Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Front Immunol ; 11: 1995, 2020.
Article in English | MEDLINE | ID: mdl-32983140

ABSTRACT

Most patients with chronic lymphocytic leukemia (CLL) exhibit an indolent disease course and unresponsive B cell receptors (BCRs) exemplified by an anergic phenotype of their leukemic cells. In up to 5% of patients, CLL transforms from an indolent subtype to an aggressive form of B cell lymphoma (Richter's syndrome), which is associated with worse disease outcome and severe downregulation of NFAT2. Here we show that ablation of the tyrosine kinase LCK, which has previously been characterized as a main NFAT2 target gene in CLL, leads to loss of the anergic phenotype, thereby restoring BCR signaling, which results in an acceleration of CLL. Our study identifies LCK as a main player in mediating BCR unresponsiveness and its role as a crucial regulator of anergy in CLL.


Subject(s)
Biomarkers, Tumor , Gene Deletion , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Disease Models, Animal , Disease Progression , Disease Susceptibility , Immunophenotyping , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Mice , Mice, Knockout , Phenotype , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism
2.
Cell Mol Immunol ; 17(2): 143-152, 2020 02.
Article in English | MEDLINE | ID: mdl-30523347

ABSTRACT

The B7-family inducible costimulator (ICOS) activates phosphoinositide-3 kinase (PI3K) and augments calcium mobilization triggered by the T-cell receptor (TCR). We surprisingly found that the entire cytoplasmic domain of ICOS is dispensable for its costimulation of calcium mobilization. This costimulatory function relies on the unique transmembrane domain (TMD) of ICOS, which promotes association with the tyrosine kinase Lck. TMD-enabled Lck association is also required for p85 recruitment to ICOS and subsequent PI3K activation, and Lck underlies both the bystander and costimulatory signaling activity of ICOS. TMD-replaced ICOS, even with an intact cytoplasmic domain, fails to support TFH development or GC formation in vivo. When transplanted onto a chimeric antigen receptor (CAR), the ICOS TMD enhances interactions between T cells and antigen-presenting target cells. Therefore, by revealing an unexpected function of the ICOS TMD, our study offers a new perspective for the understanding and potential application of costimulation biology.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Protein Domains/genetics , Receptors, Chimeric Antigen/metabolism , Signal Transduction/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , Calcium/metabolism , Cell Communication/immunology , HEK293 Cells , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Knockout , Mutant Proteins , Phosphatidylinositol 3-Kinases/metabolism , Protein Domains/immunology , Receptors, Chimeric Antigen/genetics , Signal Transduction/immunology , Transduction, Genetic
3.
Mol Cell ; 67(3): 498-511.e6, 2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28735895

ABSTRACT

The Src Family kinase Lck sets a critical threshold for T cell activation because it phosphorylates the TCR complex and the Zap70 kinase. How a T cell controls the abundance of active Lck molecules remains poorly understood. We have identified an unappreciated role for a phosphosite, Y192, within the Lck SH2 domain that profoundly affects the amount of active Lck in cells. Notably, mutation of Y192 blocks critical TCR-proximal signaling events and impairs thymocyte development in retrogenic mice. We determined that these defects are caused by hyperphosphorylation of the inhibitory C-terminal tail of Lck. Our findings reveal that modification of Y192 inhibits the ability of CD45 to associate with Lck in cells and dephosphorylate the C-terminal tail of Lck, which prevents its adoption of an active open conformation. These results suggest a negative feedback loop that responds to signaling events that tune active Lck amounts and TCR sensitivity.


Subject(s)
Leukocyte Common Antigens/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Thymocytes/enzymology , src Homology Domains , Animals , Enzyme Activation , Genotype , HEK293 Cells , Humans , Jurkat Cells , Leukocyte Common Antigens/chemistry , Leukocyte Common Antigens/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Mutation , Phenotype , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Thymocytes/immunology , Time Factors , Transfection
4.
Am J Pathol ; 186(7): 1814-1824, 2016 07.
Article in English | MEDLINE | ID: mdl-27157991

ABSTRACT

The protein Lck (p56(Lck)) is a Src family tyrosine kinase expressed at all stages of thymocyte development and is required for maturation of T cells. The targeted disruption of Lck gene in mice results in severe block in thymocyte maturation with substantial reduction in the development of CD4(+)CD8(+) thymocytes, severe reduction of peripheral T cells, and disruption of T-cell receptor signaling with defective function of T-cell responses. To investigate the role of T lymphocyte in the development of cigarette smoke-induced pulmonary changes, Lck(-/-) mice and corresponding congenic wild-type mice were chronically exposed to cigarette smoke, and their lungs were analyzed by biochemical, immunologic, and morphometric methods. Smoking mice from both genotypes showed disseminated foci of emphysema and large areas of goblet cell metaplasia in bronchial and bronchiolar epithelium. Morphometric evaluation of lung changes and lung elastin determination confirmed that mice from both genotypes showed the same degree of emphysematous lesions. Thus, cigarette smoke exposure in the presence of severe reduction in number and function of peripheral T cells does not influence the development of pulmonary changes induced by cigarette smoke. The data obtained suggest that innate immunity is a leading actor in the early development of pulmonary changes in smoking mice and that the adaptive immune response may play a role at later stages.


Subject(s)
Pulmonary Emphysema/immunology , Smoking/adverse effects , T-Lymphocytes/immunology , Animals , Bronchi/pathology , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Emphysema/pathology , Real-Time Polymerase Chain Reaction , Smoking/immunology
5.
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
6.
Apoptosis ; 14(5): 711-20, 2009 May.
Article in English | MEDLINE | ID: mdl-19343496

ABSTRACT

Ionising radiation, hypoxia, and the cyclooxygenase-2 inhibitor Celecoxib are known agonists of the intrinsic apoptosis pathway that involves mitochondrial damage upstream of caspase activation. Mitochondrial integrity is regulated by the pro-apoptotic Bcl-2 protein family members Bak and Bax. Upstream of the mitochondria, many kinases and phosphatases control the apoptotic response. However, the role of the non-receptor tyrosine kinase p56/Lck during apoptosis is controversial. The present investigation demonstrate the existence of two JCaM1.6 subclones, one expressing and one deficient for Bak. The lack of p56/Lck expression in JCaM1.6 cells per se did hardly affect apoptosis induced by ionising radiation, hypoxia, or Celecoxib. Only the additional loss of Bak expression, as observed in one JCaM1.6 subclone, rendered the cells resistant. siRNA-mediated downregulation of Bak and p56/Lck mimicked the observed effects in the subclones. Earlier experiments performed with the Bak-negative clone might have lead to the wrong assumption that lack of p56/Lck alone, and not the additonal loss of Bak, was responsible for reduced sensitivity towards stimuli of the intrinsic apoptosis pathway.


Subject(s)
Apoptosis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/deficiency , Apoptosis/drug effects , Apoptosis/radiation effects , Celecoxib , Cell Line, Tumor , Clone Cells , Gene Silencing/drug effects , Gene Silencing/radiation effects , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Pyrazoles/pharmacology , Radiation, Ionizing , Signal Transduction/drug effects , Signal Transduction/radiation effects , Sulfonamides/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism
7.
J Immunol ; 181(12): 8425-32, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050260

ABSTRACT

The Nef protein of HIV-1 is a key promoter of disease progression, owing to its dramatic yet ill-defined impact on viral replication. Previously, we have shown that Nef enhances Tat-mediated transcription in a manner depending on Lck and the cytoplasmic sequestration of the transcriptional repressor embryonic ectodermal development [corrected]. In this study, we report that Lck is activated by Nef and targets protein kinase Ctheta downstream, leading to the translocation of the kinase into membrane microdomains. Although microdomain-localized protein kinase Ctheta is thought to induce the transcription factor NFkappaB, we unexpectedly failed to correlate Nef-induced signaling events with enhanced NFkappaB activity. Instead, we observed an increase in ERK MAPK activity. We conclude that Nef-mediated signaling cooperates with Nef-induced derepression and supports HIV transcription through an ERK MAPK-dependent, but NFkappaB-independent, pathway.


Subject(s)
HIV-1/genetics , Isoenzymes/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Microdomains/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C/physiology , nef Gene Products, Human Immunodeficiency Virus/physiology , Cell Line , Enzyme Activation/genetics , Gene Expression Regulation, Viral , Humans , Isoenzymes/deficiency , Isoenzymes/genetics , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , MAP Kinase Signaling System/genetics , Membrane Microdomains/virology , NF-kappa B/metabolism , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C-theta , Protein Transport/genetics , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Up-Regulation/genetics
8.
Sci STKE ; 2006(358): pe45, 2006 Oct 24.
Article in English | MEDLINE | ID: mdl-17062896

ABSTRACT

Some bacterial and viral proteins are potent activators of the immune response, earning them the title of superantigens (SAgs). Infection with pathogens containing these proteins can produce massive T cell activation and can result in various potentially fatal conditions, such as toxic shock and food poisoning. Unlike conventional peptide antigens, SAgs bind promiscuously to the external faces of class II major histocompatibility complex (MHC) molecules and families of T cell receptors (TCRs), thereby activating large numbers of T cells simultaneously. The manner in which SAgs bind MHC and TCR differs from the way in which peptide antigens interact with these structures. Nevertheless, because they simultaneously engage MHC and TCR, SAgs were assumed to activate T cells through the canonical signaling pathway that has been described for T cell activation by TCR engagement of peptide-MHC complexes. However, recent research shows that SAgs also activate an alternative signaling pathway in T cells. This study shows that SAgs can stimulate T cells in the absence of the Src family kinase, Lck, by activating a heterotrimeric guanine nucleotide-binding protein (G protein), Galpha(11). Galpha(11) activates phospholipase C-beta (PLC-beta), rather than the more abundant PLC-gamma1, and, by this means, links SAg signaling to the phosphatidylinositol and protein kinase C signaling pathways. The discovery of a signaling pathway specifically activated by SAgs, and not by conventional peptide antigens, opens the possibility of developing therapeutic reagents that may help control diseases caused by these agents.


Subject(s)
Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology , Superantigens/physiology , Antigens, Bacterial/immunology , Antigens, Bacterial/physiology , Antigens, Viral/immunology , Antigens, Viral/physiology , Calcium Signaling , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Isoenzymes/physiology , Jurkat Cells , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Major Histocompatibility Complex/immunology , Models, Immunological , Phospholipase C beta , Receptors, Antigen, T-Cell/immunology , Superantigens/immunology , T-Lymphocyte Subsets/immunology , Type C Phospholipases/physiology
9.
Antioxid Redox Signal ; 8(9-10): 1749-56, 2006.
Article in English | MEDLINE | ID: mdl-16987028

ABSTRACT

Oxidative stress can activate a variety of intracellular signaling pathways. The authors previously reported the CaM-K inhibitor KN-93 inhibited hydrogen peroxide-induced phosphorylation of Akt on threonine 308 (T308). In this report they demonstrate that phosphorylation of T308 in response to hydrogen peroxide treatment is not inhibited by LY294002, suggesting that phosphorylation of this residue in response to oxidative stress is largely PI3K independent. In contrast, hydrogen peroxide-induced phosphorylation of Akt on serine 473 (S473) was downregulated by both PI3K and CaM-K inhibition, indicating that hydrogen peroxideinduced phosphorylation of Akt on S473 was largely dependent on both PI3K and a CaM-K activity. Further, it is reported that p56(Lck) had a substantial role in hydrogen peroxide-induced phosphorylation of S473, but only a minimal role in hydrogen peroxide-induced phosphorylation of T308. These data suggest that in response to hydrogen peroxide, two pathways are activated in Jurkat T lymphocytes that converge to result in the phosphorylation of Akt on S473 and T308. One pathway involves the CaM-Ks that may directly phosphorylate Akt on T308. In this pathway, neither the Src kinases nor PI3K are required. The other pathway mediated by hydrogen peroxide results in the phosphorylation of Akt on S473 and requires CaM-K, PI3K, and Src activity.


Subject(s)
Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carbazoles/pharmacology , Cell Line, Tumor , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Hydrogen Peroxide/pharmacology , Indole Alkaloids , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Models, Biological , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
J Immunol ; 175(11): 7398-406, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301647

ABSTRACT

The Src family kinase Lck is essential for T cell Ag receptor-mediated signaling. In this study, we report the effects of acute elimination of Lck in Jurkat TAg and primary T cells using RNA interference mediated by short-interfering RNAs. In cells with Lck knockdown (kd), proximal TCR signaling was strongly suppressed as indicated by reduced zeta-chain phosphorylation and intracellular calcium mobilization. However, we observed sustained and elevated phosphorylation of ERK1/2 in Lck kd cells 30 min to 2 h after stimulation. Downstream effects on immune function as determined by activation of a NFAT-AP-1 reporter, and TCR/CD28-stimulated IL-2 secretion were strongly augmented in Jurkat and primary T cells, respectively. As expected, overexpression of SHP-1 in Jurkat cells inhibited TCR-induced NFAT-AP-1 activation, but this effect could be overcome by simultaneous kd of Lck. Furthermore, acute elimination of Lck also suppressed TCR-mediated activation of SHP-1, suggesting the possible role of SHP-1 in a negative feedback loop originating from Lck. This report underscores Lck as an important mediator of proximal TCR signaling, but also indicates a suppressive role on downstream immune function.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Blotting, Northern , CD28 Antigens/immunology , CD28 Antigens/metabolism , Calcium/metabolism , Cell Line, Tumor , Enzyme Activation/immunology , Humans , Immunoprecipitation , Interleukin-2/immunology , Interleukin-2/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/immunology , Protein Tyrosine Phosphatases/metabolism , RNA, Small Interfering , Transfection
11.
J Immunol ; 174(12): 7807-14, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15944285

ABSTRACT

The current model of T cell activation is that TCR engagement stimulates Src family tyrosine kinases (SFK) to phosphorylate CD3zeta. CD3zeta phosphorylation allows for the recruitment of the tyrosine kinase ZAP70, which is phosphorylated and activated by SFK, leading to the phosphorylation of downstream targets. We stimulated mouse CTLs with plate-bound anti-CD3 and, after cell lysis, recovered proteins that associated with the CD3 complex. The protein complexes were not preformed, and a number of tyrosine-phosphorylated proteins were inducibly and specifically associated with the TCR/CD3 complex. These results suggest that complex formation only occurs at the site of TCR engagement. The recruitment and tyrosine phosphorylation of most proteins were abolished when T cells were stimulated in the presence of the SFK inhibitor PP2. Surprisingly, CD3zeta, but not CD3epsilon, was inducibly tyrosine phosphorylated in the presence of PP2. Furthermore, ZAP70 was recruited, but not phosphorylated, after TCR stimulation in the presence of PP2, thus confirming the phosphorylation status of CD3zeta. These data suggest that there is a differential requirement for SFK activity in phosphorylation of CD3zeta vs CD3epsilon. Consistent with this possibility, ZAP70 recruitment was also detected with anti-CD3-stimulated, Lck-deficient human Jurkat T cells. We conclude that TCR/CD3-induced CD3zeta phosphorylation and ZAP70 recruitment do not absolutely require Lck or other PP2-inhibitable SFK activity, but that SFK activity is absolutely required for CD3epsilon and ZAP70 phosphorylation. These data reveal the potential for regulation of signaling through the TCR complex by the differential recruitment or activation of SFK.


Subject(s)
CD3 Complex/metabolism , Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Animals , CD3 Complex/immunology , Clone Cells , Enzyme Activation , Humans , Immune Sera/pharmacology , Immunosuppressive Agents/pharmacology , Jurkat Cells , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tyrosine/metabolism , ZAP-70 Protein-Tyrosine Kinase , src-Family Kinases/antagonists & inhibitors
12.
J Immunol ; 175(1): 27-31, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15972627

ABSTRACT

B-1 cells constitute a unique B cell subset that is primarily responsible for producing nonimmune Ig. This natural Ig acts as a principal line of defense against infection. A key feature of B-1 cells is the failure of BCR-triggered signal transduction. Recently, defective BCR signaling in B-1 cells has been attributed to elevated expression of the canonical T cell src kinase, Lck. In the present study, we re-examined Lck expression in normal B-1 cells. We found that B-1 cells expressed less Lck at both the protein and RNA levels than did B-2 cells. The same B-1 cells manifested defective BCR-mediated induction of IKKbeta phosphorylation, IkappaBalpha degradation, and intracellular Ca(2+) mobilization. Thus, the failure of BCR signaling in B-1 cells does not relate to subset-specific elevation of Lck.


Subject(s)
B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Animals , Base Sequence , Calcium Signaling , DNA, Complementary/genetics , I-kappa B Kinase , I-kappa B Proteins/metabolism , In Vitro Techniques , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
13.
J Immunol ; 174(10): 6105-12, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879105

ABSTRACT

Tid1, a DnaJ cochaperone protein, is the mammalian homologue of the Drosophila tumor suppressor Tid56 whose antitumor function is most likely mediated through its capacity to regulate cell differentiation in imaginal discs. We suspected that the mammalian counterpart, tid1, may also be involved in regulating cell differentiation. To investigate this, we exploited the system of T cell development to examine whether tid1 plays a role in this well-defined process. Mice with tid1 specifically deleted in T cells developed thymic atrophy, with dramatic reduction of double-positive and single-positive thymocytes in the tid1(-/-) thymus. Although the subpopulations of tid1(-/-) double-negative (DN) 1-3 thymocytes were normal, the subpopulation of DN4 thymocytes was measurably smaller because of reduced proliferation and significant cell death. Immature tid1(-/-) thymocytes show normal VDJ beta-chain rearrangement and pre-TCR and CD3 expression in both DN3 and DN4 thymocytes, but in DN4 thymocytes, there was significantly reduced expression of the antiapoptotic bcl-2 gene. Restoring the expression level of Bcl-2 protein in tid1(-/-) thymus by introduction of a transgenic human bcl-2 gene resulted in reversal of the developmental defects in tid1(-/-) thymus. Together, these results demonstrate that tid1 is critical in early thymocyte development, especially during transition from the DN3 to double-positive stages, possibly through its regulation of bcl-2 expression, which provides survival signals.


Subject(s)
Cell Differentiation/immunology , Heat-Shock Proteins/physiology , T-Lymphocyte Subsets/cytology , Adjuvants, Immunologic/deficiency , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/physiology , Animals , Cell Death/genetics , Cell Death/immunology , Cell Differentiation/genetics , Cytokines/biosynthesis , Cytokines/physiology , Epitopes, T-Lymphocyte/immunology , Gene Expression Profiling , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Gene Silencing , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Growth Inhibitors/physiology , HSP40 Heat-Shock Proteins , Heat-Shock Proteins/deficiency , Heat-Shock Proteins/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Thymus Gland/cytology , Thymus Gland/metabolism
14.
Proc Natl Acad Sci U S A ; 101(41): 14859-64, 2004 Oct 12.
Article in English | MEDLINE | ID: mdl-15465914

ABSTRACT

Although there was no impairment in IL-2 secretion and proliferation of Fyn-deficient naïve CD4 cells after stimulation with antigen and antigen-presenting cells, stimulation of these cells with anti-CD3 and anti-CD28 revealed profound defects. Crosslinking of purified wild-type naïve CD4 cells with anti-CD3 activated Lck and initiated the signaling cascade downstream of Lck, including phosphorylation of ZAP-70, LAT, and PLC-gamma1; calcium flux; and dephosphorylation and nuclear translocation of the nuclear factor of activated T cells (NFAT)p. All of these signaling events were diminished severely in Fyn-deficient naïve cells activated by CD3 crosslinking. Coaggregation of CD3 and CD4 reconstituted this Lck-dependent signaling pathway in Fyn(-/-) T cells. These results suggest that when signaling of naïve T cells is restricted to the T cell antigen receptor, Fyn plays an essential role by positive regulation of Lck activity.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Calcium/metabolism , Cell Culture Techniques , Interleukin-2/metabolism , Lymph Nodes/immunology , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation , Spleen/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
15.
Biochem Pharmacol ; 67(10): 1859-72, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15130763

ABSTRACT

Induction of apoptosis is a hallmark of the cellular response of human lymphocytes and lymphoma cells to treatment with anticancer drugs and irradiation. Both treatment modalities trigger apoptosis through intrinsic, mitochondrial apoptosis pathways resulting in the activation of caspases. We and others have shown that the tyrosine kinase p56/Lck is involved in the regulation of apoptosis induced by irradiation or treatment with ceramide but dispensable for death receptor triggered cell death. However, the role of p56/Lck for apoptosis induction in response to anticancer drugs is unclear. To elucidate the putative requirement of p56/Lck for apoptosis signaling of cytotoxic drugs, activation of caspases and alteration of mitochondrial functions were determined in Jurkat T cells, the p56/Lck deficient JCaM1.6 cells and the p56/Lck retransfected JCaM1.6/Lck cells in response to chemotherapeutic drugs with different targets of their primary action. Treatment with Doxorubicin, Paclitaxel or 5-Fluorouracil induced a breakdown of the mitochondrial membrane potential and apoptotic cell death in p56/Lck expressing Jurkat and the retransfected JCaM1.6/Lck cells within 48h of treatment. However, almost no mitochondrial alterations and no induction of apoptosis could be detected in the p56/Lck deficient JCaM1.6 cells. Correspondingly, activation of caspases-9, -8, and -3 and cleavage of the caspase-3 substrate PARP (poly-(ADP-ribose)-polymerase) were almost completely absent in JCaM1.6 cells while present in p56/Lck positive Jurkat and JCaM1.6/Lck cells. In contrast, retransfection of the cells with the p56/Lck-related tyrosine kinase Src could not restore sensitivity to the treatment with cytotoxic drugs indicating a specific role of the tyrosine kinase p56/Lck in apoptosis signaling. Importantly, kinase-activity of p56/Lck may be dispensable for its pro-apoptoptic action since preincubation with the Src-kinase inhibitor PP2 did not reduce apoptosis induced by cytotoxic drugs. In conclusion, the tyrosine kinase p56/Lck is essential for apoptosis induction by Doxorubicin, Paclitaxel and 5-Fluorouracil regulating early steps of the mitochondrial apoptosis signaling cascade, including alteration of mitochondrial functions and caspase-activation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Caspase 3 , Caspases/metabolism , Doxorubicin/pharmacology , Fluorouracil/pharmacology , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mitochondria/drug effects , Mitochondria/enzymology , Paclitaxel/pharmacology , Protein-Tyrosine Kinases/physiology , Signal Transduction/drug effects
16.
J Immunol ; 172(10): 6093-100, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15128794

ABSTRACT

NK T cells are a unique lymphocyte population that have developmental requirements distinct from conventional T cells. Mice lacking the tyrosine kinase Fyn have 5- to 10-fold fewer mature NK T cells. This study shows that Fyn-deficient mice have decreased numbers of NK1.1(-) NK T cell progenitors as well. 5-Bromo-2'-deoxyuridine-labeling studies indicate that the NK T cells remaining in fyn(-/-) mice exhibit a similar turnover rate as wild-type cells. The fyn(-/-) NK T cells respond to alpha-galactosylceramide, a ligand recognized by NK T cells, and produce cytokines, but have depressed proliferative capacity. Transgenic expression of the NK T cell-specific TCR alpha-chain Valpha14Jalpha18 leads to a complete restoration of NK T cell numbers in fyn(-/-) mice. Together, these results suggest that Fyn may have a role before alpha-chain rearrangement rather than for positive selection or the peripheral upkeep of cell number. NK T cells can activate other lymphoid lineages via cytokine secretion. These secondary responses are impaired in Fyn-deficient mice, but occur normally in fyn mutants expressing the Valpha14Jalpha18 transgene. Because this transgene restores NK T cell numbers, the lack of secondary lymphocyte activation in the fyn-mutant mice is due to the decreased numbers of NK T cells present in the mutant, rather than an intrinsic defect in the ability of the other fyn(-/-) lymphoid populations to respond.


Subject(s)
Killer Cells, Natural/cytology , Killer Cells, Natural/enzymology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , Animals , Antigens/metabolism , Antigens, Ly , Antigens, Surface , B-Lymphocyte Subsets/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Ceramides/pharmacology , Cytokines/metabolism , Genes, T-Cell Receptor alpha , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lectins, C-Type , Lymphocyte Activation , Lymphocyte Count , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B , Protein-Tyrosine Kinases/deficiency , Proteins/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-fyn , Receptors, Antigen, T-Cell, alpha-beta/genetics , Stem Cells/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transgenes/immunology
17.
J Immunol ; 172(3): 1437-43, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734719

ABSTRACT

TCR engagement can induce either T cell proliferation and differentiation or activation-induced T cell death (AICD) through apoptosis. The intracellular signaling pathways that dictate such a disparate fate after TCR engagement have only been partially elucidated. Non-FcR-binding anti-CD3 mAbs induce a partial agonist TCR signaling pattern and cause AICD on Ag-activated, cycling T cells. In this study, we examined TCR signaling during the induction of AICD by anti-CD3 fos, a non-FcR-binding anti-CD3 mAb. This mAb activates Fyn, Lck, and extracellular signal-regulated kinase, and induces phosphorylation of Src-like adapter protein, despite the inability to cause calcium mobilization or TCR polarization. Anti-CD3 fos also fails to effectively activate zeta-associated protein of 70 kDa or NF-kappaB. Using Ag-specific T cells deficient for Fyn or Lck, we provide compelling evidence that activation of Lck is required for the induction of AICD. Our data indicate that a selective and distinct TCR signaling pattern is required for AICD by TCR partial agonist ligands.


Subject(s)
Apoptosis/immunology , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Receptors, Antigen, T-Cell/agonists , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antigens/pharmacology , Apoptosis/genetics , CD3 Complex/immunology , Calcium/metabolism , Calcium/physiology , Cells, Cultured , Enzyme Activation/immunology , Fas Ligand Protein , Immunoglobulin Fab Fragments/genetics , Ligands , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Ovalbumin/pharmacology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fyn , Receptors, Antigen, T-Cell/immunology , Solubility , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology , Up-Regulation/immunology , fas Receptor/physiology , src-Family Kinases/metabolism , src-Family Kinases/physiology
18.
J Immunol ; 172(1): 79-87, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14688312

ABSTRACT

Apoptosis is one way of controlling immune responses, and a variety of immunosuppressive drugs suppress harmful immune responses by inducing apoptosis of lymphocytes. In this study we observed that rosmarinic acid, a secondary metabolite of herbal plants, induced apoptosis in an p56(lck) (Lck)-dependent manner; Lck(+) Jurkat T cells undergo apoptosis in response to rosmarinic acid (RosA) treatment, whereas Lck(-) Jurkat subclone J.CaM1.6 cells do not. J.CaM1.6 cells with various Lck mutants indicated that Lck SH2 domain, but not Lck kinase activity, was required for RosA-induced apoptosis. RosA induced apoptosis in the absence of a TCR stimulus, and this was not prevented by interruption of the Fas/Fas ligand interaction. Instead, RosA-mediated apoptosis involved a mitochondrial pathway as indicated by cytochrome c release and the complete blockage of apoptosis by an inhibitor of mitochondrial membrane depolarization. Both caspase-3 and -8 were indispensable in RosA-induced apoptosis and work downstream of mitochondria and caspase-9 in the order of caspase-9/caspase-3/caspase-8. In freshly isolated human PBMC, RosA specifically induced apoptosis of Lck(+) subsets such as T and NK cells, but not Lck-deficient cells, including B cells and monocytes. Moreover, RosA's ability to kill T and NK cells was restricted to actively proliferating cells, but not to resting cells. In conclusion, Lck-dependent apoptotic activity may make RosA an attractive therapeutic tool for the treatment of diseases in which T cell apoptosis is beneficial.


Subject(s)
Apoptosis/drug effects , Cinnamates/pharmacology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Membrane Glycoproteins/physiology , Mitochondria/physiology , Signal Transduction/immunology , T-Lymphocyte Subsets/cytology , fas Receptor/physiology , Apoptosis/immunology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , CD3 Complex/physiology , Caspase 3 , Caspase 8 , Caspases/metabolism , Depsides , Fas Ligand Protein , Humans , Hydrolysis/drug effects , Intracellular Membranes/drug effects , Intracellular Membranes/enzymology , Intracellular Membranes/immunology , Jurkat Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/enzymology , Ligands , Lymphocyte Activation/drug effects , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Membrane Glycoproteins/biosynthesis , Membrane Potentials/drug effects , Membrane Potentials/immunology , Mitochondria/drug effects , Mitochondria/enzymology , Monocytes/cytology , Monocytes/drug effects , Monocytes/enzymology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/enzymology , Transfection , fas Receptor/biosynthesis , src Homology Domains/drug effects , src Homology Domains/immunology , Rosmarinic Acid
19.
J Immunol ; 171(2): 519-23, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12847211

ABSTRACT

To understand the regulatory activities of kinases in vivo requires their study across a biologically relevant window of activity. To this end, ATP analog-sensitive kinase alleles (ASKAs) specifically sensitive to a competitive inhibitor have been developed. This article tests whether ASKA technology can be applied to complex immunological systems, such as lymphoid development. The results show that when applied to reaggregate thymic organ culture, novel p56(Lck) ASKAs readily expose a dose-dependent correlation of thymocyte development with a range of p56(Lck) activity. By regulating kinase activity, rather than amounts of RNA or protein, ASKA technology offers a general means for assessing the quantitative contributions to immunology of numerous kinases emerging from genomics analyses. It can obviate the generation of multiple lines of mice expressing different levels of kinase transgenes and should permit specific biological effects to be associated with defined biochemical activities.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Adenosine Triphosphate/chemistry , Alleles , Animals , Cell Aggregation/genetics , Cell Aggregation/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Enzyme Activation/genetics , Enzyme Activation/immunology , Fetus , Genetic Complementation Test , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Mice , Mice, Knockout , Mice, Transgenic , Mutagenesis, Site-Directed , Organ Culture Techniques , Retroviridae/genetics , Substrate Specificity/genetics , Substrate Specificity/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Transduction, Genetic
20.
Eur J Immunol ; 33(2): 441-5, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12645942

ABSTRACT

Mature B cells are grouped into two major subsets, B-1 and B-2, believed to derive from separate lineages. We have recently shown that B-1 cells, which are characterized by CD5 surface expression, specifically exhibit significant levels of the tyrosine kinase Lck in man. Here we show that also in mice Lck expression is restricted to B-1 cells and address the potential role of Lck in B-1 cell development and activation. Using as a model an Lck-/- mouse, we show that, while dispensable for B-1 cell development, Lck is required for full and sustained activation of the tyrosine phosphorylation and MAP kinase cascades triggered by the BCR in CD5+, B-1 cells. The data suggest a potential role for Lck in the achievement of the higher activation threshold required for productive BCR signaling in B-1 as compared to B-2 cells.


Subject(s)
B-Lymphocyte Subsets/enzymology , Lymphocyte Activation/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Receptors, Antigen, B-Cell/immunology , Animals , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , CD5 Antigens/analysis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/deficiency , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Peritoneal Cavity/cytology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/deficiency , Phosphorylation , Protein Processing, Post-Translational/physiology , Signal Transduction , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/immunology , ras Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...