Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
J Biol Chem ; 276(48): 45207-16, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11487585

ABSTRACT

B cell linker protein (BLNK) is a SLP-76-related adaptor protein essential for signal transduction from the BCR. To identify components of BLNK-associated signaling pathways, we performed a phosphorylation-dependent yeast two-hybrid analysis using BLNK probes. Here we report that the serine/threonine kinase hematopoietic progenitor kinase 1 (HPK1), which is activated upon antigen-receptor stimulation and which has been implicated in the regulation of MAP kinase pathways, interacts physically and functionally with BLNK in B cells and with SLP-76 in T cells. This interaction requires Tyr(379) of HPK1 and the Src homology 2 (SH2) domain of BLNK/SLP-76. Via homology modeling, we defined a consensus binding site within ligands for SLP family SH2 domains. We further demonstrate that the SH2 domain of SLP-76 participates in the regulation of AP-1 and NFAT activation in response to T cell receptor (TCR) stimulation and that HPK1 inhibits AP-1 activation in a manner partially dependent on its interaction with SLP-76. Our data are consistent with a model in which full activation of HPK1 requires its own phosphorylation on tyrosine and subsequent interaction with adaptors of the SLP family, providing a mechanistic basis for the integration of this kinase into antigen receptor signaling cascades.


Subject(s)
Carrier Proteins/metabolism , Lymphocytes/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Cell Line , DNA, Complementary/metabolism , Databases as Topic , Enzyme Activation , Humans , Immunoblotting , Jurkat Cells , Mice , Mice, Inbred C57BL , Models, Biological , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , T-Lymphocytes/metabolism , Transcription Factor AP-1/metabolism , Two-Hybrid System Techniques , Up-Regulation , src Homology Domains
2.
Proc Natl Acad Sci U S A ; 98(16): 9312-7, 2001 Jul 31.
Article in English | MEDLINE | ID: mdl-11470902

ABSTRACT

In neuronal cells, presenilin-dependent gamma-secretase activity cleaves amyloid precursor proteins to release Abeta peptides, and also catalyzes the release of the intracellular domain of the transmembrane receptor Notch. Accumulation of aberrant Abeta peptides appears to be causally related to Alzheimer's disease. Inhibition of Abeta peptide production is therefore a potential target for therapeutic intervention. Notch proteins play an important role in cell fate determination in many different organisms and at different stages of development, for example in mammalian T cell development. We therefore addressed whether structurally diverse gamma-secretase inhibitors impair Notch function by studying thymocyte development in murine fetal thymic organ cultures. Here we show that high concentrations of the most potent inhibitors blocked thymocyte development at the most immature stage. In contrast, lower concentrations or less potent inhibitors impaired differentiation at a later stage, most notably suppressing the development of CD8 single-positive T cells. These phenotypes are consistent with an impairment of Notch signaling by gamma-secretase inhibitors and define a strict Notch dose dependence of consecutive stages during thymocyte development.


Subject(s)
Endopeptidases/metabolism , Membrane Proteins/physiology , Thymus Gland/embryology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Female , Flow Cytometry , Mice , Mice, Transgenic , Organ Culture Techniques , Presenilin-1 , Presenilin-2 , Thymus Gland/cytology
3.
J Biol Chem ; 276(21): 18082-9, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11278409

ABSTRACT

The lck gene encodes a protein-tyrosine kinase that plays a key role in signaling mediated through T cell receptor (TCR) and pre-TCR complexes. Transcription of the lck gene is regulated by two independent promoter elements: the proximal and distal promoters. Previous studies employing transgenic mice demonstrated that the sequence between -584 and -240 from the transcription start site in the mouse lck proximal promoter is required for its tissue-specific expression in the thymus. In this study, we demonstrate that a Krüppel-like zinc finger protein, mtbeta (BFCOL1, BERF-1, ZBP-89, ZNF148), previously cloned as a protein that binds to the CD3delta gene enhancer, binds to the -365 to -328 region of the lck proximal promoter. mtbeta is ubiquitously expressed in various cell lines and mouse tissues. Overexpressed mtbeta is more active in T-lineage cells than B-lineage cells for transactivating an artificial promoter consisting of the mtbeta binding site and a TATA box. Activity of the lck proximal promoter was significantly impaired by mutating the mtbeta binding site or by reducing mtbeta protein expression level by using antisense mRNA. Our results indicate that mtbeta activity is regulated in a tissue-specific manner and that mtbeta is a critical transactivator for the lck proximal promoter.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Animals , Cell Line , Humans , Promoter Regions, Genetic/genetics , T-Lymphocytes , Transcription, Genetic
4.
J Immunol ; 166(4): 2209-17, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160274

ABSTRACT

p56(lck) is a protein tyrosine kinase expressed throughout T cell development. It associates noncovalently with the cytoplasmic domains of the CD4 and CD8 coreceptor molecules and has been implicated in TCR signaling in mature T cells. Its role in early thymocyte differentiation has been demonstrated in vivo, both by targeted gene disruption and by transgene expression. Previously, we showed that expression of a dominant-negative form of p56(lck) in double-positive thymocytes inhibits positive selection. We now demonstrate that expression of constitutively activated p56(lck) (p56(lck)F505) accelerates the transition from the double-positive to the single-positive stage. Importantly, p56(lck)F505 drives survival and lineage commitment of thymocytes in the absence of TCR engagement by appropriate MHC molecules. These results indicate that activation of p56(lck) constitutes an early step in conveying maturational signals after TCR ligation by a positively selecting ligand. Our study provides direct in vivo evidence for the role of p56(lck) in regulating TCR signaling.


Subject(s)
CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/enzymology , Thymus Gland/cytology , Thymus Gland/enzymology , Animals , CD4-CD8 Ratio , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Division/genetics , Cell Division/immunology , Crosses, Genetic , Enzyme Activation/genetics , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Immunophenotyping , Lymphocyte Count , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Point Mutation , Promoter Regions, Genetic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Transgenes/immunology
5.
Immunity ; 13(5): 599-609, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11114373

ABSTRACT

Lnk is an SH2 domain-containing adaptor protein expressed preferentially in lymphocytes. To illuminate the importance of Lnk, we generated lnk(-/-) mice. Whereas T cell development was unaffected, pre-B and immature B cells accumulated in the spleens. In the bone marrow, B-lineage cells were proportionately increased, reflecting enhanced production of pro-B cells that resulted in part from hypersensitivity of precursors to SCF, the ligand for c-kit. Hence, Lnk ordinarily acts to regulate B cell production. Further characterization of lnk(-/-) mice also revealed that full-length Lnk is a 68 kDa protein containing a conserved proline-rich region and a PH domain. Lnk is a representative of a multigene adaptor protein family whose members act, by analogy with Lnk, to modulate intracellular signaling.


Subject(s)
B-Lymphocytes/physiology , Proteins/physiology , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/immunology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Alignment , Signal Transduction , src Homology Domains
7.
J Biol Chem ; 275(40): 31318-24, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-10913130

ABSTRACT

We have previously described a small molecule-directed protein dimerization strategy, using coumermycin to juxtapose Raf fusion proteins containing the coumermycin-binding domain GyrB. Oligomerization of cytoplasmically localized Raf-GyrB fusion proteins leads to an increase in the kinase activity of both Raf and its substrate Mek. Surprisingly, more distal targets, such as Erk1 and Erk2, are not activated using this approach. Here we report that coumermycin-induced oligomerization of a membrane-localized Raf-GyrB fusion protein potently activated Erk1 and Erk2, up-regulated Fos protein levels, and induced expression of many immediate-early response genes. Thus, both membrane localization and oligomerization of Raf-GyrB are required to target Raf signals to downstream effectors. The ability to activate the entire Raf signal transduction cascade conditionally, using coumermycin-induced oligomerization, should prove useful for dissecting Raf-mediated effects on gene expression and cellular differentiation.


Subject(s)
Cell Membrane/metabolism , Proto-Oncogene Proteins c-raf/biosynthesis , 3T3 Cells , Aminocoumarins , Animals , Carcinogens , Cell Differentiation , Coumarins/pharmacology , DNA Gyrase , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , Immediate-Early Proteins/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Novobiocin/pharmacology , Phosphorylation , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , Subcellular Fractions/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transcription, Genetic , Transfection , Up-Regulation
8.
J Immunol ; 164(8): 4071-9, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10754300

ABSTRACT

Caspases, a family of cysteine proteases, are critical mediators of apoptosis. To address the importance of caspases in thymocyte development, we have generated transgenic mice that express the baculovirus protein p35, a viral caspase inhibitor, specifically in the thymus. p35 expression inhibited Fas (CD95)-, CD3-, or peptide-induced caspase activity in vitro and conferred resistance to Fas-induced apoptosis. However, p35 did not block specific peptide-induced negative selection in OT1 and HY TCR transgenic mouse models. Even the potent pharmacological caspase inhibitor zVAD-FMK (benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl-ketone) could not prevent peptide-induced deletion of OT1 thymocytes, although it improved basal thymocyte survival in vitro. Moreover, the developmental block observed in rag1-/- thymocytes, which lack pre-TCR signaling, was also not rescued by p35 expression. These results indicate that caspase-independent signal transduction pathways can mediate thymocyte death during normal T cell development.


Subject(s)
Apoptosis/immunology , Caspases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Thymus Gland/cytology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/genetics , Caspase 3 , Caspase Inhibitors , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Clonal Deletion/immunology , Crosses, Genetic , Cysteine Proteinase Inhibitors/biosynthesis , Enzyme Activation/immunology , Female , H-Y Antigen/genetics , Inhibitor of Apoptosis Proteins , Male , Mice , Mice, Transgenic , Nucleopolyhedroviruses/genetics , Nucleopolyhedroviruses/immunology , Ovalbumin/immunology , Peptides/antagonists & inhibitors , Peptides/immunology , Receptors, Antigen, T-Cell/biosynthesis , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Viral Proteins/antagonists & inhibitors , Viral Proteins/biosynthesis , Viral Proteins/genetics
9.
Mol Cell Biol ; 19(6): 4200-8, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330160

ABSTRACT

Mice deficient in the transmembrane protein tyrosine phosphatase CD45 exhibit a block in thymocyte development. To determine whether the block in thymocyte development was due to the inability to dephosphorylate the inhibitory phosphorylation site (Y505) in p56(lck) (Lck), we generated CD45-deficient mice that express transgenes for the Lck Y505F mutation and the DO11.10 T-cell antigen receptor (TCR). CD4 single-positive T cells developed and accumulated in the periphery. Treatment with antigen resulted in thymocyte apoptosis and the loss of transgenic-TCR-bearing cells. Peripheral CD45-deficient T cells from the mice expressing both transgenes responded to antigen by increasing CD69 expression, interleukin-2 production, and proliferation. These results indicate that thymocyte development requires the dephosphorylation of the inhibitory site in Lck by CD45.


Subject(s)
Leukocyte Common Antigens/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Thymus Gland/growth & development , Animals , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Crosses, Genetic , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Gene Expression Regulation , Immunoblotting , Mice , Mice, Transgenic , Phosphotransferases/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , Spleen/metabolism , Time Factors
10.
Int Immunol ; 11(2): 169-77, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10069415

ABSTRACT

A pre-TCR-CD3 signal is required for the efficient maturation of CD4- CD8- thymocytes to the CD4+ CD8+ stage. This study addressed whether a similar signal is required for maturation of intestinal intraepithelial lymphocytes (IEL) that may develop extrathymically. We have shown previously that IEL from mice deficient for CD3- associated zeta chains include an immature population of CD3- CD8alphaalpha+ cells expressing cytoplasmic TCR beta chains but lacking detectable surface TCRalphabeta, CD16 and B220. Here we stimulated the appearance of such IEL in epsilon+/- zeta-/- mice by expression of an activated Lck transgene or in vivo treatment with anti-CD3epsilon. Anti-CD3epsilon treatment of RAG-deficient animals also yielded CD16- B220- IEL. In contrast, expression of a TCRbeta transgene in rag-1(-/-) mice did not stimulate the appearance of CD3- CD8alphaalpha+ CD16- B220- cells. Taken together these data indicate that although anti-CD3epsilon treatment and LckF505 assist in catalyzing a CD16+ B220+ --> CD16- B220- transition, these manipulations are not equivalent to a pre-TCR signal in IEL lymphocytes.


Subject(s)
Intestinal Mucosa/immunology , Signal Transduction , T-Lymphocytes/physiology , Thymus Gland/cytology , Animals , CD3 Complex/physiology , CD8 Antigens/biosynthesis , Cell Differentiation , Genes, T-Cell Receptor beta , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Leukocyte Common Antigens/biosynthesis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Receptor-CD3 Complex, Antigen, T-Cell/genetics , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, IgG/biosynthesis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/immunology , Transgenes
11.
Proc Natl Acad Sci U S A ; 96(3): 1024-9, 1999 Feb 02.
Article in English | MEDLINE | ID: mdl-9927687

ABSTRACT

The central role of type-2 helper T (Th2) cells in the development of allergic responses and immune responses against helminthic parasites is well documented. The differentiation of Th2 cells from naive T cells requires both the recognition of antigen by T cell antigen receptors (TCR) and the activation of downstream signal-transduction molecules of the interleukin 4 receptor (IL-4R) pathway, including Jak1, Jak3, and STAT6. Little is known, however, about how these two distinct pathways cooperate with each other to induce Th2 cells. Here, we use a T cell-specific H-Ras-dominant-negative transgenic mouse to show that TCR-mediated activation of the Ras/mitogen-activated protein kinase pathway alters IL-4R function and is required for Th2 cell differentiation. The enhancement of IL-4R signaling seems to be a consequence of both direct "crosstalk" with the TCR signaling pathway and increased protein expression of downstream signaling molecules of the IL-4R pathway. Therefore, successful Th2 differentiation depends on the effectiveness of the TCR-mediated activation of the Ras/mitogen-activated protein kinase pathway in modifying the IL-4R-mediated signaling pathway.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/physiology , Receptors, Interleukin-4/physiology , Th2 Cells/immunology , ras Proteins/metabolism , Animals , Antibodies, Monoclonal , Cell Differentiation , Interferon-gamma/biosynthesis , Janus Kinase 1 , Janus Kinase 3 , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein-Tyrosine Kinases/metabolism , Receptors, Interleukin-4/biosynthesis , STAT6 Transcription Factor , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trans-Activators/metabolism
12.
J Immunol ; 162(2): 677-83, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9916685

ABSTRACT

A novel cytokine from a thymic stromal cell line (thymic stromal lymphopoietin (TSLP)) promotes the development of B220+/IgM+ immature B cells when added to fetal liver cultures, long term bone marrow cultures, or bone marrow cells plated in semisolid medium. Because the activities of TSLP overlap with those of IL-7 in some in vitro assays, we compared the signaling mechanisms employed by TSLP and IL-7. Proliferation of a factor-dependent pre-B cell line (NAG8/7) in response to either TSLP or IL-7 was inhibited by anti-IL-7R alpha mAbs, suggesting that the functional TSLP receptor complex uses IL-7R alpha. In contrast, three different Abs to the common cytokine receptor gamma-chain had no effect on the response of these cells to TSLP, indicating that the functional TSLP receptor complex does not use the common cytokine receptor gamma-chain. Both cytokines induced activation of Stat5, but only IL-7 induced activation of the Janus family kinases Jak1 and Jak3. In fact, TSLP failed to activate any of the four known Janus family kinases, suggesting that Stat5 phosphorylation is mediated by a novel mechanism. Taken together, these data support the idea that TSLP can make unique contributions to B lymphopoiesis and indicate that it does so by mechanisms distinct from IL-7.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cytokines/physiology , Immunoglobulin M/biosynthesis , Interleukin-7/physiology , Milk Proteins , Proto-Oncogene Proteins , Signal Transduction/immunology , Thymus Gland/metabolism , Animals , B-Lymphocyte Subsets/metabolism , Bone Marrow Cells/cytology , Cell Differentiation/immunology , Cell Line , Cells, Cultured , Colony-Forming Units Assay , DNA-Binding Proteins/metabolism , Enzyme Activation/immunology , Fetus , Janus Kinase 1 , Janus Kinase 2 , Janus Kinase 3 , Liver/cytology , Mice , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptors, Interleukin-7/chemistry , Receptors, Interleukin-7/physiology , STAT5 Transcription Factor , Stromal Cells/metabolism , Thymus Gland/cytology , Time Factors , Trans-Activators/metabolism , Tyrosine/metabolism , Thymic Stromal Lymphopoietin
13.
J Exp Med ; 188(7): 1385-90, 1998 Oct 05.
Article in English | MEDLINE | ID: mdl-9763619

ABSTRACT

L-selectin binding activity for its ligand expressed by vascular endothelium is rapidly and transiently increased after leukocyte activation. To identify mechanisms for upregulation and assess how this influences leukocyte/endothelial cell interactions, cell-surface dimers of L-selectin were induced using the coumermycin-GyrB dimerization strategy for cross-linking L-selectin cytoplasmic domains in L-selectin cDNA-transfected lymphoblastoid cells. Coumermycin- induced L-selectin dimerization resulted in an approximately fourfold increase in binding of phosphomanan monoester core complex (PPME), a natural mimic of an L-selectin ligand, comparable to that observed after leukocyte activation. Moreover, L-selectin dimerization significantly increased (by approximately 700%) the number of lymphocytes rolling on vascular endothelium under a broad range of physiological shear stresses, and significantly slowed their rolling velocities. Therefore, L-selectin dimerization may explain the rapid increase in ligand binding activity that occurs after leukocyte activation and may directly influence leukocyte migration to peripheral lymphoid tissues or to sites of inflammation. Inducible oligomerization may also be a common mechanism for rapidly upregulating the adhesive or ligand-binding function of other cell-surface receptors.


Subject(s)
L-Selectin/metabolism , Leukocytes/metabolism , Cells, Cultured , DNA Gyrase , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Dimerization , Endothelium, Vascular , Humans , L-Selectin/genetics , Mannans/metabolism , Mannosephosphates/metabolism
14.
Int Immunol ; 10(8): 1077-82, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9723693

ABSTRACT

The stress-activated protein kinases (SAPK) are a group of dual-specificity kinases with potential roles in the control of apoptosis and proliferation. In most cells they are regulated through phosphorylation by MKK-4. We have investigated the role of MKK-4 in T cell development and function by generating transgenic animals expressing catalytically inactive MKK-4 (dMKK-4) in the thymus. Our results show that overexpression of dMKK-4 does not interfere with normal T cell development. Furthermore, expression of dMKK-4 inhibits Fas- but not phorbol ester plus ionomycin-induced activation of SAPK, suggesting that a SAPK kinase different from MKK-4 is responsible for the regulation of SAPK activation after stimulation of T cells with phorbol ester plus ionomycin. We then analyzed the effect of dMKK-4 on Fas-induced apoptosis of thymocytes. Our results show that activation of SAPK is not a necessary event in Fas-induced apoptosis of thymocytes.


Subject(s)
Apoptosis , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/analysis , Antigens, CD/immunology , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Catalytic Domain/genetics , Enzyme Activation/drug effects , Enzyme Activation/physiology , Flow Cytometry , Ionomycin/pharmacology , JNK Mitogen-Activated Protein Kinases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Precipitin Tests , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/immunology , Spleen/immunology , T-Lymphocyte Subsets , T-Lymphocytes/cytology , T-Lymphocytes/enzymology , Tetradecanoylphorbol Acetate/pharmacology , Thymus Gland/immunology , fas Receptor/immunology , fas Receptor/physiology
15.
Proc Natl Acad Sci U S A ; 95(16): 9459-64, 1998 Aug 04.
Article in English | MEDLINE | ID: mdl-9689102

ABSTRACT

The majority of T cells develop in the thymus and exhibit well characterized phenotypic changes associated with their maturation. Previous analysis of intestinal intraepithelial lymphocytes (IEL) from nude mice and a variety of experimentally manipulated models led to the view that at least a portion of these cells represent a distinct T cell population that matures extrathymically. The IEL that are postulated to mature within the intestine include both T cell receptor (TCR) alpha beta- and gamma delta-bearing subpopulations. They can be distinguished from conventional thymically derived T cells in that they express an unusual coreceptor, a CD8alpha homodimer. In addition, they can utilize the Fc receptor gamma-chain in place of the CD3-associated zeta-chain for TCR signaling and their maturation depends on the interleukin 2 receptor beta-chain. Moreover, TCRalpha beta+CD8alpha alpha+ IEL are not subject to conventional thymic selection processes. To determine whether CD3(-)CD8alpha alpha+ IEL represent precursors of T cells developing extrathymically, we examined IEL from knockout mice lacking the recombination activating gene-1 (rag-1), CD3epsilon, or both Lck and Fyn, in which thymic T cell development is arrested. CD3(-)CD8alpha alpha+CD16(+) IEL from all three mutant strains, as well as from nude mice, included cells that express pre-TCRalpha transcripts, a marker of T cell commitment. These IEL from lck-/-fyn-/- animals exhibited TCR beta-gene rearrangement. However, CD3(-)CD8alpha alpha+CD16(+) IEL from epsilon-deficient mice had not undergone Dbeta-Jbeta joining, despite normal rearrangement at the TCRbeta locus in thymocytes from these animals. These results revealed another distinction between thymocytes and IEL, and suggested an unexpectedly early role for CD3epsilon in IEL maturation.


Subject(s)
Intestines/cytology , Lymphocytes/cytology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Antigens, CD/immunology , Base Sequence , Cell Lineage , DNA Primers , Immunophenotyping , Intestines/immunology , Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/immunology
16.
J Immunol ; 160(5): 2130-8, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9498750

ABSTRACT

The tyrosine kinase Jak3 plays a key role in transducing signals from the IL-2, -4, -7, -9, and -15 receptors. Mice lacking Jak3 exhibit a profound, early block in both B and T cell development. To examine the mechanisms whereby Jak3 influences T cell function, we have reconstituted thymic development in Jak3-/- animals by introducing a Jak3 transgene in which expression was driven by the lck proximal promoter. Thymic reconstitution required Jak3 kinase activity, as catalytically inactive Jak3 did not restore early thymic development. Furthermore, the thymus-restricted expression pattern of the transgene allowed us to assess the requirement for Jak3 in peripheral T cells. In these mice, loss of Jak3 expression was associated with a failure to proliferate in response to antigen receptor crosslinking, the accumulation of T cells manifesting an activated cell surface phenotype, and an increased CD4/CD8 ratio among peripheral T cells, all of which are characteristics that were observed in Jak3-/- animals. Finally, we present data which suggest that peripheral T cells proliferate more rapidly in vivo and also undergo apoptosis more rapidly, upon loss of Jak3. Hence Jak3 exerts effects on mature peripheral T lymphocytes, as well as on thymocytes, resulting in the proper maintenance of circulating, quiescent cells.


Subject(s)
Homeostasis/immunology , Protein-Tyrosine Kinases/physiology , T-Lymphocyte Subsets/enzymology , Animals , Apoptosis/genetics , Apoptosis/immunology , CD4-CD8 Ratio , Catalysis , Cell Differentiation/genetics , Cell Differentiation/immunology , Fas Ligand Protein , Homeostasis/genetics , Immunophenotyping , Janus Kinase 3 , Ligands , Lymphocyte Activation/genetics , Lymphocyte Count , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology , Transgenes/immunology , Up-Regulation/genetics , Up-Regulation/immunology , fas Receptor/biosynthesis
17.
Immunity ; 6(6): 703-14, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9208843

ABSTRACT

Mice deficient for the pre-TCR alpha (pT alpha) chain cannot form a pre-T cell receptor (TCR) and exhibit a severe defect in early T cell development, characterized by lack of "beta selection" and impaired generation of double-positive (DP) thymocytes. Here, we demonstrate that intraperitoneal injection of CD3epsilon-specific antibodies into pT alpha-/- x RAG-/- mice or introduction of an activated p56(lck) transgene in pT alpha-/- mice fully restores the number of DP thymocytes, and that expression of a transgenic pT alpha chain lacking its cytoplasmic portion can overcome all developmental defects associated with pT alpha deficiency. These results allow a better definition of the role of pT alpha in pre-TCR signal transduction and provide conclusive evidence that the cytoplasmic tail of pT alpha is not essential for pre-TCR signaling.


Subject(s)
CD3 Complex/physiology , Receptors, Antigen, T-Cell, alpha-beta/chemistry , T-Lymphocytes/cytology , src-Family Kinases/physiology , Animals , Cell Differentiation , Cytoplasm , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Sequence Deletion , Signal Transduction , Structure-Activity Relationship , Thymus Gland/cytology , Transgenes
18.
J Biol Chem ; 272(23): 14562-70, 1997 Jun 06.
Article in English | MEDLINE | ID: mdl-9169414

ABSTRACT

Stimulation of the T cell antigen receptor (TCR) activates a set of non-receptor protein tyrosine kinases that assist in delivering signals to the cell interior. Among the presumed substrates for these kinases, adaptor proteins, which juxtapose effector enzyme systems with the antigen receptor complex, figure prominently. Previous studies suggested that Lnk, a 38-kDa protein consisting of a single SH2 domain and a region containing potential tyrosine phosphorylation sites, might serve to join Grb2, phospholipase C-gamma1, and phosphatidylinositol 3-kinase to the TCR. To elucidate the physiological roles of Lnk in T cell signal transduction, we isolated the mouse Lnk cDNA, characterized the structure of the mouse Lnk gene, and generated transgenic mice that overproduce Lnk in thymocytes. Here we report that although Lnk becomes phosphorylated during T cell activation, it plays no limiting role in the TCR signaling process. Moreover, we have distinguished p38(Lnk) from the more prominent 36-kDa tyrosine phosphoproteins that appear in activated T cells. Together these studies suggest that Lnk participates in signaling from receptors other than antigen receptors in lymphocytes.


Subject(s)
Adaptor Proteins, Signal Transducing , Protein Biosynthesis , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Exons , Flow Cytometry , GRB2 Adaptor Protein , Genomic Library , Intracellular Signaling Peptides and Proteins , Isoenzymes/metabolism , Membrane Proteins , Mice , Mice, Transgenic , Molecular Sequence Data , Molecular Weight , Phosphatidylinositol 3-Kinases , Phospholipase C gamma , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polymerase Chain Reaction , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , RNA, Messenger/biosynthesis , Receptors, Antigen, T-Cell/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Spleen/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Transcription, Genetic , Type C Phospholipases/metabolism , src Homology Domains
19.
Eur J Immunol ; 27(2): 554-62, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9045930

ABSTRACT

The developmental stages and the role of protein tyrosine kinases (PTK) in the maturation of CD3+CD8 alpha alpha+ intraepithelial lymphocytes (IEL) have not been extensively characterized. However, comparisons of thymic and extrathymic T cell development indicate that these processes involve some distinct signaling and selection events. We used mice deficient in Lck, Fyn, or both Lck and Fyn to analyze the role that these src-family PTK play in IEL development. In contrast to thymocyte development, we found that all IEL subsets develop in mice deficient for either kinase alone. However, lck-/- animals exhibited reduced numbers of TcR alphabeta+ CD8alpha alpha+ IEL, indicating that Lck is important in the development of these cells. Mice which lack both Lck and Fyn fail to generate TcR alphabeta+ IEL, suggesting that signaling through the preTcR, mediated by Lck and, to a lesser extent Fyn, is required for maturation of all TcR alphabeta+ IEL lineages. Interestingly, a small population of TcR gammadelta+ CD8 alpha alpha+ cells are apparent in lck-/-fyn-/- animals, demonstrating that TcR alphabeta+ CD8 alpha alpha+ and TcR gammadelta+ CD8alpha alpha+ IEL have distinct PTK requirements for their development or expansion. CD3-CD8alpha- CD44+ and CD3-CD8alpha alpha+ CD16/32+ B220+ cells comprise the majority of IEL in both lck-/- fyn-/- and rag -/- mice, while they are poorly represented in wildtype controls. Comparison of the cell surface phenotype of these putative precursor IEL in lck-/- fyn-/- and rag-/- animals suggests that IEL maturation in these animals is arrested at an equivalent developmental stage. Overall, the data presented demonstrate that signals mediated by Lck or Fyn direct TcR alphabeta+ CD8alpha alpha+ IEL maturation but are dispensable for the development of TcR gammadelta+ CD8 alpha alpha+ IEL.


Subject(s)
Proto-Oncogene Proteins/physiology , T-Lymphocyte Subsets/drug effects , src-Family Kinases/physiology , Animals , CD3 Complex/analysis , CD8 Antigens/analysis , Cell Differentiation/drug effects , Epithelium/drug effects , Epithelium/immunology , Hyaluronan Receptors/analysis , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Mice , Mice, Mutant Strains , Protein-Tyrosine Kinases/pharmacology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins c-fyn , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , src-Family Kinases/deficiency
20.
Int Immunol ; 9(1): 27-41, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9043945

ABSTRACT

Antibody reactivity to self-antigens is a normal component of the immune system. To study the mechanism by which self-reactive B cells are generated and maintained, we analyzed B cell development in transgenic mice that express a rearranged VH81X heavy chain from the pre-immune repertoire. In these mice, > 95% of B cells express the transgene in association with a variety of kappa light chains but V kappa 1 C being the dominant light chain. These transgenic B cells with identical V kappa 1C-J kappa 5 joins do not normally secrete IgM in vivo, but antibodies derived from these B cells, through LPS activation in vitro or after hybridoma immortalization, are self-reactive and recognize an ubiquitous epitope(s) on intracytoplasmic proteins from different tissues. They have the phenotype and localization pattern of long-lived marginal zone B cells and their development in vivo is blocked by injection of soluble VH81X-V kappa 1CJ kappa 5 IgM antibody. The observations in this transgenic mouse provide evidence for positive selection of a population of self-reactive B cells. These B cells enter the peripheral pool of B cells where they localize in the marginal zone of the spleen and, in contrast to other transgene-expressing B cells, do not secrete IgM antibody.


Subject(s)
B-Lymphocyte Subsets/immunology , Spleen/cytology , Spleen/immunology , Animals , Antibodies/blood , Autoantigens/immunology , B-Lymphocyte Subsets/classification , B-Lymphocyte Subsets/metabolism , Gene Rearrangement, B-Lymphocyte/immunology , Half-Life , Immunoglobulin Heavy Chains/blood , Immunoglobulin Idiotypes/administration & dosage , Immunoglobulin Idiotypes/biosynthesis , Immunoglobulin Idiotypes/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin kappa-Chains/genetics , Immunophenotyping , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...