Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
PLoS Biol ; 19(3): e3001063, 2021 03.
Article in English | MEDLINE | ID: mdl-33684096

ABSTRACT

The function of Sprouty2 (Spry2) in T cells is unknown. Using 2 different (inducible and T cell-targeted) knockout mouse strains, we found that Spry2 positively regulated extracellular signal-regulated kinase 1/2 (ERK1/2) signaling by modulating the activity of LCK. Spry2-/- CD4+ T cells were unable to activate LCK, proliferate, differentiate into T helper cells, or produce cytokines. Spry2 deficiency abrogated type 2 inflammation and airway hyperreactivity in a murine model of asthma. Spry2 expression was higher in blood and airway CD4+ T cells from patients with asthma, and Spry2 knockdown impaired human T cell proliferation and cytokine production. Spry2 deficiency up-regulated the lipid raft protein caveolin-1, enhanced its interaction with CSK, and increased CSK interaction with LCK, culminating in augmented inhibitory phosphorylation of LCK. Knockdown of CSK or dislodgment of caveolin-1-bound CSK restored ERK1/2 activation in Spry2-/- T cells, suggesting an essential role for Spry2 in LCK activation and T cell function.


Subject(s)
Asthma/physiopathology , CSK Tyrosine-Protein Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Proteins/metabolism , Adult , Animals , Asthma/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , MAP Kinase Signaling System/physiology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology
2.
J Immunol ; 195(9): 4218-27, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26416283

ABSTRACT

Previously, we demonstrated that CD28 and CTLA-4 signaling control Casitas-B-lineage lymphoma (Cbl)-b protein expression, which is critical for T cell activation and tolerance induction. However, the molecular mechanism(s) of this regulation remains to be elucidated. In this study, we found that Cbl-b fails to undergo tyrosine phosphorylation upon CD3 stimulation because SHP-1 is recruited to and dephosphorylates Cbl-b, whereas CD28 costimulation abrogates this interaction. In support of this finding, T cells lacking SHP-1 display heightened tyrosine phosphorylation and ubiquitination of Cbl-b upon TCR stimulation, which correlates with decreased levels of Cbl-b protein. The aberrant Th2 phenotype observed in T cell-specific Shp1(-/-) mice is reminiscent of heightened Th2 response in Cblb(-/-) mice. Indeed, overexpressing Cbl-b in T cell-specific Shp1(-/-) T cells not only inhibits heightened Th2 differentiation in vitro, but also Th2 responses and allergic airway inflammation in vivo. Therefore, SHP-1 regulates Cbl-b-mediated T cell responses by controlling its tyrosine phosphorylation and ubiquitination.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Proto-Oncogene Proteins c-cbl/metabolism , T-Lymphocytes/immunology , Animals , CD28 Antigens/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/chemistry , Th2 Cells/immunology , Ubiquitination
3.
J Immunol ; 191(12): 6208-21, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227778

ABSTRACT

Focal adhesion kinase (FAK) is a critical regulator of signal transduction in multiple cell types. Although this protein is activated upon TCR engagement, the cellular function that FAK plays in mature human T cells is unknown. By suppressing the function of FAK, we revealed that FAK inhibits TCR-mediated signaling by recruiting C-terminal Src kinase to the membrane and/or receptor complex following TCR activation. Thus, in the absence of FAK, the inhibitory phosphorylation of Lck and/or Fyn is impaired. Together, these data highlight a novel role for FAK as a negative regulator TCR function in human T cells. These results also suggest that changes in FAK expression could modulate sensitivity to TCR stimulation and contribute to the progression of T cell malignancies and autoimmune diseases.


Subject(s)
Focal Adhesion Kinase 1/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Adolescent , Adult , Amino Acid Substitution , CD4-Positive T-Lymphocytes/enzymology , CSK Tyrosine-Protein Kinase , Enzyme Activation/physiology , Female , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Male , MicroRNAs/genetics , Middle Aged , Multienzyme Complexes , Phosphorylation , Phosphotyrosine/physiology , Protein Processing, Post-Translational , Protein Transport , Proto-Oncogene Proteins c-fyn/physiology , RNA Interference , Recombinant Fusion Proteins/metabolism , Transfection , Young Adult , src-Family Kinases/metabolism
4.
J Immunol ; 190(3): 1360-71, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23293352

ABSTRACT

The 90-kDa heat shock protein (Hsp90) has become an important therapeutic target with ongoing evaluation in a number of malignancies. Although Hsp90 inhibitors have a high therapeutic index with limited effects on normal cells, they have been described to inhibit dendritic cell function. However, its effect on human immune effector cells may have significant clinical implications, but remains unexplored. In this study, we have evaluated the effects of Hsp90 inhibition on human T lymphocyte and NK cells, including their Ag expression, activation, proliferation, and functional activities. These studies demonstrate that Hsp90 inhibition irreversibly downregulates cell surface expression of critical Ags (CD3, CD4, CD8), the costimulatory molecule (CD28, CD40L), and αß receptors on T lymphocytes, as well as activating receptors (CD2, CD11a, CD94, NKp30, NKp44, NKp46, KARp50.3) on NK cells. Hsp90 inhibition significantly reduced CD4 protein expression on T lymphocytes at both the cell surface and intracellular level, which was shown to be associated with aberrant regulation of Src-kinase p56(Lck). Downregulation of the Ags triggered by Hsp90 inhibition on CD3(+) T lymphocytes, both in CD4(+) and CD8(+) T cell subsets, was associated with a disruption in their cellular activation, proliferation, and/or IFN-γ production, when the inhibition occurred either in activated or inactivated cells. In addition, downregulation of key activating receptors on NK cells following Hsp90 inhibition resulted in decreased cytotoxicity against tumor cells. Therefore, these observations demonstrate the need to closely monitor immune function in patients being treated with a Hsp90 inhibitor and may provide a potential therapeutic application in autoimmune diseases.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Apoptosis/drug effects , Benzoquinones/pharmacology , Cell Line, Tumor , Cytokines/pharmacology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Killer Cells, Natural/drug effects , Lactams, Macrocyclic/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Multiple Myeloma/pathology , Phenotype , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Natural Killer Cell/biosynthesis , Receptors, Natural Killer Cell/genetics , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects
5.
Yakugaku Zasshi ; 144(5): 497-501, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692923

ABSTRACT

Signal-transducing adaptor protein-2 (STAP-2) is a unique scaffold protein that regulates several immunological signaling pathways, including LIF/LIF receptor and LPS/TLR4 signals. STAP-2 is required for Fas/FasL-dependent T cell apoptosis and SDF-1α-induced T cell migration. Conversely, STAP-2 modulates integrin-mediated T cell adhesion, suggesting that STAP-2 is essential for several negative and positive T cell functions. However, whether STAP-2 is involved in T cell-antigen receptor (TCR)-mediated T cell activation is unknown. STAP-2 deficiency was recently reported to suppress TCR-mediated T cell activation by inhibiting LCK-mediated CD3ζ and ZAP-70 activation. Using STAP-2 deficient mice, it was demonstrated that STAP-2 is required for the pathogenesis of Propionibacterium acnes-induced granuloma formation and experimental autoimmune encephalomyelitis. Here, detailed functions of STAP-2 in TCR-mediated T cell activation, and how STAP-2 affects the pathogenesis of T cell-mediated inflammation and immune diseases, are reviewed.


Subject(s)
Adaptor Proteins, Signal Transducing , Lymphocyte Activation , Receptors, Antigen, T-Cell , Signal Transduction , T-Lymphocytes , ZAP-70 Protein-Tyrosine Kinase , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/physiology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , CD3 Complex , Cell Adhesion , Cell Movement , Chemokine CXCL12/physiology , Chemokine CXCL12/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Inflammation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Propionibacterium acnes/physiology , Propionibacterium acnes/immunology , Receptors, Antigen, T-Cell/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism , ZAP-70 Protein-Tyrosine Kinase/physiology
6.
J Neurosci ; 32(21): 7278-86, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623673

ABSTRACT

The molecular mechanisms underlying preconditioning (PC), a powerful endogenous neuroprotective phenomenon, remain to be fully elucidated. Once identified, these endogenous mechanisms could be manipulated for therapeutic gain. We investigated whether lymphocyte cell kinase (Lck), a member of the Src kinases family, mediates PC. We used both in vitro primary cortical neurons and in vivo mouse cerebral focal ischemia models of preconditioning, cellular injury, and neuroprotection. Genetically engineered mice deficient in Lck, gene silencing using siRNA, and pharmacological approaches were used. Cortical neurons preconditioned with sublethal exposure to NMDA or oxygen glucose deprivation (OGD) exhibited enhanced Lck kinase activity, and were resistant to injury on subsequent exposure to lethal levels of NMDA or OGD. Lck gene silencing using siRNA abolished tolerance against both stimuli. Lck-/- mice or neurons isolated from Lck-/- mice did not exhibit PC-induced tolerance. An Lck antagonist administered to wild-type mice significantly attenuated the neuroprotective effect of PC in the mouse focal ischemia model. Using pharmacological and gene silencing strategies, we also showed that PKCε is an upstream regulator of Lck, and Fyn is a downstream target of Lck. We have discovered that Lck plays an essential role in PC in both cellular and animal models of stroke. Our data also show that the PKCε-Lck-Fyn axis is a key mediator of PC. These findings provide new opportunities for stroke therapy development.


Subject(s)
Brain Ischemia/enzymology , Cerebral Cortex/enzymology , Ischemic Preconditioning/methods , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Neuroprotective Agents/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain Ischemia/genetics , Cell Death/physiology , Cerebral Cortex/drug effects , Disease Models, Animal , Gene Silencing/physiology , Glucose/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Knockout , N-Methylaspartate/toxicity , Neurons/enzymology , Oxygen/pharmacology , Primary Cell Culture , Protein Kinase C-epsilon/metabolism , RNA, Small Interfering/pharmacology
7.
Blood ; 117(1): 108-17, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20876849

ABSTRACT

T-cell development is critically dependent on the activities of the Src-family kinases p56(lck) and p59(fyn). While Lck plays a dominant role in the initiation of T-cell receptor (TCR) signaling and in thymocyte differentiation, Fyn plays a more subtle regulatory role. We sought to determine the role of intracellular localization in the differing functions of Lck and Fyn in T cells. By generating transgenic mice that express chimeric Lck-Fyn proteins, we showed that the N-terminal unique domain determines the intracellular localization and function of Lck in pre-TCR and mature αßTCR signaling in vivo. Furthermore, coexpression of a "domain-swap" Lck protein containing the Fyn unique domain with an inducible Lck transgene resulted in the development of thymomas. In contrast to previous reports of Lck-driven thymomas, tumor development was dependent on either pre-TCR or mature TCR signals, and was completely ablated when mice were crossed to a recombination activating gene 1 (Rag1)-deficient background. These data provide a mechanistic basis for the differing roles of Lck and Fyn in T-cell development, and show that intracellular localization as determined by the N-terminal unique domains is critical for Src-family kinase function in vivo.


Subject(s)
Cell Differentiation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Proto-Oncogene Proteins c-fyn/physiology , Thymoma/pathology , Thymus Gland/cytology , Animals , Blotting, Western , CD2 Antigens/genetics , Female , Flow Cytometry , Humans , Immunoprecipitation , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , RNA, Messenger/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes/metabolism , Thymoma/metabolism , Thymus Gland/metabolism
8.
Blood ; 117(12): 3331-42, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21200022

ABSTRACT

The integrin lymphocyte function-associated antigen 1 (LFA-1) controls many functions of T lymphocytes and is particularly essential during lymphocyte migration from blood into tissues. LFA-1 is considered to initiate "outside-in" signaling when bound to ligand intercellular adhesion molecule 1 (ICAM-1), but little is known about the proteins involved or where in the cell such LFA-1-mediated signaling might be operating. Here we show that LFA-1 is constitutively associated with the protein tyrosine kinases Lck and zeta chain-associated protein of 70 kDa (ZAP-70). When LFA-1 binds ICAM-1, both kinases become phosphorylated and the consequence of kinase activation is the conversion of intermediate- to high-affinity LFA-1 and an increase in close contact with ICAM-1. In the polarized T lymphocyte, phospho-ZAP-70 is concentrated within a region of high-affinity LFA-1 that includes talin and encompasses the lamella/lamellipodial interface as well as further back in the cell. Deficiency of ZAP-70 through inhibition or knockdown in T lymphocytes decreases the speed of migration on ICAM-1, as well as reducing firm adhesion under shear-flow conditions. Through its control of high-affinity LFA-1, the LFA-1/Lck/ZAP-70 complex is in position to initiate the rapid adhesion strengthening and migration necessary for T-lymphocyte responses when stimulated vasculature is encountered at sites of infection or injury.


Subject(s)
Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocyte Function-Associated Antigen-1/physiology , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/physiology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Chemotaxis, Leukocyte/physiology , Humans , Integrins/genetics , Integrins/metabolism , Integrins/physiology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Protein Binding/drug effects , Protein Binding/genetics , Protein Transport/drug effects , Protein Transport/genetics , Protein Transport/immunology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Substrate Specificity , T-Lymphocytes/drug effects , Transfection , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/metabolism
9.
J Immunol ; 186(2): 931-9, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21160038

ABSTRACT

The TNF-related apoptosis-inducing ligand was shown to provide a costimulatory signal that cooperates with the TCR/CD3 complex to induce T cell proliferation and cytokine production. Although a number of signaling pathways were linked to the TCR/CD3 complex, it is not known how these two receptors cooperate to induce T cell activation. In this study, we show that TRAIL-induced costimulation of T cells depends on activation of the NF-κB pathway. TRAIL induced the NF-κB pathway by phosphorylation of inhibitor of κB factor kinase and protein kinase C in conjunction with anti-CD3. Furthermore, we demonstrated that TRAIL costimulation induced phosphorylation of the upstream TCR-proximal tyrosine kinases, Lck and ZAP70. Ligation of the TRAIL by its soluble receptor, DR4-Fc, alone was able to induce the phosphorylation of Lck and ZAP70 and to activate the NF-κB pathway; however, it was insufficient to fully activate T cells to support T cell proliferation. In contrast, TRAIL engagement in conjunction with anti-CD3, but not TRAIL ligation alone, induced lipid raft assembly and recruitment of Lck and PKC. These results demonstrate that TRAIL costimulation mediates NF-κB activation and T cell proliferation by lipid raft assembly and recruitment of Lck. Our results suggest that in TRAIL costimulation, lipid raft recruitment of Lck integrates mitogenic NF-κB-dependent signals from the TCR and TRAIL in T lymphocytes.


Subject(s)
Cell Proliferation , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Microdomains/metabolism , NF-kappa B/metabolism , T-Lymphocytes/immunology , TNF-Related Apoptosis-Inducing Ligand/physiology , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Membrane Microdomains/physiology , NF-kappa B/physiology , Protein Transport/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase/biosynthesis , ZAP-70 Protein-Tyrosine Kinase/physiology
10.
Am J Physiol Cell Physiol ; 302(10): C1504-12, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22378744

ABSTRACT

The cAMP/PKA signaling system constitutes an inhibitory pathway in T cells and, although its biochemistry has been thoroughly investigated, its possible effects on ion channels are still not fully understood. K(V)1.3 channels play an important role in T-cell activation, and their inhibition suppresses T-cell function. It has been reported that PKA modulates K(V)1.3 activity. Two PKA isoforms are expressed in human T cells: PKAI and PKAII. PKAI has been shown to inhibit T-cell activation via suppression of the tyrosine kinase Lck. The aim of this study was to determine the PKA isoform modulating K(V)1.3 and the signaling pathway underneath. 8-Bromoadenosine 3',5'-cyclic monophosphate (8-BrcAMP), a nonselective activator of PKA, inhibited K(V)1.3 currents both in primary human T and in Jurkat cells. This inhibition was prevented by the PKA blocker PKI(6-22). Selective knockdown of PKAI, but not PKAII, with siRNAs abolished the response to 8-BrcAMP. Additional studies were performed to determine the signaling pathway mediating PKAI effect on K(V)1.3. Overexpression of a constitutively active mutant of Lck reduced the response of K(V)1.3 to 8-Br-cAMP. Moreover, knockdown of the scaffolding protein disc large 1 (Dlg1), which binds K(V)1.3 to Lck, abolished PKA modulation of K(V)1.3 channels. Immunohistochemistry studies showed that PKAI, but not PKAII, colocalizes with K(V)1.3 and Dlg1 indicating a close proximity between these proteins. These results indicate that PKAI selectively regulates K(V)1.3 channels in human T lymphocytes. This effect is mediated by Lck and Dlg1. We thus propose that the K(V)1.3/Dlg1/Lck complex is part of the membrane pathway that cAMP utilizes to regulate T-cell function.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cyclic AMP-Dependent Protein Kinase Type I/physiology , Kv1.3 Potassium Channel/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Membrane Proteins/physiology , T-Lymphocytes/enzymology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cells, Cultured , Discs Large Homolog 1 Protein , Humans , Immunosuppressive Agents/pharmacology , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
11.
J Immunol ; 184(8): 4178-84, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20237292

ABSTRACT

The Src family kinase Lck has been shown to be crucial in T cell signaling and development. However, its role in Th effector functions is not well understood. Lck has previously been shown to play a role in the cytokine expression of Th2 cells, but the mechanism by which Lck influences Th2 effector functions is unknown. Using a mouse model, we report that Lck is important in regulating the expression of IL-4 in Th2 skewed cells but is not as necessary for the expression of Th2 cytokines IL-5, IL-10, and IL-13. Furthermore, in the absence of Lck, T-bet and GATA-3 expression is aberrant. Moreover, this atypical expression pattern of T-bet and GATA-3 correlates with increased histone 3 acetylation at the Ifng locus and production of the Th1 cytokine IFN-gamma. We find overexpression of GATA-3 restores IL-4 expression in lck(-/-) Th2 cells; this indicates that the decreased IL-4 expression is due in part to reduced amounts of GATA-3. Taken together, these data imply that Lck mediates Th2 differentiation through effects on T-bet and GATA-3.


Subject(s)
Cell Differentiation/immunology , GATA3 Transcription Factor/antagonists & inhibitors , Gene Expression Regulation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , T-Box Domain Proteins/antagonists & inhibitors , Th2 Cells/enzymology , Th2 Cells/immunology , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Cells, Cultured , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-4/antagonists & inhibitors , Interleukin-4/biosynthesis , Interleukin-4/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Th2 Cells/metabolism
12.
J Immunol ; 185(6): 3285-94, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20729329

ABSTRACT

T cell activation and effector function is essential for robust immunity. Ag TCR signals are known to regulate T lymphocyte differentiation, but the mechanisms involved in this regulation remain unclear. Recent work has demonstrated that the Src family protein tyrosine kinase p56Lck specifically links TCR signaling to activation of the MAPK pathway through the function of its Src homology 3 (SH3) domain. The MAPK pathway is involved in T cell activation and has previously been implicated in Th2 immunity. We have used Lck SH3 mutant knockin mice (LckW97A) to investigate the potential role of this regulatory mechanism in T lymphocyte activation and effector function. Our results demonstrate that Lck SH3 domain function regulates activation of T lymphocytes as indicated by reduced IL-2 production, CD69 induction, and proliferation of LckW97A T cells following TCR stimulation. Biochemical studies confirm that activation of the MAPK pathway is selectively altered following TCR ligation in LckW97A T lymphocytes. Phospho-ERK induction is reduced, but phospho-phospholipase Cgamma1 induction and calcium mobilization are largely unaffected. Immunization with DNP-keyhole limpet hemocyanin, heat-killed Brucella abortus, or infection with Nippostrongylus brasiliensis demonstrates selectively impaired Th2 immunity with reduced serum levels of IgG1, IgE, and IL-4. In vitro studies show that LckW97A T cells can differentiate into Th2-type cells, but they form IFN-gamma-producing cells under conditions that normally favor Th2 development. These data indicate that the Lck SH3 domain controls T lymphocyte activation by regulating MAPK pathway induction and demonstrate a novel role for Lck in the regulation of Th2-type immunity.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Th2 Cells/enzymology , Th2 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Epitopes, T-Lymphocyte/genetics , Gene Knock-In Techniques , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nippostrongylus/immunology , Protein Structure, Tertiary/genetics , Strongylida Infections/enzymology , Strongylida Infections/immunology , Strongylida Infections/pathology , T-Lymphocyte Subsets/parasitology , Th2 Cells/parasitology , src-Family Kinases/chemistry , src-Family Kinases/genetics , src-Family Kinases/physiology
13.
J Immunol ; 184(3): 1436-44, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042571

ABSTRACT

Mycolactone is a diffusible lipid toxin produced by Mycobacterium ulcerans, the causative agent of a necrotizing skin disease referred to as Buruli ulcer. Intriguingly, patients with progressive lesions display a systemic suppression of Th1 responses that resolves on surgical excision of infected tissues. In this study, we examined the effects of mycolactone on the functional biology of T cells and identified two mechanisms by which mycolactone suppresses cell responsiveness to antigenic stimulation. At noncytotoxic concentrations, mycolactone blocked the activation-induced production of cytokines by a posttranscriptional, mammalian target of rapamycin, and cellular stress-independent mechanism. In addition, mycolactone triggered the lipid-raft association and activation of the Src-family kinase, Lck. Mycolactone-mediated hyperactivation of Lck resulted in the depletion of intracellular calcium stores and downregulation of the TCR, leading to impaired T cell responsiveness to stimulation. These biochemical alterations were not observed when T cells were exposed to other bacterial lipids, or to structurally related immunosuppressors. Mycolactone thus constitutes a novel type of T cell immunosuppressive agent, the potent activity of which may explain the defective cellular responses in Buruli ulcer patients.


Subject(s)
Bacterial Toxins/pharmacology , Immunosuppressive Agents/pharmacology , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Animals , Bacterial Toxins/toxicity , Buruli Ulcer/enzymology , Buruli Ulcer/immunology , Cells, Cultured , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Immunosuppressive Agents/toxicity , Intracellular Fluid/drug effects , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Jurkat Cells , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Macrolides , Mice , Mice, Inbred C57BL , Mycobacterium ulcerans/immunology , Protein Processing, Post-Translational/immunology , T-Lymphocytes/enzymology , Time Factors
14.
J Immunol ; 184(8): 4074-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20228198

ABSTRACT

Regulatory T cell (Treg) development proceeds via a two-step process in which naive CD4(+) thymocytes are first converted into CD4(+)CD25(+)CD122(+)GITR(+)Foxp3(-) Treg progenitors, followed by a second step in which IL-2 converts these Treg progenitors into CD4(+)Foxp3(+) Tregs. The costimulatory molecule CD28 is required for efficient Treg development. However, the stage at which CD28 affects Treg development remains undefined. In this article, we demonstrate that Cd28(-/-) mice lack Treg progenitors. Furthermore, the P(187)YAP motif in the cytoplasmic tail of CD28, which links CD28 to Lck activation, is required for this process. In contrast, the Y(170)MNM motif, which links CD28 to PI3K activation, is not required for Treg progenitor development. Finally, the CD28/Lck pathway was shown to activate the NF-kappaB family of transcription factors. We demonstrate that c-Rel, but not NF-kappaB1, promotes the development of Treg progenitors. Thus, a CD28/c-Rel-dependent pathway is involved in initiating Treg development.


Subject(s)
CD28 Antigens/physiology , Cell Differentiation/immunology , Proto-Oncogene Proteins c-rel/physiology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Animals , CD28 Antigens/biosynthesis , CD28 Antigens/genetics , Cell Differentiation/genetics , Cytoplasm/enzymology , Cytoplasm/genetics , Cytoplasm/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Gene Knock-In Techniques , Interleukin-2/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , NF-kappa B/metabolism , NF-kappa B p50 Subunit/deficiency , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/physiology , Peptide Fragments/genetics , Peptide Fragments/physiology , Protein Binding/genetics , Protein Binding/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Stem Cells/immunology , Stem Cells/metabolism , Stem Cells/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
15.
Nat Med ; 6(4): 429-34, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742150

ABSTRACT

Infections are thought to be important in the pathogenesis of many heart diseases. Coxsackievirus B3 (CVB3) has been linked to chronic dilated cardiomyopathy, a common cause of progressive heart disease, heart failure and sudden death. We show here that the sarcoma (Src) family kinase Lck (p56lck) is required for efficient CVB3 replication in T-cell lines and for viral replication and persistence in vivo. Whereas infection of wild-type mice with human pathogenic CVB3 caused acute and very severe myocarditis, meningitis, hepatitis, pancreatitis and dilated cardiomyopathy, mice lacking the p56lck gene were completely protected from CVB3-induced acute pathogenicity and chronic heart disease. These data identify a previously unknown function of Src family kinases and indicate that p56lck is the essential host factor that controls the replication and pathogenicity of CVB3.


Subject(s)
Cardiomyopathy, Dilated/virology , Coxsackievirus Infections/virology , Enterovirus B, Human/pathogenicity , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Chronic Disease , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/pathology , Encephalomyocarditis virus/pathogenicity , Enterovirus B, Human/physiology , HeLa Cells , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Knockout , Virus Replication , src-Family Kinases/metabolism
16.
Cell Mol Immunol ; 18(3): 644-659, 2021 03.
Article in English | MEDLINE | ID: mdl-32868912

ABSTRACT

Tcf-1 (encoded by Tcf7) not only plays critical roles in promoting T cell development and differentiation but also has been identified as a tumor suppressor involved in preventing T cell malignancy. However, the comprehensive mechanisms of Tcf-1 involved in T cell transformation remain poorly understood. In this study, Tcf7fl/fl mice were crossed with Vav-cre, Lck-cre, or Cd4-cre mice to delete Tcf-1 conditionally at the beginning of the HSC, DN2-DN3, or DP stage, respectively. The defective T cell development phenotypes became gradually less severe as the deletion stage became more advanced in distinct mouse models. Interestingly, consistent with Tcf7-/- mice, Tcf7fl/flVav-cre mice developed aggressive T cell lymphoma within 45 weeks, but no tumors were generated in Tcf7fl/flLck-cre or Tcf7fl/flCd4-cre mice. Single-cell RNA-seq (ScRNA-seq) indicated that ablation of Tcf-1 at distinct phases can subdivide DN1 cells into three clusters (C1, C2, and C3) and DN2-DN3 cells into three clusters (C4, C5, and C6). Moreover, Tcf-1 deficiency redirects bifurcation among divergent cell fates, and clusters C1 and C4 exhibit high potential for leukemic transformation. Mechanistically, we found that Tcf-1 directly binds and mediates chromatin accessibility for both typical T cell regulators and proto-oncogenes, including Myb, Mycn, Runx1, and Lyl1 in the DN1 phase and Lef1, Id2, Dtx1, Fyn, Bcl11b, and Zfp36l2 in the DN2-DN3 phase. The aberrant expression of these genes due to Tcf-1 deficiency in very early T cells contributes to subsequent tumorigenesis. Thus, we demonstrated that Tcf-1 plays stage-specific roles in regulating early thymocyte development and transformation, providing new insights and evidence for clinical trials on T-ALL leukemia.


Subject(s)
Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/pathology , Hepatocyte Nuclear Factor 1-alpha/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Lymphoma, T-Cell/pathology , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Gene Expression Profiling , Lymphocyte Activation , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Exp Med ; 198(7): 1089-102, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-14517277

ABSTRACT

The exact role of major histocompatibility complex (MHC) molecules in the peripheral survival of naive T cells is controversial, as some studies have suggested that they are critically required whereas others have suggested that they are not. Here we controlled for some of the features that differed among the earlier studies, and analyzed both the survival and expansion of naive CD4+ T cells transferred into MHC syngeneic, allogeneic, or MHC negative environments. We found that naive T cells transferred into MHC negative or allogeneic environments often fail to survive because of rejection and/or competition by natural killer (NK) cells, rather than failure to recognize a particular MHC allele. In the absence of NK cells, naive CD4+ T cells survived equally well regardless of the MHC type of the host. There was, however, an MHC requirement for extensive space-induced "homeostatic" expansion. Although the first few divisions occurred in the absence of MHC molecules, the cells did not continue to divide or transit to a CD44hi phenotype. Surprisingly, this MHC requirement could be satisfied by alleles other than the restricting haplotype. Therefore, space-induced expansion and survival are two different phenomena displaying different MHC requirements. Memory CD4+ T cells, whose survival and expansion showed no requirements for MHC molecules at all, dampened the space-induced expansion of naive cells, showing that the two populations are not independent in their requirements for peripheral niches.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Histocompatibility Antigens/physiology , Alleles , Animals , Cell Survival , Hyaluronan Receptors/analysis , Immunologic Memory , Killer Cells, Natural/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Mice , Mice, Inbred C57BL , Models, Immunological , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology
18.
J Exp Med ; 186(10): 1713-24, 1997 Nov 17.
Article in English | MEDLINE | ID: mdl-9362531

ABSTRACT

To investigate the cellular dynamics of ZAP-70, we have studied the distribution and regulation of its intracellular location using a ZAP-70 green fluorescent protein chimera. Initial experiments in epithelial cells indicated that ZAP-70 is diffusely located throughout the quiescent cell, and accumulates at the plasma membrane upon cellular activation, a phenotype enhanced by the coexpression of Lck and the initiation of ZAP-70 kinase activity. Subsequent studies in T cells confirmed this phenotype. Intriguingly, a large amount of ZAP-70, both chimeric and endogenous, resides in the nucleus of quiescent and activated cells. Nuclear ZAP-70 becomes tyrosine phosphorylated upon stimulation via the T cell receptor, indicating that it may have an important biologic function.


Subject(s)
Intracellular Fluid/enzymology , Luminescent Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antigens/genetics , COS Cells , Cell Membrane/enzymology , Cell Membrane/metabolism , Cell Nucleus/chemistry , Cell Nucleus/enzymology , Cell Nucleus/genetics , Clone Cells , Enzyme Activation/genetics , Fluorescent Antibody Technique, Direct , Green Fluorescent Proteins , Humans , Jurkat Cells , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Nuclear Envelope/chemistry , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Staining and Labeling , Tyrosine/metabolism , ZAP-70 Protein-Tyrosine Kinase
19.
J Exp Med ; 188(5): 931-9, 1998 Sep 07.
Article in English | MEDLINE | ID: mdl-9730894

ABSTRACT

The tyrosine kinase p56lck regulates the differentiation and proliferative expansion of pre-T cells. However, nothing is known about other signaling molecules that operate with p56lck to mediate the pleiotropic changes that occur at this stage of thymocyte development. We used a genetic strategy to examine the requirement for the GTPase Rho in p56lck-mediated signals in the thymus. By generating mice double transgenic for a constitutively activated form of p56lck (p56lckF505) and the Rho inhibitor C3 transferase we were able to compare thymocyte development in mice expressing active p56lck on a wild-type or Rho- background. Thymocytes expressing active p56lck show enhanced proliferation of pre-T cells resulting in increased numbers of late pre-T cells, however, this dramatic effect on pre-T cell proliferation is lost when the p56lck transgene is expressed in thymocytes lacking endogenous Rho GTPase function. Expression of active p56lck also generates double positive (DP) thymocytes with low levels of CD2 antigen expression. Again, p56lck cannot prevent expression of CD2 when expressed on a Rho- background. CD4(+)CD8(+) DP cells expressing active p56lck have been shown to lack functional alpha/beta-T cell receptor (TCR) complexes due to p56lck-mediated inhibition of TCR gene Vbeta-Dbeta rearrangement. This inhibition of TCR expression by active p56lck is unimpaired in the absence of Rho function. The signaling pathways that are mediated by p56lck and control thymocyte proliferation, alpha/beta-TCR and CD2 antigen expression can thus be distinguished by their dependency on Rho function.


Subject(s)
GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/physiology , GTPase-Activating Proteins , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Animals , CD2 Antigens/biosynthesis , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Differentiation/immunology , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/antagonists & inhibitors , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Stem Cells/cytology , Stem Cells/enzymology , Stem Cells/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
20.
J Virol ; 83(22): 11528-39, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726522

ABSTRACT

Nef, an important pathogenicity factor of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV), elevates virus replication in vivo. Among other activities, Nef affects T-cell receptor (TCR) signaling via several mechanisms. For HIV-1 Nef these include alteration of the organization and function of the immunological synapse (IS) such as relocalization of the Lck kinase, as well as early inhibition of TCR/CD3 complex (TCR-CD3)-mediated actin rearrangements and tyrosine phosphorylation. Although most SIV and HIV-2 Nef alleles (group 2) potently downregulate cell surface TCR-CD3, this activity was lost in the viral lineage that gave rise to HIV-1 and its SIV counterparts (group 1). To address the contribution of TCR-CD3 downregulation to Nef effects on TCR signal initiation, we compared the activities of 18 group 1 and group 2 Nef proteins, as well as SIV Nef mutants with defects in TCR-CD3 downmodulation. We found that alteration of Lck's subcellular localization is largely conserved and occurs independently of actin remodeling inhibition or TCR-CD3 downregulation. Surprisingly, Nef proteins of both groups also strongly reduced TCR-induced actin remodeling and tyrosine phosphorylation on TCR-stimulatory surfaces and TCR-CD3 downmodulation competence by group 2 Nef proteins only slightly elevated these effects. Furthermore, Nef proteins from HIV-1 and SIV reduced conjugation between infected primary human T lymphocytes and Raji B cells and potently prevented F-actin polarization at the IS independently of their ability to downmodulate TCR-CD3. These results establish alterations of early TCR signaling events at the IS, including F-actin remodeling and relocalization of Lck, as evolutionary conserved activities of highly divergent lentiviral Nef proteins.


Subject(s)
Actins/physiology , HIV-1/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Receptors, Antigen, T-Cell/physiology , nef Gene Products, Human Immunodeficiency Virus/physiology , Conserved Sequence , Down-Regulation , Humans , Jurkat Cells , Receptor-CD3 Complex, Antigen, T-Cell/physiology , Viral Regulatory and Accessory Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL