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










Publication year range
1.
Front Oncol ; 5: 58, 2015.
Article in English | MEDLINE | ID: mdl-25815272

ABSTRACT

Maintenance of immunological tolerance is a critical hallmark of the immune system. Several signaling checkpoints necessary to balance activating and inhibitory input to immune cells have been described so far, among which the E3 ligase Cbl-b appears to be a central player. Cbl-b is expressed in all leukocyte subsets and regulates several signaling pathways in T cells, NK cells, B cells, and different types of myeloid cells. In most cases, Cbl-b negatively regulates activation signals through antigen or pattern recognition receptors and co-stimulatory molecules. In line with this function, cblb-deficient immune cells display lower activation thresholds and cblb knockout mice spontaneously develop autoimmunity and are highly susceptible to experimental autoimmunity. Interestingly, genetic association studies link CBLB-polymorphisms with autoimmunity also in humans. Vice versa, the increased activation potential of cblb-deficient cells renders them more potent to fight against malignancies or infections. Accordingly, several reports have shown that cblb knockout mice reject tumors, which mainly depends on cytotoxic T and NK cells. Thus, targeting Cbl-b may be an interesting strategy to enhance anti-cancer immunity. In this review, we summarize the findings on the molecular function of Cbl-b in different cell types and illustrate the potential of Cbl-b as target for immunomodulatory therapies.

3.
PLoS One ; 8(6): e65178, 2013.
Article in English | MEDLINE | ID: mdl-23762309

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells with a promising potential in cancer immunotherapy. Cbl proteins are E3 ubiquitin ligases and have been implicated in regulating the functional activity of various immune cells. As an example, c-Cbl negatively affects DC activation. We here describe that another member of the Cbl-protein family (i.e. Cbl-b) is highly expressed in murine bone-marrow-derived DCs (BMDCs). Differentiation of cblb-/- bone marrow mononuclear cells into classical BMDCs is unaltered, except enhanced induction of DEC-205 (CD205) expression. When tested in mixed-lymphocyte reaction (MLR), cblb-/- BMDCs exhibit increased allo-stimulatory capacity in vitro. BMDCs were next in vitro stimulated by various toll like receptor (TLR)-agonists (LPS, Poly(I:C), CpG) and exposed to FITC-labeled dextran. Upon TLR-stimulation, cblb-/- BMDCs produce higher levels of proinflammatory cytokines (IL-1α, IL-6 and TNF-α) and exhibit a slightly higher level of FITC-dextran uptake. To further characterize the functional significance of cblb-/- BMDCs we tested them in antigen-specific T cell responses against ovalbumin (OVA) protein and peptides, activating either CD8(+) OT-I or CD4(+) OT-II transgenic T cells. However, cblb-/- BMDCs are equally effective in inducing antigen-specific T cell responses when compared to wildtype BMDCs both in vitro and in vivo. The migratory capacity into lymph nodes during inflammation was similarly not affected by the absence of Cbl-b. In line with these observations, cblb-/- peptide-pulsed BMDCs are equally effective vaccines against OVA-expressing B16 tumors in vivo when compared to wildtype BMDCs. We conclude that in contrast to c-Cbl, Cbl-b plays only a limited role in the induction of Ag-specific T cell responses by murine BMDCs in vitro and in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Proto-Oncogene Proteins c-cbl/immunology , Ubiquitin-Protein Ligases/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Biological Transport , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dextrans/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Gene Expression , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Culture Test, Mixed , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Ovalbumin/immunology , Peptides/pharmacology , Poly I-C/pharmacology , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
4.
Immunol Lett ; 151(1-2): 31-8, 2013 03.
Article in English | MEDLINE | ID: mdl-23439007

ABSTRACT

The physiological functions of PKCα and PKCθ isotypes downstream of the antigen receptor have been defined in CD3(+) T cells. In contrast, no function of the second conventional PKC member, PKCß, has been described yet in T cell antigen receptor signalling. To investigate the hypothesis that both conventional PKCα and PKCß isotypes may have overlapping functions in T cell activation signalling, we generated mice that lacked the genes for both isotypes. We found that PKCα(-/-)/ß(-/-) animals are viable, live normal life spans and display normal T cell development. However, these animals possess additive defects in T cell responses in comparison to animals that carry single mutations in these genes. Our studies demonstrate that the activities of PKCα and PKCß converge to regulate IL-2 cytokine responses in anti-CD3 stimulated primary mouse T cells. Here, we present genetic evidence that PKCα and PKCß cooperate in IL-2 transcriptional transactivation in primary mouse T cells independently of the actions of PKCθ.


Subject(s)
CD3 Complex/immunology , Interleukin-2/genetics , Protein Kinase C-alpha/metabolism , Protein Kinase C/metabolism , RNA, Messenger/genetics , Transcription, Genetic , Animals , CD3 Complex/metabolism , Immunologic Memory , Immunophenotyping , Interleukin-2/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Phenotype , Protein Kinase C/genetics , Protein Kinase C beta , Protein Kinase C-alpha/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription Factor AP-1/metabolism , Transcriptional Activation
5.
PLoS One ; 8(1): e53709, 2013.
Article in English | MEDLINE | ID: mdl-23335970

ABSTRACT

In T cells PKCθ mediates the activation of critical signals downstream of TCR/CD28 stimulation. We investigated the molecular mechanisms by which PKCθ regulates NFκB transactivation by examining PKCθ/ß single and double knockout mice and observed a redundant involvement of PKCθ and PKCß in this signaling pathway. Mechanistically, we define a PKCθ-CYLD protein complex and an interaction between the positive PKCθ/ß and the negative CYLD signaling pathways that both converge at the level of TAK1/IKK/I-κBα/NFκB and NFAT transactivation. In Jurkat leukemic T cells, CYLD is endoproteolytically processed in the initial minutes of stimulation by the paracaspase MALT1 in a PKC-dependent fashion, which is required for robust IL-2 transcription. However, in primary T cells, CYLD processing occurs with different kinetics and an altered dependence on PKC. The formation of a direct PKCθ/CYLD complex appears to regulate the short-term spatial distribution of CYLD, subsequently affecting NFκB and NFAT repressional activity of CYLD prior to its MALT1-dependent inactivation. Taken together, our study establishes CYLD as a new and critical PKCθ interactor in T cells and reveals that antagonistic PKCθ/ß-CYLD crosstalk is crucial for the adjustment of immune thresholds in primary mouse CD3(+) T cells.


Subject(s)
CD3 Complex , Cysteine Endopeptidases/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Protein Kinase C/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , CD3 Complex/metabolism , Caspases/metabolism , Cell Line , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Enzyme Activation , Gene Expression Regulation , Humans , Jurkat Cells , Lymphocyte Activation , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/metabolism , Phenotype , Protein Binding , Protein Kinase C/genetics , Proteolysis , T-Lymphocytes/immunology , Transcriptional Activation
6.
PLoS One ; 7(9): e44295, 2012.
Article in English | MEDLINE | ID: mdl-22962608

ABSTRACT

The ubiquitin ligase Cbl-b is an established regulator of T cell immune response thresholds. We recently showed that adoptive cell transfer (ACT) of cblb(-/-) CD8(+) T cells enhances dendritic cell (DC) immunization-mediated anti-tumor effects in immune-competent recipients. However, translation of cblb targeting to clinically applicable concepts requires that inhibition of cblb activity be transient and reversible. Here we provide experimental evidence that inhibition of cblb using chemically synthesized siRNA has such potential. Silencing cblb expression by ex vivo siRNA transfection of polyclonal CD8(+) T cells prior to ACT increased T cell tumor infiltration, significantly delayed tumor outgrowth, and increased survival rates of tumor-bearing mice. As shown by ex vivo recall assays, cblb silencing resulted in significant augmentation of intratumoral T cell cytokine response. ACT of cblb-silenced polyclonal CD8(+) T cells combined with DC-based tumor vaccines predominantly mediated anti-tumor immune responses, whereas no signs of autoimmunity could be detected. Importantly, CBLB silencing in human CD8(+) T cells mirrored the effects observed for cblb-silenced and cblb-deficient murine T cells. Our data validate the concept of enhanced anti-tumor immunity by repetitive ACT of ex vivo cblb siRNA-silenced hyper-reactive CD8(+) T cells as add-on adjuvant therapy to augment the efficacy of existing cancer immunotherapy regimens in clinical practice.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Melanoma, Experimental/therapy , Proto-Oncogene Proteins c-cbl/immunology , Skin Neoplasms/therapy , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Silencing , Humans , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Transforming Growth Factor beta/immunology
7.
Immunol Cell Biol ; 90(1): 130-4, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21383769

ABSTRACT

The success of cancer immunotherapy is limited by potent endogenous immune-evasion mechanisms, which are at least in part mediated by transforming growth factor-ß (TGF-ß). The E3 ubiquitin ligase Cbl-b is a key regulator of T cell activation and is established to regulate TGF-ß sensitivity. cblb-deficient animals reject tumors via CD8(+) T cells, which make Cbl-b an ideal target for improvement of adoptive T-cell transfer (ATC) therapy. In this study, we show that cblb-deficient CD8(+) T cells are hyper-responsive to T-cell receptor (TCR)/CD28-stimulation and are in part protected against the negative cues induced by TGF-ß in vitro. Notably, adoptive transfer of polyclonal, non-TCR transgenic cblb-deficient CD8(+) T cells is not sufficient to reject B16-ova or EG7 tumors in vivo. Thus, cblb-deficient ATC requires proper in vivo re-activation by a dendritic cell (DC) vaccine. In strict contrast to ATC monotherapy, this approach delayed tumor outgrowth and significantly increased survival rates, which is paralleled by increased CD8(+) T-cells infiltration to the tumor site and enrichment of ova-specific and interferon-γ (IFN-γ)-secreting CD8(+) T cell in the draining lymph node (LN). Moreover, CD8(+) T cells from cblb-deficient mice vaccinated with the DC vaccine show increased cytolytic activity in vivo. In summary, our data using cblb-deficient polyclonal, non-TCR-transgenic adoptively transferred CD8(+) T cells into immuno-competent non-lymphodepleted recipients suggest that targeting Cbl-b might serve as a novel 'adjuvant approach', suitable to augment the effectiveness of established anti-cancer immunotherapies.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/therapeutic use , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-cbl/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Combined Modality Therapy , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology
8.
Immunol Lett ; 126(1-2): 54-9, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19682494

ABSTRACT

PKCtheta serine/threonine and Itk tyrosine protein kinases have been implicated in T lymphocyte signal transmission. We observed a PKCtheta/Itk complex after T cell activation, raising the possibility that PKCtheta and Itk might interact functionally during T cell development and response. To address this question PKCtheta/Itk double knockout mice were generated and T cell activation responses were compared to single deficiencies as well as to wild type controls. Consistent with previous reports, Itk and PKCtheta are required in modulating CD3(+) T cell cytokine secretion responses ex vivo. Itk- and PKCtheta-deficient cells show impaired NFAT/AP-1 and NF-kappaB transactivation responses, however the combined loss, did not exceed but partially rescue the strong NFAT and NF-kappaB activation defects observed in Itk(-/-) single-deficient T cells. Taken together, this provides evidence for a more complex functional crosstalk between Itk and PKCtheta during T cell receptor signalling then previously anticipated.


Subject(s)
Isoenzymes/metabolism , Lymphocyte Activation , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Blotting, Western , CD3 Complex/metabolism , Cell Proliferation , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Hyaluronan Receptors/metabolism , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Isoenzymes/genetics , Lectins, C-Type , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Phosphorylation , Protein Binding , Protein Kinase C/genetics , Protein Kinase C-theta , Protein-Tyrosine Kinases/genetics , T-Lymphocytes/cytology , Transcription Factor AP-1/metabolism , Transcriptional Activation
9.
J Pharmacol Exp Ther ; 330(3): 792-801, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19491325

ABSTRACT

There is a pressing need for immunosuppressants with an improved safety profile. The search for novel approaches to blocking T-cell activation led to the development of the selective protein kinase C (PKC) inhibitor AEB071 (sotrastaurin). In cell-free kinase assays AEB071 inhibited PKC, with K(i) values in the subnanomolar to low nanomolar range. Upon T-cell stimulation, AEB071 markedly inhibited in situ PKC catalytic activity and selectively affected both the canonical nuclear factor-kappaB and nuclear factor of activated T cells (but not activator protein-1) transactivation pathways. In primary human and mouse T cells, AEB071 treatment effectively abrogated at low nanomolar concentration markers of early T-cell activation, such as interleukin-2 secretion and CD25 expression. Accordingly, the CD3/CD28 antibody- and alloantigen-induced T-cell proliferation responses were potently inhibited by AEB071 in the absence of nonspecific antiproliferative effects. Unlike former PKC inhibitors, AEB071 did not enhance apoptosis of murine T-cell blasts in a model of activation-induced cell death. Furthermore, AEB071 markedly inhibited lymphocyte function-associated antigen-1-mediated T-cell adhesion at nanomolar concentrations. The mode of action of AEB071 is different from that of calcineurin inhibitors, and AEB071 and cyclosporine A seem to have complementary effects on T-cell signaling pathways.


Subject(s)
Immunosuppressive Agents/pharmacology , Macrophage Activation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrroles/antagonists & inhibitors , Quinazolines/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , CD28 Antigens/metabolism , Calcium/metabolism , Cell Adhesion/drug effects , Cytokines/biosynthesis , Electrophoretic Mobility Shift Assay , Flow Cytometry , Genes, Reporter , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Jurkat Cells , Mice , Mice, Knockout , NF-kappa B/drug effects , NFATC Transcription Factors/metabolism , Protein Kinase C/genetics , Receptors, Antigen, T-Cell/drug effects , Signal Transduction/drug effects
10.
Mol Immunol ; 46(10): 2071-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19356803

ABSTRACT

The physiological roles of PKC alpha and PKC theta were defined in T cell immune functions downstream of the antigen receptor. To investigate the hypothesis that both PKC isotypes may have overlapping functions, we generated mice lacking both genes. We find that PKC alpha(-/-)/theta(-/-) animals have additive T cell response defects in comparison to animals carrying single mutations in these genes. Our studies demonstrate that the activities of PKC alpha and PKC theta converge to regulate both IL-2 cytokine responses and T cell intrinsic alloreactivity in vivo. Mechanistically, this PKC alpha/theta crosstalk primarily affects the NFAT transactivation pathway in T lymphocytes, as observed by decreased phosphorylation of Ser-9 on GSK3 beta, reduced nuclear translocation and DNA binding of NFAT in isolated PKC alpha(-/-)/theta(-/-) CD3(+) T cells. This additive defect proved to be of physiological relevance, because PKC alpha(-/-)/theta(-/-) mice demonstrated significantly prolonged allograft survival in heart transplantation experiments, whereas both PKC alpha(-/-) and PKC theta(-/-) mice showed only minimal graft prolongation when compared to wild type controls. While PKC theta appears to be the rate-limiting PKC isotype mediating T lymphocyte activation, we here provide genetic evidence that PKC alpha and PKC theta have overlapping functions in alloimmunoreactivity in vivo and both PKC theta and PKC alpha isotypes must be targeted to prevent organ allograft rejection.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C-alpha/metabolism , Protein Kinase C/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , CD3 Complex/metabolism , Cell Death , Cell Proliferation , Enzyme Activation , Flow Cytometry , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Heart Transplantation , Interleukin-2/metabolism , Isoenzymes/deficiency , Lymphocyte Subsets/enzymology , Mice , NFATC Transcription Factors/metabolism , Protein Kinase C/deficiency , Protein Kinase C-alpha/deficiency , Protein Kinase C-theta , T-Lymphocytes/cytology , Transcriptional Activation , Transplantation, Homologous
11.
Blood ; 112(12): 4617-27, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18796635

ABSTRACT

The antigen-specific interaction of a T cell with an antigen-presenting cell (APC) results in the formation of an immunologic synapse (IS) between the membranes of the 2 cells. beta(2) integrins on the T cell, namely, leukocyte function-associated antigen 1 (LFA-1) and its counter ligand, namely, immunoglobulin-like cell adhesion molecule 1 (ICAM-1) on the APC, critically stabilize this intercellular interaction. The small GTPase Rap1 controls T-cell adhesion through modulating the affinity and/or spatial organization of LFA-1; however, the upstream regulatory components triggered by the T-cell receptor (TCR) have not been resolved. In the present study, we identified a previously unknown function of a protein kinase C- theta (PKC-theta)/RapGEF2 complex in LFA-1 avidity regulation in T lymphocytes. After T-cell activation, the direct phosphorylation of RapGEF2 at Ser960 by PKC- theta regulates Rap1 activation as well as LFA-1 adhesiveness to ICAM-1. In OT-II TCR-transgenic CD4(+) T cells, clustering of LFA-1 after antigen activation was impaired in the absence of PKC- theta. These data define that, among other pathways acting on LFA-1 regulation, PKC- theta and its effector RapGEF2 are critical factors in TCR signaling to Rap1. Taken together, PKC- theta sets the threshold for T-cell activation by positively regulating both the cytokine responses and the adhesive capacities of T lymphocytes.


Subject(s)
Cell Adhesion/genetics , Isoenzymes/physiology , Protein Kinase C/physiology , T-Lymphocytes/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Adhesion/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , RNA, Small Interfering/pharmacology , Substrate Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Up-Regulation/genetics , Up-Regulation/physiology
12.
Immunity ; 29(2): 205-16, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701084

ABSTRACT

The protein kinase C (PKC) family of serine-threonine kinases plays a central role in T lymphocyte activation. Here, we identify NR2F6, a nuclear zinc-finger orphan receptor, as a critical PKC substrate and essential regulator of CD4(+) T cell activation responses. NR2F6 potently antagonized the ability of T helper 0 (Th0) and Th17 CD4(+) T cells to induce expression of key cytokine genes such as interleukin-2 (IL-2) and IL-17. Mechanistically, NR2F6 directly interfered with the DNA binding of nuclear factor of activated T cells (NF-AT):activator protein 1 (AP-1) but not nuclear factor kappaB (NF-kappa B) and, subsequently, transcriptional activity of the NF-AT-dependent IL-17A cytokine promoter. Consistent with our model, Nr2f6-deficient mice had hyperreactive lymphocytes, developed a late-onset immunopathology, and were hypersusceptible to Th17-dependent experimental autoimmune encephalomyelitis. Our study establishes NR2F6 as a transcriptional repressor of IL-17 expression in Th17-differentiated CD4(+) T cells in vitro and in vivo.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Interleukin-17/metabolism , Lymphocyte Activation , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/metabolism , COUP Transcription Factors , DNA-Binding Proteins/deficiency , Interleukin-17/immunology , Interleukin-2/immunology , Interleukin-2/metabolism , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/deficiency , Repressor Proteins , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factor AP-1/metabolism
13.
J Immunol ; 176(10): 6004-11, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16670309

ABSTRACT

Using model tumor T cell lines, protein kinase C (PKC) alpha has been implicated in IL-2 cytokine promoter activation in response to Ag receptor stimulation. In this study, for the first time, PKCalpha null mutant mice are analyzed and display normal T and B lymphocyte development. Peripheral CD3(+) PKCalpha-deficient T cells show unimpaired activation-induced IL-2 cytokine secretion, surface expression of CD25, CD44, and CD69, as well as transactivation of the critical transcription factors NF-AT, NF-kappaB, AP-1, and STAT5 in vitro. Nevertheless, CD3/CD28 Ab- and MHC alloantigen-induced T cell proliferation and IFN-gamma production are severely impaired in PKCalpha(-/-) CD3(+) T cells. Consistently, PKCalpha-deficient CD3(+) T cells from OVA-immunized PKCalpha-deficient mice exhibit markedly reduced recall proliferation to OVA in in vitro cultures. In vivo, PKCalpha-deficient mice give diminished OVA-specific IgG2a and IgG2b responses following OVA immunization experiments. In contrast, OVA-specific IgM and IgG1 responses and splenic PKCalpha(-/-) B cell proliferation are unimpaired. Our genetic data, thus, define PKCalpha as the physiological and nonredundant PKC isotype in signaling pathways that are necessary for T cell-dependent IFN-gamma production and IgG2a/2b Ab responses.


Subject(s)
Immunoglobulin Class Switching/genetics , Immunoglobulin G/classification , Immunoglobulin G/genetics , Immunoglobulin Isotypes/genetics , Protein Kinase C-alpha/deficiency , Protein Kinase C-alpha/genetics , Animals , CD3 Complex/biosynthesis , Cell Proliferation , Cells, Cultured , Interferon-gamma/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C-alpha/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...