Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Immunity ; 56(9): 2054-2069.e10, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37597518

ABSTRACT

Ligation of retinoic acid receptor alpha (RARα) by RA promotes varied transcriptional programs associated with immune activation and tolerance, but genetic deletion approaches suggest the impact of RARα on TCR signaling. Here, we examined whether RARα would exert roles beyond transcriptional regulation. Specific deletion of the nuclear isoform of RARα revealed an RARα isoform in the cytoplasm of T cells. Extranuclear RARα was rapidly phosphorylated upon TCR stimulation and recruited to the TCR signalosome. RA interfered with extranuclear RARα signaling, causing suboptimal TCR activation while enhancing FOXP3+ regulatory T cell conversion. TCR activation induced the expression of CRABP2, which translocates RA to the nucleus. Deletion of Crabp2 led to increased RA in the cytoplasm and interfered with signalosome-RARα, resulting in impaired anti-pathogen immunity and suppressed autoimmune disease. Our findings underscore the significance of subcellular RA/RARα signaling in T cells and identify extranuclear RARα as a component of the TCR signalosome and a determinant of immune responses.


Subject(s)
Autoimmune Diseases , Lymphocyte Activation , Humans , Retinoic Acid Receptor alpha/genetics , Cell Membrane , Receptors, Antigen, T-Cell
2.
J Vis Exp ; (192)2023 02 24.
Article in English | MEDLINE | ID: mdl-36912535

ABSTRACT

Fms-like tyrosine kinase 3 ligand (Flt3L) is a hematopoietic cytokine that promotes the survival and differentiation of dendritic cells (DCs). It has been used in tumor vaccines to activate innate immunity and enhance antitumor responses. This protocol demonstrates a therapeutic model using cell-based tumor vaccine consisting of Flt3L-expressing B16-F10 melanoma cells along with phenotypic and functional analysis of immune cells in the tumor microenvironment (TME). Procedures for cultured tumor cell preparation, tumor implantation, cell irradiation, tumor size measurement, intratumoral immune cell isolation, and flow cytometry analysis are described. The overall goal of this protocol is to provide a preclinical solid tumor immunotherapy model, and a research platform to study the relationship between tumor cells and infiltrating immune cells. The immunotherapy protocol described here can be combined with other therapeutic modalities, such as immune checkpoint blockade (anti-CTLA-4, anti-PD-1, anti-PD-L1 antibodies) or chemotherapy in order to improve the cancer therapeutic effect of melanoma.


Subject(s)
Cancer Vaccines , Melanoma, Experimental , Animals , Humans , Melanoma, Experimental/therapy , Dendritic Cells , Immunotherapy/methods , Cytokines , Vaccination , Tumor Microenvironment
3.
J Immunother Cancer ; 9(9)2021 09.
Article in English | MEDLINE | ID: mdl-34588224

ABSTRACT

BACKGROUND: Our previous studies revealed a critical role of a novel CTLA4-protein kinase C-eta (PKCη) signaling axis in mediating the suppressive activity of regulatory T cells (Tregs) in antitumor immunity. These studies have employed adoptive transfer of germline PKCη-deficient (Prkch-/-) Tregs into Prkch+/+ mice prior to tumor implantation. Here, we extended these findings into a biologically and clinically more relevant context. METHODS: We have analyzed the role of PKCη in antitumor immunity and the tumor microenvironment (TME) in intact tumor-bearing mice with Treg-specific or CD8+ T cell-specific Prkch deletion, including in a therapeutic model of combinatorial treatment. In addition to measuring tumor growth, we analyzed the phenotype and functional attributes of tumor-infiltrating immune cells, particularly Tregs and dendritic cells (DCs). RESULTS: Using two models of mouse transplantable cancer and a genetically engineered autochthonous hepatocellular carcinoma (HCC) model, we found, first, that mice with Treg-specific Prkch deletion displayed a significantly reduced growth of B16-F10 melanoma and TRAMP-C1 adenocarcinoma tumors. Tumor growth reduction was associated with a less immunosuppressive TME, indicated by increased numbers and function of tumor-infiltrating CD8+ effector T cells and elevated expression of the costimulatory ligand CD86 on intratumoral DCs. In contrast, CD8+ T cell-specific Prkch deletion had no effect on tumor growth or the abundance and functionality of CD8+ effector T cells, consistent with findings that Prkch-/- CD8+ T cells proliferated normally in response to in vitro polyclonal or specific antigen stimulation. Similar beneficial antitumor effects were found in mice with germline or Treg-specific Prkch deletion that were induced to develop an autochthonous HCC. Lastly, using a therapeutic model, we found that monotherapies consisting of Treg-specific Prkch deletion or vaccination with irradiated Fms-like tyrosine kinase 3 ligand (Flt3L)-expressing B16-F10 tumor cells post-tumor implantation significantly delayed tumor growth. This effect was more pronounced in mice receiving a combination of the two immunotherapies. CONCLUSION: These findings demonstrate the potential utility of PKCη inhibition as a viable clinical approach to treat patients with cancer, especially when combined with adjuvant therapies.


Subject(s)
CTLA-4 Antigen/metabolism , Immunotherapy/methods , Neoplasms/drug therapy , Animals , Humans , Mice , Neoplasms/genetics , T-Lymphocytes, Regulatory
4.
J Immunol ; 204(9): 2439-2446, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32198145

ABSTRACT

We reported that protein kinase C-η (PKCη) forms a novel (to our knowledge) signaling complex with the checkpoint inhibitory protein CTLA-4 in regulatory T cells (Tregs). This complex is required for the contact-dependent suppressive activity of Tregs, including suppression of antitumor immunity. However, the importance of PKCη in protective immunity mediated by T effector cells remains unclear. We used mice with germline or conditional Treg-specific deletion of Prkch, the PKCη-encoding gene, to explore CD8+ T cell-dependent antiviral immunity using the lymphocytic choriomeningitis virus Armstrong strain acute infection model as well as the in vitro activation of murine or human CD8+ T cells. Five days following infection, germline Prkch -/- mice displayed enhanced viral clearance compared with control mice. Similarly, Prkch Treg-specific conditional knockout mice also showed improved viral clearance and displayed enhanced expression of granzyme B and IFN-γ by both virus-specific and total CD8+ T cells, demonstrating that enhanced viral clearance in germline Prkch -/- mice is caused by PKCη deficiency in Tregs and the resulting functional defect of Prkch -/- Tregs. In addition, purified Prkch -/- mouse CD8+ T cells as well as PRKCH knockdown human CD8+ T cells displayed intact, or even enhanced, T cell activation in vitro as measured by proliferation and expression of granzyme B and IFN-γ. Thus, global PKCη deletion does not impair overall CD8+ T cell-mediated immunity, including antiviral immunity, implying that selective pharmacological PKCη inhibition could be safely used in vivo to inhibit undesired contact-dependent suppression by Tregs and, thus, enhance tumor-specific and, likely, virus-specific immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation , Protein Kinase C , T-Lymphocytes, Regulatory , Virus Diseases , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation/drug effects , CTLA-4 Antigen/immunology , Granzymes/immunology , HEK293 Cells , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , Mice, Knockout , Protein Kinase C/deficiency , Protein Kinase C/immunology , Protein Kinase Inhibitors/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Virus Diseases/immunology
5.
Sci Signal ; 12(577)2019 04 16.
Article in English | MEDLINE | ID: mdl-30992398

ABSTRACT

Protein kinase C-θ (PKCθ) is an important component of proximal T cell receptor (TCR) signaling. We previously identified the amino-terminal C2 domain of PKCθ as a phosphotyrosine (pTyr)-binding domain. Using a mutant form of PKCθ that cannot bind pTyr (PKCθHR2A), we showed that pTyr binding by PKCθ was required for TCR-induced T cell activation, proliferation, and TH2 cell differentiation but not for T cell development. Using tandem mass spectrometry and coimmunoprecipitation, we identified the kinase ζ-associated protein kinase of 70 kDa (Zap70) as a binding partner of the PKCθ pTyr-binding pocket. Tyr126 of Zap70 directly bound to PKCθ, and the interdomain B residues Tyr315 and Tyr319 were indirectly required for binding to PKCθ, reflecting their role in promoting the open conformation of Zap70. PKCθHR2A-expressing CD4+ T cells displayed defects not only in known PKCθ-dependent signaling events, such as nuclear factor κB (NF-κB) activation and TH2 cell differentiation, but also in full activation of Zap70 itself and in the activating phosphorylation of linker of activation of T cells (LAT) and phospholipase C-γ1 (PLCγ1), signaling proteins that are traditionally considered to be activated independently of PKC. These findings demonstrate that PKCθ plays an important role in a positive feedback regulatory loop that modulates TCR-proximal signaling and, moreover, provide a mechanistic explanation for earlier reports that documented an important role for PKCθ in T cell Ca2+ signaling. This PKCθ-Zap70 interaction could potentially serve as a promising and highly selective immunosuppressive drug target in autoimmunity and organ transplantation.


Subject(s)
Calcium Signaling , Protein Kinase C-theta/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/enzymology , ZAP-70 Protein-Tyrosine Kinase/metabolism , Animals , Humans , Jurkat Cells , Mice , Mice, Knockout , Protein Kinase C-theta/genetics , Receptors, Antigen, T-Cell/genetics , ZAP-70 Protein-Tyrosine Kinase/genetics
6.
JCI Insight ; 2(23)2017 12 07.
Article in English | MEDLINE | ID: mdl-29212947

ABSTRACT

The ability of Tregs to control the development of immune responses is essential for maintaining immune system homeostasis. However, Tregs also inhibit the development of efficient antitumor responses. Here, we explored the characteristics and mechanistic basis of the Treg-intrinsic CTLA4/PKCη signaling pathway that we recently found to be required for contact-dependent Treg-mediated suppression. We show that PKCη is required for the Treg-mediated suppression of tumor immunity in vivo. The presence of PKCη-deficient (Prkch-/-) Tregs in the tumor microenvironment was associated with a significantly increased expression of the costimulatory molecule CD86 on intratumoral CD103+ DCs, enhanced priming of antigen-specific CD8+ T cells, and greater levels of effector cytokines produced by these cells. Similar to mouse Tregs, the GIT/PAK/PIX complex also operated downstream of CTLA4 and PKCη in human Tregs, and GIT2 knockdown in Tregs promoted antitumor immunity. Collectively, our data suggest that targeting the CTLA4/PKCη/GIT/PAK/PIX signaling pathway in Tregs could represent a novel immunotherapeutic strategy to alleviate the negative impact of Tregs on antitumor immune responses.


Subject(s)
CTLA-4 Antigen/immunology , Protein Kinase C/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , B7-2 Antigen/metabolism , Female , Heterografts , Humans , Immune Tolerance , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Prostatic Neoplasms/immunology , Signal Transduction/immunology
8.
Nat Immunol ; 17(7): 825-33, 2016 07.
Article in English | MEDLINE | ID: mdl-27135603

ABSTRACT

Signaling via the inducible costimulator ICOS fuels the stepwise development of follicular helper T cells (TFH cells). However, a signaling pathway unique to ICOS has not been identified. We found here that the kinase TBK1 associated with ICOS via a conserved motif, IProx, that shares homology with the tumor-necrosis-factor receptor (TNFR)-associated factors TRAF2 and TRAF3. Disruption of this motif abolished the association of TBK1 with ICOS, TRAF2 and TRAF3, which identified a TBK1-binding consensus. Alteration of this motif in ICOS or depletion of TBK1 in T cells severely impaired the differentiation of germinal center (GC) TFH cells and the development of GCs, interfered with B cell differentiation and disrupted the development of antibody responses, but the IProx motif and TBK1 were dispensable for the early differentiation of TFH cells. These results reveal a previously unknown ICOS-TBK1 signaling pathway that specifies the commitment of GC TFH cells.


Subject(s)
B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/physiology , Germinal Center/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Antibody Formation/genetics , Cell Differentiation/genetics , Cells, Cultured , Inducible T-Cell Co-Stimulator Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding , Protein Serine-Threonine Kinases/genetics , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 3/genetics
9.
J Cell Sci ; 128(23): 4341-52, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26483383

ABSTRACT

SLAT (also known as DEF6) promotes T cell activation and differentiation by regulating NFAT-Ca(2+) signaling. However, its role in TCR-mediated inside-out signaling, which induces integrin activation and T cell adhesion, a central process in T cell immunity and inflammation, has not been explored. Here, we show that SLAT is crucial for TCR-induced adhesion to ICAM-1 and affinity maturation of LFA-1 in CD4(+) T cells. Mechanistic studies revealed that SLAT interacts, through its PH domain, with a key component of inside-out signaling, namely the active form of the small GTPase Rap1 (which has two isoforms, Rap1A and Rap1B). This interaction has been further shown to facilitate the interdependent recruitment of Rap1 and SLAT to the T cell immunological synapse upon TCR engagement. Furthermore, a SLAT mutant lacking its PH domain drastically inhibited LFA-1 activation and CD4(+) T cell adhesion. Finally, we established that a constitutively active form of Rap1, which is present at the plasma membrane, rescues the defective LFA-1 activation and ICAM-1 adhesion in SLAT-deficient (Def6(-/-)) T cells. These findings ascribe a new function to SLAT, and identify Rap1 as a target of SLAT function in TCR-mediated inside-out signaling.


Subject(s)
Cell Adhesion/physiology , DNA-Binding Proteins/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Nuclear Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , rap GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Guanine Nucleotide Exchange Factors , Lymphocyte Function-Associated Antigen-1/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/genetics , rap GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/genetics
10.
Sci Signal ; 7(345): ra93, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25270259

ABSTRACT

The guanine nucleotide exchange factor SLAT (SWAP-70-like adaptor of T cells) regulates T cell activation and differentiation by enabling Ca(2+) release from intracellular stores in response to stimulation of the T cell receptor (TCR). We found a TCR-induced association between SLAT and inositol 1,4,5-trisphosphate (IP3) receptor type 1 (IP3R1). The N-terminal region of SLAT, which contains two EF-hand motifs that we determined bound Ca(2+), and the SLAT pleckstrin homology (PH) domain independently bound to IP3R1 by associating with a conserved motif within the IP3R1 ligand-binding domain. Disruption of the SLAT-IP3R1 interaction with cell-permeable, IP3R1-based fusion peptides inhibited TCR-stimulated Ca(2+) signaling, activation of the transcription factor NFAT (nuclear factor of activated T cells), and production of cytokines, suggesting that this interaction is required for optimal T cell activation. The finding that SLAT is an IP3R1-interacting protein required for T cell activation suggests that this interaction could be a potential target for a selective immunosuppressive drug.


Subject(s)
Calcium Signaling/immunology , DNA-Binding Proteins/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Lymphocyte Activation/immunology , Nuclear Proteins/metabolism , T-Lymphocytes/immunology , Animals , Blotting, Western , Cell Fractionation , DNA-Binding Proteins/genetics , Genetic Vectors/genetics , Guanine Nucleotide Exchange Factors , Immunoprecipitation , Interferon-gamma/metabolism , Luciferases , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , T-Lymphocytes/metabolism
11.
Nat Immunol ; 15(5): 465-72, 2014 May.
Article in English | MEDLINE | ID: mdl-24705298

ABSTRACT

Regulatory T (Treg) cells, which maintain immune homeostasis and self-tolerance, form an immunological synapse (IS) with antigen-presenting cells (APCs). However, signaling events at the Treg cell IS remain unknown. Here we show that the kinase PKC-η associated with CTLA-4 and was recruited to the Treg cell IS. PKC-η-deficient Treg cells displayed defective suppressive activity, including suppression of tumor immunity but not of autoimmune colitis. Phosphoproteomic and biochemical analysis revealed an association between CTLA-4-PKC-η and the GIT2-αPIX-PAK complex, an IS-localized focal adhesion complex. Defective activation of this complex in PKC-η-deficient Treg cells was associated with reduced depletion of CD86 from APCs by Treg cells. These results reveal a CTLA-4-PKC-η signaling axis required for contact-dependent suppression and implicate this pathway as a potential cancer immunotherapy target.


Subject(s)
CTLA-4 Antigen/metabolism , Immunological Synapses/metabolism , Immunotherapy/trends , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Multiprotein Complexes/metabolism , Protein Kinase C/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immune Tolerance/genetics , Jurkat Cells , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Protein Binding , Protein Kinase C/genetics , Proteomics , Signal Transduction
12.
Nat Immunol ; 12(11): 1105-12, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964608

ABSTRACT

Protein kinase C-θ (PKC-θ) translocates to the center of the immunological synapse, but the underlying mechanism and its importance in T cell activation are unknown. Here we found that the V3 domain of PKC-θ was necessary and sufficient for localization to the immunological synapse mediated by association with the coreceptor CD28 and dependent on the kinase Lck. We identified a conserved proline-rich motif in V3 required for association with CD28 and immunological synapse localization. We found association with CD28 to be essential for PKC-θ-mediated downstream signaling and the differentiation of T helper type 2 cells (T(H)2 cells) and interleukin 17-producing helper T cells (T(H)17 cells) but not of T helper type 1 cells (T(H)1 cells). Ectopic expression of V3 sequestered PKC-θ from the immunological synapse and interfered with its functions. Our results identify a unique mode of CD28 signaling, establish a molecular basis for the immunological synapse localization of PKC-θ and indicate V3-based 'decoys' may be therapeutic modalities for T cell-mediated inflammatory diseases.


Subject(s)
CD28 Antigens/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , T-Lymphocyte Subsets/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Amino Acid Motifs/genetics , Animals , CD28 Antigens/immunology , Cell Differentiation/immunology , Cells, Cultured , Immunological Synapses , Immunomodulation , Isoenzymes/genetics , Isoenzymes/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Proline-Rich Protein Domains/genetics , Protein Binding/immunology , Protein Kinase C/genetics , Protein Kinase C/immunology , Protein Kinase C-theta , Protein Transport/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
13.
J Immunol ; 183(11): 7259-67, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19915062

ABSTRACT

SWAP-70-like adapter of T cells (SLAT; also known as Def6) is a novel guanine nucleotide exchange factor for Rho GTPases that has been previously shown to play a role in CD4+ T cell activation and Th1/Th2 differentiation. However, the role of SLAT/Def6 in autoimmunity and its associated Th1- and Th17-specific responses has not yet been clearly elucidated. We used a prototypical and pathologically relevant Th1/Th17-mediated autoimmune model, that is, experimental autoimmune encephalomyelitis, to assess the role of SLAT/Def6 in autoantigen-specific T cell response. We found that T cell-expressed SLAT/Def6 was critical for experimental autoimmune encephalomyelitis development and pathogenesis, as evidenced by the resistance of Def6-deficient (Def6(-/-)) mice to clinical signs of the disease associated with a lack of CNS inflammation and demyelination in myelin oligodendrocyte glycoprotein-immunized Def6(-/-) mice. Moreover, Def6 deficiency resulted in a severely diminished myelin oligodendrocyte glycoprotein-specific CD4+ T cell proliferation as well as a defect in IFN-gamma and IL-17 production in secondary lymphoid organs and the CNS. Lastly, Def6(-/-) CD4+ T cells were grossly deficient in their ability to differentiate into Th17 cells both in vitro and in vivo in a T cell-intrinsic manner. Therefore, our study establishes T cell-expressed SLAT/Def6 as a pivotal positive regulator of Th17 inflammatory responses and, thus, essential in controlling autoimmune and inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Nuclear Proteins/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Guanine Nucleotide Exchange Factors , Interleukin-17/biosynthesis , Mice , Mice, Knockout , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
14.
Immunity ; 29(5): 704-19, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18976935

ABSTRACT

SWAP-70-like adaptor of T cells (SLAT) is a guanine nucleotide exchange factor for Rho GTPases that regulates the development of T helper 1 (Th1) and Th2 cell inflammatory responses by controlling the Ca(2+)-NFAT signaling pathway. However, the mechanism used by SLAT to regulate these events is unknown. Here, we report that the T cell receptor (TCR)-induced translocation of SLAT to the immunological synapse required Lck-mediated phosphorylation of two tyrosine residues located in an immunoreceptor tyrosine-based activation motif-like sequence but was independent of the SLAT PH domain. This subcellular relocalization was coupled to, and necessary for, activation of the NFAT pathway. Furthermore, membrane targeting of the SLAT Dbl-homology (catalytic) domain was sufficient to trigger TCR-mediated NFAT activation and Th1 and Th2 differentiation in a Cdc42-dependent manner. Therefore, tyrosine-phosphorylation-mediated relocalization of SLAT to the site of antigen recognition is required for SLAT to exert its pivotal role in NFAT-dependent CD4(+) T cell differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Immunological Synapses/immunology , NFATC Transcription Factors/metabolism , Nuclear Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Line, Tumor , DNA-Binding Proteins/deficiency , Guanine Nucleotide Exchange Factors , Humans , Immunological Synapses/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/deficiency , Phosphorylation , Receptors, Antigen, T-Cell/immunology , Th1 Cells/metabolism , Th2 Cells/metabolism , Transfection , Tyrosine/metabolism , cdc42 GTP-Binding Protein/immunology , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/immunology , rac1 GTP-Binding Protein/metabolism
15.
J Clin Invest ; 117(8): 2164-75, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17657315

ABSTRACT

SWAP-70-like adapter of T cells (SLAT) is a novel guanine nucleotide exchange factor for Rho GTPases that is upregulated in Th2 cells, but whose physiological function is unclear. We show that SLAT(-/-) mice displayed a developmental defect at one of the earliest stages of thymocyte differentiation, the double-negative 1 (DN1) stage, leading to decreased peripheral T cell numbers. SLAT(-/-) peripheral CD4(+) T cells demonstrated impaired TCR/CD28-induced proliferation and IL-2 production, which was rescued by the addition of exogenous IL-2. Importantly, SLAT(-/-) mice were grossly impaired in their ability to mount not only Th2, but also Th1-mediated lung inflammatory responses, as evidenced by reduced airway neutrophilia and eosinophilia, respectively. Levels of Th1 and Th2 cytokine in the lungs were also markedly reduced, paralleling the reduction in pulmonary inflammation. This defect in mounting Th1/Th2 responses, which was also evident in vitro, was traced to a severe reduction in Ca(2+) mobilization from ER stores, which consequently led to defective TCR/CD28-induced translocation of nuclear factor of activated T cells 1/2 (NFATc1/2). Thus, SLAT is required for thymic DN1 cell expansion, T cell activation, and Th1 and Th2 inflammatory responses.


Subject(s)
Calcium Signaling/immunology , DNA-Binding Proteins/immunology , NFATC Transcription Factors/immunology , Nuclear Proteins/immunology , Pneumonia/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , CD28 Antigens/genetics , CD28 Antigens/immunology , Calcium Signaling/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , DNA-Binding Proteins/deficiency , Guanine Nucleotide Exchange Factors , Inflammation/genetics , Inflammation/immunology , Interleukin-2/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , Nuclear Proteins/deficiency , Pneumonia/genetics , Pneumonia/pathology , Pulmonary Eosinophilia/genetics , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/pathology , Th2 Cells/pathology , Thymus Gland/immunology , Thymus Gland/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...