Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(8): 5692-9, 2001 Feb 23.
Article in English | MEDLINE | ID: mdl-11069896

ABSTRACT

The previously cloned rat nerve growth factor-regulated G protein-coupled receptor NRG-1 (Glickman, M., Malek, R. L., Kwitek-Black, A. E., Jacob, H. J., and Lee N. H. (1999) Mol. Cell. Neurosci. 14, 141-52), also known as EDG-8, binds sphingosine-1-phosphate (S1P) with high affinity and specificity. In this paper we examined the signal transduction pathways regulated by the binding of S1P to EDG-8. In Chinese hamster ovary cells heterologously expressing EDG-8, S1P inhibited forskolin-induced cAMP accumulation and activated c-Jun NH2-terminal kinase. Surprisingly, S1P inhibited serum-induced activation of extracellular regulated protein kinase 1 and 2 (ERK1/2). Treatment with pertussis toxin, which ADP-ribosylates and inactivates G(i), blocked S1P-mediated inhibition of cAMP accumulation, but had no effect on c-Jun NH2-terminal kinase activation or inhibition of ERK1/2. The inhibitory effect of S1P on ERK1/2 activity was abolished by treatment with orthovanadate, suggesting the involvement of a tyrosine phosphatase. A subunit selective [35S] guanosine 5'-3-O-(thio)triphosphate binding assay demonstrates that EDG-8 activated G(i/o) and G12 but not Gs and G(q/11) in response to S1P. In agreement, EDG-8 did not stimulate phosphoinositide turnover or cAMP accumulation. The ability of S1P to induce mitogenesis in cells expressing the EDG-1 subfamily of G protein-coupled receptors is well characterized. In contrast, S1P inhibited proliferation in Chinese hamster ovary cells expressing EDG-8 but not empty vector. The antiproliferative effect, like S1P-mediated ERK1/2 inhibition, was orthovanadate-sensitive and pertussis toxin-insensitive. Our results indicate that EDG-8, a member of the EDG-1 subfamily, couples to unique signaling pathways.


Subject(s)
GTP-Binding Proteins/metabolism , Immediate-Early Proteins/classification , Lysophospholipids , Multigene Family , Neuregulin-1/classification , Receptors, Cell Surface/classification , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Cell Division/physiology , Immediate-Early Proteins/genetics , Neuregulin-1/genetics , Pertussis Toxin , Protein Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Lysophospholipid , Recombinant Proteins/metabolism , Signal Transduction , Vanadates/pharmacology , Virulence Factors, Bordetella/pharmacology
3.
J Biol Chem ; 275(50): 39379-84, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10982820

ABSTRACT

Originating from its DNA sequence, a computational model of the Edg1 receptor has been developed that predicts critical interactions with its ligand, sphingosine 1-phosphate. The basic amino acids Arg(120) and Arg(292) ion pair with the phosphate, whereas the acidic Glu(121) residue ion pairs with the ammonium moiety of sphingosine 1-phosphate. The requirement of these interactions for specific ligand recognition has been confirmed through examination of site-directed mutants by radioligand binding, ligand-induced [(35)S]GTPgammaS binding, and receptor internalization assays. These ion-pairing interactions explain the ligand specificity of the Edg1 receptor and provide insight into ligand specificity differences within the Edg receptor family. This computational map of the ligand binding pocket provides information necessary for understanding the molecular pharmacology of this receptor, thus underlining the potential of the computational method in predicting ligand-receptor interactions.


Subject(s)
Immediate-Early Proteins/metabolism , Lysophospholipids , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Amino Acid Sequence , Animals , Arginine/chemistry , Binding Sites , Blotting, Western , Cell Line , Computer Simulation , Glutamic Acid/chemistry , Humans , Immediate-Early Proteins/genetics , Immunohistochemistry , Ions , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Structure, Secondary , Rats , Receptors, Cell Surface/metabolism , Receptors, Lysophospholipid , Sequence Homology, Amino Acid , Sphingosine/genetics , Transfection , Tumor Cells, Cultured
4.
Arch Biochem Biophys ; 375(1): 69-77, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10683250

ABSTRACT

Sphingosine-1-phosphate (SPP) acts as a first messenger in immortalized human airway epithelial cells (CFNPE9o(-)), possibly interacting with an Edg family receptor. Expression of the SPP receptors Edg-1 and Edg-3, as well as a low level of Edg-5/H218, was detected in these cells, in agreement with their ability to specifically bind SPP. The related lipids, lysophosphatidic acid and sphingosylphosphorylcholine, were unable to displace SPP from its high affinity binding sites, suggesting that the biological responses to these different lysolipids are mediated by distinct receptors. SPP markedly inhibited forskolin-stimulated cAMP accumulation in a dose-dependent manner and caused a remarkable elevation of intracellular calcium, both effects being sensitive to pertussis toxin treatment. Most importantly, SPP stimulated phosphatidic acid formation, which was maximal after 2 min and decreased within 8-10 min. In the presence of butan-1-ol, suppression of SPP-induced phosphatidic acid formation and production of phosphatidylbutanol were found, clearly indicating activation of phospholipase D (PLD). This finding was also confirmed by analysis of the fatty acid composition of phosphatidic acid, showing an increase in the monounsaturated oleic acid only. The decrease of phosphatidic acid level after 8-10 min incubation with SPP was accompanied by a parallel increase of diacylglycerol production, which was abolished in the presence of butan-1-ol. This result indicates that activation of phospholipase D is followed by stimulation of phosphatidate phosphohydrolase activity. Phosphatidic acid formation was insensitive to protein kinase C inhibitors and almost completely inhibited by pertussis toxin treatment, suggesting that SPP activates phospholipase D via a G(i/o) protein-coupled receptor.


Subject(s)
Epithelial Cells/enzymology , I-kappa B Proteins , Lysophospholipids , Phospholipase D/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Respiratory System/enzymology , Sphingosine/analogs & derivatives , 3T3 Cells , Animals , Binding, Competitive/drug effects , Calcium/metabolism , Cell Line, Transformed , Colforsin/pharmacology , Cyclic AMP/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diglycerides/biosynthesis , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , GTP-Binding Proteins/metabolism , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , NF-KappaB Inhibitor alpha , Pertussis Toxin , Phosphatidic Acids/biosynthesis , RNA, Messenger/biosynthesis , Receptors, Lysophospholipid , Respiratory System/cytology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/metabolism , Sphingosine/pharmacology , Virulence Factors, Bordetella/pharmacology
5.
Blood ; 95(8): 2624-9, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10753843

ABSTRACT

EDG-6 is a recently cloned member of the endothelial differentiation gene (EDG) G protein-coupled receptor family that is expressed in lymphoid and hematopoietic tissue and in the lung. Homology of EDG-6 to the known sphingosine-1-phosphate (SPP) receptors EDG-1, EDG-3, and EDG-5 and lysophosphatidic acid (LPA) receptors EDG-2 and EDG-4 suggested that its ligand may be a lysophospholipid or lysosphingolipid. We examined the binding of [(32)P]SPP to HEK293 cells, transiently transfected with cDNA encoding EDG-6. Binding of [(32)P]SPP was saturable, demonstrating high affinity (K(D) = 63 nmol/L). Binding was also specific for SPP, as only unlabeled SPP and sphinganine-1-phosphate, which lacks the trans double bond at the 4 position, potently displaced radiolabeled SPP. LPA did not compete for binding of SPP at any concentration tested, whereas sphingosylphosphorylcholine competed for binding to EDG-6, but only at very high concentrations. In addition, SPP activated extracellular signal-regulated kinase (Erk) in EDG-6 transfected cells in a pertussis toxin-sensitive manner. These results indicate that EDG-6 is a high affinity receptor for SPP, which couples to a G(i/o) protein, resulting in the activation of growth-related signaling pathways. (Blood. 2000;95:2624-2629)


Subject(s)
Lysophospholipids , Receptors, Cell Surface/metabolism , Sphingosine/analogs & derivatives , Animals , CHO Cells , Cricetinae , Gene Transfer Techniques , Humans , Ligands , Protein Binding , Radioligand Assay , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Sphingosine/metabolism , Sphingosine/pharmacology
6.
J Biol Chem ; 274(50): 35343-50, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10585401

ABSTRACT

Sphingosine 1-phosphate (SPP) has been shown to inhibit chemotaxis of a variety of cells, in some cases through intracellular actions, while in others through receptor-mediated effects. Surprisingly, we found that low concentrations of SPP (10-100 nM) increased chemotaxis of HEK293 cells overexpressing the G protein-coupled SPP receptor EDG-1. In agreement with previous findings in human breast cancer cells (Wang, F., Nohara, K., Olivera, O., Thompson, E. W., and Spiegel, S. (1999) Exp. Cell Res. 247, 17-28), SPP, at micromolar concentrations, inhibited chemotaxis of both vector- and EDG-1-overexpressing HEK293 cells. Nanomolar concentrations of SPP also induced a marked increase in chemotaxis of human umbilical vein endothelial cells (HUVEC) and bovine aortic endothelial cells (BAEC), which express the SPP receptors EDG-1 and EDG-3, while higher concentrations of SPP were less effective. Treatment with pertussis toxin, which ADP-ribosylates and inactivates G(i)-coupled receptors, blocked SPP-induced chemotaxis. Checkerboard analysis indicated that SPP stimulates both chemotaxis and chemokinesis. Taken together, these data suggest that SPP stimulates cell migration by binding to EDG-1. Similar to SPP, sphinganine 1-phosphate (dihydro-SPP), which also binds to this family of SPP receptors, enhanced chemotaxis; whereas, another structurally related lysophospholipid, lysophosphatidic acid, did not compete with SPP for binding nor did it have significant effects on chemotaxis of endothelial cells. Furthermore, SPP increased proliferation of HUVEC and BAEC in a pertussis toxin-sensitive manner. SPP and dihydro-SPP also stimulated tube formation of BAEC grown on collagen gels (in vitro angiogenesis), and potentiated tube formation induced by basic fibroblast growth factor. Pertussis toxin treatment blocked SPP-, but not bFGF-stimulated in vitro angiogenesis. Our results suggest that SPP may play a role in angiogenesis through binding to endothelial cell G(i)-coupled SPP receptors.


Subject(s)
Chemotaxis/physiology , DNA-Binding Proteins/physiology , Endothelium, Vascular/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , I-kappa B Proteins , Immediate-Early Proteins/physiology , Lysophospholipids , Neovascularization, Physiologic/physiology , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Animals , Aorta , Cattle , Cell Division/drug effects , Cell Line , Cells, Cultured , Chemotaxis/drug effects , DNA/biosynthesis , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Immediate-Early Proteins/genetics , Kinetics , NF-KappaB Inhibitor alpha , Neovascularization, Physiologic/drug effects , Receptors, Lysophospholipid , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/pharmacokinetics , Sphingosine/pharmacology , Transfection , Umbilical Veins
7.
Mol Biol Cell ; 10(4): 1179-90, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10198065

ABSTRACT

The endothelial-derived G-protein-coupled receptor EDG-1 is a high-affinity receptor for the bioactive lipid mediator sphingosine-1-phosphate (SPP). In the present study, we constructed the EDG-1-green fluorescent protein (GFP) chimera to examine the dynamics and subcellular localization of SPP-EDG-1 interaction. SPP binds to EDG-1-GFP and transduces intracellular signals in a manner indistinguishable from that seen with the wild-type receptor. Human embryonic kidney 293 cells stably transfected with the EDG-1-GFP cDNA expressed the receptor primarily on the plasma membrane. Exogenous SPP treatment, in a dose-dependent manner, induced receptor translocation to perinuclear vesicles with a tau1/2 of approximately 15 min. The EDG-1-GFP-containing vesicles are distinct from mitochondria but colocalize in part with endocytic vesicles and lysosomes. Neither the low-affinity agonist lysophosphatidic acid nor other sphingolipids, ceramide, ceramide-1-phosphate, or sphingosylphosphorylcholine, influenced receptor trafficking. Receptor internalization was completely inhibited by truncation of the C terminus. After SPP washout, EDG-1-GFP recycles back to the plasma membrane with a tau1/2 of approximately 30 min. We conclude that the high-affinity ligand SPP specifically induces the reversible trafficking of EDG-1 via the endosomal pathway and that the C-terminal intracellular domain of the receptor is critical for this process.


Subject(s)
Immediate-Early Proteins/physiology , Lysophospholipids , Receptors, Cell Surface/physiology , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Amino Acid Sequence , Cell Line , Cell Membrane/physiology , Cell Membrane/ultrastructure , Green Fluorescent Proteins , Humans , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/drug effects , Kidney , Kinetics , Ligands , Luminescent Proteins/genetics , Molecular Sequence Data , Protein Structure, Secondary , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/drug effects , Receptors, Lysophospholipid , Recombinant Fusion Proteins/metabolism , Sphingosine/pharmacology , Subcellular Fractions/metabolism , Transfection
8.
J Biol Chem ; 274(8): 4626-32, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-9988698

ABSTRACT

Sphingosine 1-phosphate (SPP) is a lipid second messenger that also acts as a first messenger through the G protein-coupled receptor Edg-1. Here we show that SPP also binds to the related receptors H218 and Edg-3 with high affinity and specificity. SPP and sphinganine 1-phosphate bind to these receptors, whereas neither sphingosylphosphorylcholine nor lysophosphatidic acid compete with SPP for binding to either receptor. Transfection of HEK293 cells with H218 or edg-3, but not edg-1, induces rounded cell morphology in the presence of serum, which contains high levels of SPP. SPP treatment of cells overexpressing H218 cultured in delipidated serum causes cell rounding. A similar but less dramatic effect was observed in cells overexpressing Edg-3 but not with Edg-1. Cell rounding was correlated with apoptotic cell death, probably as a result of loss of attachment. Nerve growth factor-induced neuritogenesis in PC12 cells was inhibited by overexpression of H218 and to a lesser extent Edg-3. SPP treatment rapidly enhanced neurite retraction in PC12 cells overexpressing Edg-1, Edg-3, or H218. Thus, H218, and possibly Edg-3, may be the cell surface receptors responsible for cell rounding and neurite retraction induced by SPP. Moreover, the identification of these two additional SPP receptors indicates that a family of highly specific receptors exists that mediate different responses to SPP.


Subject(s)
GTP-Binding Proteins/metabolism , Lysophospholipids , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Animals , Base Sequence , Cell Line , DNA Primers , Humans , Immediate-Early Proteins/metabolism , PC12 Cells , Protein Binding , Rats , Receptors, Lysophospholipid , Sphingosine/metabolism
9.
Cancer Res ; 59(24): 6185-91, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10626811

ABSTRACT

Exogenous sphingosine-1-phosphate (SPP) inhibits chemotactic motility of several transformed cell lines. We have found that SPP at high micromolar concentrations decreased chemotaxis of estrogen-independent (MDA-MB-231 and BT 549) and estrogen-dependent (MCF-7 and ZR-75-1) human breast cancer cells. Because SPP has been implicated as a lipid-signaling molecule with novel dual intra- and intercellular actions, it was of interest to determine whether the effect of SPP on chemotactic motility of human breast cancer cells is mediated intracellularly or through the recently identified endothelial differentiation gene (EDG) family of G protein-coupled SPP receptors. There was no detectable specific binding of [32P]SPP to MDA-MB-231 or MCF-7 cells; however, reverse transcription-PCR analysis revealed that both MDA-MB-231 and MCF-7 cells expressed moderate levels of EDG-3, neither expressed EDG-1, and EDG-5 mRNA was expressed in MCF-7 but not in MDA-MB-231 cells. In contrast to SPP, sphinganine-1-phosphate, which binds to and signals through SPP receptors EDG-1, EDG-3, and EDG-5, had no effect on chemotactic motility of MDA-MB-231 or MCF-7 cells. To further discriminate between intracellular and receptor-mediated actions of SPP, we used caged SPP, a photolyzable derivative of SPP that elevates intracellular levels of SPP after illumination. Caged SPP inhibited chemotactic motility of MDA-MB-231 cells only upon UV irradiation. In addition, in MCF-7 cells, overexpression of sphingosine kinase, the enzyme that produces SPP, inhibited chemotactic motility compared with vector-transfected cells and markedly increased cellular SPP levels in the absence of detectable secretion. Our results suggest that the inhibitory effect of SPP on chemotactic motility of human breast cancer cells is likely mediated through intracellular actions of SPP rather than through cell surface receptors.


Subject(s)
Chemotaxis/drug effects , Lysophospholipids , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Breast Neoplasms , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Photolysis , Receptors, Lysophospholipid , Sphingosine/pharmacology , Tumor Cells, Cultured
10.
Biochemistry ; 37(37): 12892-8, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9737868

ABSTRACT

Sphingosine 1-phosphate (SPP), a lipid second messenger formed by the action of sphingosine kinase, has been implicated in regulating diverse biological processes, including growth, survival, and differentiation. N,N-Dimethylsphingosine (DMS) inhibits sphingosine kinase and has been used to investigate the biological roles of SPP; however, little is known of the mechanism of inhibition of sphingosine kinase by DMS. In addition, DMS has been shown to inhibit protein kinase C in vitro. Here we report that DMS is a competitive inhibitor of sphingosine kinase from U937 monoblastic leukemia cells, Swiss 3T3 fibroblasts, and PC12 pheochromocytoma cells. DMS decreases basal levels of SPP and prevents increases in SPP in response to physiological stimuli known to activate sphingosine kinase. DMS also effectively increases cellular levels of ceramide in a variety of cell types, and resetting of the ceramide/SPP rheostat may account for the pro-apoptotic effects of DMS. Moreover, DMS, at concentrations which effectively inhibit sphingosine kinase, has no effect on protein kinase C activity or its membrane translocation. Thus, DMS acts as a specific competitive inhibitor of sphingosine kinase in diverse cell types and is a useful tool to elucidate the role of SPP as an intracellular second messenger.


Subject(s)
Ceramides/metabolism , Enzyme Inhibitors/pharmacology , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase C/metabolism , Sphingosine/analogs & derivatives , 3T3 Cells , Animals , Enzyme Activation/drug effects , Humans , Mice , PC12 Cells , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Rats , Sphingosine/metabolism , Sphingosine/pharmacology , Tumor Cells, Cultured
11.
J Cell Biol ; 142(1): 229-40, 1998 Jul 13.
Article in English | MEDLINE | ID: mdl-9660876

ABSTRACT

Sphingosine-1-phosphate (SPP), a bioactive lipid, acts both intracellularly and extracellularly to cause pleiotropic biological responses. Recently, we identified SPP as a ligand for the G protein-coupled receptor Edg-1 (Lee, M.-J., J.R. Van Brocklyn, S. Thangada, C.H. Liu, A.R. Hand, R. Menzeleev, S. Spiegel, and T. Hla. 1998. Science. 279:1552-1555). Edg-1 binds SPP with remarkable specificity as only sphinganine-1-phosphate displaced radiolabeled SPP, while other sphingolipids did not. Binding of SPP to Edg-1 resulted in inhibition of forskolin-stimulated cAMP accumulation, in a pertussis toxin-sensitive manner. In contrast, two well-characterized biological responses of SPP, mitogenesis and prevention of apoptosis, were clearly unrelated to binding to Edg-1 and correlated with intracellular uptake. SPP also stimulated signal transduction pathways, including calcium mobilization, activation of phospholipase D, and tyrosine phosphorylation of p125(FAK), independently of edg-1 expression. Moreover, DNA synthesis in Swiss 3T3 fibroblasts was significantly and specifically increased by microinjection of SPP. Finally, SPP suppresses apoptosis of HL-60 and pheochromocytoma PC12 cells, which do not have specific SPP binding or expression of Edg-1 mRNA. Conversely, sphinganine-1-phosphate, which binds to and signals via Edg-1, does not have any significant cytoprotective effect. Thus, SPP is a prototype for a novel class of lipid mediators that act both extracellularly as ligands for cell surface receptors and intracellularly as second messengers.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Immediate-Early Proteins/metabolism , Lysophospholipids , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , 3T3 Cells , Animals , Cell Division , Cell Line , Cell Survival , Cyclic AMP/metabolism , HL-60 Cells , Humans , Immediate-Early Proteins/genetics , Mice , PC12 Cells , Rats , Receptors, Lysophospholipid , Signal Transduction , Sphingosine/metabolism , Sphingosine/physiology
12.
Ann N Y Acad Sci ; 845: 11-8, 1998 Jun 19.
Article in English | MEDLINE | ID: mdl-9668339

ABSTRACT

Recent evidence suggests that branching pathways of sphingolipid metabolism may mediate either apoptotic or mitogenic responses depending on the cell type and the nature of the stimulus. While ceramide has been shown to be an important regulatory component of apoptosis induced by tumor necrosis factor alpha and Fas ligand, sphingosine-1-phosphate (SPP), a further metabolite of ceramide, has been implicated as a second messenger in cellular proliferation and survival induced by platelet-derived growth factor, nerve growth factor, and serum. SPP protects cells from apoptosis resulting from elevations of ceramide. Inflammatory cytokines stimulate sphingomyelinase, but not ceramidase, leading to accumulation of ceramide, whereas growth signals also leading to accumulation of ceramide, whereas growth signals also stimulate ceramidase and sphingosine kinase leading to increased SPP levels. We propose that the dynamic balance between levels of sphingolipid metabolites, ceramide, and SPP, and consequent regulation of different family members of mitogen-activated protein kinases (JNK versus ERK), is an important factor that determines whether a cell survives or dies.


Subject(s)
Apoptosis/physiology , Cell Division/physiology , Lysophospholipids , Receptors, G-Protein-Coupled , Signal Transduction , Sphingosine/analogs & derivatives , Animals , Apoptosis/drug effects , Ceramides/pharmacology , Ceramides/physiology , Humans , Nerve Growth Factors/pharmacology , Receptors, Cell Surface/physiology , Receptors, Lysophospholipid , Sphingosine/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology
14.
Science ; 279(5356): 1552-5, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9488656

ABSTRACT

The sphingolipid metabolite sphingosine-1-phosphate (SPP) has been implicated as a second messenger in cell proliferation and survival. However, many of its biological effects are due to binding to unidentified receptors on the cell surface. SPP activated the heterotrimeric guanine nucleotide binding protein (G protein)-coupled orphan receptor EDG-1, originally cloned as Endothelial Differentiation Gene-1. EDG-1 bound SPP with high affinity (dissociation constant = 8.1 nM) and high specificity. Overexpression of EDG-1 induced exaggerated cell-cell aggregation, enhanced expression of cadherins, and formation of well-developed adherens junctions in a manner dependent on SPP and the small guanine nucleotide binding protein Rho.


Subject(s)
Cadherins/biosynthesis , Cell Aggregation , Immediate-Early Proteins/metabolism , Intercellular Junctions/ultrastructure , Lysophospholipids , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Cell Differentiation , Cell Line , Cloning, Molecular , GTP-Binding Proteins/metabolism , Gene Expression , Genes, Immediate-Early , Humans , Immediate-Early Proteins/genetics , Ligands , Mitogen-Activated Protein Kinase 1/metabolism , Morphogenesis , Receptors, Cell Surface/genetics , Receptors, Lysophospholipid , Signal Transduction , Sphingosine/metabolism , Transfection , rho GTP-Binding Proteins
15.
J Neurochem ; 69(1): 116-25, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9202301

ABSTRACT

Gangliosides are implicated in the regulation of cellular proliferation as evidenced by differences in ganglioside composition associated with malignant transformation and density of cells in culture, as well as their inhibitory effects when added to cells growing in culture. Exogenously added gangliosides have a bimodal effect on proliferation in U-1242 MG glioma cells, inhibiting DNA synthesis in growing cells and stimulating it in quiescent cells. We investigated the mechanisms involved in stimulation of DNA synthesis using [3H]thymidine incorporation and immune complex kinase assays to identify responsible signal transduction pathways. Treatment of quiescent U-1242 MG cells with GM1 caused activation of the mitogen-activated protein (MAP) kinase isoform Erk2. Pretreatment with the specific MAP kinase kinase inhibitor PD98059 prevented the GM1-stimulated Erk2 activation and GM1-stimulated DNA synthesis. GM1 treatment stimulated another distinct signaling pathway leading to activation of p70 S6 kinase (p70s6k), and this was prevented by pretreatment with rapamycin. Rapamycin also inhibited GM1-stimulated DNA synthesis. Activation of both pathways and stimulation of DNA synthesis were inhibited by forskolin treatment; however, GM1 had no effect on cyclic AMP levels. Platelet-derived growth factor also activated both Erk2 and p70s6k but did not cause DNA synthesis, suggesting that GM1 may stimulate additional cascades, which also contribute to GM1-mediated DNA synthesis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , G(M1) Ganglioside/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Colforsin/pharmacology , Cyclic AMP/pharmacology , DNA/biosynthesis , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , G(M1) Ganglioside/metabolism , Glioma , Humans , Immunosuppressive Agents/pharmacology , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase Kinases , Mitogens/metabolism , Platelet-Derived Growth Factor/pharmacology , Polyenes/pharmacology , Protein Kinase Inhibitors , Ribosomal Protein S6 Kinases , Signal Transduction/immunology , Sirolimus , Tumor Cells, Cultured
16.
J Biol Chem ; 272(26): 16211-5, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9195921

ABSTRACT

The proto-oncogene molecule c-Crk plays a role in growth factor-induced activation of Ras. Sphingosine 1-phosphate (SPP), a metabolite of cellular sphingolipids, has previously been shown to play a role in growth factor receptor signaling (Olivera, A., and Spiegel, S. (1993) Nature 365, 557-560). SPP was found to strongly induce tyrosine phosphorylation of Crk, but not Shc, in NIH-3T3 parental, insulin-like growth factor-I receptor-overexpressing and Crk-overexpressing (3T3-Crk) fibroblasts. Sphingosine, a metabolic precursor of SPP, also produced a slight increase in tyrosine phosphorylation of Crk. In contrast, other sphingolipid metabolites including ceramide did not alter Crk tyrosine phosphorylation. Furthermore, Crk enhanced SPP-induced mitogenesis, as measured by SPP-stimulated [3H]thymidine incorporation in a manner proportional to the level of Crk expression in 3T3-Crk cells. This stimulation appears to be Ras-dependent, whereas SPP stimulation of MAP kinase activity is Ras-independent. These data indicate that SPP activates a tyrosine kinase that phosphorylates Crk and that Crk is a positive effector of SPP-induced mitogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Lysophospholipids , Proto-Oncogene Proteins/metabolism , Sphingosine/analogs & derivatives , Tyrosine/metabolism , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , GRB2 Adaptor Protein , Mice , Phosphorylation , Proteins/metabolism , Proto-Oncogene Proteins c-crk , Sphingosine/pharmacology , src Homology Domains
17.
J Immunol ; 158(3): 1125-32, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9013951

ABSTRACT

To elucidate the molecular basis for inhibition of B cell proliferation and differentiation by the Fc receptor for IgG (Fc(gamma)RII), we compared the signaling events in B cells stimulated by cross-linking surface Ig alone (positive signaling), or by co-cross-linking surface Ig and Fc(gamma)RII (negative signaling). Both modes of stimulation induced tyrosine kinase activation. Positive signaling induced activation of Ras, Raf-1 kinase, and mitogen-activated protein kinase; these events were significantly attenuated during negative signaling. Since Ras is activated by SOS and Vav, two known guanine nucleotide exchange factors, activation events associated with these molecules using the two different stimuli were examined. Results of these experiments indicated that tyrosine phosphorylation of Vav did not change upon co-cross-linking. In contrast, the association of Shc and Grb2 was abrogated under negative and induced under positive signaling conditions. Concomitantly, Shc was observed to associate with a tyrosine-phosphorylated 145-kDa protein, previously identified as Src homology 2-containing inositol phosphatase, only under conditions of negative signaling. Based on these results, we hypothesize that negative signaling via the Fc(gamma)RII in B cells is at least partly the result of a block in Ras activation, and that SOS, but not Vav, is the major guanine nucleotide exchange factor in B cells for Ras activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , B-Lymphocytes/physiology , Cell Cycle Proteins , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Antigen, B-Cell/physiology , Receptors, IgG/physiology , Animals , Cells, Cultured , GRB2 Adaptor Protein , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoproteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-vav , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL
...