Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Nat Genet ; 24(3): 296-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10700187

ABSTRACT

Atrioventricular and semilunar valve abnormalities are common birth defects, but how cardiac valvulogenesis is directed remains largely unknown. During studies of genetic interaction between Egfr, encoding the epidermal growth factor receptor, and Ptpn11, encoding the protein-tyrosine-phosphatase Shp2, we discovered that Egfr is required for semilunar, but not atrioventricular, valve development. Although unnoticed in earlier studies, mice homozygous for the hypomorphic Egfr allele waved-2 (Egfrwa2/wa2) exhibit semilunar valve enlargement resulting from over-abundant mesenchymal cells. Egfr-/- mice (CD1 background) have similar defects. The penetrance and severity of the defects in Egfrwa2/wa2 mice are enhanced by heterozygosity for a targeted mutation of exon 2 of Ptpn11 (ref. 3). Compound (Egfrwa2/wa2:Ptpn11+/-) mutant mice also show premature lethality. Electrocardiography, echocardiography and haemodynamic analyses showed that affected mice develop aortic stenosis and regurgitation. Our results identify the Egfr and Shp2 as components of a growth-factor signalling pathway required specifically for semilunar valvulogenesis, support the hypothesis that Shp2 is required for Egfr signalling in vivo, and provide an animal model for aortic valve disease.


Subject(s)
Aortic Valve/abnormalities , ErbB Receptors/physiology , Protein Tyrosine Phosphatases/physiology , Pulmonary Valve/abnormalities , Abnormalities, Multiple/genetics , Animals , Aortic Valve/embryology , Aortic Valve/pathology , Aortic Valve Insufficiency/genetics , Aortic Valve Stenosis/genetics , Epistasis, Genetic , ErbB Receptors/deficiency , ErbB Receptors/genetics , Genotype , Heart Conduction System/physiopathology , Hyperplasia , Intracellular Signaling Peptides and Proteins , Mesoderm/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/genetics , Pulmonary Valve/embryology , Pulmonary Valve/pathology , Sequence Deletion , Ventricular Dysfunction, Left/genetics
2.
Diabetologia ; 55(2): 468-78, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22124607

ABSTRACT

AIMS/HYPOTHESIS: Insulin activates insulin receptor protein tyrosine kinase and downstream phosphatidylinositol-3-kinase (PI3K)/Akt signalling in muscle to promote glucose uptake. The insulin receptor can serve as a substrate for the protein tyrosine phosphatase (PTP) 1B and T cell protein tyrosine phosphatase (TCPTP), which share a striking 74% sequence identity in their catalytic domains. PTP1B is a validated therapeutic target for the alleviation of insulin resistance in type 2 diabetes. PTP1B dephosphorylates the insulin receptor in liver and muscle to regulate glucose homeostasis, whereas TCPTP regulates insulin receptor signalling and gluconeogenesis in the liver. In this study we assessed for the first time the role of TCPTP in the regulation of insulin receptor signalling in muscle. METHODS: We generated muscle-specific TCPTP-deficient (Mck-Cre;Ptpn2(lox/lox)) mice (Mck, also known as Ckm) and assessed the impact on glucose homeostasis and muscle insulin receptor signalling in chow-fed versus high-fat-fed mice. RESULTS: Blood glucose and insulin levels, insulin and glucose tolerance, and insulin-induced muscle insulin receptor activation and downstream PI3K/Akt signalling remained unaltered in chow-fed Mck-Cre;Ptpn2(lox/lox) versus Ptpn2(lox/lox) mice. In addition, body weight, adiposity, energy expenditure, insulin sensitivity and glucose homeostasis were not altered in high-fat-fed Mck-Cre;Ptpn2(lox/lox) versus Ptpn2(lox/lox) mice. CONCLUSIONS/INTERPRETATION: These results indicate that TCPTP deficiency in muscle has no effect on insulin signalling and glucose homeostasis, and does not prevent high-fat diet-induced insulin resistance. Thus, despite their high degree of sequence identity, PTP1B and TCPTP contribute differentially to insulin receptor regulation in muscle. Our results are consistent with the notion that these two highly related PTPs make distinct contributions to insulin receptor regulation in different tissues.


Subject(s)
Glucose/metabolism , Muscles/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Animals , Diabetes Mellitus, Type 2/blood , Glucose Tolerance Test , Homeostasis , Insulin/metabolism , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , Receptor, Insulin/metabolism , Signal Transduction , Time Factors , Tissue Distribution
3.
Curr Opin Cell Biol ; 13(2): 182-95, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11248552

ABSTRACT

Protein tyrosine phosphatases (PTPs), the enzymes that dephosphorylate tyrosyl phosphoproteins, were initially believed to be few in number and serve a 'housekeeping' role in signal transduction. Recent work indicates that this is totally incorrect. Instead, PTPs comprise a large superfamily whose members play critical roles in a wide variety of cellular processes. Moreover, PTPs exhibit exquisite substrate specificity in vivo. Recent evidence has led us to propose that members of the PTP family achieve selectivity through different combinations of specific targeting strategies and intrinsic catalytic domain specificity.


Subject(s)
Protein Tyrosine Phosphatases/physiology , Signal Transduction , Animals , Humans , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Substrate Specificity
4.
Curr Opin Cell Biol ; 9(2): 193-204, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9069265

ABSTRACT

Protein-tyrosyl phosphorylation, regulated by protein tyrosine kinases and protein tyrosine phosphatases (PTPs), is a key cellular control mechanism. Until recently, little was known about PTPs. However, the past two years have witnessed an explosion of information about PTP structure, regulation and function. Crystal structures of several PTPs have provided insights into enzymatic mechanisms and regulation and suggested the design of 'substrate-trapping' mutants. Candidate homophilic and heterophilic ligands for transmembrane PTPs have been identified, and roles for transmembrane PTPs in regulating cell-cell interactions have been suggested. Finally, progress has been made in understanding signaling by Src homology 2 domain containing PTPs and PTPs controlling yeast osmoregulatory pathways.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Animals , Cell Adhesion , Cytokines/metabolism , Growth Substances/metabolism , Humans , Phosphorylation , Protein Conformation , Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , src Homology Domains
5.
Proc Natl Acad Sci U S A ; 105(44): 16940-5, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18957543

ABSTRACT

Tumors derived from rat LA7 cancer stem cells (CSCs) contain a hierarchy of cells with different capacities to generate self-renewing spheres and tubules serially ex vivo and to evoke tumors in vivo. We isolated two morphologically distinct cell types with distinct tumorigenic potential from LA7-evoked tumors: cells with polygonal morphology that are characterized by expression of p21/(WAF1) and p63 and display hallmarks of CSCs and elongated epithelial cells, which generate tumors with far less heterogeneity than LA7 CSCs. Serial transplantation of elongated epithelial cells results in progressive loss of tumorigenic potential; tumor heterogeneity; CD44, E-cadherin, and epithelial cytokeratin expression and increased alpha-smooth muscle actin I and vimentin expression. In contrast, serial transplantation of LA7 CSCs can be performed indefinitely and results in tumors that maintain their heterogeneity, consistent with self-renewal and multilineage differentiation potential. Collectively, our data show that polygonal cells are CSCs, whereas epithelial elongated cells are lineage-committed progenitors with tumorigenic potential, and suggest that tumor progenitors, although lacking indefinite self-renewal potential, nevertheless may make a substantial contribution to tumor development. Because LA7 cells can switch between conditions that favor maintenance of pure CSCs vs. differentiation into other tumor cell types, this cell system provides the opportunity to study factors that influence CSC self-renewal and differentiation. One factor, p63, was identified as a key gene regulating the transition between CSCs and early progenitor cells.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/cytology , Animals , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Clone Cells , Disease Models, Animal , Female , Immunohistochemistry , Mammary Glands, Animal/metabolism , Mice , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Rats , Stem Cells/cytology
6.
J Exp Med ; 184(3): 1111-26, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-9064328

ABSTRACT

Receptor protein tyrosine kinases (RTKs) transmit downstream signals via interactions with secondary signaling molecules containing SH2 domains. Although many SH2-phosphotyrosyl interactions have been defined in vitro, little is known about the physiological significance of specific RTK/SH2 interactions in vivo. Also, little is known about the mechanisms by which specific RTKs interact with and/or are regulated by specific protein tyrosine phosphatases (PTPs). To address such issue, we carried out a genetic analysis of the previously reported biochemical interaction between the RTK c-Kit, encoded at the W locus, and the SH2-containing non-transmembrane PTP SHP1, encoded at the motheaten (me) locus (1). Mice carrying a kinase-defective allele of c-Kit (Wv/+) were crossed with me/+ mice, which carry one effectively null allele of SHP1, and then backcrossed to generate all possible allelic combinations. Our results indicate strong intergenic complementation between these loci in hematopoietic progenitor cells. Compared to progenitors purified from normal mice, bone marrow progenitor cells (lin-) from me/me mice markedly hyper-proliferated in response to Kit ligand (KL). stimulation. Superimposition of the me/me genotype increased the number of one marrow-derived CFU-E from Wv/+ mice. Conversely, the presence of one or two copies of Wv decreased the number of macrophages and granulocytes in me/me lung, skin, peripheral blood and bone marrow, thereby decreasing the severity of the me/me phenotype. The decrease in dermal mast cells in Wv/Wv mice was rescued to levels found in Wv/+mice by superimposition of the me/me genotype. Surprisingly, however, the presence or absence of SHP1 had no effect on the proliferative response of bone marrow-derived cultured mast cells to KL or IL3 ex vivo. Nevertheless, the immediate-early response to KL stimulation, as measured by KL-induced tyrosyl phosphorylation, was substantially increased in mast cells from Wv/+:me/me compared to Wv/ +:+/+ mice, strongly suggesting that SHP1 directly dephosphorylates and regulates c-Kit. Taken together, our results establish that SHP1 negatively regulates signaling from c-Kit in vivo, but in a cell type-specific manner.


Subject(s)
Protein Tyrosine Phosphatases/physiology , Proto-Oncogene Proteins c-kit/physiology , Animals , Cell Count , Female , Genotype , Hematopoietic Stem Cells/physiology , Intracellular Signaling Peptides and Proteins , Male , Mast Cells/cytology , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 6
7.
Curr Opin Genet Dev ; 8(1): 112-26, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9529614

ABSTRACT

Genetic analysis has enhanced our understanding of the biological roles of many protein tyrosine kinases (PTKs). More recently, studies utilizing both spontaneous mutants and mutants induced by homologous recombination techniques have begun to yield key insights into the role of specific protein tyrosine phosphatases (PTPs) and to suggest how PTKs and PTPs interact. Specific PTPs in Saccharomyces cerevesiae and Schizomyces pombe regulate MAP kinase pathways. Several Drosophila receptor PTPs control axonal targeting pathways, whereas the non-receptor PTP Corkscrew (Csw), plays an essential positive signaling role in multiple developmental pathways directed by receptor PTKs. The vertebrate homolog of Csw, SHP-2, also is required for growth factor signaling and normal development. Finally, very recent studies of other mammalian PTPs suggest that they have critical roles in processes as diverse as hematopoiesis and liver and pituitary development.


Subject(s)
Protein Tyrosine Phosphatases/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Humans
8.
Mol Cell Biol ; 25(18): 8052-63, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16135797

ABSTRACT

SKAP-HOM is a cytosolic adaptor protein representing a specific substrate for the Src family protein tyrosine kinase Fyn. Previously, several groups have provided experimental evidence that SKAP-HOM (most likely in cooperation with the cytosolic adaptor protein ADAP) is involved in regulating leukocyte adhesion. To further assess the physiological role of SKAP-HOM, we investigated the immune system of SKAP-HOM-deficient mice. Our data show that T-cell responses towards a variety of stimuli are unaffected in the absence of SKAP-HOM. Similarly, B-cell receptor (BCR)-mediated total tyrosine phosphorylation and phosphorylation of Erk, p38, and JNK, as well as immunoreceptor-mediated Ca(2+) responses, are normal in SKAP-HOM(-/-) animals. However, despite apparently normal membrane-proximal signaling events, BCR-mediated proliferation is strongly attenuated in the absence of SKAP-HOM(-/-). In addition, adhesion of activated B cells to fibronectin (a ligand for beta1 integrins) as well as to ICAM-1 (a ligand for beta2 integrins) is strongly reduced. In vivo, the loss of SKAP-HOM results in a less severe clinical course of experimental autoimmune encephalomyelitis following immunization of mice with the encephalitogenic peptide of MOG (myelin oligodendrocyte glycoprotein). This is accompanied by strongly reduced serum levels of MOG-specific antibodies and lower MOG-specific T-cell responses. In summary, our data suggest that SKAP-HOM is required for proper activation of the immune system, likely by regulating the cross-talk between immunoreceptors and integrins.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , B-Lymphocytes/immunology , Phosphoproteins/physiology , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion/immunology , Cytosol/chemistry , Cytosol/metabolism , Hematopoietic System/cytology , Hematopoietic System/metabolism , Immunoglobulins/blood , Integrins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/metabolism , Myelin Proteins , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Phosphoproteins/analysis , Phosphoproteins/genetics , Receptors, Antigen, B-Cell/metabolism , Signal Transduction
9.
Leukemia ; 32(1): 203-213, 2018 01.
Article in English | MEDLINE | ID: mdl-28804122

ABSTRACT

BCR-ABL1-targeting tyrosine kinase inhibitors (TKIs) have revolutionized treatment of Philadelphia chromosome-positive (Ph+) hematologic neoplasms. Nevertheless, acquired TKI resistance remains a major problem in chronic myeloid leukemia (CML), and TKIs are less effective against Ph+ B-cell acute lymphoblastic leukemia (B-ALL). GAB2, a scaffolding adaptor that binds and activates SHP2, is essential for leukemogenesis by BCR-ABL1, and a GAB2 mutant lacking SHP2 binding cannot mediate leukemogenesis. Using a genetic loss-of-function approach and bone marrow transplantation models for CML and BCR-ABL1+ B-ALL, we show that SHP2 is required for BCR-ABL1-evoked myeloid and lymphoid neoplasia. Ptpn11 deletion impairs initiation and maintenance of CML-like myeloproliferative neoplasm, and compromises induction of BCR-ABL1+ B-ALL. SHP2, and specifically, its SH2 domains, PTP activity and C-terminal tyrosines, are essential for BCR-ABL1+, but not WT, pre-B-cell proliferation. The mitogen-activated protein kinase kinase (MEK) / extracellular signal-regulated kinase (ERK) pathway is regulated by SHP2 in WT and BCR-ABL1+ pre-B cells, but is only required for the proliferation of BCR-ABL1+ cells. SHP2 is required for SRC family kinase (SFK) activation only in BCR-ABL1+ pre-B cells. RNAseq reveals distinct SHP2-dependent transcriptional programs in BCR-ABL1+ and WT pre-B cells. Our results suggest that SHP2, via SFKs and ERK, represses MXD3/4 to facilitate a MYC-dependent proliferation program in BCR-ABL1-transformed pre-B cells.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , HEK293 Cells , Hematologic Neoplasms/drug therapy , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred C57BL , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , src Homology Domains/genetics
10.
Curr Biol ; 9(16): 927-30, 1999 Aug 26.
Article in English | MEDLINE | ID: mdl-10469599

ABSTRACT

Inhibitory immunoreceptors downregulate signaling by recruiting Src homology 2 (SH2) domain-containing tyrosine and/or lipid phosphatases to activating receptor complexes [1]. There are indications that some inhibitory receptors might also perform other functions [2] [3]. In adherent macrophages, two inhibitory receptors, SHPS-1 and PIR-B, are the major proteins binding to the tyrosine phosphatase SHP-1. SHPS-1 also associates with two tyrosine-phosphorylated proteins (pp55 and pp130) and a protein tyrosine kinase [4]. Here, we have identified pp55 and pp130 as the adaptor molecules SKAP55hom/R (Src-kinase-associated protein of 55 kDa homologue) and FYB/SLAP-130 (Fyn-binding protein/SLP-76-associated protein of 130 kDa), respectively, and the tyrosine kinase activity as PYK2. Two distinct SHPS-1 complexes were formed, one containing SKAP55hom/R and FYB/SLAP-130, and the other containing PYK2. Recruitment of FYB/SLAP-130 to SHPS-1 required SKAP55hom/R, whereas PYK2 associated with SHPS-1 independently. Formation of both complexes was independent of SHP-1 and tyrosine phosphorylation of SHPS-1. Finally, tyrosine phosphorylation of members of the SHPS-1 complexes was regulated by integrin-mediated adhesion. Thus, SHPS-1 provides a scaffold for the assembly of multi-protein complexes that might both transmit adhesion-regulated signals and help terminate such signals through SHP-1-directed dephosphorylation. Other inhibitory immunoreceptors might have similar scaffold-like functions.


Subject(s)
Antigens, Differentiation , Bone Marrow Cells/chemistry , Macrophages/chemistry , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/metabolism , Protein Folding , Receptors, Immunologic , Animals , COS Cells , Cell Adhesion , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Immunoblotting , Macrophages/drug effects , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Mice , Mice, Mutant Strains , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/metabolism , Sulfones/analysis , Sulfones/metabolism , Uridine/analogs & derivatives , Uridine/analysis , Uridine/metabolism
11.
Curr Biol ; 8(18): 1009-17, 1998 Sep 10.
Article in English | MEDLINE | ID: mdl-9740800

ABSTRACT

BACKGROUND: Signals from the B-cell antigen receptor (BCR) help to determine B-cell fate, directing either proliferation, differentiation, or growth arrest/apoptosis. The protein tyrosine phosphatase SHP-1 is known to regulate the strength of BCR signaling. Although the B-cell co-receptor CD22 binds SHP-1, B cells in CD22-deficient mice are much less severely affected than those in SHP-1-deficient mice, suggesting that SHP-1 may also regulate B-cell signaling by affecting other signaling molecules. Moreover, direct substrates of SHP-1 have not been identified in any B-cell signaling pathway. RESULTS: We identified the B-cell transmembrane protein CD72 as a new SHP-1 binding protein and as an in vivo substrate of SHP-1 in B cells. We also defined the binding sites for SHP-1 and the adaptor protein Grb2 on CD72. Tyrosine phosphorylation of CD72 correlated strongly with BCR-induced growth arrest/apoptosis in B-cell lines and in primary B cells. Preligation of CD72 attenuated BCR-induced growth arrest/death signals in immature and mature B cells or B-cell lines, whereas preligation of CD22 enhanced BCR-induced growth arrest/apoptosis. CONCLUSIONS: We have identified CD72 as the first clear in vivo substrate of SHP-1 in B cells. Our results suggest that tyrosine-phosphorylated CD72 may transmit signals for BCR-induced apoptosis. By dephosphorylation CD72. SHP-1 may have a positive role in B-cell signaling. These results have potentially important implications for the involvement of CD72 and SHP-1 in B-cell development and autoimmunity.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/physiology , Protein Tyrosine Phosphatases/metabolism , Receptors, Antigen, B-Cell/physiology , Animals , B-Lymphocytes/immunology , Cell Division , Cell Line , Cells, Cultured , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Phosphorylation , Phosphotyrosine , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Recombinant Fusion Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction , Spleen/immunology , Substrate Specificity , Transfection , src Homology Domains
12.
Curr Opin Immunol ; 9(3): 405-20, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9203419

ABSTRACT

Many lymphocyte signaling pathways are regulated by protein tyrosyl phosphorylation, which is controlled by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs). Substantial progress has been made in defining the functions of lymphocyte PTPs. Individual PTPs can enhance or diminish cell signaling levels. The transmembrane PTP CD45 is a key positive element in multiple lymphocyte signaling pathways in vivo. New insights into the function of individual CD45 isoforms have emerged. Anti-CD45 antibodies with potent immunosuppressant activity have been identified, suggesting that CD45 may be a propitious target for drug design. Progress has also been made in elucidating the function and targets of specific nontransmembrane PTPs, particularly those with Src homology 2 domains.


Subject(s)
Leukocyte Common Antigens/immunology , Lymphocyte Activation/immunology , Protein Tyrosine Phosphatases/immunology , Signal Transduction/immunology , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data
13.
Mol Cell Biol ; 18(11): 6666-78, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9774681

ABSTRACT

Epithelial cell differentiation is regulated by specific combinations of growth factors, hormones, and extracellular matrix (ECM). How these divergent signals are integrated is largely unknown. We used primary cultures of normal human bronchial epithelial cells (NHBEs) to investigate mechanisms of signal integration. In defined, serum-free media, NHBEs undergo mucosecretory differentiation only when grown in the presence of retinoids and on the appropriate substratum (collagen gels). We identified the retinoic acid receptor beta (RARbeta) gene as an early marker of NHBE differentiation. In contrast to immortalized cell lines, in NHBEs strong retinoid-induced RARbeta transcription occurs only when cells are grown on collagen gels, and it requires new protein synthesis and a cis-acting element that maps outside the known RARbeta promoter elements. NHBEs grown on collagen gels exhibit reduced epidermal growth factor (EGF)-induced Raf, MEK, and mitogen-activated protein kinase (MAPK) activity. This correlates with a specific inability to achieve high levels of p66(SHC) tyrosyl phosphorylation and association of p66(SHC) with GRB2, despite high levels of EGF receptor (EGFR) autophosphorylation. Notably, inhibition of EGFR or MEK/MAPK activation replaces the ECM requirement for RARbeta induction. Our results strongly suggest that a key mechanism by which specific ECMs facilitate retinoid-induced mucosecretory differentiation of NHBEs is by restricting the level of EGFR-dependent MEK/MAPK activation evoked by autocrine and/or paracrine EGFR ligands.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Bronchi/physiology , Cell Differentiation/physiology , Extracellular Matrix/physiology , Growth Substances/physiology , Mitogen-Activated Protein Kinase Kinases , Tretinoin/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Collagen/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , GRB2 Adaptor Protein , Humans , Immunohistochemistry , MAP Kinase Kinase 1 , Mucins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Retinoic Acid/metabolism , Shc Signaling Adaptor Proteins , Signal Transduction/physiology , Src Homology 2 Domain-Containing, Transforming Protein 1
14.
Mol Cell Biol ; 15(7): 3945-59, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7791800

ABSTRACT

Retinoic acid (RA) is required for normal airway epithelial cell growth and differentiation both in vivo and in vitro. One of the earliest events following the exposure of bronchial epithelial cells to RA is the strong induction of RA receptor beta (RAR beta) mRNA. Previous work established that many lung cancer cell lines and primary tumors display abnormal RAR beta mRNA expression, most often absence or weak expression of the RAR beta 2 isoform, even after RA treatment. Restoration of RAR beta 2 into RAR beta-negative lung cancer cell lines has been reported to inhibit tumorigenicity. Since RAR beta 2 inactivation may contribute to lung cancer, we have investigated the molecular mechanism of defective RAR beta 2 expression. Nuclear run-on assays and transient transfections with RAR beta 2 promoter constructs indicate the presence of trans-acting transcriptional defects in most lung cancer cell lines, which map to the RA response element (RARE). These defects cannot be complemented by RAR-retinoid X receptor cotransfection and can be separated into two types: (i) one affecting transcription from direct repeat RAREs, but not palindromic RAREs, and (ii) another affecting transcription from both types of RARE. Studies using chimeras between RAR alpha, TR alpha, and other transcription factors suggest the existence of novel RAR-thyroid hormone receptor AF-2-specific cofactors, which are necessary for high levels of transcription. Furthermore, these factors may be frequently inactivated in human lung cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Regulatory Sequences, Nucleic Acid/genetics , Adenovirus E1A Proteins/metabolism , Base Sequence , DNA-Binding Proteins/metabolism , Genetic Complementation Test , Humans , Isomerism , Lung Neoplasms/etiology , Models, Genetic , Molecular Sequence Data , Mutation , TATA-Box Binding Protein , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection
15.
Mol Cell Biol ; 18(1): 161-77, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418864

ABSTRACT

SHP-2 is a positive component of many receptor tyrosine kinase signaling pathways. The related protein-tyrosine phosphatase (PTP) SHP-1 usually acts as a negative regulator. The precise domains utilized by SHP-2 to transmit positive signals in vivo and the basis for specificity between SHP-1 and SHP-2 are not clear. In Xenopus, SHP-2 is required for mesoderm induction and completion of gastrulation. We investigated the effects of SHP-2 mutants and SHP-2/SHP-1 chimeras on basic fibroblast growth factor-induced mesoderm induction. Both SH2 domains and the PTP domain are required for normal SHP-2 function in this pathway. The N-terminal SH2 domain is absolutely required, whereas the C-terminal SH2 contributes to wild-type function. The C-terminal tyrosyl phosphorylation sites and proline-rich region are dispensable, arguing against adapter models of SHP-2 function. Although the SH2 domains contribute to SHP-2 specificity, studies of SHP chimeras reveal that substantial specificity resides in the PTP domain. Thus, PTP domains exhibit biologically relevant specificity in vivo, and noncatalytic and catalytic domains of PTPs contribute to specificity in a combinatorial fashion.


Subject(s)
Mesoderm/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Xenopus/metabolism , Animals , Intracellular Signaling Peptides and Proteins , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/genetics , SH2 Domain-Containing Protein Tyrosine Phosphatases , Structure-Activity Relationship , Substrate Specificity , Xenopus/embryology
16.
Mol Cell Biol ; 12(9): 4164-9, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1508211

ABSTRACT

Early-growth-response genes, also known as immediate-early genes, play important roles in regulating cell proliferation. We have identified a new type of early-growth-response gene product, a 77,811-Da putative serine/threonine kinase, which is highly inducible by serum and phorbol ester. mRNA encoding this putative kinase is markedly elevated within 1 h after treatment with mitogen, and this induction is synergistically increased by cycloheximide. Dexamethasone blocks serum induction of the kinase mRNA, as does transformation by v-Ki-ras. The kinase mRNA was detected in mouse brain, lung, and heart. This new putative kinase, which we term Snk, for serum-inducible kinase, showed similarity in its proposed catalytic domain to many other protein kinases; however, no other kinase showed enough sequence similarity with Snk to suggest the existence of a common function. Hence, Snk represents a new type of protein kinase involved in the early mitogenic response whose activity is transcriptionally and posttranscriptionally regulated.


Subject(s)
Protein Kinases/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , DNA , Gene Expression Regulation , Glucocorticoids/physiology , Male , Mice , Mitogens/pharmacology , Molecular Sequence Data , Organ Specificity/genetics , Protein Kinases/metabolism , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid , Transcription, Genetic
17.
Mol Cell Biol ; 20(1): 299-311, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10594032

ABSTRACT

In Xenopus ectodermal explants (animal caps), fibroblast growth factor (FGF) evokes two major events: induction of ventrolateral mesodermal tissues and elongation. The Xenopus FGF receptor (XFGFR) and certain downstream components of the XFGFR signal transduction pathway (e.g., members of the Ras/Raf/MEK/mitogen-activated protein kinase [MAPK] cascade) are required for both of these processes. Likewise, activated versions of these signaling components induce mesoderm and promote animal cap elongation. Previously, using a dominant negative mutant approach, we showed that the protein-tyrosine phosphatase SHP-2 is necessary for FGF-induced MAPK activation, mesoderm induction, and elongation of animal caps. Taking advantage of recent structural information, we now have generated novel, activated mutants of SHP-2. Here, we show that expression of these mutants induces animal cap elongation to an extent comparable to that evoked by FGF. Surprisingly, however, activated mutant-induced elongation can occur without mesodermal cytodifferentiation and is accompanied by minimal activation of the MAPK pathway and mesodermal marker expression. Our results implicate SHP-2 in a pathway(s) directing cell movements in vivo and identify potential downstream components of this pathway. Our activated mutants also may be useful for determining the specific functions of SHP-2 in other signaling systems.


Subject(s)
Protein Tyrosine Phosphatases/genetics , Signal Transduction , Xenopus laevis/embryology , Xenopus laevis/genetics , Animals , Ectoderm , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Intracellular Signaling Peptides and Proteins , Mutation , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction/drug effects , src Homology Domains/genetics
18.
Mol Cell Biol ; 16(7): 3685-97, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668185

ABSTRACT

SHPTP1 (PTP1C, HCP, SHP) is an SH2 domain-containing tyrosine phosphatase expressed predominantly in hematopoietic cells. A frameshift mutation in the SHPTP1 gene causes the motheaten (me/me) mouse. These mice are essentially SHPTP1 null and display multiple hematopoietic abnormalities, most prominently hyperproliferation and inappropriate activation of granulocytes and macrophages. The me/me phenotype suggests that SHPTP1 negatively regulates macrophage proliferative pathways. Using primary bone marrow-derived macrophages from me/me mice and normal littermates, we examined the role of SHPTP1 in regulating signaling by the major macrophage mitogen colony-stimulating factor 1 (CSF-1) (also known as macrophage colony-stimulating factor). Macrophages from me/me mice hyperproliferate in response to CSF-1. In the absence of SHPTP1, the CSF-1 receptor (CSF-1R) is hyperphosphorylated upon CSF-1 stimulation, suggesting that SHPTP1 dephosphorylates the CSF-1R. At least some CSF-1R-associated proteins also are hyperactivated. SHPTP1 is associated constitutively, via its SH2 domains, with an unidentified 130-kDa phosphotyrosyl protein (P130). P130 and SHPTP1 are further tyrosyl phosphorylated upon CSF-1 stimulation. Tyrosyl-phosphorylated SHPTP1 binds to Grb2 via the Grb2 SH2 domain. Moreover, in me/me macrophages, Grb2 is associated, via its SH3 domains, with several tyrosyl phosphoproteins. These proteins are hyperphosphorylated on tyrosyl residues in me/me macrophages, suggesting that Grb2 may recruit substrates for SHPTP1. Our results indicate that SHPTP1 is a critical negative regulator of CSF-1 signaling in vivo and suggest a potential new function for Grb2.


Subject(s)
Hematopoietic Stem Cells/cytology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/physiology , Protein Tyrosine Phosphatases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Bone Marrow Cells , Cell Division/drug effects , Cells, Cultured , Frameshift Mutation , Hematopoietic Stem Cells/physiology , Intracellular Signaling Peptides and Proteins , Kinetics , Macrophage Activation , Macrophages/drug effects , Mice , Mice, Inbred C3H , Mice, Knockout , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/isolation & purification , Receptor, Macrophage Colony-Stimulating Factor/genetics , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction , src Homology Domains
19.
Mol Cell Biol ; 15(12): 7050-8, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8524272

ABSTRACT

Interferons (IFNs) induce early-response genes by stimulating Janus family (Jak) tyrosine kinases, leading to tyrosine phosphorylation of Stat transcription factors. Previous studies implicated protein-tyrosine phosphatase (PTP) activity in the control of IFN-regulated Jak/Stat signaling, but the specific PTPs responsible remained unidentified. We have found that SH2 domain-containing PTP1 (SHPTP1; also called PTP1C, HCP, or SHP) reversibly associates with the IFN-alpha receptor complex upon IFN addition. Compared with macrophages from normal littermate controls, macrophages from motheaten mice, which lack SHPTP1, show dramatically increased Jak1 and Stat1 alpha tyrosine phosphorylation, whereas Tyk2 and Stat2 activation is largely unaffected. These findings correlate with selectively increased complex formation on a gamma response element, but not an IFN-stimulated response element, in motheaten macrophages. Our results establish that SHPTP1 selectively regulates distinct components of Jak/Stat signal transduction pathways in vivo.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Interferon Type I/pharmacology , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Base Sequence , Cell Line , DNA-Binding Proteins/isolation & purification , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Macrophages/drug effects , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Molecular Sequence Data , Oligodeoxyribonucleotides , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/isolation & purification , Protein-Tyrosine Kinases/isolation & purification , SH2 Domain-Containing Protein Tyrosine Phosphatases , STAT2 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/isolation & purification
20.
Mol Cell Biol ; 14(3): 1824-34, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8114715

ABSTRACT

The phosphorylation and dephosphorylation of proteins on tyrosyl residues are key regulatory mechanisms in T-cell signal transduction and are controlled by the opposing activities of protein tyrosine kinases and phosphotyrosyl phosphatases (PTPs). In T cells, several nontransmembrane protein tyrosine kinases are associated with receptors; for example, Lck is bound to the coreceptors CD4 and CD8 and becomes activated upon their stimulation. In comparison, little is known about the role of nontransmembrane PTPs in early T-cell signaling. SH-PTP1 (PTP1C, HCP, SHP) is a nontransmembrane PTP expressed primarily in hematopoietic cells, including T cells. We have found that SH-PTP1 is basally phosphorylated on serine in resting T cells. Upon stimulation of CD4 or CD8 either in a T-cell hybridoma cell line or in primary thymocytes, SH-PTP1 becomes tyrosyl phosphorylated. Moreover, SH-PTP1 is constitutively phosphorylated on tyrosine in the Lck-overexpressing lymphoma cell line LSTRA. SH-PTP1 is also a good substrate for recombinant Lck in vitro. Comparisons of the tryptic phosphopeptide maps of wild-type SH-PTP1 and deletion and point mutations establish that the two sites (Y-536 and Y-564) which are directly phosphorylated by Lck in vitro are also phosphorylated in vivo in LSTRA cells. One of these sites (Y-564) is phosphorylated in T cells in response to Lck activation. We conclude that SH-PTP1 undergoes Lck-dependent tyrosyl phosphorylation in T cells and likely plays a role in early T-cell signaling.


Subject(s)
Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/enzymology , Amino Acid Sequence , Animals , Base Sequence , CD4 Antigens/physiology , CD8 Antigens/physiology , DNA Primers/chemistry , Humans , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Mice , Molecular Sequence Data , Peptide Mapping , Phosphorylation , Receptors, Immunologic/physiology , Recombinant Proteins , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL