Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Science ; 364(6436): 188-193, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30975888

ABSTRACT

Notch signaling is a core patterning module for vascular morphogenesis that codetermines the sprouting behavior of endothelial cells (ECs). Tight quantitative and temporal control of Notch activity is essential for vascular development, yet the details of Notch regulation in ECs are incompletely understood. We found that ubiquitin-specific peptidase 10 (USP10) interacted with the NOTCH1 intracellular domain (NICD1) to slow the ubiquitin-dependent turnover of this short-lived form of the activated NOTCH1 receptor. Accordingly, inactivation of USP10 reduced NICD1 abundance and stability and diminished Notch-induced target gene expression in ECs. In mice, the loss of endothelial Usp10 increased vessel sprouting and partially restored the patterning defects caused by ectopic expression of NICD1. Thus, USP10 functions as an NICD1 deubiquitinase that fine-tunes endothelial Notch responses during angiogenic sprouting.


Subject(s)
Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Proteolysis , Receptor, Notch1/metabolism , Ubiquitin Thiolesterase/physiology , Animals , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Protein Domains , Protein Stability , RNA, Small Interfering/genetics , Signal Transduction , Ubiquitin Thiolesterase/genetics
2.
Cell Death Differ ; 20(1): 21-30, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22722335

ABSTRACT

Ubiquitination has long been recognised as a key determinator of protein fate by tagging proteins for proteasomal degradation. Most recently, the ability of conjugated ubiquitin chains to confer selectivity to autophagy was demonstrated. Although autophagy was first believed to be a bulk, non-selective 'self-eating' degradative process, the molecular mechanisms of selectivity are now starting to emerge. With the discovery of autophagy receptors - which bind both ubiquitinated substrates and autophagy specific light chain 3 (LC3) modifier on the inner sheath of autophagosomes - a new pathway of selective autophagy is being unravelled. In this review, we focus on the special role of ubiquitin signals and selective autophagy receptors in sorting a variety of autophagic cargos.


Subject(s)
Autophagy/physiology , Ubiquitin/metabolism , Ubiquitination/physiology , Humans , Signal Transduction
3.
Pathologica ; 104(2): 56-64, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22953501

ABSTRACT

AIMS: To study the expression of CD2-associated protein (CD2AP), an adaptor protein involved in T-cell signalling and renal function, in normal, reactive and neoplastic human lymphoid tissues. METHODS AND RESULTS: We used immunohistochemical techniques to evaluate monoclonal antibodies against CD2AP on over 400 formalin fixed paraffin embedded tissue blocks retrieved from the host institutions of three authors. The samples tested included normal, reactive and neoplastic lymphoid tissue. In lymphoid tissues, strong CD2AP staining was observed in plasmacytoid dendritic cells (pDCs), weak and variable in mantle zone B cells and moderate in rare germinal center cells. CD2AP labeled cortical and rare medullary thymocytes and isolated mononuclear cells in bone marrow trephines. Furthermore, epithelial and endothelial cells expressed CD2AP. Among neoplasms, the greatest number of CD2AP-positive cases were found in diffuse large B cell (21/94), NK T-cell lymphomas (7/67), "blastic plasmacytoid dendritic cell neoplasms" (9/10) and some types of solid tumor. CONCLUSIONS: Our finding that mature peripheral T cells are CD2AP-negative but immature cortical thymocytes are positive may prove useful for diagnostic purposes. Moreover, our results demonstrate that CD2AP represents a useful marker of normal and neoplastic pDC and may be used in a diagnostic panel in reactive or neoplastic lymphoid proliferations.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/metabolism , Cytoskeletal Proteins/metabolism , Dendritic Cells/metabolism , Lymphoma/diagnosis , Lymphoma/metabolism , Thymocytes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Biomarkers/metabolism , Cell Line , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/immunology , Humans , Immunohistochemistry , Lymphocytes/cytology
4.
Oncogene ; 31(43): 4599-608, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22179831

ABSTRACT

Once stimulated, the epidermal growth factor receptor (EGFR) undergoes self-phosphorylation, which, on the one hand, instigates signaling cascades, and on the other hand, recruits CBL ubiquitin ligases, which mark EGFRs for degradation. Using RNA interference screens, we identified a deubiquitinating enzyme, Cezanne-1, that opposes receptor degradation and enhances EGFR signaling. These functions require the catalytic- and ubiquitin-binding domains of Cezanne-1, and they involve physical interactions and transphosphorylation of Cezanne-1 by EGFR. In line with the ability of Cezanne-1 to augment EGF-induced growth and migration signals, the enzyme is overexpressed in breast cancer. Congruently, the corresponding gene is amplified in approximately one third of mammary tumors, and high transcript levels predict an aggressive disease course. In conclusion, deubiquitination by Cezanne-1 curtails degradation of growth factor receptors, thereby promotes oncogenic growth signals.


Subject(s)
Endopeptidases/metabolism , ErbB Receptors/metabolism , Neoplasms/pathology , Catalysis , Disease Progression , Humans , Neoplasms/metabolism , Phosphorylation , RNA, Small Interfering , Ubiquitin/metabolism , Ubiquitination
5.
Oncogene ; 27(26): 3739-45, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18212736

ABSTRACT

Deregulated nuclear factor kappaB (NF-kappaB) activation plays an important role in inflammation and tumorigenesis. ABIN proteins have been characterized as negative regulators of NF-kappaB signaling. However, their mechanism of NF-kappaB inhibition remained unclear. With the help of a yeast two-hybrid screen, we identified ABIN proteins as novel ubiquitin-interacting proteins. The minimal ubiquitin-binding domain (UBD) corresponds to the ABIN homology domain 2 (AHD2) and is highly conserved in ABIN-1, ABIN-2 and ABIN-3. Moreover, this region is also present in NF-kappaB essential modulator/IkappaB kinase gamma (NEMO/IKKgamma) and the NEMO-like protein optineurin, and is therefore termed UBD in ABIN proteins and NEMO (UBAN). Nuclear magnetic resonance studies of the UBAN domain identify it as a novel type of UBD, with the binding surface on ubiquitin being significantly different from the binding surface of other UBDs. ABIN-1 specifically binds ubiquitinated NEMO via a bipartite interaction involving its UBAN and NEMO-binding domain. Mutations in the UBAN domain led to a loss of ubiquitin binding and impaired the NF-kappaB inhibitory potential of ABINs. Taken together, these data illustrate an important role for ubiquitin binding in the negative regulation of NF-kappaB signaling by ABINs and identify UBAN as a novel UBD.


Subject(s)
DNA-Binding Proteins/physiology , NF-kappa B/antagonists & inhibitors , Ubiquitin/metabolism , Binding Sites , Cell Line , DNA-Binding Proteins/chemistry , Humans , NF-kappa B/physiology , Protein Structure, Tertiary , Signal Transduction , Two-Hybrid System Techniques
6.
Biochem Soc Trans ; 35(Pt 5): 942-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17956251

ABSTRACT

The NF-kappaB (nuclear factor kappaB) transcription factors control cell survival, proliferation and innate and adaptive immune response. Post-translational modifications of key components of the NF-kappaB pathway provide the molecular basis for signal transmission from the cell membrane to the nucleus. Here, we describe the involvement of different types of ubiquitin modification in the regulation of the NF-kappaB signalling pathway.


Subject(s)
NF-kappa B/metabolism , Signal Transduction , Ubiquitin/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism
7.
Oncogene ; 25(21): 3071-8, 2006 05 18.
Article in English | MEDLINE | ID: mdl-16407834

ABSTRACT

The Pix/Cool proteins are involved in the regulation of cell morphology by binding to small Rho GTPases and kinases of the Pak family. Recently, it has been shown that betaPix/Cool-1 associates with the ubiquitin ligase Cbl, which appears to be a critical step in Cdc42-mediated inhibition of epidermal-growth-factor-receptor (EGFR) ubiquitylation and downregulation. Here we show that the SH3 domain of betaPix specifically interacts with a proline-arginine motif (PxxxPR) present within the ubiquitin ligase Cbl and Pak1 kinase. Owing to targeting of the same sequence, Cbl and Pak1 compete for binding to betaPix. In this complex, Cbl mediates ubiquitylation and subsequent degradation of betaPix. Our findings reveal a double feedback loop in which the Cdc42/betaPix complex blocks Cbl's ability to downregulate EGFR, while Cbl in turn promotes degradation of betaPix in order to escape this inhibition. Such a relationship provides a mechanism to fine-tune the kinetics of RTK endocytosis and degradation depending on the pool of active Cdc42 and the duration of EGFR signaling.


Subject(s)
Cell Cycle Proteins/physiology , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/physiology , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-cbl/physiology , cdc42 GTP-Binding Protein/physiology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Binding, Competitive , Breast Neoplasms/pathology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line/metabolism , Cell Line, Tumor/metabolism , Endocytosis , Epithelial Cells/metabolism , ErbB Receptors/biosynthesis , ErbB Receptors/physiology , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Genes, erbB-1 , Guanine Nucleotide Exchange Factors/biosynthesis , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Kidney , Neoplasm Proteins/physiology , Protein Binding , Protein Interaction Mapping , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/chemistry , Proto-Oncogene Proteins c-cbl/genetics , Recombinant Fusion Proteins/physiology , Rho Guanine Nucleotide Exchange Factors , Transfection , Ubiquitin/metabolism , p21-Activated Kinases , rac1 GTP-Binding Protein/metabolism , src Homology Domains
8.
Biochem Soc Trans ; 31(Pt 6): 1178-81, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14641021

ABSTRACT

Activated EGF (epidermal growth factor) receptors are removed from the cell surface via endocytosis and subsequent degradation in the lysosome. This ultimately attenuates EGF receptor signals and diminishes the level of cell activation. The mechanisms underlying the EGF receptor down-regulation are beginning to be elucidated at the molecular level. Recent reports have indicated that receptor monoubiquitination and networks of protein-protein interactions control distinct steps in EGF receptor internalization, endosomal trafficking and sorting for lysosomal degradation. The emerging importance of the ubiquitin ligase Cbl and the adaptor molecule CIN85 (Cbl-interacting protein of 85 kDa) in the regulation of these pathways is discussed in detail.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , Hydrolysis
9.
Cell Mol Life Sci ; 60(9): 1805-27, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14523545

ABSTRACT

Cbl proteins control multiple cellular processes by acting as ubiquitin ligases and multifunctional adaptor molecules. They are involved in the control of cell proliferation, differentiation and cell morphology, as well as in pathologies such as autoimmune diseases, inflammation and cancer. Here we review recent advances in understanding the role of Cbl and the importance of a growing repertoire of Cbl-interacting proteins in the regulation of signaling pathways triggered by growth factors, antigens, cell adhesion, cytokines and hormones. We also address key issues of the nature of proteins that bind Cbl in particular cells, where they are located, and how they are altered or traffic within cells upon stimulation. It is becoming obvious that temporal and spatial changes in Cbl signaling networks are essential for the control of physiological processes in a variety of cells and organs and that their deregulation can result in the development of human diseases.


Subject(s)
Retroviridae Proteins, Oncogenic/metabolism , Signal Transduction/physiology , Animals , Endocytosis/physiology , Humans , Ligases/metabolism , Oncogene Protein v-cbl , Protein Binding , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Retroviridae Proteins, Oncogenic/genetics , Ubiquitin/metabolism
10.
Biochemistry ; 40(51): 15743-51, 2001 Dec 25.
Article in English | MEDLINE | ID: mdl-11747451

ABSTRACT

To investigate the palmitoylation of the human bradykinin B2 receptor, we have mutated individually or simultaneously into glycine two potential acylation sites (cysteines 324 and 329) located in the carboxyl terminus of the receptor and evaluated the effects of these mutations by transfection in COS-7, CHO-K1, and HEK 293T. The wild-type receptor and the single mutants, but not the double mutant, incorporated [3H]palmitate, indicating that the receptor carboxyl tail can be palmitoylated at both sites. The mutants did not differ from the wild-type receptor for the kinetics of [3H]bradykinin binding, the basal and bradykinin-stimulated coupling to phospholipases C and A2, and agonist-induced phosphorylation. The nonpalmitoylated receptor had a 30% reduced capacity to internalize [3H]bradykinin. This indicates that palmitoylation does not influence the basal activity of the receptor and its agonist-driven activation. However, the mutants triggered phospholipid metabolism and MAP kinase activation in response to B2 receptor antagonists. Pseudopeptide and nonpeptide compounds that behaved as antagonists on the wild-type receptor became agonists on the nonpalmitoylated receptor and produced phospholipases C and A2 responses of 25-50% as compared to that of bradykinin. These results suggest that palmitoylation is required for the stabilization of the receptor-ligand complex in an uncoupled conformation.


Subject(s)
Bradykinin/analogs & derivatives , Palmitic Acid/metabolism , Receptors, Bradykinin/metabolism , Amidines/pharmacology , Amino Acid Sequence , Animals , Bradykinin/pharmacology , Bradykinin Receptor Antagonists , CHO Cells , COS Cells , Cell Line , Cricetinae , Gene Expression Regulation/drug effects , Humans , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Peptide Fragments/metabolism , Piperazines/pharmacology , Receptor, Bradykinin B2 , Receptors, Bradykinin/biosynthesis , Receptors, Bradykinin/genetics , Transfection
11.
J Biol Chem ; 276(44): 40431-40, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11517230

ABSTRACT

Reversible phosphorylation plays important roles in G protein-coupled receptor signaling, desensitization, and endocytosis, yet the precise location and role of in vivo phosphorylation sites is unknown for most receptors. Using metabolic 32P labeling and phosphopeptide sequencing we provide a complete phosphorylation map of the human bradykinin B2 receptor in its native cellular environment. We identified three serine residues, Ser(339), Ser(346), and Ser(348), at the C-terminal tail as principal phosphorylation sites. Constitutive phosphorylation occurs at Ser(348), while ligand-induced phosphorylation is found at Ser(339) and Ser(346)/Ser(348) that could be executed by several G protein-coupled receptor kinases. In addition, we found a protein kinase C-dependent phosphorylation of Ser(346) that was mutually exclusive with the basal phosphorylation at Ser(348) and therefore may be implicated in differential regulation of B2 receptor activation. Functional analysis of receptor mutants revealed that a low phosphorylation stoichiometry is sufficient to initiate receptor sequestration while a clustered phosphorylation around Ser(346) is necessary for desensitization of the B2 receptor-induced phospholipase C activation. This was further supported by the specifically reduced Ser(346)/Ser(348) phosphorylation observed upon stimulation with a nondesensitizing B2 receptor agonist. The differential usage of clustered phosphoacceptor sites points to distinct roles of multiple kinases in controlling G protein-coupled receptor function.


Subject(s)
Receptors, Bradykinin/metabolism , Serine/metabolism , Amino Acid Sequence , Cell Line , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Phosphorylation , Receptor, Bradykinin B2 , Receptors, Bradykinin/chemistry , Receptors, Bradykinin/genetics , Receptors, Bradykinin/physiology
12.
J Mol Med (Berl) ; 78(12): 708-20, 2001.
Article in English | MEDLINE | ID: mdl-11434724

ABSTRACT

Natural silicate materials, including zeolite clinoptilolite, have been shown to exhibit diverse biological activities and have been used successfully as a vaccine adjuvant and for the treatment of diarrhea. We report a novel use of finely ground clinoptilolite as a potential adjuvant in anticancer therapy. Clinoptilolite treatment of mice and dogs suffering from a variety of tumor types led to improvement in the overall health status, prolongation of life-span, and decrease in tumors size. Local application of clinoptilolite to skin cancers of dogs effectively reduced tumor formation and growth. In addition, toxicology studies on mice and rats demonstrated that the treatment does not have negative effects. In vitro tissue culture studies showed that finely ground clinoptilolite inhibits protein kinase B (c-Akt), induces expression of p21WAF1/CIP1 and p27KIP1 tumor suppressor proteins, and blocks cell growth in several cancer cell lines. These data indicate that clinoptilolite treatment might affect cancer growth by attenuating survival signals and inducing tumor suppressor genes in treated cells.


Subject(s)
Adjuvants, Pharmaceutic/therapeutic use , Neoplasms/drug therapy , Protein Serine-Threonine Kinases , Zeolites/therapeutic use , Adjuvants, Pharmaceutic/adverse effects , Adjuvants, Pharmaceutic/pharmacology , Aging/physiology , Animals , Apoptosis/drug effects , Body Weight/drug effects , Cell Cycle Proteins/analysis , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/analysis , Dog Diseases/drug therapy , Dog Diseases/pathology , Dogs , Female , HeLa Cells , Humans , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Neoplasms/pathology , Neoplasms/veterinary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Signal Transduction/drug effects , Tumor Cells, Cultured , Tumor Suppressor Proteins/analysis , Zeolites/adverse effects , Zeolites/pharmacology
13.
J Biol Chem ; 276(38): 35537-45, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11463795

ABSTRACT

Insulin controls glucose uptake by translocating GLUT4 and other glucose transporters to the plasma membrane in muscle and adipose tissues by a mechanism that appears to require protein kinase C (PKC)-zeta/lambda operating downstream of phosphatidylinositol 3-kinase. In diabetes mellitus, insulin-stimulated glucose uptake is diminished, but with hyperglycemia, uptake is maintained but by uncertain mechanisms. Presently, we found that glucose acutely activated PKC-zeta/lambda in rat adipocytes and rat skeletal muscle preparations by a mechanism that was independent of phosphatidylinositol 3-kinase but, interestingly, dependent on the apparently sequential activation of the dantrolene-sensitive, nonreceptor proline-rich tyrosine kinase-2; components of the extracellular signal-regulated kinase (ERK) pathway, including, GRB2, SOS, RAS, RAF, MEK1 and ERK1/2; and, most interestingly, phospholipase D, thus yielding increases in phosphatidic acid, a known activator of PKC-zeta/lambda. This activation of PKC-zeta/lambda, moreover, appeared to be required for glucose-induced increases in GLUT4 translocation and glucose transport in adipocytes and muscle cells. Our findings suggest the operation of a novel pathway for activating PKC-zeta/lambda and glucose transport.


Subject(s)
Glucose/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Muscle Proteins , Phospholipase D/metabolism , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Adipocytes/enzymology , Adipocytes/metabolism , Androstadienes/pharmacology , Animals , Dantrolene/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Focal Adhesion Kinase 2 , Glucose Transporter Type 4 , Isoenzymes , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Protein Transport , Rats , Wortmannin
14.
Oncogene ; 20(31): 4180-7, 2001 Jul 12.
Article in English | MEDLINE | ID: mdl-11464284

ABSTRACT

The Cdx1 homeobox gene encodes for an intestine-specific transcription factor involved in the control of proliferation and differentiation of epithelial cells. Although it has been indicated that Cdx1 may act as a proto-oncogene in cultured fibroblasts, its direct role in the regulation of intestinal tumorigenesis has not been demonstrated. Here we show that expression of Cdx1 in an intestinal epithelial cell line (IEC-6) induces anchorage-independent growth in soft agar and promotes the formation of adenocarcinoma in vivo. The phenotype of Cdx1-induced tumors was exacerbated when IEC-6/Cdx1 cells were injected together with matrigel containing mitogens and extracellular matrix components. These changes were correlated with an increase in the GTP-bound form of Ras, modulation of Cdc42 and Rho-A activities, and accumulation of phosphatidyl inositol 3 (PI3) kinase products. Moreover, combined inhibition of Ras/Rho and PI3 kinase signaling by synthethic inhibitors blocked colony formation of IEC-6/Cdx1 cells in soft agar. Taken together, these results demonstrate a direct involvement of Cdx1, and its collaboration with Ras, Rho and PI3 kinase pathways, in transformation and tumorigenesis of intestinal epithelial cells.


Subject(s)
Avian Proteins , Cell Transformation, Neoplastic/genetics , GTP-Binding Proteins/metabolism , Genes, Homeobox , Homeodomain Proteins/genetics , Intestinal Mucosa/pathology , Phosphatidylinositol 3-Kinases/metabolism , ras Proteins/metabolism , Animals , Cell Line , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Rats
15.
Biol Chem ; 382(1): 135-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11258664

ABSTRACT

Sphingosine kinase phosphorylates sphingosine to generate sphingosine 1-phosphate, a phospholipid that has been implicated in signaling by a number of transmembrane receptors and was recently shown to act as a ligand for a specific class of G protein-coupled receptors. Here we show that the G protein-coupled bradykinin B2 receptor activates sphingosine kinase leading to a time- and dose-dependent elevation of cellular sphingosine 1-phosphate levels that was blocked by the sphingosine kinase inhibitor dihydrosphingosine. Furthermore, increasing doses of this inhibitor partially affected the bradykinin-mediated ERK/MAP kinase activation and fully blocked the protein kinase C-independent component of the signaling pathway from the B2 receptor to the ERK/MAP kinase cascade. Overexpression of sphingosine kinase did not additionally increase the bradykinin-induced ERK/MAP kinase activity, indicating a permissive rather than activating role of sphingosine 1-phosphate in B2 receptor-mediated mitogenic signaling.


Subject(s)
Lysophospholipids , Mitogen-Activated Protein Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Bradykinin/physiology , Sphingosine/analogs & derivatives , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/genetics , Humans , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/genetics , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Receptor, Bradykinin B2 , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/biosynthesis , Sphingosine/pharmacology
16.
J Biol Chem ; 275(52): 40817-26, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11007796

ABSTRACT

Glucose serves as both a nutrient and regulator of physiological and pathological processes. Presently, we found that glucose and certain sugars rapidly activated extracellular signal-regulated kinase (ERK) by a mechanism that was: (a) independent of glucose uptake/metabolism and protein kinase C but nevertheless cytochalasin B-inhibitable; (b) dependent upon proline-rich tyrosine kinase-2 (PYK2), GRB2, SOS, RAS, RAF, and MEK1; and (c) amplified by overexpression of the Glut1, but not Glut2, Glut3, or Glut4, glucose transporter. This amplifying effect was independent of glucose uptake but dependent on residues 463-468, IASGFR, in the Glut1 C terminus. Accordingly, glucose effects on ERK were amplified by expression of Glut4/Glut1 or Glut2/Glut1 chimeras containing IASGFR but not by Glut1/Glut4 or Glut1/Glut2 chimeras lacking these residues. Also, deletion of Glut1 residues 469-492 was without effect, but mutations involving serine 465 or arginine 468 yielded dominant-negative forms that inhibited glucose-dependent ERK activation. Glucose stimulated the phosphorylation of tyrosine residues 402 and 881 in PYK2 and binding of PYK2 to Myc-Glut1. Our findings suggest that: (a) glucose activates the GRB2/SOS/RAS/RAF/MEK1/ERK pathway by a mechanism that requires PYK2 and residues 463-468, IASGFR, in the Glut1 C terminus and (b) Glut1 serves as a sensor, transducer, and amplifier for glucose signaling to PYK2 and ERK.


Subject(s)
Glucose/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Monosaccharide Transport Proteins/physiology , Protein-Tyrosine Kinases/physiology , 3T3 Cells , Adipocytes/metabolism , Animals , Deoxyglucose/metabolism , Disaccharides/pharmacology , Focal Adhesion Kinase 2 , Glucose Transporter Type 1 , Glucose Transporter Type 2 , Mice , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats
17.
Nat Cell Biol ; 2(9): 574-81, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980697

ABSTRACT

Integration of signalling pathways initiated by receptor tyrosine kinases and integrins is essential for growth-factor-mediated biological responses. Here we show that co-stimulation of growth-factor receptors and integrins activates the focal-adhesion kinase (FAK) family to promote outgrowth of neurites in PC12 and SH-SY5Y cells. Pyk2 and FAK associate with adhesion-based complexes that contain epidermal growth factor (EGF) receptors, through their carboxy- and amino-terminal domains. Expression of the C-terminal domain of Pyk2 or of FAK is sufficient to block neurite outgrowth, but not activation of extracellular-signal-regulated kinase (ERK). Moreover, activation and autophosphorylation of Pyk2/FAK, as well as of effectors of their adhesion-targeting domains, such as paxillin, are important for propagation of signals that control neurite formation. Thus, Pyk2/FAK have important functions in signal integration proximal to integrin/growth-factor receptor complexes in neurons.


Subject(s)
Growth Substances/metabolism , Integrins/metabolism , Neurites/physiology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Animals , Cytoskeletal Proteins/metabolism , Enzyme Activation , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Kinase 2 , Focal Adhesion Protein-Tyrosine Kinases , Growth Substances/pharmacology , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Paxillin , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Tumor Cells, Cultured
18.
Mol Cell Biol ; 20(18): 6837-48, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10958680

ABSTRACT

G protein-coupled receptors (GPCRs) have been shown to stimulate extracellular regulated kinases (ERKs) through a number of linear pathways that are initiated by G(q/11) or G(i) proteins. We studied signaling to the ERK cascade by receptors that simultaneously activate both G protein subfamilies. In HEK293T cells, bradykinin B(2) receptor (B(2)R)-induced stimulation of ERK2 and transcriptional activity of Elk1 are dependent on Galpha(q)-mediated protein kinase C (PKC) and on Galpha(i)-induced Ras activation, while they are independent of Gbetagamma subunits, phosphatidylinositol 3-kinase, and tyrosine kinases. Similar results were obtained with m(1) and m(3) muscarinic receptors in HEK293T cells and with the B(2)R in human and mouse fibroblasts, indicating a general mechanism in signaling toward the ERK cascade. Furthermore, the bradykinin-induced activation of ERK is strongly reduced in Galpha(q/11)-deficient fibroblasts. In addition, we found that constitutively active mutants of Galpha(q/11) or Galpha(i) proteins alone poorly stimulate ERK2, whereas a combination of both led to synergistic effects. We conclude that dually coupled GPCRs require a cooperation of Galpha(i)- and G(q/11)-mediated pathways for efficient stimulation of the ERK cascade. Cooperative signaling by multiple G proteins thus might represent a novel concept implicated in the regulation of cellular responses by GPCRs.


Subject(s)
DNA-Binding Proteins , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins , Transcription Factors , Animals , Cell Line , Cell Line, Transformed , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gq-G11 , GTP-Binding Proteins/genetics , Humans , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Receptor, Bradykinin B2 , Receptor, Muscarinic M1 , Receptor, Muscarinic M3 , Receptors, Bradykinin/metabolism , Receptors, Cell Surface/metabolism , Receptors, Muscarinic/metabolism , Transcriptional Activation , Virulence Factors, Bordetella/pharmacology , beta-Adrenergic Receptor Kinases , ets-Domain Protein Elk-1 , ras Proteins/metabolism
19.
J Biol Chem ; 275(38): 29338-47, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10882725

ABSTRACT

The cytokine-inducible SH2 protein-3 (CIS3/SOCS-3/SSI-3) has been shown to inhibit the JAK/STAT pathway and act as a negative regulator of fetal liver erythropoiesis. Here, we studied the molecular mechanisms by which CIS3 regulates the erythropoietin (EPO) receptor (EPOR) signaling in erythroid progenitors and Ba/F3 cells expressing the EPOR (BF-ER). CIS3 binds directly to the EPOR as well as JAK2 and inhibits EPO-dependent proliferation and STAT5 activation. We have identified the region containing Tyr(401) in the cytoplasmic domain of the EPOR as a direct binding site for CIS3. Deletion of the Tyr(401) region of the EPOR reduced the inhibitory effect of CIS3, suggesting that binding of CIS3 to the EPOR augmented the negative effect of CIS3. Both N- and C-terminal regions adjacent to the SH2 domain of CIS3 were necessary for binding to EPOR and JAK2. In the N-terminal region of CIS3, the amino acid Gly(45) was critical for binding to the EPOR but not to JAK2, while Leu(22) was critical for binding to JAK2. The mutation of G45A partially reduced ability of CIS3 to inhibit EPO-dependent proliferation and STAT5 activation, while L22D mutant CIS3 was completely unable to suppress EPOR signaling. Moreover, overexpression of STAT5, which also binds to Tyr(401), reduced the binding of CIS3 to the EPOR, and the inhibitory effect of CIS3 against EPO signaling, while it did not affect JAB/SOCS-1/SSI-1. These data demonstrate that binding of CIS3 to the EPOR augments the inhibitory effect of CIS3. CIS3 binding to both EPOR and JAK2 may explain a specific regulatory role of CIS3 in erythropoiesis.


Subject(s)
Erythropoietin/physiology , Protein-Tyrosine Kinases/physiology , Proteins/physiology , Proto-Oncogene Proteins , Receptors, Erythropoietin/physiology , Repressor Proteins , Signal Transduction/physiology , Transcription Factors , Animals , Cells, Cultured , Erythroblasts/physiology , Erythropoietin/pharmacology , Janus Kinase 2 , Mice , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
20.
Gastroenterology ; 117(6): 1326-38, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10579974

ABSTRACT

BACKGROUND & AIMS: Homeobox genes are involved in establishing and maintaining differentiated patterns in adult tissues. Cdx1 might carry out that function in the intestinal epithelium because its expression is specific to that tissue and increases during development. METHODS: Cdx1 expression was induced in IEC-6 intestinal epithelial cells by stable transfection, and subsequent changes in cell growth, resistance to apoptosis, migration, and differentiation were monitored. RESULTS: Compared with control, IEC-6/Cdx1 cells proliferated more rapidly, were more resistant to apoptosis, and migrated 3-4 times faster, as shown by an in vitro wound assay. IEC-6/Cdx1 cells in culture formed multilayers. Morphology of the top layer was similar to that of columnar epithelium, with cells showing typical features of differentiated enterocytes, including complex junctions and well-developed microvilli with glycocalix. Expression of 2 markers of enterocyte differentiation, aminopeptidase N and villin, was induced in IEC-6/Cdx1 cells. Aminopeptidase N was targeted to the basolateral membrane, and villin was localized to the cytoplasm. Actin filaments, which were mostly present in transcytoplasmic stress fibers in control cells, were redistributed to the cortex in Cdx1-transfected cells. CONCLUSIONS: Cdx1 expression in IEC-6 cells induces phenotypic changes characteristic of differentiating enterocytes, suggesting an important role for Cdx1 in the transition from stem cells to proliferating/transit cells.


Subject(s)
Avian Proteins , Homeodomain Proteins/physiology , Intestinal Mucosa/cytology , Actins/metabolism , Animals , Apoptosis/physiology , CD13 Antigens/metabolism , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Cell Movement/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Microfilament Proteins/metabolism , Phenotype , Rats , Stem Cells/cytology , Transfection , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...