Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Mucosal Immunol ; 11(2): 462-473, 2018 03.
Article in English | MEDLINE | ID: mdl-28745326

ABSTRACT

Protection against Mycobacterium tuberculosis (Mtb) infection requires CD4 T cells to migrate into the lung and interact with infected macrophages. In mice, less-differentiated CXCR3+ CD4 T cells migrate into the lung and suppress growth of Mtb, whereas CX3CR1+ terminally differentiated Th1 cells accumulate in the blood vasculature and do not control pulmonary infection. Here we examine CD4 T-cell differentiation and lung homing during primary Mtb infection of rhesus macaques. Mtb-specific CD4 T cells simultaneously appeared in the airways and blood ∼21-28 days post exposure, indicating that recently primed effectors are quickly recruited into the lungs after entering circulation. Mtb-specific CD4 T cells in granulomas display a tissue-parenchymal CXCR3+CX3CR1-PD-1hiCTLA-4+ phenotype. However, most granuloma CD4 T cells are found within the outer lymphocyte cuff and few localize to the myeloid cell core containing the bacilli. Using the intravascular stain approach, we find essentially all Mtb-specific CD4 T cells in granulomas have extravasated across the vascular endothelium into the parenchyma. Therefore, it is unlikely to be that lung-homing defects introduced by terminal differentiation limit the migration of CD4 T cells into granulomas following primary Mtb infection of macaques. However, intralesional positioning defects within the granuloma may pose a major barrier to T-cell-mediated immunity during tuberculosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Granuloma, Respiratory Tract/immunology , Lung/immunology , Macaca mulatta/immunology , Macrophages, Alveolar/immunology , Mycobacterium tuberculosis/physiology , Tuberculosis/immunology , Animals , Antigens, Bacterial/immunology , Cell Communication , Cell Movement , Cells, Cultured , Humans , Immune Evasion , Immunity, Cellular , Lung/microbiology , Macaca mulatta/microbiology , Mice , Mice, Inbred C57BL , Receptors, CXCR3/metabolism
2.
Mucosal Immunol ; 10(2): 318-321, 2017 03.
Article in English | MEDLINE | ID: mdl-27966550

ABSTRACT

Development of effective tuberculosis vaccines is hampered by insufficient understanding of protective immunity. Here, Woodworth et al.1 show secondary effector CD4 T cells generated after Mtb challenge of H56/CAF01 vaccinated mice display superior lung homing compared with primary effectors. Vaccination generates large populations of parenchymal lung effector cells by inducing CXCR3+KLRG1- cells that continuously migrate from lymph nodes to lung, and limiting the generation of non-protective CX3CR1+KLRG1+ intravascular effectors, providing insight vaccine-mediated protection against tuberculosis.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines , Animals , CD4-Positive T-Lymphocytes/immunology , Lung/immunology , Mice , Vaccination , Vaccines, Subunit
3.
Leukemia ; 27(8): 1666-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23399893

ABSTRACT

Philadelphia chromosome-positive leukemias, including chronic myeloid leukemia and B-cell acute lymphoblastic leukemia (B-ALL), are driven by the oncogenic BCR-ABL fusion protein. Animal modeling experiments utilizing retroviral transduction and subsequent bone marrow transplantation have demonstrated that BCR-ABL generates both myeloid and lymphoid disease in mice receiving whole bone marrow transduced with BCR-ABL. Y177 of BCR-ABL is critical to the development of myeloid disease, and phosphorylation of Y177 has been shown to induce GRB2 binding to BCR-ABL, followed by activation of the Ras and phosphoinositide 3 kinase signaling pathways. We show that the GRB2-related adapter protein, GADS, also associates with BCR-ABL, specifically through Y177 and demonstrate that BCR-ABL-driven lymphoid disease requires Gads. BCR-ABL transduction of Gads(-/-) bone marrow results in short latency myeloid disease within 3-4 weeks of transplant, while wild-type mice succumb to both a longer latency lymphoid and myeloid diseases. We report that GADS mediates a unique BCR-ABL complex with SLP-76 in BCR-ABL-positive cell lines and B-ALL patient samples. These data suggest that GADS mediates lymphoid disease downstream of BCR-ABL through the recruitment of specific signaling intermediates.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fusion Proteins, bcr-abl/metabolism , Leukemia, Lymphoid/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/metabolism , Blood Cells/metabolism , Bone Marrow/metabolism , Bone Marrow Transplantation , CD48 Antigen , Cell Line , Disease Models, Animal , Fusion Proteins, bcr-abl/genetics , Gene Expression , Hematopoiesis, Extramedullary , Humans , Immunophenotyping , Leukemia, B-Cell/genetics , Leukemia, B-Cell/metabolism , Leukemia, Lymphoid/genetics , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Protein Binding , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
4.
Leukemia ; 27(5): 1146-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23160449

ABSTRACT

BCR-ABL induces chronic myeloid leukemia (CML) through the aberrant regulation of multiple signaling substrates. Previous research has shown that BCR-ABL mediates down-modulation of CBL-B protein levels. A murine bone marrow transplantation (BMT) study was performed to assess the contribution of Cbl-b to BCR-ABL-induced disease. The predominant phenotype in the Cbl-b(-/-) recipients was a CML-like myeloproliferative disease (MPD) similar to that observed in the wild-type animals, but with a longer latency, diminished circulating leukocyte numbers and reduced spleen weights. Despite the decreased leukemic burden in comparison to their wild-type counterparts, the Cbl-b(-/-) animals displayed enhanced numbers of Gr-1(+)/Mac-1(+) spleen cells and neutrophilia. On the basis of prior evidence of CBL-B-dependent motility toward SDF-1α, we hypothesized that Cbl-b deficiency might impair bone marrow localization during transplantation. Homing experiments showed reduced migration of Cbl-b(-/-) cells to the bone marrow. Intrafemoral transplantation of BCR-ABL-transduced Cbl-b(-/-) cells revealed equivalent latency of disease development to the wild-type transplants, supporting the conclusion that Cbl-b deficiency diminishes homing of leukemic cells to the bone marrow, and perturbs the proliferation of BCR-ABL-expressing malignant clones during CML development.


Subject(s)
Bone Marrow Cells/physiology , Fusion Proteins, bcr-abl/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Proto-Oncogene Proteins c-cbl/physiology , Animals , Bone Marrow Transplantation , Cell Movement , Cells, Cultured , Mice , Mice, Inbred BALB C , Receptors, Chemokine/analysis
5.
Proc Natl Acad Sci U S A ; 105(38): 14436-41, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18780792

ABSTRACT

Actin filament binding by the focal adhesion (FA)-associated protein talin stabilizes cell-substrate adhesions and is thought to be rate-limiting in cell migration. Although F-actin binding by talin is known to be pH-sensitive in vitro, with lower affinity at higher pH, the functional significance of this pH dependence is unknown. Because increased intracellular pH (pH(i)) promotes cell migration and is a hallmark of metastatic carcinomas, we asked whether it increases FA remodeling through lower-affinity talin-actin binding. Talin contains several actin binding sites, but we found that only the COOH-terminal USH-I/LWEQ module showed pH-dependent actin binding, with lower affinity and decreased maximal binding at higher pH. Molecular dynamics simulations and NMR of this module revealed a structural mechanism for pH-dependent actin binding. A cluster of titratable amino acids with upshifted pK(a) values, including His-2418, was identified at one end of the five-helix bundle distal from the actin binding site. Protonation of His-2418 induces changes in the conformation and dynamics of the remote actin binding site. Structural analyses of a mutant talin-H2418F at pH 6.0 and 8.0 suggested changes different from the WT protein, and we confirmed that actin binding by talin-H2418F was relatively pH-insensitive. In motile fibroblasts, increasing pH(i) decreased FA lifetime and increased the migratory rate. However, expression of talin-H2418F increased lifetime 2-fold and decreased the migratory rate. These data identify a molecular mechanism for pH-sensitive actin binding by talin and suggest that FA turnover is pH-dependent and in part mediated by pH-dependent affinity of talin for binding actin.


Subject(s)
Actins/metabolism , Focal Adhesions/metabolism , Models, Molecular , Talin/chemistry , Talin/metabolism , Actins/chemistry , Animals , Cell Line , Computer Simulation , Hydrogen-Ion Concentration , Mice , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Tertiary , Talin/genetics
6.
Proc Natl Acad Sci U S A ; 103(45): 16930-5, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-17077140

ABSTRACT

Activation of JAK2 by chromosomal translocation or point mutation is a recurrent event in hematopoietic malignancies, including acute leukemias and myeloproliferative disorders. Although the effects of activated JAK2 signaling have been examined in cell lines and murine models, the functional consequences of deregulated JAK2 in the context of human hematopoietic cells are currently unknown. Here we report that expression of TEL-JAK2, a constitutively active variant of the JAK2 kinase, in lineage-depleted human umbilical cord blood cells results in erythropoietin-independent erythroid differentiation in vitro and induces the rapid development of myelofibrosis in an in vivo NOD/SCID xenotransplantation assay. These studies provide functional evidence that activated JAK2 signaling in primitive human hematopoietic cells is sufficient to drive key processes implicated in the pathophysiology of polycythemia vera and idiopathic myelofibrosis. Furthermore, they describe an in vivo model of myelofibrosis initiated with primary cells, highlighting the utility of the NOD/SCID xenotransplant system for the development of experimental models of human hematopoietic malignancies.


Subject(s)
Erythropoiesis/physiology , Hematopoietic Stem Cells/metabolism , Oncogene Proteins, Fusion/metabolism , Primary Myelofibrosis/etiology , Animals , Erythropoietin/metabolism , Gene Expression , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oncogene Proteins, Fusion/genetics , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transduction, Genetic , Transplantation, Heterologous
7.
Annu Rev Pharmacol Toxicol ; 42: 527-52, 2002.
Article in English | MEDLINE | ID: mdl-11807182

ABSTRACT

The NHE family of ion exchangers includes six isoforms (NHE1-NHE6) that function in an electroneutral exchange of intracellular H(+) for extracellular Na(+). This review focuses on the only ubiquitously expressed isoform, NHE1, which is localized at the plasma membrane where it plays a critical role in intracellular pH (pHi) and cell volume homeostasis. All NHE isoforms share a similar topology: an N-terminus of 12 transmembrane (TM) alpha-helices that collectively function in ion exchange, and a C-terminal cytoplasmic regulatory domain that modulates transport activity by the TM domain. Extracellular signals, mediated by diverse classes of cell-surface receptors, regulate NHE1 activity through distinct signaling networks that converge to directly modify the C-terminal regulatory domain. Modifications in the C-terminus, including phosphorylation and the binding of regulatory proteins, control transport activity by altering the affinity of the TM domain for intracellular H(+). Recently, it was determined that NHE1 also functions as a membrane anchor for the actin-based cytoskeleton, independently of its role in ion translocation. Through its effects on pHi homeostasis, cell volume, and the actin cortical network, NHE1 regulates a number of cell behaviors, including adhesion, shape determination, migration, and proliferation.


Subject(s)
Sodium-Hydrogen Exchangers/physiology , Animals , Apoptosis , Calmodulin/metabolism , Cell Division , Cell Survival , Cytoskeleton/physiology , Humans , Phosphorylation , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/chemistry
8.
J Immunol ; 167(4): 2030-9, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11489985

ABSTRACT

The in vivo T cell response to persistent Ag contains a hyporesponsive phase following an initial expansion and subsequent partial deletion of the responding cells. The mechanism(s) responsible for this tolerance process is poorly understood. In this study, we describe a new paired transgenic model (TCR and Ag), which within 7-14 days produces 20-40 million hyporesponsive T cells. This state is characterized by an 85-95% reduction in all cytokine production, an impairment of re-expression of CD25 and CD69, and a desensitization of the proliferative response to Ag. TCR levels were normal, and in vivo mixing experiments showed no evidence for active suppression. The hyporesponsiveness partially dissipated without proliferation when the cells were transferred into a non-Ag-bearing host. If the second host expressed Ag, the T cells initially regained responsiveness, but then slowly entered an even deeper state of tolerance characterized by an additional 7- to 10-fold lowering of cytokine production and a greater desensitization of proliferation. Surprisingly, this readaptation took place with the same level of Ag presentation, suggesting that other parameters can influence the tolerance threshold. Both the readjustment in sensitivity and the reversal without Ag convincingly demonstrate for the first time a truly adaptive tolerance process in CD4+ T cells in vivo.


Subject(s)
Adoptive Transfer , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Immune Tolerance , Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Adoptive Transfer/methods , Animals , Antigen Presentation/genetics , Antigen-Presenting Cells/immunology , CD3 Complex/biosynthesis , CD3 Complex/genetics , Columbidae , Cytochrome c Group/genetics , Cytochrome c Group/immunology , Dose-Response Relationship, Immunologic , Immune Tolerance/genetics , Interphase/genetics , Interphase/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics
9.
J Biol Chem ; 276(35): 32704-13, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11435425

ABSTRACT

A subset of chromosomal translocations that participate in leukemia involve activated tyrosine kinases. The ets transcription factor, TEL, undergoes translocations with several distinct tyrosine kinases including JAK2. TEL-JAK2 transforms cell lines to factor independence, and constitutive tyrosine kinase activity results in the phosphorylation of several substrates including STAT1, STAT3, and STAT5. In this study we have shown that TEL-JAK2 can constitutively activate the phosphatidylinositol 3'-kinase (PI 3'-kinase) signaling pathway. The regulatory subunit of PI 3'-kinase, p85, associates with TEL-JAK2 in immunoprecipitations, and this was shown to be mediated by the amino-terminal SH2 domain of p85 but independent of a putative p85-binding motif within TEL-JAK2. The scaffolding protein Gab2 can also mediate the association of p85. TEL-JAK2 constitutively phosphorylates the downstream substrate protein kinase B/AKT. Importantly, the pharmacologic PI 3'-kinase inhibitor, LY294002, blocked TEL-JAK2 factor-independent growth and phosphorylation of protein kinase B. However, LY294002 did not alter STAT5 tyrosine phosphorylation, indicating that STAT5 and protein kinase B activation mediated by TEL-JAK2 are independent signaling pathways. Therefore, activation of the PI 3'-kinase signaling pathway is an important event mediated by TEL-JAK2 chromosomal translocations.


Subject(s)
DNA-Binding Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Amino Acid Substitution , Animals , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Chromones/pharmacology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Interleukin-3/pharmacology , Janus Kinase 2 , Kinetics , Mice , Morpholines/pharmacology , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-ets , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/pharmacology , Repressor Proteins/chemistry , Repressor Proteins/genetics , Transfection , Translocation, Genetic , ETS Translocation Variant 6 Protein
10.
J Biol Chem ; 276(33): 31349-56, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11369779

ABSTRACT

NIK, a recently identified Nck-interacting kinase, acts upstream of the MEK kinase MEKK1 to activate the c-Jun N-terminal kinase JNK. We now show that NIK binds to and divergently activates the plasma membrane Na(+)-H(+) exchanger NHE1. In a genetic screen, NHE1 interacted with NIK at a site N-terminal (amino acids 407-502) to the Nck-binding domain, and this site is critical for its association with NHE1 in vivo. NIK also phosphorylates NHE1; however, the phosphorylation sites, which are distal to amino acid 638, are distinct from the NIK-binding site on NHE1 (amino acids 538-638). Expression of wild-type, but not a kinase-inactive, NIK in fibroblasts increased NHE1 phosphorylation and activity. The kinase domain of NIK, however, was not sufficient for this response in vivo. Full phosphorylation and activation of NHE1 required both the kinase and the NHE1-binding domains of NIK, suggesting that the NHE1-binding site functions as a targeting signal. The functional significance of an interaction between NIK and NHE1 was confirmed by the ability of a kinase-inactive NIK to selectively inhibit activation of NHE1 by platelet-derived growth factor but not by thrombin. Moreover, although NIK activates JNK through a mechanism dependent on MEKK1, it phosphorylated and activated NHE1 independently of MEKK1. These findings indicate that NIK acts downstream of platelet-derived growth factor receptors to phosphorylate and activate NHE1 divergently of its activation of JNK.


Subject(s)
Platelet-Derived Growth Factor/pharmacology , Protein Serine-Threonine Kinases/physiology , Sodium-Hydrogen Exchangers/metabolism , Animals , Cricetinae , Cricetulus , Hydrogen-Ion Concentration , Phosphorylation , NF-kappaB-Inducing Kinase
11.
Blood ; 97(8): 2230-7, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290583

ABSTRACT

Erythropoietin (EPO) specifically activates the Janus kinase JAK2 and the transcription factor signal transducer and activator of transcription-5 (STAT5). All members of the STAT family are tyrosine phosphorylated in response to cytokine stimulation at a conserved carboxy-terminal tyrosine, Y694, in the case of STAT5. To determine structural features important for STAT signaling, we generated an activation-specific STAT5 antibody using a phosphopeptide containing amino acids 687 to 698 of STAT5 as antigen. This antibody specifically recognizes tyrosine- phosphorylated STAT5 but not nonphosphorylated STAT5. In immunoprecipitation reactions from cell lines and primary erythroblasts, 2 distinct polyclonal activation-specific STAT5 antibodies selectively immunoprecipitate the tyrosine phosphorylated EPO receptor (EPO-R) in addition to STAT5 under native and denaturing conditions. We propose that the activation-specific STAT5 antibody recognizes the 2 substrates to which the STAT5 SH2 domain interacts, namely, the tyrosine- phosphorylated EPO-R and STAT5 itself. Several studies have implicated EPO-R Y343, Y401, Y431, and Y479 in the recruitment of STAT5. Using a series of EPO-R tyrosine mutants expressed in Ba/F3 cells, we have shown that the activation-specific STAT5 antibody immunoprecipitates an EPO-R containing only 2 tyrosines at positions 343 and 401, confirming the importance of these tyrosines in STAT5 recruitment. These data uncover a novel aspect of STAT SH2 domain recognition and demonstrate the utility of activation-specific antibodies for examining the specificity of STAT-cytokine receptor interactions.


Subject(s)
DNA-Binding Proteins/immunology , Epitopes/immunology , Erythropoietin/pharmacology , Milk Proteins , Receptors, Erythropoietin/immunology , Trans-Activators/immunology , Animals , Antibody Specificity , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/chemistry , Epitopes/chemistry , Mice , Mice, Inbred C57BL , Models, Biological , Peptide Fragments/immunology , Phosphorylation , Phosphotyrosine/chemistry , Precipitin Tests , Protein Denaturation , Protein Processing, Post-Translational , Rabbits , Receptors, Erythropoietin/chemistry , STAT5 Transcription Factor , Sheep , Trans-Activators/chemistry , src Homology Domains
12.
Exp Hematol ; 29(4): 432-40, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11301183

ABSTRACT

Erythropoietin (EPO) is a lineage-restricted growth factor that is required for erythroid proliferation and differentiation. EPO stimulates the phosphorylation and activation of p70 S6 kinase (p70 S6K), which is required for cell cycle progression. Here, the minimal cytoplasmic domains of the EPO receptor (EPO-R) required for p70 S6K activation were determined.Ba/F3 cells were stably transfected with wild-type (WT) EPO-R or EPO-R carboxyl-terminal deletion mutants, designated by the number of amino acids deleted from the cytoplasmic tail (-99, -131, -221). Transfected cells were growth factor deprived and then stimulated with EPO. p70 S6K, JAK2, IRS-2, and ERK1/2 phosphorylation/activation were examined. The ability of transfected 3-phosphoinositide-dependent protein kinase 1 (PDK1) to reconstitute p70 S6K phosphorylation in EPO-R mutants also was determined. Phosphorylation and activation of p70 S6K, JAK2, IRS-2, and ERK1/2 in Ba/F3 cells transfected with EPO-R-99 or EPO-R-99Y343F were similar to WT EPO-R. In contrast, EPO-dependent p70 S6K phosphorylation/activation, as well as IRS-2 and ERK1/2 phosphorylation, were minimal or absent in cells transfected with EPO-R-131 or EPO-R-221. JAK2 phosphorylation was reduced significantly in cells transfected with EPO-R-131 and abolished with EPO-R-221. To examine the role of PDK1, a kinase known to phosphorylate p70 S6K, Ba/F3 EPO-R-131 cells were transiently transfected with PDK1. WT constitutively active PDK1 restored p70 S6K phosphorylation in Ba/F3 EPO-R-131 cells but not in Ba/F3 EPO-R-221 cells. The results demonstrate that a minimal cytoplasmic subdomain of the EPO-R extending between -99 and -131 is required for p70 S6K phosphorylation and activation. The results also demonstrate that PDK1 is a critical component in this signaling pathway, which requires the presence of domains between -131 and -221 for its activation of p70 S6K.


Subject(s)
Cytoplasm/chemistry , Proto-Oncogene Proteins , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/physiology , Ribosomal Protein S6 Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Binding Sites , Cell Line , Erythropoietin/pharmacology , Gene Deletion , Gene Expression , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Janus Kinase 2 , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Phosphoproteins/metabolism , Phosphorylation , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Erythropoietin/genetics , Structure-Activity Relationship , Transfection
14.
Eur J Immunol ; 30(8): 2157-63, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10940906

ABSTRACT

Recent experiments have suggested that the IL-2 locus is monoallelically expressed. We tested this hypothesis using TCR-transgenic mice carrying one inactivated IL-2 allele. The frequency in single-cell assays of IL-2-producing cells following optimal stimulation by antigen and antigen-presenting cells was equivalent to that from wild-type mice, but the amount of IL-2 produced per cell was twofold less. Similar observations were made by intracellular staining for IL-2, although stimulation in bulk culture was less optimal, showing only a 1.7-fold difference. Importantly, the frequency of responding cells from the heterozygotes was less than from the wild-type mice if the IL-2 assay was performed after only 24 - 30 h of activation, suggesting that the targeted allele could compete with the normal allele early after stimulation and give the misimpression that the heterozygotes had fewer IL-2-producing cells. These data strongly argue that the IL-2 locus can be expressed biallelically under optimum stimulation conditions.


Subject(s)
Alleles , Chromosome Mapping , Interleukin-2/genetics , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/metabolism , Animals , Interleukin-2/biosynthesis , Lymph Nodes/metabolism , Mice , Mice, Transgenic
15.
J Biol Chem ; 275(6): 4398-406, 2000 Feb 11.
Article in English | MEDLINE | ID: mdl-10660611

ABSTRACT

Ship1 (SH2 inositol 5-phosphatase 1) has been shown to be a target of tyrosine phosphorylation downstream of cytokine and immunoregulatory receptors. In addition to its catalytic activity on phosphatidylinositol substrates, it can serve as an adaptor protein in binding Shc and Grb2. Erythropoietin (EPO), the primary regulator of erythropoiesis, has been shown to activate the tyrosine phosphorylation of Shc, resulting in recruitment of Grb2. However, the mechanism by which the erythropoietin receptor (EPO-R) recruits Shc remains unknown. EPO activates the tyrosine phosphorylation of Ship1, resulting in the interdependent recruitment of Shc and Grb2. Ship1 is recruited to the EPO-R in an SH2-dependent manner. Utilizing a panel of EPO-R deletion and tyrosine mutants, we have discovered remarkable redundancy in Ship1 recruitment. EPO-R Tyr(401) appears to be a major site of Ship1 binding; however, Tyr(429) and Tyr(431) can also serve to recruit Ship1. In addition, we have shown that EPO stimulates the formation of a ternary complex consisting of Ship1, Shc, and Grb2. Ship1 may modulate several discrete signal transduction pathways. EPO-dependent activation of ERK1/2 and protein kinase B (PKB)/Akt was examined utilizing a panel of EPO-R deletion mutants. Activation of ERK1/2 was observed in EPO-RDelta99, which retains only the most proximal tyrosine, Tyr(343). In contrast, EPO-dependent PKB activation was observed in EPO-RDelta43, but not in EPO-RDelta99. It appears that EPO-dependent PKB activation is downstream of a region that indirectly couples to phosphatidylinositol 3-kinase.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases , Receptors, Erythropoietin/metabolism , src Homology Domains , Animals , Cell Line , Enzyme Activation/drug effects , Erythropoietin/pharmacology , GRB2 Adaptor Protein , Humans , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation , Phosphotyrosine/analysis , Protein Binding , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Erythropoietin/genetics , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrosine/genetics , Tyrosine/metabolism
16.
Blood ; 95(3): 992-8, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10648414

ABSTRACT

The t(4;14) translocation occurs in 25% of multiple myeloma (MM) and results in both the ectopic expression of fibroblast growth factor receptor 3 (FGFR3) from der4 and immunoglobulin heavy chain-MMSET hybrid messenger RNA transcripts from der14. The subsequent selection of activating mutations of the translocated FGFR3 by MM cells indicates an important role for this signaling pathway in tumor development and progression. To investigate the mechanism by which FGFR3 overexpression promotes MM development, interleukin-6 (IL-6)-dependent murine B9 cells were transduced with retroviruses expressing functional wild-type or constitutively activated mutant FGFR3. Overexpression of mutant FGFR3 resulted in IL-6 independence, decreased apoptosis, and an enhanced proliferative response to IL-6. In the presence of ligand, wild-type FGFR3-expressing cells also exhibited enhanced proliferation and survival in comparison to controls. B9 clones expressing either wild-type FGFR3 at high levels or mutant FGFR3 displayed increased phosphorylation of STAT3 and higher levels of bcl-x(L) expression than did parental B9 cells after cytokine withdrawal. The mechanism of the enhanced cell responsiveness to IL-6 is unknown at this time, but does not appear to be mediated by the mitogen-activated protein kinases SAPK, p38, or ERK. These findings provide a rational explanation for the mechanism by which FGFR3 contributes to both the viability and propagation of the myeloma clone and provide a basis for the development of therapies targeting this pathway.


Subject(s)
Apoptosis , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Gene Expression Regulation, Neoplastic , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/metabolism , Myeloma Proteins/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/biosynthesis , Signal Transduction/physiology , Translocation, Genetic/genetics , Cell Division/drug effects , Chromosomes, Human, Pair 14/ultrastructure , Chromosomes, Human, Pair 4/ultrastructure , DNA-Binding Proteins/metabolism , Germinal Center/pathology , Humans , Interleukin-6/pharmacology , MAP Kinase Signaling System , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Myeloma Proteins/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , STAT3 Transcription Factor , Trans-Activators/metabolism , bcl-X Protein
17.
Mol Cell ; 6(6): 1425-36, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11163215

ABSTRACT

The association of actin filaments with the plasma membrane maintains cell shape and adhesion. Here, we show that the plasma membrane ion exchanger NHE1 acts as an anchor for actin filaments to control the integrity of the cortical cytoskeleton. This occurs through a previously unrecognized structural link between NHE1 and the actin binding proteins ezrin, radixin, and moesin (ERM). NHE1 and ERM proteins associate directly and colocalize in lamellipodia. Fibroblasts expressing NHE1 with mutations that disrupt ERM binding, but not ion translocation, have impaired organization of focal adhesions and actin stress fibers, and an irregular cell shape. We propose a structural role for NHE1 in regulating the cortical cytoskeleton that is independent of its function as an ion exchanger.


Subject(s)
Cytoskeleton/metabolism , DNA-Binding Proteins/metabolism , Hydrogen/metabolism , Sodium-Hydrogen Exchangers/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Blood Proteins/metabolism , Cell Division , Cell Line , Cell Size , Cytoskeletal Proteins/metabolism , Focal Adhesions/metabolism , Humans , Ion Transport , Membrane Proteins/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Phosphoproteins/metabolism , Precipitin Tests , Protein Binding , Protons , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics , Stress Fibers/metabolism
18.
J Biol Chem ; 274(51): 36125-31, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593895

ABSTRACT

Calcineurin, a Ca(2+)/calmodulin-stimulated protein phosphatase, plays a key role in T-cell activation by regulating the activity of NFAT (nuclear factor of activated T cells), a family of transcription factors required for the synthesis of several cytokine genes. Calcineurin is the target of the immunosuppressive drugs cyclosporin A and FK506 complexed with their cytoplasmic receptors cyclophilin and FKBP12, respectively. In this study we report that calcineurin is also the target of a recently identified Ca(2+)-binding protein, CHP (for calcineurin homologous protein), which shares a high degree of homology with the regulatory B subunit of calcineurin and with calmodulin. In Jurkat and HeLa cells, overexpression of CHP specifically impaired the nuclear translocation and transcriptional activity of NFAT but had no effect on AP-1 transcriptional activity and only a small (<25%) inhibitory effect on the transcriptional activity of NFkappaB. Further study indicated that CHP inhibits calcineurin activity. In cells overexpressing CHP, the phosphatase activity of immunoprecipitated calcineurin was inhibited by approximately 50%; and in a reconstituted assay, the activity of purified calcineurin was inhibited up to 97% by the addition of purified recombinant CHP in a dose-dependent manner. Moreover, prolonged activation of Jurkat cells was associated with a decreased abundance of CHP, suggesting a possible regulatory mechanism allowing activation of calcineurin. CHP, therefore, is a previously unrecognized endogenous inhibitor of calcineurin activity.


Subject(s)
Calcineurin/pharmacology , Calcium-Binding Proteins/pharmacology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Calcineurin/genetics , Calcium-Binding Proteins/genetics , Enzyme Activation/drug effects , Humans , Jurkat Cells , Lymphocyte Activation/drug effects , T-Lymphocytes/metabolism , Transfection
19.
J Am Anim Hosp Assoc ; 35(4): 341-7, 1999.
Article in English | MEDLINE | ID: mdl-10416780

ABSTRACT

The influence of urinary diversion procedures on urethral healing was studied in 15 male dogs following transection and anastomosis of the intrapelvic portions of their urethras. Dogs were randomly assigned to one of three treatment groups and had urine diverted from the surgical site by indwelling transurethral catheter, cystostomy catheter, or a combination of transurethral catheter and cystostomy catheter. There were no statistically significant differences in urethral healing when considering the different diversion methods, based on clinical, radiographic, and urodynamic parameters evaluated.


Subject(s)
Cystostomy/veterinary , Dogs/physiology , Dogs/surgery , Urethra/physiology , Urethra/surgery , Urinary Catheterization/veterinary , Wound Healing , Animals , Male
20.
J Biol Chem ; 274(29): 20465-72, 1999 Jul 16.
Article in English | MEDLINE | ID: mdl-10400674

ABSTRACT

Erythropoietin (Epo) activates a voltage-independent Ca2+ channel that is dependent on tyrosine phosphorylation. To identify the domain(s) of the Epo receptor (Epo-R) required for Epo-induced Ca2+ influx, Chinese hamster ovary (CHO) cells were transfected with wild-type or mutant Epo receptors subcloned into pTracer-cytomegalovirus vector. This vector contains an SV40 early promoter, which drives expression of the green fluorescent protein (GFP) gene, and a cytomegalovirus immediate-early promoter driving expression of the Epo-R. Successful transfection was verified in single cells by detection of GFP, and intracellular Ca2+ ([Ca]i) changes were simultaneously monitored with rhod-2. Transfection of CHO cells with pTracer encoding wild-type Epo-R, but not pTracer alone, resulted in an Epo-induced [Ca]i increase that was abolished in cells transfected with Epo-R F8 (all eight cytoplasmic tyrosines substituted). Transfection with carboxyl-terminal deletion mutants indicated that removal of the terminal four tyrosine phosphorylation sites, but not the tyrosine at position 479, abolished Epo-induced [Ca]i increase, suggesting that tyrosines at positions 443, 460, and/or 464 are important. In CHO cells transfected with mutant Epo-R in which phenylalanine was substituted for individual tyrosines, a significant increase in [Ca]i was observed with mutants Epo-R Y443F and Epo-R Y464F. The rise in [Ca]i was abolished in cells transfected with Epo-R Y460F. Results were confirmed with CHO cells transfected with plasmids expressing Epo-R mutants in which individual tyrosines were added back to Epo-R F8 and in stably transfected Ba/F3 cells. These results demonstrate a critical role for the Epo-R cytoplasmic tyrosine 460 in Epo-stimulated Ca2+ influx.


Subject(s)
Calcium Channels/metabolism , Receptors, Erythropoietin/metabolism , Animals , CHO Cells , Calcium/metabolism , Calcium Channels/chemistry , Cricetinae , Green Fluorescent Proteins , Ion Transport , Luminescent Proteins/metabolism , Mutation , Receptors, Erythropoietin/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...