Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 281(42): 31399-407, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16926156

ABSTRACT

Although several studies have shown that a subset of insulin-like growth factor (IGF) signals require the activation of heterotrimeric G proteins, the molecular mechanisms underlying IGF-stimulated G protein signaling remain poorly understood. Here, we have studied the mechanism by which endogenous IGF receptors activate the ERK1/2 mitogen-activated protein kinase cascade in HEK293 cells. In these cells, treatment with pertussis toxin and expression of a Galpha(q/11)-(305-359) peptide that inhibits G(q/11) signaling additively inhibited IGF-stimulated ERK1/2 activation, indicating that the signal was almost completely G protein-dependent. Treatment with IGF-1 or IGF-2 promoted translocation of green fluorescent protein (GFP)-tagged sphingosine kinase (SK) 1 from the cytosol to the plasma membrane, increased endogenous SK activity within 30 s of stimulation, and caused a statistically significant increase in intracellular and extracellular sphingosine 1-phosphate (S1P) concentration. Using a GFP-tagged S1P1 receptor as a biological sensor for the generation of physiologically relevant S1P levels, we found that IGF-1 and IGF-2 induced GFP-S1P receptor internalization and that the effect was blocked by pretreatment with the SK inhibitor, dimethylsphingosine. Treating cells with dimethylsphingosine, silencing SK1 expression by RNA interference, and blocking endogenous S1P receptors with the competitive antagonist VPC23019 all significantly inhibited IGF-stimulated ERK1/2 activation, suggesting that IGFs elicit G protein-dependent ERK1/2 activation by stimulating SK1-dependent transactivation of S1P receptors. Given the ubiquity of SK and S1P receptor expression, S1P receptor transactivation may represent a general mechanism for G protein-dependent signaling by non-G protein-coupled receptors.


Subject(s)
Lysophospholipids/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Somatomedins/physiology , Sphingosine/analogs & derivatives , Animals , Enzyme Activation , Green Fluorescent Proteins/metabolism , Humans , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Structure, Tertiary , Signal Transduction , Somatomedins/metabolism , Sphingosine/metabolism , Transcriptional Activation
2.
Prostaglandins Other Lipid Mediat ; 79(1-2): 126-40, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16516816

ABSTRACT

The transformation of macrophages into foam cells is a critical event in the development of atherosclerosis. The most studied aspect of this process is the uptake of modified LDL through the scavenger receptors. Another salient aspect is the effect of modified LDL immune complexes on macrophages activation and foam cell formation. Macrophages internalize oxidized LDL immune complexes (oxLDL-IC) via the Fc-gamma receptor and transform into activated foam cells. In this study we examined the effect of oxLDL-IC on sphingosine kinase 1 (SK1), an enzyme implicated in mediating pro-survival and inflammatory responses through the generation of the signaling molecule sphingosine-1-phosphate (S1P). Intriguingly, oxLDL-IC, but not oxLDL alone, induced an immediate translocation and release of SK1 into the conditioned medium as evidenced by fluorescence confocal microscopy. Immunoblot analysis of cell lysates and conditioned medium revealed a decrease in intracellular SK1 protein levels accompanied by a concomitant increase in extracellular SK1 levels. Furthermore, measurement of S1P formation showed that the activity of cell-associated SK decreased in response to oxLDL-IC compared to oxLDL alone, whereas the activity of SK increased extracellularly. Blocking oxLDL-IC binding to Fc-gamma receptors resulted in decreased levels of extracellular S1P. The data also show that cell survival of human U937 cells exposed to oxLDL-IC increased compared to oxLDL alone. Exogenously added S1P further increased cell survival induced by oxLDL-IC. Taken together, these findings indicate that S1P may be generated extracellularly in response to modified LDL immune complexes and may therefore promote cell survival and prolong cytokine release by activated macrophages.


Subject(s)
Antigen-Antibody Complex/metabolism , Atherosclerosis/enzymology , Foam Cells/enzymology , Lipoproteins, LDL/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/pharmacology , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Survival/drug effects , Cell Survival/immunology , Foam Cells/immunology , Foam Cells/pathology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/pathology , Lipoproteins, LDL/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , U937 Cells
3.
FASEB J ; 20(2): 386-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16319132

ABSTRACT

Sphingosine kinase 1 (SK1) phosphorylates sphingosine to form sphingosine 1-phosphate (S1P), which has the ability to promote cell proliferation and survival and stimulate angiogenesis. The SK1/S1P pathway also plays a critical role in regulation of cyclooxygenase-2 (COX-2), a well-established pathogenic factor in colon carcinogenesis. Therefore, we examined the expression of SK1 and COX-2 in rat colon tumors induced by azoxymethane (AOM) and the relationship of these two proteins in normal and malignant intestinal epithelial cells. Strongly positive SK1 staining was found in 21/28 (75%) of rat colon adenocarcinomas induced by AOM, whereas no positive SK1 staining was observed in normal mucosa. The increase in SK1 and COX-2 expression in AOM-induced rat colon adenocarcinoma was confirmed at the level of mRNA by real-time RT-PCR. In addition, it was found that 1) down-regulation of SK1 in HT-29 human colon cancer cells by small interfering RNA (siRNA) decreases COX-2 expression and PGE2 production; 2) overexpression of SK1 in RIE-1 rat intestinal epithelial cells induces COX-2 expression; and 3) S1P stimulates COX-2 expression and PGE2 production in HT-29 cells. These results suggest that the SK1/S1P pathway may play an important role in colon carcinogenesis, in part, by regulating COX-2 expression and PGE2 production.


Subject(s)
Colonic Neoplasms/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Up-Regulation , Animals , Azoxymethane/pharmacology , Cell Line, Tumor , Colon/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred BALB C , Phosphotransferases (Alcohol Group Acceptor)/genetics , Rats , Rats, Inbred F344 , Sphingosine/analogs & derivatives , Sphingosine/metabolism
4.
J Biol Chem ; 280(52): 43030-8, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16243846

ABSTRACT

Sphingosine 1-phosphate is a bioactive sphingolipid that regulates cell growth and suppresses programmed cell death. The biosynthesis of sphingosine 1-phosphate is catalyzed by sphingosine kinase (SK) but the mechanism by which the subcellular localization and activity of SK is regulated in response to various stimuli is not fully understood. To elucidate the origin and structural determinant of the specific subcellular localization of SK, we performed biophysical and cell studies of human SK1 (hSK1) and selected mutants. In vitro measurements showed that hSK1 selectively bound phosphatidylserine over other anionic phospholipids and strongly preferred the plasma membrane-mimicking membrane to other cellular membrane mimetics. Mutational analysis indicates that conserved Thr54 and Asn89 in the putative membrane-binding surface are essential for lipid selectivity and membrane targeting both in vitro and in the cell. Also, phosphorylation of Ser225 enhances the membrane affinity and plasma membrane selectivity of hSK1, presumably by modulating the interaction of Thr54 and Asn89 with the membrane. Collectively, these studies suggest that the specific plasma membrane localization and activation of SK1 is mediated largely by specific lipid-protein interactions.


Subject(s)
Cell Membrane/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Amino Acid Sequence , Asparagine/chemistry , Cell Line , DNA Mutational Analysis , Green Fluorescent Proteins/chemistry , Humans , Kinetics , Lipids/chemistry , Lysophospholipids/chemistry , Mass Spectrometry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Molecular Sequence Data , Mutation , Peptides/chemistry , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Phosphatidylserines/chemistry , Phospholipids/chemistry , Phosphorylation , Pressure , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Serine/chemistry , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Surface Plasmon Resonance , Threonine/chemistry , Time Factors , Transfection
5.
J Histochem Cytochem ; 53(9): 1159-66, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15923363

ABSTRACT

Sphingosine kinase 1 (SK1) is a key enzyme critical to the sphingolipid metabolic pathway responsible for catalyzing the formation of the bioactive lipid sphingosine-1-phosphate. SK1-mediated production of sphingosine-1-phosphate has been shown to stimulate such biological processes as cell growth, differentiation, migration, angiogenesis, and inhibition of apoptosis. In this study, cell type-specific immunolocalization of SK1 was examined in the bronchus/terminal bronchiole of the lung. Strong immunopositive staining was evident at the apical surface of pseudostratified epithelial cells of the bronchus and underlying smooth muscle cells, submucosal serous glands, immature chondrocytes, type II alveolar cells, foamy macrophages, endothelial cells of blood vessels, and neural bundles. Immunohistochemical screening for SK1 expression was performed in 25 samples of normal/tumor patient matched non-small-cell lung cancer tissue and found that 25 of 25 tumor samples (carcinoid [5 samples], squamous [10 samples], and adenocarcinoma tumors [10 samples]), exhibited overwhelmingly positive immunostaining for SK1 as compared with patient-matched normal tissue. In addition, an approximately 2-fold elevation of SK1 mRNA expression was observed in lung cancer tissue versus normal tissue, as well as in several other solid tumors. Taken together, these findings define the localization of SK1 in lung and provide clues as to how SK1 may play a role in normal lung physiology and the pathophysiology of lung cancer.


Subject(s)
Adenocarcinoma/enzymology , Carcinoid Tumor/enzymology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Squamous Cell/enzymology , Lung Neoplasms/enzymology , Lung/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Antibody Specificity , Bronchi/enzymology , Humans , Immunohistochemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/immunology , RNA, Messenger/metabolism , Rabbits
6.
J Biol Chem ; 279(19): 20546-54, 2004 May 07.
Article in English | MEDLINE | ID: mdl-14988393

ABSTRACT

Sphingosine kinase 1 (SK1), a key enzyme in sphingosine 1-phosphate (S1P) synthesis, regulates various aspects of cell behavior, including cell survival and proliferation. DNA damaging anti-neoplastic agents have been shown to induce p53, ceramide levels, and apoptosis; however, the effects of anti-neoplastic agents on SK have not been assessed. In this study, we investigated the effects of a DNA damaging agent, actinomycin D (Act D), on the function of sphingosine kinase (SK1). Act D caused a reduction in the protein levels of SK1, as indicated by Western blot analysis, with a concomitant decrease in SK activity. The down-regulation was post-transcriptional, because the mRNA levels of SK1 remained unchanged. Similar decreases in SK1 protein were observed with other DNA damaging agents such as doxorubicin, etoposide, and gamma-irradiation. ZVAD, the pancaspase inhibitor, and Bcl-2 annulled the effect of Act D on SK1, demonstrating a role for cysteine proteases downstream of Bcl-2 in the down-regulation of SK1. Inhibition of caspases 3, 6, 7, and 9 only partially reversed Act D-induced SK1 loss. Inhibition of cathepsin B, a lysosomal protease, produced a significant reversal of SK1 decline by Act D, suggesting that a multitude of ZVAD-sensitive cysteine proteases downstream of Bcl-2 mediated the SK1 decrease. When p53 up-regulation after Act D treatment was inhibited, SK1 down-regulation was rescued, demonstrating p53 dependence of SK1 modulation. Treatment of cells with S1P, the product of SK1, partially inhibited Act D-induced cell death, raising the possibility that a decrease in SK1 may be in part necessary for cell death to occur. Furthermore, the knockdown of SK1 by small interfering RNA in MCF-7 cells resulted in a significant reduction in cell viability. These studies demonstrate that SK1 is down-regulated by genotoxic stress, and that basal SK1 function may be necessary for the maintenance of tumor cell growth.


Subject(s)
DNA Damage , Down-Regulation , Endopeptidases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Tumor Suppressor Protein p53/metabolism , Apoptosis , Blotting, Western , Cell Division , Cell Line, Tumor , Cell Survival , Coloring Agents/pharmacology , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Gamma Rays , Humans , Mass Spectrometry , Models, Biological , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Transcription, Genetic , Up-Regulation
7.
FASEB J ; 17(11): 1411-21, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12890694

ABSTRACT

In this study we addressed the role of sphingolipid metabolism in the inflammatory response. In a L929 fibroblast model, tumor necrosis factor-alpha (TNF) induced prostaglandin E2 (PGE2) production by 4 h and cyclooxygenase-2 (COX-2) induction as early as 2 h. This TNF-induced PGE2 production was inhibited by NS398, a COX-2 selective inhibitor. GC-MS analysis revealed that only COX-2-generated prostanoids were produced in response to TNF, thus providing further evidence of COX-2 selectivity. As sphingolipids have been implicated in mediating several actions of TNF, their role in COX-2 induction and PGE2 production was evaluated. Sphingosine-1-phosphate (S1P) induced both COX-2 and PGE2 in a dose-responsive manner with an apparent ED50 of 100-300 nM. The related sphingolipid sphingosine also induced PGE2, though with much less efficacy. TNF induced a 3.5-fold increase in sphingosine-1-phosphate levels at 10 min that rapidly returned to baseline by 40 min. Small interfering RNAs (siRNAs) directed against mouse SK1 decreased (typically by 80%) SK1 protein and inhibited TNF-induced SK activity. Treatment of cells with RNAi to SK1 but not SK2 almost completely abolished the ability of TNF to induce COX-2 or generate PGE2. By contrast, cells treated with RNAi to S1P lyase or S1P phosphatase enhanced COX-2 induction leading to enhanced generation of PGE2. Treatment with SK1 RNAi also abolished the effects of exogenous sphingosine and ceramide on PGE2, revealing that the action of sphingosine and ceramide are due to intracellular metabolism into S1P. Collectively, these results provide novel evidence that SK1 and S1P are necessary for TNF to induce COX-2 and PGE2 production. Based on these findings, this study indicates that SK1 and S1P could be implicated in pathological inflammatory disorders and cancer.


Subject(s)
Dinoprostone/biosynthesis , Isoenzymes/biosynthesis , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor)/physiology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , Cyclooxygenase 2 , Enzyme Induction , Humans , Membrane Proteins , Mice , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , Signal Transduction , Sphingosine/pharmacology , Tumor Cells, Cultured
8.
J Biol Chem ; 278(36): 34541-7, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12815058

ABSTRACT

Sphingosine-1-phosphate (S1P) is a highly bioactive lipid that exerts numerous biological effects both intracellularly as a second messenger and extracellularly by binding to its G-protein-coupled receptors of the endothelial differentiation gene family (S1P receptors-(1-5)). Intracellularly, at least two enzymes, sphingosine kinase and S1P phosphatase, regulate the activity of S1P by governing the phosphorylation status of S1P. To study the regulation of S1P levels, we cloned the human isoform of S1P phosphatase 1 (hSPPase1). The hSPPase1 has 78% homology to the mouse SPPase at the amino acid level with 6-8 possible transmembrane domains. Confocal microscopy revealed green fluorescent protein-tagged hSPPase1, expressed in either MCF7 or HEK293 cells, co-localized to endoplasmic reticulum with calreticulin. According to Northern blot analysis, hSPPase1 is expressed in most tissues, with the strongest levels found in the highly vascular tissues of placenta and kidney. Transient overexpression of hSPPase1 exhibited a 2-fold increase in phosphatase activity against S1P and dihydro-S1P, indicating that the expressed protein was functional. Small interfering RNA (siRNA) knockdown of endogenous hSPPase1 drastically reduced hSPPase1 mRNA levels, as confirmed by reverse transcription PCR, and resulted in an overall 25% reduction of in vitro phosphatase activity in the membrane fractions. Sphingolipid mass measurements in hSPPase1 siRNA knockdown cells revealed a 2-fold increase of S1P levels and concomitant decrease in sphingosine. In vivo labeling of hSPPase1 siRNA-treated cells showed accumulation of S1P within cells, as well as significantly increased secretion of S1P into the media, indicating that hSPPase1 regulates secreted S1P. In addition, siRNA-induced knockdown of hSPPase1 endowed resistance to tumor necrosis factor-alpha and the chemotherapeutic agent daunorubicin. Collectively, these data suggest that regulation of hSPPase1 with the resultant changes in cellular and secreted S1P could have important implications to cell proliferation, angiogenesis, and apoptosis.


Subject(s)
Lysophospholipids , Membrane Proteins , Phosphoric Monoester Hydrolases/chemistry , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , Cell Division , Cell Line , Cell Survival , Cloning, Molecular , Coloring Agents/pharmacology , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Enzymologic , Humans , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Molecular Sequence Data , Protein Isoforms , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , Sphingolipids/chemistry , Sphingolipids/metabolism , Sphingosine/chemistry , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Tissue Distribution , Tumor Cells, Cultured
9.
J Biol Chem ; 277(38): 35257-62, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12124383

ABSTRACT

Sphingosine-1-phosphate (S1P) is a highly bioactive sphingolipid involved in diverse biological processes leading to changes in cell growth, differentiation, motility, and survival. S1P generation is regulated via sphingosine kinase (SK), and many of its effects are mediated through extracelluar action on G-protein-coupled receptors. In this study, we have investigated the mechanisms regulating SK, where this occurs in the cell, and whether this leads to release of S1P extracellularly. The protein kinase C (PKC) activator, phorbol 12-myristate 13-acetate (PMA), induced early activation of SK in HEK 293 cells, and this activation was more specific to the membrane-associated SK. Therefore, we next investigated whether PMA induced translocation of SK to the plasma membrane. PMA induced translocation of both endogenous and green fluorescent protein (GFP)-tagged human SK1 (hSK1) to the plasma membrane. PMA also induced phosphorylation of GFP-hSK1. The PMA-induced translocation was abrogated by preincubation with known PKC inhibitors (bisindoylmaleimide and calphostin-c) as well as by the indirect inhibitor of PKC, C(6)-ceramide, supporting a role for PKC in mediating translocation of SK to the plasma membrane. SK activity was not necessary for translocation, because a dominant negative G82D mutation also translocated in response to PMA. Importantly, PKC regulation of SK was accompanied by a 4-fold increase in S1P in the media. These results demonstrate a novel mechanism by which PKC regulates SK and increases secretion of S1P, allowing for autocrine/paracrine signaling.


Subject(s)
Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Amino Acid Sequence , Base Sequence , Cell Line , Cell Membrane/enzymology , Cloning, Molecular , DNA Primers , Enzyme Activation , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Transport , Recombinant Fusion Proteins/metabolism
10.
Anticancer Res ; 22(6A): 3197-204, 2002.
Article in English | MEDLINE | ID: mdl-12530065

ABSTRACT

BACKGROUND: The favorable clinical activity of paclitaxel has prompted considerable interest in combining this agent with other clinically effective antineoplastic agents including the antimetabolite 5-fluorouracil (5-FU). Our previous studies indicated that simultaneous exposure or pretreatment with 5-FU could significantly interfere with the cytotoxic effects of paclitaxel on both mitotic arrest and apoptosis. Biochemical examination also revealed that 5-FU inhibited expression of p21WAF1/CIP1 that may contribute to paclitaxel cytotoxicity. MATERIALS AND METHODS: In this study, human breast cancer BCap37 cells were transfected with either sense or antisense p53 or p21WAF1/CIP1. The established stable transfectants were then analyzed for an altered sensitivity to paclitaxel, 5-FU or the combinations of these drugs using a series of cytotoxic and apoptosis assays. RESULTS: Tumor cells transfected with antisense p53 or p21WAF1/CIP1 exhibited a significant increase in their sensitivity to paclitaxel. The reduced protein levels of p53 and p21WAF1/CIP1 were also found to abrogate the inhibitory effects of 5-FU on paclitaxel-induced mitotic arrest and apoptosis. CONCLUSION: These findings suggest that the status of p53 and p21WAF1/CIP1 might play an important role in tumor cell susceptibility to paclitaxel and its combination with other drugs such as 5-FU.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclins/biosynthesis , Fluorouracil/pharmacology , Paclitaxel/pharmacology , Tumor Suppressor Protein p53/biosynthesis , Apoptosis/drug effects , Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA, Antisense/administration & dosage , DNA, Antisense/genetics , Drug Interactions , Drug Screening Assays, Antitumor , Fluorouracil/administration & dosage , Humans , Paclitaxel/administration & dosage , Paclitaxel/antagonists & inhibitors , Prohibitins , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...