Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
J Immunol ; 181(3): 2056-64, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641343

ABSTRACT

IFN-gamma production by T cells is pivotal for defense against many pathogens, and the proximal promoter of IFN-gamma, -73 to -48 bp upstream of the transcription start site, is essential for its expression. However, transcriptional regulation mechanisms through this promoter in primary human cells remain unclear. We studied the effects of cAMP response element binding protein/activating transcription factor (CREB/ATF) and AP-1 transcription factors on the proximal promoter of IFN-gamma in human T cells stimulated with Mycobacterium tuberculosis. Using EMSA, supershift assays, and promoter pulldown assays, we demonstrated that CREB, ATF-2, and c-Jun, but not cyclic AMP response element modulator, ATF-1, or c-Fos, bind to the proximal promoter of IFN-gamma upon stimulation, and coimmunoprecipitation indicated the possibility of interaction among these transcription factors. Chromatin immunoprecipitation confirmed the recruitment of these transcription factors to the IFN-gamma proximal promoter in live Ag-activated T cells. Inhibition of ATF-2 activity in T cells with a dominant-negative ATF-2 peptide or with small interfering RNA markedly reduced the expression of IFN-gamma and decreased the expression of CREB and c-Jun. These findings suggest that CREB, ATF-2, and c-Jun are recruited to the IFN-gamma proximal promoter and that they up-regulate IFN-gamma transcription in response to microbial Ag. Additionally, ATF-2 controls expression of CREB and c-Jun during T cell activation.


Subject(s)
Activating Transcription Factor 2/metabolism , Antigens, Bacterial/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Interferon-gamma/metabolism , T-Lymphocytes/metabolism , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/pharmacology , CD3 Complex/immunology , Cell Survival , Cells, Cultured , Electrophoretic Mobility Shift Assay , Humans , Interferon-gamma/genetics , Mycobacterium tuberculosis/immunology , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Small Interfering/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Eur J Immunol ; 38(2): 459-69, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18203140

ABSTRACT

We evaluated the role of regulatory T cells (CD4(+) CD25(+) Foxp3(+) cells, Tregs) in human Mycobacterium tuberculosis infection. Tregs were expanded in response to M. tuberculosis in healthy tuberculin reactors, but not in tuberculin-negative individuals. The M. tuberculosis mannose-capped lipoarabinomannan (ManLAM) resulted in regulatory T cell expansion, whereas the M. tuberculosis 19-kDa protein and heat shock protein 65 had no effect. Anti-IL-10 and anti-TGF-beta alone or in combination, did not reduce expansion of Tregs. In contrast, the cyclooxygenase enzyme-2 inhibitor NS398 significantly inhibited expansion of Tregs, indicating that prostaglandin E2 (PGE2) contributes to Treg expansion. Monocytes produced PGE2 upon culturing with heat-killed M. tuberculosis or ManLAM, and T cells from healthy tuberculin reactors enhanced PGE2 production by monocytes. Expanded Tregs produced significant amounts of TGF-beta and IL-10 and depletion of Tregs from PBMC of these individuals increased the frequency of M. tuberculosis-responsive CD4(+) IFN-gamma cells. Culturing M. tuberculosis-expanded Tregs with autologous CD8(+) cells decreased the frequency of IFN-gamma(+)cells. Freshly isolated PBMC from tuberculosis patients had increased percentages of Tregs, compared to healthy tuberculin reactors. These findings demonstrate that Tregs expand in response to M. tuberculosis through mechanisms that depend on ManLAM and PGE2.


Subject(s)
Cell Proliferation , Dinoprostone/physiology , Lipopolysaccharides/pharmacology , Mannose/physiology , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Antigens, Bacterial/pharmacology , Cells, Cultured , Dinoprostone/biosynthesis , Growth Inhibitors/pharmacology , Growth Inhibitors/physiology , Humans , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Mannose/chemistry , Monocytes/immunology , Monocytes/microbiology , T-Lymphocytes, Regulatory/cytology , Tuberculosis/immunology , Tuberculosis/pathology
4.
J Immunol ; 174(10): 6357-63, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879136

ABSTRACT

IFN-gamma is essential for resistance to many intracellular pathogens, including Mycobacterium tuberculosis. Transcription of the IFN-gamma gene in activated T cells is controlled by the proximal promoter element (-73 to -48 bp). CREB binds to the IFN-gamma proximal promoter, and binding is enhanced by phosphorylation of CREB. Studies in human T cell lines and in transgenic mice have yielded conflicting results about whether CREB is a positive or a negative regulator of IFN-gamma transcription. To determine the role of CREB in mediating IFN-gamma production in response to a microbial pathogen, we evaluated the peripheral blood T cell response to M. tuberculosis in healthy tuberculin reactors. EMSAs, chromatin immunoprecipitation, and Western blotting demonstrated that stimulation of PBMC with M. tuberculosis induced phosphorylation and enhanced binding of CREB to the IFN-gamma proximal promoter. Neutralization of CREB with intracellular Abs or down-regulation of CREB levels with small interfering RNA decreased M. tuberculosis-induced production of IFN-gamma and IFN-gamma mRNA expression. In addition, M. tuberculosis-stimulated T cells from tuberculosis patients, who have ineffective immunity, showed diminished IFN-gamma production, reduced amounts of CREB binding to the IFN-gamma proximal promoter, and absence of phosphorylated CREB. These findings demonstrate that CREB positively regulates IFN-gamma production by human T cells that respond to M. tuberculosis.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Interferon-gamma/biosynthesis , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Up-Regulation/immunology , CREB-Binding Protein , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation/immunology , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Intracellular Fluid/microbiology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/microbiology , Mycobacterium tuberculosis/pathogenicity , Nuclear Proteins , Phosphorylation , Promoter Regions, Genetic/immunology , Protein Binding/immunology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Serine/metabolism , T-Lymphocytes/metabolism , Trans-Activators
5.
J Immunol ; 173(3): 1966-77, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15265931

ABSTRACT

The secreted Mycobacterium tuberculosis 10-kDa culture filtrate protein (CFP)10 is a potent T cell Ag that is recognized by a high percentage of persons infected with M. tuberculosis. We determined the molecular basis for this widespread recognition by identifying and characterizing a 15-mer peptide, CFP10(71-85), that elicited IFN-gamma production and CTL activity by both CD4(+) and CD8(+) T cells from persons expressing multiple MHC class II and class I molecules, respectively. CFP10(71-85) contained at least two epitopes, one of 10 aa (peptide T1) and another of 9 aa (peptide T6). T1 was recognized by CD4(+) cells in the context of DRB1*04, DR5*0101, and DQB1*03, and by CD8(+) cells of A2(+) donors. T6 elicited responses by CD4(+) cells in the context of DRB1*04 and DQB1*03, and by CD8(+) cells of B35(+) donors. Deleting a single amino acid from the amino or carboxy terminus of either peptide markedly reduced IFN-gamma production, suggesting that they are minimal epitopes for both CD4(+) and CD8(+) cells. As far as we are aware, these are the shortest microbial peptides that have been found to elicit responses by both T cell subpopulations. The capacity of CFP10(71-85) to stimulate IFN-gamma production and CTL activity by CD4(+) and CD8(+) cells from persons expressing a spectrum of MHC molecules suggests that this peptide is an excellent candidate for inclusion in a subunit antituberculosis vaccine.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HLA Antigens/immunology , Mycobacterium tuberculosis/immunology , Peptide Fragments/immunology , T-Cell Antigen Receptor Specificity , Alleles , Amino Acid Substitution , Bacterial Proteins/chemistry , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , Epitopes/chemistry , Epitopes/immunology , HLA-DQ Antigens/immunology , HLA-DQ beta-Chains , HLA-DR Antigens/immunology , HLA-DRB1 Chains , Humans , Interferon-gamma/biosynthesis , Peptide Fragments/chemistry , Sequence Deletion , Tuberculosis Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL