Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Oncogene ; 37(49): 6368-6382, 2018 12.
Article in English | MEDLINE | ID: mdl-30061636

ABSTRACT

The life expectancy for pancreatic cancer patients has seen no substantial changes in the last 40 years as very few and mostly just palliative treatments are available. As the five years survival rate remains around 5%, the identification of novel pharmacological targets and development of new therapeutic strategies are urgently needed. Here we demonstrate that inhibition of the G protein-coupled receptor GPR55, using genetic and pharmacological approaches, reduces pancreatic cancer cell growth in vitro and in vivo and we propose that this may represent a novel strategy to inhibit pancreatic ductal adenocarcinoma (PDAC) progression. Specifically, we show that genetic ablation of Gpr55 in the KRASWT/G12D/TP53WT/R172H/Pdx1-Cre+/+ (KPC) mouse model of PDAC significantly prolonged survival. Importantly, KPC mice treated with a combination of the GPR55 antagonist Cannabidiol (CBD) and gemcitabine (GEM, one of the most used drugs to treat PDAC), survived nearly three times longer compared to mice treated with vehicle or GEM alone. Mechanistically, knockdown or pharmacologic inhibition of GPR55 reduced anchorage-dependent and independent growth, cell cycle progression, activation of mitogen-activated protein kinase (MAPK) signalling and protein levels of ribonucleotide reductases in PDAC cells. Consistent with this, genetic ablation of Gpr55 reduced proliferation of tumour cells, MAPK signalling and ribonucleotide reductase M1 levels in KPC mice. Combination of CBD and GEM inhibited tumour cell proliferation in KPC mice and it opposed mechanisms involved in development of resistance to GEM in vitro and in vivo. Finally, we demonstrate that the tumour suppressor p53 regulates GPR55 protein expression through modulation of the microRNA miR34b-3p. Our results demonstrate the important role played by GPR55 downstream of p53 in PDAC progression. Moreover our data indicate that combination of CBD and GEM, both currently approved for medical use, might be tested in clinical trials as a novel promising treatment to improve PDAC patients' outcome.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Receptors, Cannabinoid/metabolism , Animals , Antineoplastic Agents/pharmacology , Cannabidiol/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Mice , Mice, Knockout , Pancreatic Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Gemcitabine
2.
J Thromb Haemost ; 11(2): 345-56, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23216754

ABSTRACT

BACKGROUND: The proline-rich tyrosine kinase Pyk2 is a focal adhesion kinase expressed in blood platelets, and is activated downstream of G-protein coupled receptors as well as integrin α2ß1. OBJECTIVE: In this study we have investigated the involvement of Pyk2 in integrin αIIbß3 outside-in signaling in human and murine platelets. METHODS: We analyzed the stimulation of intracellular signaling pathways in platelets from Pyk2 knockout mice adherent to immobilized fibrinogen. RESULTS: Pyk2 was rapidly phosphorylated and activated in human and murine platelets adherent to fibrinogen through integrin αIIbß3. Activation of Pyk2 was Src-dependent, but did not require phospholipase Cγ2 activity. Platelets from Pyk2 knockout mice showed a defective ability to adhere and spread on fibrinogen, in association with a dramatic reduction of phosphatidylinositol 3-kinase (PI3K) activation and Akt phosphorylation. Pharmacological and genetic analysis demonstrated that integrin αIIbß3 engagement selectively stimulated the ß-isoform of PI3K (PI3Kß), and that, as for Pyk2, PI3Kß activation required Src family kinases activity, but not phospholipase Cγ2. In fibrinogen-adherent platelets, both Pyk2 and PI3Kß were necessary for stimulation of the small GTPase Rap1b, a regulator of cell adhesion and spreading. Integrin αIIbß3 engagement triggered the association of the PI3Kß regulatory subunit p85 with the adaptor protein c-Cbl, which was mediated by the p85 SH3 domain, and was independent of c-Cbl tyrosine phosphorylation. However, p85-associated c-Cbl was tyrosine phosphorylated by activated Pyk2 in fibrinogen adherent platelets. CONCLUSIONS: These results identify a novel pathway of integrin αIIbß3 outside-in signaling and recognize the tyrosine kinase Pyk2 as a major regulator of platelet adhesion and spreading on fibrinogen.


Subject(s)
Blood Platelets/enzymology , Focal Adhesion Kinase 2/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction , Animals , Cell Shape , Enzyme Activation , Fibrinogen/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/deficiency , Focal Adhesion Kinase 2/genetics , Humans , Integrin alpha2/metabolism , Integrin beta3/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phosphorylation , Platelet Adhesiveness , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , rap GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism
3.
Curr Med Chem ; 18(18): 2763-9, 2011.
Article in English | MEDLINE | ID: mdl-21568903

ABSTRACT

Abnormal activation of phosphoinositide 3-kinase (PI3K) signalling is very common in cancer, leading to deregulation of several intracellular processes normally controlled by this enzyme, including cell survival, growth, proliferation and migration. Mutations in the gene encoding the tumour suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which leads to uncontrolled activation of the PI3K pathway, are reported in different cancers. Among the downstream effectors of PI3Ks, 3- phosphoinositide-dependent protein kinase 1 (PDK1) and protein kinase B (PKB)/Akt have a key role in several cancer types. More recent data indicate that alteration of PDK1 is a critical component of oncogenic PI3K signalling in breast cancer, suggesting that inhibition of PDK1 can inhibit breast cancer progression. PDK1 has an essential role in regulating cell migration especially in the context of PTEN deficiency. Downregulation of PDK1 levels inhibits migration and experimental metastasis of human breast cancer cells. PDK1 activates a large number of proteins, including Akt, some PKC isoforms, S6K and SGK. Data also reveal that PDK1 is oncogenic and this is dependent on PI3K pathway. Therefore, accumulating evidence demonstrates that PDK1 is a valid therapeutic target and suggests that PDK1 inhibitors may be useful to prevent cancer progression and abnormal tissue dissemination. This review will focus on published data on the role of PDK1 in cancer and approaches used to inhibit PDK1.


Subject(s)
Breast Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases , Breast Neoplasms/enzymology , Cell Movement/drug effects , Female , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects
4.
Oncogene ; 30(2): 142-52, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20838378

ABSTRACT

Recently, the orphan receptor G protein-coupled receptor 55 (GPR55) has been proposed as a potential cannabinoid receptor, although controversy remains on its physiological roles. Current evidence suggests a role for GPR55 as a receptor for the lysophospholipid lysophosphatidylinositol (LPI). In this study, we show that GPR55 is expressed in several prostate and ovarian cancer cell lines, both at the mRNA and at the protein level, and that it has a critical role in regulating proliferation and anchorage-independent growth. We further show that GPR55 mediates the effects of LPI in prostate and ovarian cancer cells. Indeed we demonstrate that LPI is able to induce calcium mobilization and activation of Akt and extracellular signal-regulated kinase (ERK)1/2 in these cells and that both pharmacological blockade of GPR55 and its downregulation using specific small interfering RNA strongly inhibits these processes. We further identify an autocrine loop by which LPI is synthesized by cytosolic phospholipase A2, pumped out of the cell by the ATP-binding cassette transporter ABCC1/MRP1, and is then able to initialize cascades downstream of GPR55. All together, these data demonstrate a role of LPI and its receptor GPR55 in cancer cells in activating an autocrine loop that regulates cell proliferation. These findings may have important implications for LPI as a novel cancer biomarker and for its receptor GPR55 as a potential therapeutic target.


Subject(s)
Autocrine Communication , Biomarkers, Tumor/metabolism , Cell Proliferation , Lysophospholipids/metabolism , Ovarian Neoplasms/pathology , Prostatic Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/analysis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Male , Multidrug Resistance-Associated Proteins/analysis , Multidrug Resistance-Associated Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Phospholipases A2/analysis , Phospholipases A2/metabolism , Piperidines/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , RNA, Small Interfering/genetics , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics , Rimonabant
5.
Br J Cancer ; 102(1): 104-14, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20051961

ABSTRACT

BACKGROUND: Owing to its role in cancer, the phosphoinositide 3-kinase (PI3K)/Akt pathway is an attractive target for therapeutic intervention. We previously reported that the inhibition of Akt by inositol 1,3,4,5,6-pentakisphosphate (InsP(5)) results in anti-tumour properties. To further develop this compound we modified its structure to obtain more potent inhibitors of the PI3K/Akt pathway. METHODS: Cell proliferation/survival was determined by cell counting, sulphorhodamine or acridine orange/ethidium bromide assay; Akt activation was determined by western blot analysis. In vivo effect of compounds was tested on PC3 xenografts, whereas in vitro activity on kinases was determined by SelectScreen Kinase Profiling Service. RESULTS: The derivative 2-O-benzyl-myo-inositol 1,3,4,5,6-pentakisphosphate (2-O-Bn-InsP(5)) is active towards cancer types resistant to InsP(5) in vitro and in vivo. 2-O-Bn-InsP(5) possesses higher pro-apoptotic activity than InsP(5) in sensitive cells and enhances the effect of anti-cancer compounds. 2-O-Bn-InsP(5) specifically inhibits 3-phosphoinositide-dependent protein kinase 1 (PDK1) in vitro (IC(50) in the low nanomolar range) and the PDK1-dependent phosphorylation of Akt in cell lines and excised tumours. It is interesting to note that 2-O-Bn-InsP(5) also inhibits the mammalian target of rapamycin (mTOR) in vitro. CONCLUSIONS: InsP(5) and 2-O-Bn-InsP(5) may represent lead compounds to develop novel inhibitors of the PI3K/Akt pathway (including potential dual PDK1/mTOR inhibitors) and novel potential anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Inositol Phosphates/chemistry , Inositol Phosphates/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Cell Line, Tumor/transplantation , Drug Delivery Systems , Drug Design , Enzyme Activation/drug effects , Female , Humans , Inositol Phosphates/chemical synthesis , Inositol Phosphates/therapeutic use , Intracellular Signaling Peptides and Proteins/drug effects , Male , Mice , Mice, Nude , Molecular Structure , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/drug effects , Structure-Activity Relationship , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
6.
Biochem Soc Trans ; 35(Pt 2): 211-4, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371240

ABSTRACT

Although it is now well established that PI3K (phosphoinositide 3-kinase) is a key enzyme in several intracellular processes, there are still relatively few reports that precisely identify the specific isoforms of PI3K actually involved in such events. The lack of specific inhibitors has made it particularly difficult to address the physiological roles of some isoforms, such as the members of class II. As a consequence, there is still relatively little understanding of the role of these enzymes and the question about the intracellular role of these isoforms still waits for more answers.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Androstadienes/pharmacology , Animals , Cell Physiological Phenomena , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/metabolism , Microsomes/enzymology , Phosphatidylinositol 3-Kinases/classification , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Wortmannin
7.
Biochem Soc Trans ; 35(Pt 2): 229-30, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371245

ABSTRACT

Activation of the enzyme PLC (phospholipase C) leads to the formation of second messengers Ins(1,4,5)P(3) and diacylglycerol. RTKs (receptor tyrosine kinases) activate this reaction through PLCgamma isoenzymes. It has been shown that PI3K (phosphoinositide 3-kinase) may regulate PLCgamma activity through the interaction of PI3K product PtdIns(3,4,5)P(3) and the PLCgamma PH domain (pleckstrin homology domain). Here, we analyse the potential functional roles of the PI3K/PLC pathway.


Subject(s)
Phosphatidylinositol 3-Kinases/physiology , Phospholipase C gamma/metabolism , Animals , Blood Vessels/physiology , Enzyme Activation , Kinetics , Membrane Lipids/metabolism , Phospholipids/metabolism , Second Messenger Systems , Veins
8.
Proc Natl Acad Sci U S A ; 100(19): 10740-5, 2003 Sep 16.
Article in English | MEDLINE | ID: mdl-12960403

ABSTRACT

The biological role of the protein tyrosine kinase, Pyk2, was explored by targeting the Pyk2 gene by homologous recombination. Pyk2-/- mice are viable and fertile, without overt impairment in development or behavior. However, the morphology and behavior of Pyk2-/- macrophages were impaired. Macrophages isolated from mutant mice failed to become polarized, to undergo membrane ruffling, and to migrate in response to chemokine stimulation. Moreover, the contractile activity in the lamellipodia of Pyk2-/- macrophages was impaired, as revealed by measuring the rearward movement toward the nucleus of fibronectin-coated beads on the lamellipodia in opposition to an immobilizing force generated by optical tweezers. Consistently, the infiltration of macrophages into a carageenan-induced inflammatory region was strongly inhibited in Pyk2-/- mice. In addition, chemokine stimulation of inositol (1, 4, 5) triphosphate production and Ca2+ release, as well as integrin-induced activation of Rho and phosphatidyl inositol 3 kinase, were compromised in Pyk2-/- macrophages. These experiments reveal a role for Pyk2 in cell signaling in macrophages essential for cell migration and function.


Subject(s)
Cell Movement/physiology , Macrophages, Peritoneal/cytology , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Actins/metabolism , Animals , Focal Adhesion Kinase 2 , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism
9.
FEBS Lett ; 506(3): 173-9, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11602240

ABSTRACT

Pleckstrin homology (PH) domains are protein modules found in proteins involved in many cellular processes. The majority of PH domain-containing proteins require membrane association for their function. It has been shown that most PH domains interact directly with the cell membrane by binding to phosphoinositides with a broad range of specificity and affinity. While a highly specific binding of the PH domain to a phosphoinositide can be necessary and sufficient for the correct recruitment of the host protein to the membrane, a weaker and less specific interaction may be necessary but not sufficient, thus probably requiring alternative, co-operative mechanisms.


Subject(s)
Blood Proteins/chemistry , Phosphatidylinositols/metabolism , Phosphoproteins/chemistry , Proteins/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Models, Molecular , Phospholipase C delta , Protein Binding , Type C Phospholipases/chemistry , Type C Phospholipases/metabolism
10.
J Biol Chem ; 276(22): 19524-31, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11278560

ABSTRACT

The Dbl family guanine nucleotide exchange factors (GEFs) contain a region of sequence similarity consisting of a catalytic Dbl homology (DH) domain in tandem with a pleckstrin homology (PH) domain. PH domains are involved in the regulated targeting of signaling molecules to plasma membranes by protein-protein and/or protein-lipid interactions. Here we show that Dbl PH domain binding to phosphatidylinositol 4,5-bisphosphate and phosphatidylinositol 3,4,5-triphosphate results in the inhibition of Dbl GEF activity on Rho family GTPase Cdc42. Phosphatidylinositol 4,5-bisphosphate binding to the PH domain significantly inhibits the Cdc42 interactive activity of the DH domain suggesting that the DH domain is subjected to the PH domain modulation under the influence of phosphoinositides (PIPs). We generated Dbl mutants unable to interact with PIPs. These mutants retained GEF activity on Cdc42 in the presence of PIPs and showed a markedly enhanced activating potential for both Cdc42 and RhoA in vivo while displaying decreased cellular transforming activity. Immunofluorescence analysis of NIH3T3 transfectants revealed that whereas the PH domain localizes to actin stress fibers and plasma membrane, the PH mutants are no longer detectable on the plasma membrane. These results suggest that modulation of PIPs in both the GEF catalytic activity and the targeting to plasma membrane determines the outcome of the biologic activity of Dbl.


Subject(s)
Blood Proteins/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/chemistry , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , COS Cells , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Mice , Microscopy, Fluorescence , Microscopy, Immunoelectron , Mutation , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Signal Transduction , Time Factors , Transfection , cdc42 GTP-Binding Protein/metabolism
11.
FASEB J ; 14(15): 2618-22, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11099481

ABSTRACT

Signaling pathways involving the inositol polyphosphates and the polyphosphoinositides have become intricately linked with a number of disease states. More recently, this has principally involved the 3-phosphorylated products of phosphoinositide 3-kinase, an enzyme that itself shows oncogenic activity and has hence become of interest in the design of antitumorigenic drugs. The downstream effectors of phosphoinositide 3-kinase are involved in different aspects of cellular signaling and cytoskeleton and trafficking events that are linked to specific polyphosphoinositide binding properties of specific protein domains, which themselves have emerging roles in specific disease states. Our recent findings have demonstrated that there is a selectivity of the intracellular effects of extracellularly applied inositol polyphosphates in their abilities to inhibit a range of growth-related in vivo assay conditions, and that these can themselves be linked to the inhibition of the membrane localization of a green fluorescent protein (GFP) -tagged PH domain. We propose that GFP fusions of the polyphosphoinositides binding domains of specific proteins of interest can be used in high-throughput investigations of the therapeutic value of specific inositol polyphosphates analogs. Inhibition of in vivo membrane targeting of these domains from proteins involved in cell growth and tumorigenesis can thus be used in the search for new anticancer drugs.


Subject(s)
Antineoplastic Agents , Drug Design , Inositol Phosphates/metabolism , Phosphatidylinositol Phosphates/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phytic Acid/metabolism , Signal Transduction
12.
FASEB J ; 14(9): 1179-87, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10834940

ABSTRACT

New efforts in cancer therapy are being focused at various levels of signaling pathways. With phosphoinositide 3-kinase (PI3-K) potentially being necessary for a range of cancer-related functions, we have investigated the influence of selected inositol tris- to hexakisphosphates on cell growth and tumorigenicity. We show that micromolar concentrations of inositol 1,3,4,5,6-pentakisphosphate and inositol 1,4,5,6-tetrakisphosphate [Ins(1,4,5,6)P(4)] inhibit IGF-1-induced [(3)H]-thymidine incorporation in human breast cancer (MCF-7) cells and the ability to grow in liquid medium and form colonies in agarose semisolid medium by small cell lung cancer (SCLC) cells, a human cancer cell line containing a constitutively active PI3-K. In an ovarian cancer cell line that also contains a constitutively active PI3-K (SKOV-3 cells), Ins(1,4,5,6)P(4) again inhibited liquid medium growth. Furthermore, when applied extracellularly, inositol 1,3,4,5-tetrakisphosphate was shown indeed to enter SCLC cells. These effects appeared specifically related to PH domains known to bind to phosphatidylinositol 3,4-bisphosphate [PtdIns(3,4)P(2)] and phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P(3)], indicating involvement of the PI3-K downstream target protein kinase B (PKB/Akt). This was further supported by inhibition of PKB/Akt PH domain membrane targeting in COS-7 cells by Ins(1,4,5,6)P(4). Thus, we propose that specific inositol polyphosphates inhibit PI3-K by competing with PtdIns(3,4, 5)P(3)-binding PH domains and that this occurs mainly at the level of the downstream PI3-K target, PKB/Akt.


Subject(s)
Enzyme Inhibitors/pharmacology , Inositol Phosphates/pharmacology , Neoplasms/enzymology , Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins , Animals , Binding Sites , COS Cells , Cell Division/drug effects , Cell Membrane/enzymology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA, Neoplasm/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Female , Humans , Inositol Phosphates/antagonists & inhibitors , Inositol Phosphates/metabolism , Inositol Phosphates/therapeutic use , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt , Tumor Cells, Cultured
13.
Mol Endocrinol ; 14(6): 823-36, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10847585

ABSTRACT

Insulin evokes diverse biological effects through receptor-mediated tyrosine phosphorylation of the insulin receptor substrate (IRS) proteins. Here, we show that, in vitro, the IRS-1, -2 and -3 pleckstrin homology (PH) domains bind with different specificities to the 3-phosphorylated phosphoinositides. In fact, the IRS-1 PH domain binds preferentially to phosphatidylinositol 3,4,5-trisphosphate (PtdIns-3,4,5-P3), the IRS-2 PH domain to phosphatidylinositol 3,4-bisphosphate (PtdIns-3,4-P2), and the IRS-3 PH domain to phosphatidylinositol 3-phosphate. When expressed in NIH-IR fibroblasts and L6 myocytes, the IRS-1 and -2 PH domains tagged with green fluorescent protein (GFP) are localized exclusively in the cytoplasm. Stimulation with insulin causes a translocation of the GFP-IRS-1 and -2 PH domains to the plasma membrane within 3-5 min. This translocation is blocked by the phosphatidylinositol 3-kinase (PI 3-K) inhibitors, wortmannin and LY294002, suggesting that this event is PI 3-K dependent. Interestingly, platelet-derived growth factor (PDGF) did not induce translocation of the IRS-1 and -2 PH domains to the plasma membrane, indicating the existence of specificity for insulin. In contrast, the GFP-IRS-3 PH domain is constitutively localized to the plasma membrane. These results reveal a differential regulation of the IRS PH domains and a novel positive feedback loop in which PI 3-K functions as both an upstream regulator and a downstream effector of IRS-1 and -2 signaling.


Subject(s)
Blood Proteins/chemistry , Phosphatidylinositols/metabolism , Phosphoproteins/analysis , Phosphoproteins/chemistry , Subcellular Fractions/chemistry , Animals , Cell Membrane/chemistry , Cytoplasm/chemistry , Fibroblasts/ultrastructure , Green Fluorescent Proteins , Humans , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Luminescent Proteins , Mice , Muscles/ultrastructure , Mutagenesis, Site-Directed , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/metabolism , Polymerase Chain Reaction , Sequence Homology
14.
J Biol Chem ; 275(20): 14873-81, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10809731

ABSTRACT

Current studies involve an investigation of the role of the pleckstrin homology (PH) domain in membrane targeting and activation of phospholipase Cbeta(1) (PLCbeta(1)). Here we report studies on the membrane localization of the isolated PH domain from the amino terminus of PLCbeta(1) (PLCbeta(1)-PH) using fluorescence microscopy of a green fluorescent protein fusion protein. Whereas PLCbeta(1)-PH does not localize to the plasma membrane in serum-starved cells, it undergoes a rapid but transient migration to the plasma membrane upon stimulation of cells with serum or lysophosphatidic acid (LPA). Regulation of the plasma membrane localization of PLCbeta(1)-PH by phosphoinositides was also investigated. PLCbeta(1)-PH was found to bind phosphatidylinositol 3-phosphate most strongly, whereas other phosphoinositides were bound with lower affinity. The plasma membrane localization of PLCbeta(1)-PH induced by serum and LPA was blocked by wortmannin pretreatment and by LY294002. In parallel, activation of PLCbeta by LPA was inhibited by wortmannin, by LY294002, or by the overexpression of PLCbeta(1)-PH. Microinjection of betagamma subunits of G proteins in serum-starved cells induced the translocation of PLCbeta(1)-PH to the plasma membrane. These results demonstrate that a cooperative mechanism involving phosphatidylinositol 3-phosphate and the Gbetagamma subunit regulates the plasma membrane localization and activation of PLCbeta(1)-PH.


Subject(s)
Cell Membrane/enzymology , Isoenzymes/chemistry , Isoenzymes/metabolism , Phosphatidylinositols/metabolism , Type C Phospholipases/chemistry , Type C Phospholipases/metabolism , 3T3 Cells , Androstadienes/pharmacology , Animals , COS Cells , Chromones/pharmacology , Culture Media, Serum-Free , Enzyme Activation , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/metabolism , Glutathione Transferase/analysis , Green Fluorescent Proteins , Growth Substances/pharmacology , HeLa Cells , Humans , Luminescent Proteins/analysis , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Morpholines/pharmacology , Phospholipase C beta , Polymerase Chain Reaction , Rats , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Transfection , Wortmannin , src Homology Domains
15.
Mol Cell Biol ; 20(4): 1448-59, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10648629

ABSTRACT

The Gab1 protein is tyrosine phosphorylated in response to various growth factors and serves as a docking protein that recruits a number of downstream signaling proteins, including phosphatidylinositol 3-kinase (PI-3 kinase). To determine the role of Gab1 in signaling via the epidermal growth factor (EGF) receptor (EGFR) we tested the ability of Gab1 to associate with and modulate signaling by this receptor. We show that Gab1 associates with the EGFR in vivo and in vitro via pTyr sites 1068 and 1086 in the carboxy-terminal tail of the receptor and that overexpression of Gab1 potentiates EGF-induced activation of the mitogen-activated protein kinase and Jun kinase signaling pathways. A mutant of Gab1 unable to bind the p85 subunit of PI-3 kinase is defective in potentiating EGFR signaling, confirming a role for PI-3 kinase as a downstream effector of Gab1. Inhibition of PI-3 kinase by a dominant-interfering mutant of p85 or by Wortmannin treatment similarly impairs Gab1-induced enhancement of signaling via the EGFR. The PH domain of Gab1 was shown to bind specifically to phosphatidylinositol 3,4,5-triphosphate [PtdIns(3,4,5)P3], a product of PI-3 kinase, and is required for activation of Gab1-mediated enhancement of EGFR signaling. Moreover, the PH domain mediates Gab1 translocation to the plasma membrane in response to EGF and is required for efficient tyrosine phosphorylation of Gab1 upon EGF stimulation. In addition, overexpression of Gab1 PH domain blocks Gab1 potentiation of EGFR signaling. Finally, expression of the gene for the lipid phosphatase PTEN, which dephosphorylates PtdIns(3,4, 5)P3, inhibits EGF signaling and translocation of Gab1 to the plasma membrane. These results reveal a novel positive feedback loop, modulated by PTEN, in which PI-3 kinase functions as both an upstream regulator and a downstream effector of Gab1 in signaling via the EGFR.


Subject(s)
ErbB Receptors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Tumor Suppressor Proteins , Adaptor Proteins, Signal Transducing , Animals , Biological Transport, Active , COS Cells , Cell Line , Cell Membrane/metabolism , Enzyme Activation , ErbB Receptors/chemistry , ErbB Receptors/genetics , Feedback , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , PTEN Phosphohydrolase , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Tyrosine/metabolism
16.
J Biol Chem ; 273(46): 30497-508, 1998 Nov 13.
Article in English | MEDLINE | ID: mdl-9804818

ABSTRACT

Pleckstrin homology (PH) domains are small protein modules involved in recruitment of signaling molecules to cellular membranes, in some cases by binding specific phosphoinositides. We describe use of a convenient "dot-blot" approach to screen 10 different PH domains for those that recognize particular phosphoinositides. Each PH domain bound phosphoinositides in the assay, but only two (from phospholipase C-delta1 and Grp1) showed clear specificity for a single species. Using soluble inositol phosphates, we show that the Grp1 PH domain (originally cloned on the basis of its phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) binding) binds specifically to D-myo-inositol 1,3,4,5-tetrakisphosphate (Ins(1,3,4,5)P4) (the PtdIns(3,4,5)P3 headgroup) with KD = 27.3 nM, but binds D-myo-inositol 1,3,4-trisphosphate (Ins(1,3,4)P3) or D-myo-inositol 1,4,5-trisphosphate (Ins(1,4,5)P3) over 80-fold more weakly. We show that this specificity allows localization of the Grp1 PH domain to the plasma membrane of mammalian cells only when phosphatidylinositol 3-kinase (PI 3-K) is activated. The presence of three adjacent equatorial phosphate groups was critical for inositol phosphate binding by the Grp1 PH domain. By contrast, another PH domain capable of PI 3-K-dependent membrane recruitment (encoded by EST684797) does not distinguish Ins(1,3,4)P3 from Ins(1,3,4,5)P3 (binding both with very high affinity), despite selecting strongly against Ins(1,4,5)P3. The remaining PH domains tested appear significantly less specific for particular phosphoinositides. Together with data presented in the literature, our results suggest that many PH domains bind similarly to multiple phosphoinositides (and in some cases phosphatidylserine), and are likely to be regulated in vivo by the most abundant species to which they bind. Thus, using the same simple approach to study several PH domains simultaneously, our studies suggest that highly specific phosphoinositide binding is a characteristic of relatively few cases.


Subject(s)
Phosphatidylinositols/metabolism , Sequence Homology, Amino Acid , Animals , Binding Sites , Calorimetry , Humans , Inositol Phosphates/metabolism , Isoenzymes/metabolism , Kinetics , Ligands , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C delta , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Type C Phospholipases/metabolism
17.
Cell Growth Differ ; 9(7): 513-22, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9690619

ABSTRACT

Many growth factors rapidly induce transcription of the c-fos proto-oncogene. We have investigated the pathways for induction of the c-fos promoter by serum and epidermal growth factor (EGF) in HeLa cells. Induction of the serum response element (SRE) of the c-fos promoter could be split into two parts, one involving the serum response factor-associated ternary complex factor (TCF) factors and the second mediated by core SRE sequences. Serum induction was mediated primarily by the core SRE, whereas EGF used both the TCF and core SRE pathways. Using activated and inhibitory signaling proteins, we found that phosphatidyl inositol 3-kinase (PI3K) and rho family members could mediate activation by serum. Activation by PI3K was mediated by core SRE sequences and was dependent upon rac and rho, suggesting a PI3K-to-rac-to-rho pathway for core SRE activation. The PI3K target Akt was also capable of activating the SRE but functioned through the TCF pathway, suggesting that Akt does not mediate the primary PI3K pathway to the SRE and that Akt is capable of activating TCF family members. Serum and EGF induction of the core SRE was partially inhibited by rho and PI3K inhibitors. The use of these inhibitors demonstrates the complexity of signaling pathways to the SRE and suggests that serum activates rho by PI3K-dependent and -independent pathways.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/genetics , 3T3 Cells , Animals , Enzyme Activation , Epidermal Growth Factor/metabolism , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Mice , Proto-Oncogene Mas , Signal Transduction , rac GTP-Binding Proteins , rho GTP-Binding Proteins
18.
Oncogene ; 16(18): 2357-65, 1998 May 07.
Article in English | MEDLINE | ID: mdl-9620553

ABSTRACT

Lysophosphatidylinositol (LysoPtdIns) is formed by a constitutively-active phosphoinositide-specific phospholipase A2 in Ras-transformed cells and can stimulate cell proliferation. To evaluate whether LysoPtdIns could function as an autocrine modulator of cell growth, we examined whether LysoPtdIns can be released in the medium of Ras-transformed FRT-Fibro fibroblasts and thyroid cells. Here, we report that LysoPtdIns accumulates in the extracellular space of these lines and reaches levels up to tenfold higher than in the case of normal cells. Moreover, the ionophore A23187 increased the levels of the lysolipid in the extracellular medium. Extracellular LysoPtdIns was rapidly hydrolyzed to inositol 1:2-cyclic phosphate. LysoPtdIns induced thymidine incorporation in FRT-Fibro Ha-Ras fibroblasts, whereas inositol cyclic 1:2-cyclic phosphate did not affect cell growth per se, nor did it interfere with the LysoPtdIns mitogenic activity. We hypothesize that in Ras-transformed fibroblasts the formation and release of LysoPtdIns may function as an autocrine mechanism that participates in the Ras-dependent stimulation of cell growth.


Subject(s)
Autocrine Communication , Cell Transformation, Neoplastic/genetics , Genes, ras , Lysophospholipids/metabolism , Mitogens/metabolism , Animals , Cell Division , Fibroblasts/cytology , Fibroblasts/metabolism , Rats , Thyroid Gland/cytology , Thyroid Gland/metabolism
19.
EMBO J ; 17(2): 414-22, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9430633

ABSTRACT

Signaling via growth factor receptors frequently results in the concomitant activation of phospholipase C gamma (PLC gamma) and phosphatidylinositol (PI) 3-kinase. While it is well established that tyrosine phosphorylation of PLC gamma is necessary for its activation, we show here that PLC gamma is regulated additionally by the lipid products of PI 3-kinase. We demonstrate that the pleckstrin homology (PH) domain of PLC gamma binds to phosphatidylinositol 3,4,5-trisphosphate [PdtIns(3,4,5)P3], and is targeted to the membrane in response to growth factor stimulation, while a mutated version of this PH domain that does not bind PdtIns(3,4,5)P3 is not membrane targeted. Consistent with these observations, activation of PI 3-kinase causes PLC gamma PH domain-mediated membrane targeting and PLC gamma activation. By contrast, either the inhibition of PI 3-kinase by overexpression of a dominant-negative mutant or the prevention of PLC gamma membrane targeting by overexpression of the PLC gamma PH domain prevents growth factor-induced PLC gamma activation. These experiments reveal a novel mechanism for cross-talk and mutual regulation of activity between two enzymes that participate in the control of phosphoinositide metabolism.


Subject(s)
Blood Proteins/physiology , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins , Type C Phospholipases/metabolism , Amino Acid Sequence , Androstadienes/pharmacology , Animals , Blood Platelets , Blood Proteins/genetics , Blood Proteins/metabolism , COS Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/metabolism , Enzyme Activation/drug effects , HeLa Cells , Humans , Isoenzymes/genetics , Molecular Sequence Data , Mutagenesis , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phospholipase C gamma , Protein Binding/genetics , Protein Structure, Tertiary , Rats , Signal Transduction , Type C Phospholipases/genetics , Wortmannin
20.
Mol Cell Biol ; 17(10): 5784-90, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9315636

ABSTRACT

The signaling events which mediate activation of c-Jun N-terminal kinase (JNK) are not yet well characterized. To broaden our understanding of upstream mediators which link extracellular signals to the JNK pathway, we investigated the role of phosphatidylinositol (PI) 3-kinase in epidermal growth factor (EGF)-mediated JNK activation. In this report we demonstrate that a dominant negative form of PI 3-kinase as well as the inhibitor wortmannin blocks EGF-induced JNK activation dramatically. However, wortmannin does not have an effect on JNK activation induced by UV irradiation or osmotic shock. In addition, a membrane-targeted, constitutively active PI 3-kinase (p110beta) was shown to produce in vivo products and to activate JNK, while a kinase-mutated form of this protein showed no activation. On the basis of these experiments, we propose that PI 3-kinase activity plays a role in EGF-induced JNK activation in these cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epidermal Growth Factor/pharmacology , Mitogen-Activated Protein Kinases , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/physiology , Androstadienes/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Membrane/enzymology , Enzyme Activation , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 3 , Mutation , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Fusion Proteins , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL
...