lymphoid tissue (tissue-resident) Tregs expressing Th17-related genes, Helios, tissue-resident markers including CD103 and CD69, and a prominent NF-κB transcriptome. Thus, IKK2 regulates tissue-resident Treg differentiation, and overactivity drives dose-dependent skin and systemic inflammation.


Subject(s)
Gain of Function Mutation , I-kappa B Kinase , T-Lymphocytes, Regulatory , Animals , Mice , Forkhead Transcription Factors/genetics , I-kappa B Kinase/genetics , Inflammation/genetics
2.
EMBO Rep ; 24(6): e54600, 2023 06 05.
Article in English | MEDLINE | ID: mdl-37073791

ABSTRACT

Inflammasome signaling is a central pillar of innate immunity triggering inflammation and cell death in response to microbes and danger signals. Here, we show that two virulence factors from the human bacterial pathogen Clostridium perfringens are nonredundant activators of the NLRP3 inflammasome in mice and humans. C. perfringens lecithinase (also known as phospolipase C) and C. perfringens perfringolysin O induce distinct mechanisms of activation. Lecithinase enters LAMP1+ vesicular structures and induces lysosomal membrane destabilization. Furthermore, lecithinase induces the release of the inflammasome-dependent cytokines IL-1ß and IL-18, and the induction of cell death independently of the pore-forming proteins gasdermin D, MLKL and the cell death effector protein ninjurin-1 or NINJ1. We also show that lecithinase triggers inflammation via the NLRP3 inflammasome in vivo and that pharmacological blockade of NLRP3 using MCC950 partially prevents lecithinase-induced lethality. Together, these findings reveal that lecithinase activates an alternative pathway to induce inflammation during C. perfringens infection and that this mode of action can be similarly exploited for sensing by a single inflammasome.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Clostridium perfringens/metabolism , Virulence Factors , Inflammation , Interleukin-1beta/metabolism , Nerve Growth Factors , Cell Adhesion Molecules, Neuronal
3.
Vaccine ; 39(12): 1746-1757, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33618946

ABSTRACT

Merozoite surface protein 2 (MSP2) is a highly abundant, GPI-anchored surface antigen on merozoites of the malaria parasite Plasmodium falciparum. It consists of highly conserved N- and C-terminal domains, and a central polymorphic region that allows all MSP2 alleles to be categorized into the 3D7 or FC27 family. Previously it has been shown that epitope accessibility differs between lipid-bound and lipid-free MSP2, suggesting that lipid interactions modulate the conformation and antigenicity in a way that may better mimic native MSP2 on the merozoite surface. Therefore, we have immunised mice with MSP2 engrafted onto liposomes using a C-terminal tether that mimics the native GPI anchor. To improve the immunogenicity of the formulated antigen, liposomes were supplemented with Pathogen Associated Molecular Pattern molecules, specifically agonists of the Toll-like receptor 4 (TLR4) or TLR2. Induced antibodies were directed mostly towards conserved epitopes, predominantly in the conserved C-terminal region of MSP2. We also found that immunisation with a combination of 3D7 and FC27 MSP2 enhanced antibody responses to conserved epitopes, and that the overall responses of mice immunised with MSP2-engrafted liposomes were comparable in magnitude to those of mice immunised with MSP2 formulated in Montanide ISA720. The antibodies elicited in mice by immunising with MSP2-engrafted liposomes recognised the native form of parasite MSP2 on western blots and were found to be cross-reactive with isolated 3D7 and FC27 merozoites when investigated by ELISA. The liposome-tethered MSP2 induced higher titres of complement-fixing antibodies to 3D7 and FC27 MSP2 than did MSP2 formulated in Montanide ISA720. Our results indicate that liposomal formulation represents a viable strategy for eliciting a strong immune response that favours conserved epitopes in MSP2 and thus a strain-transcendent immune response.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Animals , Antibodies, Protozoan , Antigens, Protozoan/genetics , Epitopes , Immunity , Liposomes , Malaria, Falciparum/prevention & control , Membrane Proteins , Merozoites , Mice , Plasmodium falciparum , Protozoan Proteins/genetics
4.
Nat Commun ; 11(1): 760, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029733

ABSTRACT

Inflammasomes are important for host defence against pathogens and homeostasis with commensal microbes. Here, we show non-haemolytic enterotoxin (NHE) from the neglected human foodborne pathogen Bacillus cereus is an activator of the NLRP3 inflammasome and pyroptosis. NHE is a non-redundant toxin to haemolysin BL (HBL) despite having a similar mechanism of action. Via a putative transmembrane region, subunit C of NHE initiates binding to the plasma membrane, leading to the recruitment of subunit B and subunit A, thus forming a tripartite lytic pore that is permissive to efflux of potassium. NHE mediates killing of cells from multiple lineages and hosts, highlighting a versatile functional repertoire in different host species. These data indicate that NHE and HBL operate synergistically to induce inflammation and show that multiple virulence factors from the same pathogen with conserved function and mechanism of action can be exploited for sensing by a single inflammasome.


Subject(s)
Bacillus cereus/pathogenicity , Enterotoxins/toxicity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Bacterial Proteins/toxicity , Cell Line , Enterotoxins/chemistry , Female , Hemolysin Proteins/toxicity , Host Microbial Interactions , Host Specificity , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroptosis/drug effects , Virulence Factors/toxicity
5.
Nat Microbiol ; 4(2): 362-374, 2019 02.
Article in English | MEDLINE | ID: mdl-30531979

ABSTRACT

Host recognition of microbial components is essential in mediating an effective immune response. Cytosolic bacteria must secure entry into the host cytoplasm to facilitate replication and, in doing so, liberate microbial ligands that activate cytosolic innate immune sensors and the inflammasome. Here, we identified a multicomponent enterotoxin, haemolysin BL (HBL), that engages activation of the inflammasome. This toxin is highly conserved among the human pathogen Bacillus cereus. The three subunits of HBL bind to the cell membrane in a linear order, forming a lytic pore and inducing activation of the NLRP3 inflammasome, secretion of interleukin-1ß and interleukin-18, and pyroptosis. Mechanistically, the HBL-induced pore results in the efflux of potassium and triggers the activation of the NLRP3 inflammasome. Furthermore, HBL-producing B. cereus induces rapid inflammasome-mediated mortality. Pharmacological inhibition of the NLRP3 inflammasome using MCC950 prevents B. cereus-induced lethality. Overall, our results reveal that cytosolic sensing of a toxin is central to the innate immune recognition of infection. Therapeutic modulation of this pathway enhances host protection against deadly bacterial infections.


Subject(s)
Bacillus cereus/immunology , Bacterial Proteins/immunology , Enterotoxins/immunology , Hemolysin Proteins/immunology , Inflammasomes/metabolism , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Cells, Cultured , Culture Media, Conditioned , Enterotoxins/chemistry , Enterotoxins/metabolism , Female , Hemolysin Proteins/metabolism , Immunity, Innate , Macrophages/immunology , Macrophages/pathology , Macrophages/ultrastructure , Male , Mice , Mice, Mutant Strains , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Potassium/metabolism , Protein Multimerization , Pyroptosis , Survival Analysis
6.
Cancer Immunol Immunother ; 67(9): 1461-1472, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30014244

ABSTRACT

INTRODUCTION: In this phase I study using a 3 + 3 dose escalation design, the safety, dose-limiting toxicity (DLT), immunogenicity and efficacy of intravenous Lipovaxin-MM-a multi-component dendritic cell-targeted liposomal vaccine against metastatic melanoma-was investigated. METHODS: Twelve subjects with metastatic cutaneous melanoma were recruited in three cohorts. Patients in Cohort A (n = 3) and Cohort B (n = 3) received three doses of 0.1 and 1 mL of Lipovaxin-MM, respectively, every 4 weeks. Patients in Cohort C (n = 6) received four doses of 3 mL vaccine weekly. Immunologic assessments of peripheral blood were made at regular intervals and included leukocyte subsets, cytokine levels, and Lipovaxin-MM-specific T-cell and antibody reactivities. Tumor responses were assessed by RECIST v1.0 at screening, then 8 weekly in Cohorts A and B and 6 weekly in Cohort C. RESULTS: Of a total of 94 adverse events (AEs) reported in ten subjects, 43 AEs in six subjects were considered to be possibly or probably vaccine-related. Most (95%) vaccine-related AEs were grade 1 or 2, two (5%) grade 3 vaccine-related AEs of anemia and lethargy were recorded, and higher grade AEs and DLTs were not observed. No consistent evidence of vaccine-specific humoral or cellular immune responses was found in post-immunization blood samples. One patient had a partial response, two patients had stable disease, and the remaining patients had progressive disease. CONCLUSIONS: Lipovaxin-MM was well tolerated and without clinically significant toxicity. Immunogenicity of Lipovaxin-MM was not detected. Partial response and stable disease were observed in one and two patients, respectively.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Aged , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Dendritic Cells/drug effects , Dose-Response Relationship, Immunologic , Female , Humans , Liposomes/administration & dosage , Liposomes/immunology , Male , Melanoma/immunology , Middle Aged , Skin Neoplasms/immunology , Melanoma, Cutaneous Malignant
7.
PLoS One ; 8(3): e59934, 2013.
Article in English | MEDLINE | ID: mdl-23544110

ABSTRACT

Cholera toxin (CT) is a mucosal adjuvant capable of inducing strong immune responses to co-administered antigens following oral or intranasal immunization of mice. To date, the direct effect of CT on antigen-specific CD4(+) T cell migration and proliferation profiles in vivo is not well characterized. In this study, the effect of CT on the migration pattern and proliferative responses of adoptively transferred, CD4(+) TCR transgenic T cells in orally or intranasally vaccinated mice, was analyzed by flow cytometry. GFP-expressing or CFSE-labeled OT-II lymphocytes were adoptively transferred to naïve C57BL/6 mice, and mice were subsequently vaccinated with OVA with or without CT via the oral or intranasal route. CT did not alter the migration pattern of antigen-specific T cells, regardless of the route of immunization, but increased the number of transgenic CD4(+) T cells in draining lymphoid tissue. This increase in the number of transgenic CD4(+) T cells was not due to cells undergoing more rounds of cellular division in vivo, suggesting that CT may exert an indirect adjuvant effect on CD4(+) T cells. The findings reported here suggest that CT functions as a mucosal adjuvant by increasing the number of antigen specific CD4(+) T cells independent of their migration pattern or kinetics of cellular division.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Division/immunology , Cell Movement/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/pharmacology , Epitopes/immunology , Mucous Membrane/immunology , Administration, Intranasal , Administration, Oral , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Division/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Evans Blue/metabolism , Fluoresceins/metabolism , Green Fluorescent Proteins/metabolism , Histocompatibility Antigens Class II/metabolism , Kinetics , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/drug effects , Ovalbumin/immunology , Peptides/pharmacology , Succinimides/metabolism
8.
J Immunol ; 182(12): 7440-50, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494267

ABSTRACT

Celiac disease is a chronic inflammatory enteropathy caused by cellular immunity to dietary gluten. More than 90% of patients carry HLA-DQ2 encoded by HLA-DQA1*05 and DQB1*02, and gluten-specific CD4(+) T cells from intestinal biopsies of these patients are HLA-DQ2-restricted, produce Th1 cytokines and preferentially recognize gluten peptides deamidated by tissue transglutaminase. We generated mice lacking murine MHC class II genes that are transgenic for human CD4 and the autoimmunity and celiac disease-associated HLA-DR3-DQ2 haplotype. Immunization with the alpha-gliadin 17-mer that incorporates the overlapping DQ2-alpha-I and DQ2-alpha-II epitopes immunodominant in human celiac disease generates peptide-specific HLA-DQ2-restricted CD4(+) T cells. When exposed to dietary gluten, naive or gliadin-primed mice do not develop pathology. Coincident introduction of dietary gluten and intestinal inflammation resulted in low-penetrance enteropathy and tissue transglutaminase-specific IgA. Two further strains of transgenic mice expressing HLA-DR3-DQ2 and human CD4, one with a NOD background and another TCR transgenic having over 90% of CD4(+) T cells specific for the DQ2-alpha-II epitope with a Th1 phenotype, were also healthy when consuming gluten. These humanized mouse models indicate that gluten ingestion can be tolerated without intestinal pathology even when HLA-DQ2-restricted CD4(+) T cell immunity to gluten is established, thereby implicating additional factors in controlling the penetrance of celiac disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Celiac Disease/immunology , Gliadin/immunology , HLA-DQ Antigens/immunology , HLA-DR3 Antigen/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Celiac Disease/genetics , Celiac Disease/metabolism , Celiac Disease/pathology , Cell Separation , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , Forkhead Transcription Factors/immunology , HLA-DQ Antigens/genetics , HLA-DQ Antigens/metabolism , HLA-DR3 Antigen/genetics , HLA-DR3 Antigen/metabolism , Health , Hybridomas , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Substrate Specificity
9.
Infect Immun ; 76(5): 2235-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18332213

ABSTRACT

Helicobacter pylori is the etiological agent of human chronic gastritis, a condition seen as a precursor to the development of gastrointestinal ulcers or gastric cancer. This study utilized the murine model of chronic H. pylori infection to characterize the role of macrophages in the induction of specific immune responses and gastritis and in the control of the bacterial burden following H. pylori infection and vaccination. Drug-loaded liposomes were injected intravenously to deplete macrophages from C57BL/6 mice, and effective removal of CD11b+ cells from the spleens and stomachs of mice was confirmed by immunofluorescence microscopy. Transient elimination of macrophages from C57BL/6 mice during the early period of infection reduced the gastric pathology induced by H. pylori SS1 but did not affect the bacterial load in the stomach. These data suggest that macrophages are important to the severity of gastric inflammation during H. pylori infection.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Macrophages/immunology , Macrophages/microbiology , Animals , CD11b Antigen/analysis , Colony Count, Microbial , Female , Gastritis/immunology , Helicobacter pylori/isolation & purification , Leukocyte Reduction Procedures , Macrophages/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Spleen/cytology , Stomach/cytology , Stomach/microbiology , Stomach/pathology
10.
Infect Immun ; 75(12): 5753-62, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17875635

ABSTRACT

Interleukin-12 (IL-12) and IL-18 are both central to the induction of gamma interferon (IFN-gamma), and various roles for IL-12 and IL-18 in control of intracellular microbial infections have been demonstrated. We used IL-12p40(-/-) and IL-18(-/-) mice to further investigate the role of IL-12 and IL-18 in control of Salmonella enterica serovar Typhimurium. While C57BL/6 and IL-18(-/-) mice were able to resolve attenuated S. enterica serovar Typhimurium infections, the IL-12p40(-/-) mice succumbed to a high bacterial burden after 60 days. Using ovalbumin (OVA)-specific T-cell receptor transgenic T cells (OT-II cells), we demonstrated that following oral infection with recombinant S. enterica serovar Typhimurium expressing OVA, the OT-II cells proliferated in the mesenteric lymph nodes of C57BL/6 and IL-18(-/-) mice but not in IL-12p40(-/-) mice. In addition, we demonstrated by flow cytometry that equivalent or increased numbers of T cells produced IFN-gamma in IL-12p40(-/-) mice compared with the numbers of T cells that produced IFN-gamma in C57BL/6 and IL-18(-/-) mice. Finally, we demonstrated that removal of macrophages from S. enterica serovar Typhimurium-infected C57BL/6 and IL-12p40(-/-) mice did not affect the bacterial load, suggesting that impaired control of S. enterica serovar Typhimurium infection in the absence of IL-12p40 is not due to reduced macrophage bactericidal activities, while IL-18(-/-) mice did rely on the presence of macrophages for control of the infection. Our results suggest that IL-12p40, but not IL-18, is critical to resolution of infections with attenuated S. enterica serovar Typhimurium and that especially the effects of IL-12p40 on proliferative responses of CD4+ T cells, but not the ability of these cells to produce IFN-gamma, are important in the resolution of infection by this intracellular bacterial pathogen.


Subject(s)
Interferon-gamma/immunology , Interleukin-12 Subunit p40/immunology , Interleukin-18/immunology , Salmonella typhi/immunology , Typhoid Fever/immunology , Animals , Antigen Presentation/immunology , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Epitopes, T-Lymphocyte/immunology , Female , Interferon-gamma/biosynthesis , Interleukin-12 Subunit p40/deficiency , Interleukin-18/deficiency , Macrophages/immunology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Receptors, Interferon/biosynthesis , Receptors, Interferon/immunology , Interferon gamma Receptor
11.
Int Immunol ; 19(3): 257-65, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17255112

ABSTRACT

The humoral response to the gastrointestinal (GI) flora was analyzed in secretory Ig (sIg)-deficient polymeric IgR (pIgR)(-/-) mice and otherwise congenic C57BL/6 mice. While both strains carried an ileal flora of similar size and composition, increased bacterial translocation to mesenteric lymph node was demonstrated in pIgR(-/-) mice. Serum IgA was greatly increased in pIgR(-/-) mice compared with C57BL/6 mice and reacted with commensal organisms and food. Serum IgG levels in pIgR(-/-) mice were increased to 6-fold above that of C57BL/6 mice and included specificities that bound to selected flora antigens. The enhanced recognition of flora antigens in pIgR(-/-) mice was explored using ovalbumin (OVA)-specific CD4(+) T cells and feeding of low concentrations of OVA. Increased proliferation of transgenic T cells was observed in pIgR(-/-) mice, relative to C57BL/6 mice, suggesting elevated net uptake of protein antigens from the GI tract in the absence of sIg. These studies suggest that there is increased recognition of GI flora antigens by systemic antibodies in pIgR(-/-) mice, most probably as a result of increased access of antigens from the GI flora to the systemic immune compartment, and support the hypothesis that a major function of the secretory immune system is to return environmental antigens to mucosal surfaces.


Subject(s)
Antigens, Bacterial/immunology , Bacteria/immunology , Gastrointestinal Tract/microbiology , Immunoglobulin A, Secretory/blood , Immunoglobulin G/blood , Receptors, Polymeric Immunoglobulin/immunology , Administration, Oral , Adoptive Transfer , Animals , Antibody Formation , Antigens, Bacterial/metabolism , Bacteria/classification , Bacterial Translocation , Female , Gastrointestinal Tract/metabolism , Ileum/microbiology , Intestinal Absorption , Lymph Nodes/microbiology , Lymphocyte Activation , Mesentery , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Ovalbumin/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
12.
J Immunol ; 175(6): 3810-8, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148127

ABSTRACT

A major hurdle in designing successful epitope-based vaccines resides in the delivery, stability, and immunogenicity of the peptide immunogen. The short-lived nature of unmodified peptide-based vaccines in vivo limits their therapeutic application in the immunotherapy of cancers and chronic viral infections as well as their use in generating prophylactic immunity. The incorporation of beta-amino acids into peptides decreases proteolysis, yet its potential application in the rational design of T cell mimotopes is poorly understood. To address this, we have replaced each residue of the SIINFEKL epitope individually with the corresponding beta-amino acid and examined the resultant efficacy of these mimotopes. Some analogs displayed similar MHC binding and superior protease stability compared with the native epitope. Importantly, these analogs were able to generate cross-reactive CTLs in vivo that were capable of lysing tumor cells that expressed the unmodified epitope as a surrogate tumor Ag. Structural analysis of peptides in which anchor residues were substituted with beta-amino acids revealed the basis for enhanced MHC binding and retention of immunogenicity observed for these analogs and paves the way for future vaccine design using beta-amino acids. We conclude that the rational incorporation of beta-amino acids into T cell determinants is a powerful alternative to the traditional homologous substitution of randomly chosen naturally occurring alpha-amino acids, and these mimotopes may prove particularly useful for inclusion in epitope-based vaccines.


Subject(s)
Drug Stability , Epitopes, T-Lymphocyte/chemistry , Vaccines, Synthetic/chemistry , Amino Acid Sequence , Amino Acids , Animals , Antigens, Neoplasm , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line , Cross-Priming , Epitopes, T-Lymphocyte/immunology , H-2 Antigens/immunology , Immunization , Male , Mice , Mice, Inbred C57BL , Molecular Mimicry , Peptide Hydrolases/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
13.
Infect Immun ; 73(3): 1714-22, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731072

ABSTRACT

Gene expression in murine dendritic cells (DCs) infected with green fluorescent protein-expressing Salmonella enterica serovar Typhimurium BRD509 was studied by mRNA differential display. Infected DCs were sorted from uninfected cells by flow cytometry. The mRNA expression patterns of infected and uninfected cells revealed a number of differentially expressed transcripts, which included the macrophage-derived chemokine (MDC). Up-regulation of MDC transcription in infected DCs was confirmed by Northern blotting, and the kinetics of MDC expression was examined by real-time reverse transcription-PCR, with which 31- and 150-fold increases were detected at 2 and 6 h postinfection, respectively. The increased release by DCs of MDC into culture media was detected by an enzyme-linked immunosorbent assay. The biological activity of MDC was investigated in in vitro and in vivo assays. In vitro, supernatants from S. enterica serovar Typhimurium-infected DCs were chemoattractive to T cells, and neutralization of MDC in these supernatants inhibited T-cell migration. Passive transfer of anti-MDC antibody to mice infected with BRD509 revealed that neither growth of the bacterium nor resistance of the mice to reinfection was affected and that in vivo inhibition of MDC did not affect T-cell responses, as measured by the gamma interferon ELISPOT method 3 days after challenge infection.


Subject(s)
Chemokines, CC/metabolism , Dendritic Cells/microbiology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/pathogenicity , Up-Regulation , Animals , Antibodies/administration & dosage , Antibodies/blood , Antibodies/immunology , Chemokine CCL22 , Chemokines, CC/genetics , Chemokines, CC/immunology , Dendritic Cells/immunology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neutralization Tests , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections, Animal/microbiology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...