Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Immunol ; 202(5): 1397-1405, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30692213

ABSTRACT

In chronic lymphocytic leukemia (CLL), signaling through several prosurvival B cell surface receptors activates the PI3K signaling pathway. Idelalisib is a highly selective PI3K (PI3Kδ) isoform-specific inhibitor effective in relapsed/refractory CLL and follicular lymphoma. However, severe autoimmune adverse effects in association with the use of idelalisib in the treatment of CLL, particularly as a first-line therapy, gave indications that idelalisib may preferentially target the suppressive function of regulatory T cells (Tregs). On this background, we examined the effect of idelalisib on the function of human Tregs ex vivo with respect to proliferation, TCR signaling, phenotype, and suppressive function. Our results show that human Tregs are highly susceptible to PI3Kδ inactivation using idelalisib compared with CD4+ and CD8+ effector T cells (Teffs) as evident from effects on anti-CD3/CD28/CD2-induced proliferation (order of susceptibility [IC50]: Treg [.5 µM] > CD4+ Teff [2.0 µM] > CD8+ Teff [6.5 µM]) and acting at the level of AKT and NF-κB phosphorylation. Moreover, idelalisib treatment of Tregs altered their phenotype and reduced their suppressive function against CD4+ and CD8+ Teffs. Phenotyping Tregs from CLL patients treated with idelalisib supported our in vitro findings. Collectively, our data show that human Tregs are more dependent on PI3Kδ-mediated signaling compared with CD4+ and CD8+ Teffs. This Treg-preferential effect could explain why idelalisib produces adverse autoimmune effects by breaking Treg-mediated tolerance. However, balancing effects on Treg sensitivity versus CD8+ Teff insensitivity to idelalisib could still potentially be exploited to enhance inherent antitumor immune responses in patients.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Quinazolinones/pharmacology , T-Lymphocytes, Regulatory/drug effects , Aged , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Structure-Activity Relationship , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
2.
Oncoimmunology ; 5(4): e1102828, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141387

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is highly infiltrated by CD4+T cells that express RORγt and IL-17 (TH17). Compelling evidence from the tumor microenvironment suggest that regulatory T cells (Treg) contribute to TH17 mediated inflammation. Concurrently, PDAC patients have elevated levels of pro-inflammatory cytokines that may lead to TH17 associated functional plasticity in Treg. In this study, we investigated the phenotype and functional properties of Treg in patients with PDAC. We report that PDAC patients have elevated frequency of FOXP3+Treg, which exclusively occurred within the FOXP3+RORγt+Treg compartment. The FOXP3+RORγt+Treg retained FOXP3+Treg markers and represented an activated subset. The expression of RORγt in Treg may indicate a phenotypic switch toward TH17 cells. However, the FOXP3+RORγt+Treg produced both TH17 and TH2 associated pro-inflammatory cytokines, which corresponded with elevated TH17 and TH2 immune responses in PDAC patients. Both the FOXP3+Treg and FOXP3+RORγt+Treg from PDAC patients strongly suppressed T cell immune responses, but they had impaired anti-inflammatory properties. We conclude that FOXP3+RORγt+Treg have a dual phenotype with combined pro-inflammatory and immunosuppressive activity, which may be involved in the pathogenesis of PDAC.

3.
J Leukoc Biol ; 100(1): 5-16, 2016 07.
Article in English | MEDLINE | ID: mdl-26715685

ABSTRACT

Human CD4(+)CD25(hi)FOXP3(+) regulatory T cells maintain immunologic tolerance and prevent autoimmune and inflammatory immune responses. Regulatory T cells undergo a similar activation cycle as conventional CD4(+) T cells upon antigen stimulation. Here, we demonstrate that T cell receptors and costimulation are required to activate the regulatory T cell suppressive function. Regulatory T cells suppressed the T cell receptor signaling in effector T cells in a time-dependent manner that corresponded with inhibition of cytokine production and proliferation. Modulation of the activation level and thereby the suppressive capacity of regulatory T cells imposed distinct T cell receptor signaling signatures and hyporesponsiveness in suppressed and proliferating effector T cells and established a threshold for effector T cell proliferation. The immune suppression of effector T cells was completely reversible upon removal of regulatory T cells. However, the strength of prior immune suppression by regulatory T cells and corresponding T cell receptor signaling in effector T cells determined the susceptibility to suppression upon later reexposure to regulatory T cells. These findings demonstrate how the strength of the regulatory T cell suppressive function determines intracellular signaling, immune responsiveness, and the later susceptibility of effector T cells to immune suppression and contribute to unveiling the complex interactions between regulatory T cells and effector T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Immunosuppression Therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Lymphocyte Activation , Signal Transduction
4.
Biochem J ; 460(3): 399-410, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24665965

ABSTRACT

Full T-cell activation critically depends on the engagement of the TCR (T-cell receptor) in conjunction with a second signal by co-stimulatory receptors that boost the immune response. In the present study we have compared signalling patterns induced by the two co-receptors CD2 and CD28 in human peripheral blood T-cells. These co-receptors were previously suggested to be redundant in function. By a combination of multi-parameter phosphoflow cytometry, phosphokinase arrays and Western blot analyses, we demonstrate that CD2 co-stimulation induces phosphorylation of the TCR-proximal signalling complex, whereas CD28 activates distal signalling molecules, including the transcription factors NF-κB (nuclear factor κB), ATF (activating transcription factor)-2, STAT3/5 (signal transducer and activator of transcription 3/5), p53 and c-Jun. These signalling patterns were conserved in both naïve and effector/memory T-cell subsets. We show that free intracellular Ca(2+) and signalling through the PI3K (phosphoinositide 3-kinase)/Akt pathway are required for proper CD28-induced NF-κB activation. The signalling patterns induced by CD2 and CD28 co-stimulation lead to distinct functional immune responses in T-cell proliferation and cytokine production. In conclusion, CD2 and CD28 co-stimulation induces distinct signalling responses and functional outcomes in T-cells.


Subject(s)
CD2 Antigens/immunology , CD28 Antigens/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/physiology , Activating Transcription Factor 2/metabolism , Calcium/metabolism , Humans , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
5.
Mol Cancer ; 10: 149, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22168384

ABSTRACT

BACKGROUND: The adenomatous polyposis coli (APC) protein is part of the destruction complex controlling proteosomal degradation of ß-catenin and limiting its nuclear translocation, which is thought to play a gate-keeping role in colorectal cancer. The destruction complex is inhibited by Wnt-Frz and prostaglandin E(2) (PGE(2)) - PI-3 kinase pathways. Recent reports show that PGE(2)-induced phosphorylation of ß-catenin by protein kinase A (PKA) increases nuclear translocation indicating two mechanisms of action of PGE(2) on ß-catenin homeostasis. FINDINGS: Treatment of Apc(Min/+) mice that spontaneously develop intestinal adenomas with a PKA antagonist (Rp-8-Br-cAMPS) selectively targeting only the latter pathway reduced tumor load, but not the number of adenomas. Immunohistochemical characterization of intestines from treated and control animals revealed that expression of ß-catenin, ß-catenin nuclear translocation and expression of the ß-catenin target genes c-Myc and COX-2 were significantly down-regulated upon Rp-8-Br-cAMPS treatment. Parallel experiments in a human colon cancer cell line (HCT116) revealed that Rp-8-Br-cAMPS blocked PGE(2)-induced ß-catenin phosphorylation and c-Myc upregulation. CONCLUSION: Based on our findings we suggest that PGE(2) act through PKA to promote ß-catenin nuclear translocation and tumor development in Apc(Min/+) mice in vivo, indicating that the direct regulatory effect of PKA on ß-catenin nuclear translocation is operative in intestinal cancer.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/pathology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclooxygenase 2/genetics , Proto-Oncogene Proteins c-myc/genetics , beta Catenin/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Active Transport, Cell Nucleus , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Down-Regulation , HCT116 Cells , Humans , Mice , Proto-Oncogene Proteins c-myc/metabolism , Thionucleotides/pharmacology
6.
J Virol ; 85(13): 6557-66, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490090

ABSTRACT

Chronic HIV infection is characterized by chronic immune activation and dysfunctional T cells with elevated intracellular cyclic AMP (cAMP), which inhibits the T cell activation capability. cAMP may be induced by prostaglandin E(2) following lipopolysaccharide (LPS)-induced upregulation of cyclooxygenase type 2 (COX-2) in monocytes due to the elevated LPS levels in patients with chronic HIV infection. This hypothesis was tested using celecoxib, a COX-2 inhibitor, for 12 weeks in HIV-infected patients without antiretroviral treatment in a prospective, open, randomized exploratory trial. Thirty-one patients were randomized in the trial; 27 completed the study, including 13 patients on celecoxib. Celecoxib reduced chronic immune activation in terms of CD38 density on CD8(+) T cells (-24%; P = 0.04), IgA levels (P = 0.04), and a combined score for inflammatory markers (P < 0.05). Celecoxib further reduced the inhibitory surface receptor programmed death 1 (PD-1) on CD8(+) T cells (P = 0.01), including PD-1 on the HIV Gag-specific subset (P = 0.02), enhanced the number of CD3(+) CD4(+) CD25(+) CD127(lo/-) Treg or activated cells (P = 0.02), and improved humoral memory recall responses to a T cell-dependent vaccine (P = 0.04). HIV RNA (P = 0.06) and D dimers (P = 0.07) tended to increase in the controls, whereas interleukin-6 (IL-6) possibly decreased in the treatment arm (P = 0.10). In conclusion, celecoxib downmodulated the immune activation related to clinical progression of chronic HIV infection and improved T cell-dependent functions in vivo.


Subject(s)
AIDS Vaccines/immunology , Cyclooxygenase 2 Inhibitors/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/drug effects , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , T-Lymphocytes/immunology , AIDS Vaccines/administration & dosage , Adult , Celecoxib , Chronic Disease , Cyclooxygenase 2 Inhibitors/administration & dosage , Disease Progression , Down-Regulation , Female , HIV-1/immunology , Humans , Male , Middle Aged , Pyrazoles/administration & dosage , Sulfonamides/administration & dosage , Treatment Outcome
7.
Eur J Immunol ; 40(1): 134-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19877008

ABSTRACT

The complexity of immunoregulation has focused attention on the CD4+ T "suppressor" regulatory cell (Treg), which helps maintain balance between immunity and tolerance. An immunoregulatory T-cell population that upon activation amplifies cellular immune responses was described in murine models more than 30 years ago; however, no study has yet identified a naturally occurring T "inducer" cell type. Here, we report that the ectoenzyme CD39/NTPDase1 (ecto-nucleoside triphosphate diphosphohydrolase 1) helps to delineate a novel population of human "inducer" CD4+ T cells (Tind) that significantly increases the proliferation and cytokine production of responder T cells in a dose-dependent manner. Furthermore, this unique Tind subset produces a distinct repertoire of cytokines in comparison to the other CD4+ T-cell subsets. We propose that this novel CD4+ T-cell population counterbalances the suppressive activity of suppressor Treg in peripheral blood and serves as a calibrator of immunoregulation.


Subject(s)
Antigens, CD/immunology , Apyrase/immunology , CD4-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Cytokines/biosynthesis , Cytokines/immunology , Humans , T-Lymphocyte Subsets/cytology
8.
Eur J Immunol ; 39(5): 1280-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19384871

ABSTRACT

Recent studies have indicated that Treg contribute to the HIV type 1 (HIV-1)-related immune pathogenesis. However, it is not clear whether T cells with suppressive properties reside within the HIV-1-specific T-cell population. Here, PBMC from HIV-1-infected individuals were stimulated with a 15-mer Gag peptide pool, and HIV-1-specific T cells were enriched by virtue of their secretion of IL-10 or IFN-gamma using immunomagnetic cell-sorting. Neither the IL-10-secreting cells nor the IFN-gamma-secreting cells expressed the Treg marker FOXP3, yet the IL-10-secreting cells potently suppressed anti-CD3/CD28-induced CD4(+) as well as CD8(+) T-cell proliferative responses. As shown by intracellular cytokine staining, IL-10- and IFN-gamma-producing T cells represent distinct subsets of the HIV-1-specific T cells. Our data collectively suggest that functionally defined HIV-1-specific T-cell subsets harbor potent immunoregulatory properties that may contribute to HIV-1-associated T-cell dysfunction.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Interleukin-10/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Cell Proliferation , Flow Cytometry , Forkhead Transcription Factors/blood , Forkhead Transcription Factors/immunology , HIV Infections/blood , Humans , Immunomagnetic Separation , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-10/blood , Leukocytes, Mononuclear/immunology , Statistics, Nonparametric , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology
10.
J Leukoc Biol ; 83(5): 1111-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18270250

ABSTRACT

Human CD4+CD25+ regulatory T (T(R)) cells express the transcription factor forkhead box p3 (FOXP3) and have potent immunosuppressive properties. While naturally occurring T(R) cells develop in the thymus, adaptive T(R) cells develop in the periphery from naive CD4+ T cells. Adaptive T(R) cells may express cyclooxygenase type 2 (COX-2) and suppress effector T cells by a PGE(2)-dependent mechanism, which is reversible with COX inhibitors. In this study we have characterized the differentiation of naive CD4+ T cells into adaptive T(R) cells in detail during 7 days of continuous antigen stimulation. After 2 days of stimulation of CD4+CD25- T cells, the cells expressed FOXP3 and COX-2 and displayed potent immunosuppressive properties. The suppressive phenotype was present at all observed time-points from Day 2, although suppression was merely present at Day 7. The adaptive T(R) cells expressed cell surface markers consistent with an activated phenotype and secreted high levels of TGF-beta, IL-10, and PGE(2). However, the suppressive phenotype was found exclusively in cells that proliferated upon activation. These data support the notion that activation of naive CD4+ T cells leads to concomitant acquisition of effector and suppressive properties.


Subject(s)
CD4 Antigens/analysis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/analysis , Interleukin-2 Receptor alpha Subunit/analysis , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Bacterial/pharmacology , Blood Donors , CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation , Cell Division , Coculture Techniques , Enterotoxins/pharmacology , Flow Cytometry , Forkhead Transcription Factors/genetics , Humans , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
11.
Int Immunol ; 20(2): 235-45, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18156149

ABSTRACT

Monocytes initiate innate immune responses and interact with T cells to induce antigen-specific immune responses by antigen presentation and secretion of humoral factors. We have previously shown that adaptive regulatory T cells inhibit T-cell effector functions in a cyclooxygenase (COX)-2-prostaglandin E(2) (PGE(2))-dependent manner and that PGE(2) converts resting CD4+CD25- T cells into FOXP3+ T cells with a suppressive phenotype. Here, we demonstrate that stimulation of monocytes with LPS leads to suppression of T-cell immune responses by a COX-2-PGE(2)-dependent mechanism that is reversible with COX-2 inhibitors as well as PGE(2)-neutralizing antibody and cAMP antagonist. Furthermore, we show that LPS-activated monocytes induce FOXP3 expression in resting CD4+CD25- T cells by the same pathway. These results suggest that monocytes are able to efficiently suppress T-cell immune responses in a regulatory manner and elicit an inhibitory immune profile.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Forkhead Transcription Factors/metabolism , Lipopolysaccharides/pharmacology , Monocytes/immunology , T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cytokines/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/deficiency , Lymphocyte Activation/immunology
12.
Cancer Immunol Immunother ; 57(6): 813-21, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17962941

ABSTRACT

OBJECTIVE: Naturally occurring regulatory T (T(R)) cells suppress autoreactive T cells whereas adaptive T(R) cells, induced in the periphery, play an important role in chronic viral diseases and cancer. Several studies indicate that cyclooxygenase (COX) inhibitors prevent cancer development of colon adenomas and delay disease progression in patients with colorectal cancer (CRC). We have shown that adaptive T(R) cells express COX-2 and produce PGE(2) that suppress effector T cells in a manner that is reversed by COX-inhibitors. METHODS AND RESULTS: Here we demonstrate that CRC patients have elevated levels of PGE(2) in peripheral blood, and CRC tissue samples and draining lymph nodes display increased numbers of FOXP3+ T(R) cells. Depletion of T(R) cells from PBMC enhanced anti-tumor T-cell responses to peptides from carcinoembryonic antigen. Furthermore, the COX inhibitor indomethacin and the PKA type I antagonist Rp-8-Br-cAMPS significantly improved the anti-tumor immune activity. CONCLUSION: We suggest that adaptive T(R) cells contribute to an immunosuppressive microenvironment in CRC and inhibit effector T cells by a COX-2-PGE(2)-dependent mechanism and thereby facilitate tumor growth. Therapeutic strategies targeting T(R) cells and the PGE(2)-cAMP pathway may be interesting to pursue to enhance anti-tumor immune activity in CRC patients.


Subject(s)
Colorectal Neoplasms/immunology , Cyclooxygenase 2/metabolism , Immunotherapy/methods , T-Lymphocytes, Regulatory/cytology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/pathology , Dinoprostone/metabolism , Female , Humans , Lymph Nodes/pathology , Male , Middle Aged , Models, Biological , T-Lymphocytes, Regulatory/immunology
13.
J Immunol ; 179(8): 5159-68, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911601

ABSTRACT

cAMP negatively regulates T cell immune responses by activation of type I protein kinase A (PKA), which in turn phosphorylates and activates C-terminal Src kinase (Csk) in T cell lipid rafts. Using yeast two-hybrid screening, far-Western blot, immunoprecipitation and immunofluorescense analyses, and small interfering RNA-mediated knockdown, we identified Ezrin as the A-kinase anchoring protein that targets PKA type I to lipid rafts. Furthermore, Ezrin brings PKA in proximity to its downstream substrate Csk in lipid rafts by forming a multiprotein complex consisting of PKA/Ezrin/Ezrin-binding protein 50, Csk, and Csk-binding protein/phosphoprotein associated with glycosphingolipid-enriched microdomains. The complex is initially present in immunological synapses when T cells contact APCs and subsequently exits to the distal pole. Introduction of an anchoring disruptor peptide (Ht31) into T cells competes with Ezrin binding to PKA and thereby releases the cAMP/PKA type I-mediated inhibition of T cell proliferation. Finally, small interfering RNA-mediated knockdown of Ezrin abrogates cAMP regulation of IL-2. We propose that Ezrin is essential in the assembly of the cAMP-mediated regulatory pathway that modulates T cell immune responses.


Subject(s)
A Kinase Anchor Proteins/physiology , Cyclic AMP-Dependent Protein Kinase Type I/metabolism , Cyclic AMP/pharmacology , Cytoskeletal Proteins/physiology , Immunosuppressive Agents/pharmacology , Membrane Microdomains/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , A Kinase Anchor Proteins/chemistry , A Kinase Anchor Proteins/metabolism , Actins/metabolism , Cells, Cultured , Cloning, Molecular , Cyclic AMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Type I/genetics , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Cytoskeleton/immunology , Cytoskeleton/metabolism , Humans , Jurkat Cells , Membrane Microdomains/metabolism , Peptide Mapping , Phosphoproteins/physiology , Protein Binding , RNA, Small Interfering/pharmacology , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Sodium-Hydrogen Exchangers/physiology , T-Lymphocytes/enzymology
14.
J Immunol ; 177(1): 246-54, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16785520

ABSTRACT

CD4+CD25+ regulatory T (T(R)) cells suppress effector T cells by partly unknown mechanisms. In this study, we describe a population of human suppressive CD4+CD25+ adaptive T(R) (T(R)(adapt)) cells induced in vitro that express cyclooxygenase 2 (COX-2) and the transcription factor FOXP3. T(R)(adapt) cells produce PGE(2) and suppress effector T cell responses in a manner that is reversed by COX inhibitors and PGE(2) receptor-specific antagonists. In resting CD4+CD25- T cells, treatment with PGE(2) induced FOXP3 expression. Thus, autocrine and paracrine effects of PGE(2) produced by COX-2-positive T(R)(adapt) cells may be responsible for both the FOXP3+ phenotype and the mechanism used by these cells to suppress effector T cells.


Subject(s)
Cyclooxygenase 2/biosynthesis , Dinoprostone/physiology , Forkhead Transcription Factors/biosynthesis , Receptors, Interleukin-2/biosynthesis , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Cell Communication/immunology , Cells, Cultured , Cyclic AMP/biosynthesis , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Enterotoxins/pharmacology , Forkhead Transcription Factors/physiology , Humans , Immunity, Innate , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/immunology , Prostaglandin Antagonists/physiology , Receptors, Prostaglandin E/antagonists & inhibitors , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/microbiology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology
15.
Microbes Infect ; 7(7-8): 1063-5, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15893493

ABSTRACT

The immune system faces the difficult task of discerning between foreign, potentially pathogen-derived antigens and self-antigens. Several mechanisms, including deletion of self-reactive T cells in the thymus, have been shown to contribute to the acceptance of self-antigens and the reciprocal reactivity to foreign antigens. Over the last decade it has become increasingly clear that CD4(+)CD25(+) T(Reg) cells are crucial for maintenance of T cell tolerance to self-antigens in the periphery, and to avoid development of autoimmune disorders. Recently, evidence has also emerged that demonstrates that CD4(+)CD25(+) T(Reg) cells can also suppress T cell responses to foreign pathogens, including viruses such as HIV. In this article we review the current knowledge and potential role of CD4(+)CD25(+) T(Reg) cells in HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , HIV Infections/immunology , Receptors, Interleukin-2/physiology , CD4-Positive T-Lymphocytes/virology , Humans
16.
Blood ; 104(12): 3672-8, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15308569

ABSTRACT

The antiviral response of CD8 T cells involves the differentiation of naive T cells into distinct types of effector and memory cells, which may be distinguished by the level of CD7 expression. We have investigated CD8 T cells in adults and children infected with HIV-1 to determine the disease relevance of cell subsets defined by CD7. CD8 T cells from patients infected with HIV-1 displayed profound down-modulation of CD7 expression as compared with healthy subjects, with expansion of both CD7(low) and CD7(negative) effector subsets. Loss of CD7(high) cells correlated directly with HIV-1 load and was particularly pronounced in patients with rapid disease progression. CD8 T cells specific for HIV-1, as well as Epstein-Barr virus (EBV) and cytomegalovirus (CMV) were predominantly found in the CD7(low) effector cell subset. Furthermore, recovery of CD4 counts on antiretroviral therapy was associated with reversion of the skewed CD7 profile in CD8 T cells. Thus, effector CD8 T-cell subsets distinguished by lowered CD7 expression expand in a manner that correlates with the magnitude of HIV-1, EBV, and CMV antigenic challenge and contract in response to successful antiretroviral treatment. The results are discussed in relation to the dual roles of CD7 as a receptor of both costimulation and cell death.


Subject(s)
Anti-Retroviral Agents/pharmacology , Antigens, CD7/analysis , CD8-Positive T-Lymphocytes/cytology , HIV Infections/immunology , Adult , Anti-Retroviral Agents/therapeutic use , Antigens, Viral/immunology , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Case-Control Studies , Cell Proliferation , Disease Progression , HIV Infections/drug therapy , Humans , Middle Aged , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , Viral Load
18.
J Virol ; 78(5): 2454-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14963140

ABSTRACT

Regulatory T (T(R)) cells maintain tolerance to self-antigens and control immune responses to alloantigens after organ transplantation. Here, we show that CD4(+) CD25(+) human T(R) cells suppress virus-specific T-cell responses. Depletion of T(R) cells from peripheral blood mononuclear cells enhances T-cell responses to cytomegalovirus and human immunodeficiency virus antigens. We propose that chronic viral infections lead to induction of suppressive T(R) cells that inhibit the antiviral immune response.


Subject(s)
Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytomegalovirus/immunology , HIV/immunology , Receptors, Interleukin-2/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation , Superantigens/immunology
19.
J Biol Chem ; 278(19): 17573-9, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12621056

ABSTRACT

Forkhead/winged helix (FOX) transcription factors are essential for control of the cell cycle and metabolism. Here, we show that spleens from Mf2-/- (FOXD2-/-) mice have reduced mRNA (50%) and protein (35%) levels of the RIalpha subunit of the cAMP-dependent protein kinase. In T cells from Mf2-/- mice, reduced levels of RIalpha translates functionally into approximately 2-fold less sensitivity to cAMP-mediated inhibition of proliferation triggered through the T cell receptor-CD3 complex. In Jurkat T cells, FOXD2 overexpression increased the endogenous levels of RIalpha through induction of the RIalpha1b promoter. FOXD2 overexpression also increased the sensitivity of the promoter to cAMP. Finally, co-expression experiments demonstrated that protein kinase Balpha/Akt1 work together with FOXD2 to induce the RIalpha1b promoter (10-fold) and increase endogenous RIalpha protein levels further. Taken together, our data indicate that FOXD2 is a physiological regulator of the RIalpha1b promoter in vivo working synergistically with protein kinase B to induce cAMP-dependent protein kinase RIalpha expression, which increases cAMP sensitivity and sets the threshold for cAMP-mediated negative modulation of T cell activation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Trans-Activators/metabolism , B-Lymphocytes/metabolism , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit , Forkhead Transcription Factors , Humans , Jurkat Cells , Lymphocyte Activation/drug effects
20.
J Immunol ; 170(5): 2349-55, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12594257

ABSTRACT

The adaptive immune response of human CD8 T cells to invading pathogens involves the differentiation of naive cells into memory and effector cells. However, the lineage relationship between memory and effector cells and the differentiation of CD8 T cells into distinct subsets of effector cell subpopulations are subjects of considerable debate. CD7 identifies three populations of CD8 T cells: CD7 high (CD7(high)), low (CD7(low)), and negative (CD7(neg)) that translate into subsets with distinct functional properties. The CD7(high) subset contains naive and memory cells and the CD7(low) and CD7(neg) subsets contain effector cells. The effector cells can functionally be divided into cytokine-secreting effector CD8 T cells and lytic effector CD8 T cells. These data provide a model of human CD8 T cell differentiation in which specialized distinct subpopulations can be identified by expression of CD7.


Subject(s)
Antigens, CD7/analysis , CD8-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , Antigens, CD7/biosynthesis , Biomarkers/analysis , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Division/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Down-Regulation/immunology , Epitopes, T-Lymphocyte/analysis , Humans , Immunologic Memory , Immunophenotyping , Interphase/immunology , Lymphocyte Count , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...