Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochim Biophys Acta ; 1831(2): 251-62, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085009

ABSTRACT

Sphingoid base-1-phosphates represent a very low portion of the sphingolipid pool but are potent bioactive lipids in mammals. This study was undertaken to determine whether these lipids are produced in palmitate-treated pancreatic ß cells and what role they play in palmitate-induced ß cell apoptosis. Our lipidomic analysis revealed that palmitate at low and high glucose supplementation increased (dihydro)sphingosine-1-phosphate levels in INS-1 ß cells. This increase was associated with an increase in sphingosine kinase 1 (SphK1) mRNA and protein levels. Over-expression of SphK1 in INS-1 cells potentiated palmitate-induced accumulation of dihydrosphingosine-1-phosphate. N,N-dimethyl-sphingosine, a potent inhibitor of SphK, potentiated ß-cell apoptosis induced by palmitate whereas over-expression of SphK1 significantly reduced apoptosis induced by palmitate with high glucose. Endoplasmic reticulum (ER)-targeted SphK1 also partially inhibited apoptosis induced by palmitate. Inhibition of INS-1 apoptosis by over-expressed SphK1 was independent of sphingosine-1-phosphate receptors but was associated with a decreased formation of pro-apoptotic ceramides induced by gluco-lipotoxicity. Moreover, over-expression of SphK1 counteracted the defect in the ER-to-Golgi transport of proteins that contribute to the ceramide-dependent ER stress observed during gluco-lipotoxicity. In conclusion, our results suggest that activation of palmitate-induced SphK1-mediated sphingoid base-1-phosphate formation in the ER of ß cells plays a protective role against palmitate-induced ceramide-dependent apoptotic ß cell death.


Subject(s)
Islets of Langerhans/drug effects , Lysophospholipids/biosynthesis , Sphingosine/analogs & derivatives , Animals , Apoptosis/drug effects , Base Sequence , Cell Line, Tumor , Chromatography, Liquid , DNA Primers , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lysophospholipids/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Molecular Sequence Data , Palmitic Acid , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/biosynthesis , Sphingosine/genetics , Tandem Mass Spectrometry
2.
Biochem J ; 438(1): 177-89, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21592087

ABSTRACT

Pancreatic ß-cell apoptosis induced by palmitate requires high glucose concentrations. Ceramides have been suggested to be important mediators of glucolipotoxicity-induced ß-cell apoptosis. In INS-1 ß-cells, 0.4 mM palmitate with 5 mM glucose increased the levels of dihydrosphingosine and dihydroceramides, two lipid intermediates in the de novo biosynthesis of ceramides, without inducing apoptosis. Increasing glucose concentrations to 30 mM amplified palmitate-induced accumulation of dihydrosphingosine and the formation of (dihydro)ceramides. Of note, glucolipotoxicity specifically induced the formation of C(18:0), C(22:0) and C(24:1) (dihydro)ceramide molecular species, which was associated with the up-regulation of CerS4 (ceramide synthase 4) levels. Fumonisin-B1, a ceramide synthase inhibitor, partially blocked apoptosis induced by glucolipotoxicity. In contrast, apoptosis was potentiated in the presence of D,L-threo-1-phenyl-2-palmitoylamino-3-morpholinopropan-1-ol, an inhibitor of glucosylceramide synthase. Moreover, overexpression of CerS4 amplified ceramide production and apoptosis induced by palmitate with 30 mM glucose, whereas down-regulation of CerS4 by siRNA (short interfering RNA) reduced apoptosis. CerS4 also potentiates ceramide accumulation and apoptosis induced by another saturated fatty acid: stearate. Collectively, our results suggest that glucolipotoxicity induces ß-cell apoptosis through a dual mechanism involving de novo ceramide biosynthesis and the formation of ceramides with specific N-acyl chain lengths rather than an overall increase in ceramide content.


Subject(s)
Apoptosis/drug effects , Ceramides/metabolism , Fumonisins/toxicity , Glucose/toxicity , Insulin-Secreting Cells/metabolism , Oxidoreductases/metabolism , Palmitates/toxicity , Animals , Blotting, Western , Cell Proliferation/drug effects , Enzyme Inhibitors/toxicity , Humans , Insulin-Secreting Cells/drug effects , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sweetening Agents/toxicity
3.
Adv Enzyme Regul ; 51(1): 229-44, 2011.
Article in English | MEDLINE | ID: mdl-21075134

ABSTRACT

The sphingosine kinases (sphingosine kinase-1 and -2) have been implicated in a variety of physiological functions. Discerning their mechanism of action is complicated because in addition to producing the potent lipid second messenger sphingosine-1-phosphate, sphingosine kinases, both by producing sphingosine-1-phosphate and consuming sphingosine, have profound effects on sphingolipid metabolism. Sphingosine kinase-1 translocates to the plasma membrane upon agonist stimulation and this translocation is essential for the pro-oncogenic properties of this enzyme. Many of the enzymes of sphingolipid metabolism, including the enzymes that degrade sphingosine-1-phosphate, are membrane bound with restricted subcellular distributions. In the work described here we explore how subcellular localization of sphingosine kinase-1 affects the downstream metabolism of sphingosine-1-phosphate and the access of sphingosine kinase to its substrates. We find, surprisingly, that restricting sphingosine kinase to either the plasma membrane or the endoplasmic reticulum has a negligible effect on the rate of degradation of the sphingosine-1-phosphate that is produced. This suggests that sphingosine-1-phosphate is rapidly transported between membranes. However we also find that cytosolic or endoplasmic-reticulum targeted sphingosine kinase expressed at elevated levels produces extremely high levels of dihydrosphingosine-1-phosphate. Dihydrosphingosine is a proximal precursor in ceramide biosynthesis. Our data indicate that sphingosine kinase can divert substrate from the ceramide de novo synthesis pathway. However plasma membrane-restricted sphingosine kinase cannot access the pool of dihydrosphingosine. Therefore whereas sphingosine kinase localization does not affect downstream metabolism of sphingosine-1-phosphate, localization has an important effect on the pools of substrate to which this key signaling enzyme has access.


Subject(s)
Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , HEK293 Cells , HeLa Cells , Humans , Intracellular Membranes/chemistry , Intracellular Membranes/metabolism , Isoenzymes/metabolism , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingolipids/chemistry , Sphingosine/metabolism
4.
Cell Signal ; 22(9): 1300-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20406683

ABSTRACT

The role of ceramide neo-genesis in cellular stress response signaling is gaining increasing attention with recent progress in elucidating the novel roles and biochemical properties of the ceramide synthase (CerS) enzymes. Selective tissue and subcellular distribution of the six mammalian CerS isoforms, combined with distinct fatty acyl chain length substrate preferences, implicate differential functions of specific ceramide species in cellular signaling. We report here that ionizing radiation (IR) induces de novo synthesis of ceramide to influence HeLa cell apoptosis by specifically activating CerS isoforms 2, 5, and 6 that generate opposing anti- and pro-apoptotic ceramides in mitochondrial membranes. Overexpression of CerS2 resulted in partial protection from IR-induced apoptosis whereas overexpression of CerS5 increased apoptosis in HeLa cells. Knockdown studies determined that CerS2 is responsible for all observable IR-induced C(24:0) CerS activity, and while CerS5 and CerS6 each confer approximately 50% of the C(16:0) CerS baseline synthetic activity, both are required for IR-induced activity. Additionally, co-immunoprecipitation studies suggest that CerS2, 5, and 6 might exist as heterocomplexes in HeLa cells, providing further insight into the regulation of CerS proteins. These data add to the growing body of evidence demonstrating interplay among the CerS proteins in a stress stimulus-, cell type- and subcellular compartment-specific manner.


Subject(s)
Apoptosis , Membrane Proteins/metabolism , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , Acyl Coenzyme A/metabolism , Ceramides/biosynthesis , HeLa Cells , Humans , Isoenzymes/metabolism , Membrane Proteins/physiology , Mitochondria/metabolism , Mitochondria/radiation effects , Palmitoyl Coenzyme A/metabolism , Radiation, Ionizing , Sphingosine N-Acyltransferase/physiology , Tumor Suppressor Proteins/physiology
5.
J Lipid Res ; 51(9): 2546-59, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20386061

ABSTRACT

Sphingosine kinase 1 (SK1) produces sphingosine-1-phosphate (S1P), a potent signaling lipid. The subcellular localization of SK1 can dictate its signaling function. Here, we use artificial targeting of SK1 to either the plasma membrane (PM) or the endoplasmic reticulum (ER) to test the effects of compartmentalization of SK1 on substrate utilization and downstream metabolism of S1P. Expression of untargeted or ER-targeted SK1, but surprisingly not PM-targeted SK1, results in a dramatic increase in the phosphorylation of dihydrosphingosine, a metabolic precursor in de novo ceramide synthesis. Conversely, knockdown of endogenous SK1 diminishes both dihydrosphingosine-1-phosphate and S1P levels. We tested the effects of SK1 localization on degradation of S1P by depletion of the ER-localized S1P phosphatases and lyase. Remarkably, S1P produced at the PM was degraded to the same extent as that produced in the ER. This indicates that there is an efficient mechanism for the transport of S1P from the PM to the ER. In acute labeling experiments, we find that S1P degradation is primarily driven by lyase cleavage of S1P. Counterintuitively, when S1P-specific phosphatases are depleted, acute labeling of S1P is significantly reduced, indicative of a phosphatase-dependent recycling process. We conclude that the localization of SK1 influences the substrate pools that it has access to and that S1P can rapidly translocate from the site where it is synthesized to other intracellular sites.


Subject(s)
Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , Cell Membrane/metabolism , Ceramides/metabolism , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sphingosine/metabolism
6.
Am J Respir Crit Care Med ; 181(4): 344-52, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19965812

ABSTRACT

RATIONALE: Vascular endothelial growth factor receptor (VEGFR) inhibition increases ceramides in lung structural cells of the alveolus, initiating apoptosis and alveolar destruction morphologically resembling emphysema. The effects of increased endogenous ceramides could be offset by sphingosine 1-phosphate (S1P), a prosurvival by-product of ceramide metabolism. OBJECTIVES: The aims of our work were to investigate the sphingosine-S1P-S1P receptor axis in the VEGFR inhibition model of emphysema and to determine whether stimulation of S1P signaling is sufficient to functionally antagonize alveolar space enlargement. METHODS: Concurrent to VEGFR blockade in mice, S1P signaling augmentation was achieved via treatment with the S1P precursor sphingosine, S1P agonist FTY720, or S1P receptor-1 (S1PR1) agonist SEW2871. Outcomes included sphingosine kinase-1 RNA expression and activity, sphingolipid measurements by combined liquid chromatography-tandem mass spectrometry, immunoblotting for prosurvival signaling pathways, caspase-3 activity and terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling assays, and airspace morphometry. MEASUREMENTS AND MAIN RESULTS: Consistent with previously reported de novo activation of ceramide synthesis, VEGFR inhibition triggered increases in lung ceramides, dihydroceramides, and dihydrosphingosine, but did not alter sphingosine kinase activity or S1P levels. Administration of sphingosine decreased the ceramide-to-S1P ratio in the lung and inhibited alveolar space enlargement, along with activation of prosurvival signaling pathways and decreased lung parenchyma cell apoptosis. Sphingosine significantly opposed ceramide-induced apoptosis in cultured lung endothelial cells, but not epithelial cells. FTY720 or SEW2871 recapitulated the protective effects of sphingosine on airspace enlargement concomitant with attenuation of VEGFR inhibitor-induced lung apoptosis. CONCLUSIONS: Strategies aimed at augmenting the S1P-S1PR1 signaling may be effective in ameliorating the apoptotic mechanisms of emphysema development.


Subject(s)
Pulmonary Alveoli/drug effects , Pulmonary Emphysema/drug therapy , Receptors, Lysosphingolipid/drug effects , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cells, Cultured , Ceramides/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Fingolimod Hydrochloride , Indoles/pharmacology , Lysophospholipids/biosynthesis , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polymerase Chain Reaction , Propylene Glycols/pharmacology , Pulmonary Alveoli/physiopathology , Pulmonary Emphysema/physiopathology , Pyrroles/pharmacology , Receptors, Lysosphingolipid/physiology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/drug effects , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/pharmacology
7.
Biochem J ; 419(3): 611-8, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19215222

ABSTRACT

LPA (lysophosphatidic acid) is a lipid mediator that stimulates cell proliferation and growth, and is involved in physiological and pathological processes such as wound healing, platelet activation, angiogenesis and the growth of tumours. Therefore defining the mechanisms of LPA production and degradation are of interest in understanding the regulation of these processes. Extracellular LPA synthesis is relatively well understood, whereas the mechanisms of its degradation are not. One route of LPA degradation is dephosphorylation. A candidate enzyme is the integral membrane exophosphatase LPP1 (lipid phosphate phosphohydrolase type 1). In the present paper, we report the development of a mouse wherein the LPP1 gene (Ppap2a) was disrupted. The homozygous mice, which are phenotypically unremarkable, generally lack Ppap2a mRNA, and multiple tissues exhibit a substantial (35-95%) reduction in LPA phosphatase activity. Compared with wild-type littermates, Ppap2a(tr/tr) animals have increased levels of plasma LPA, and LPA injected intravenously is metabolized at a 4-fold lower rate. Our results demonstrate that LPA is rapidly metabolized in the bloodstream and that LPP1 is an important determinant of this turnover. These results indicate that LPP1 is a catabolic enzyme for LPA in vivo.


Subject(s)
Extracellular Space/metabolism , Lysophospholipids/metabolism , Phosphatidate Phosphatase/metabolism , Animals , Base Sequence , Gene Expression Regulation, Enzymologic , Lysophospholipids/blood , Mice , Molecular Sequence Data , Organ Specificity , Phosphatidate Phosphatase/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
J Biol Chem ; 284(9): 5467-77, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19119142

ABSTRACT

Novel immunomodulatory molecule FTY720 is a synthetic analog of myriocin, but unlike myriocin FTY720 does not inhibit serine palmitoyltransferase. Although many of the effects of FTY720 are ascribed to its phosphorylation and subsequent sphingosine 1-phosphate (S1P)-like action through S1P(1,3-5) receptors, studies on modulation of intracellular balance of signaling sphingolipids by FTY720 are limited. In this study, we used stable isotope pulse labeling of human pulmonary artery endothelial cells with l-[U-(13)C, (15)N]serine as well as in vitro enzymatic assays and liquid chromatography-tandem mass spectrometry methodology to characterize FTY720 interference with sphingolipid de novo biosynthesis. In human pulmonary artery endothelial cells, FTY720 inhibited ceramide synthases, resulting in decreased cellular levels of dihydroceramides, ceramides, sphingosine, and S1P but increased levels of dihydrosphingosine and dihydrosphingosine 1-phosphate (DHS1P). The FTY720-induced modulation of sphingolipid de novo biosynthesis was similar to that of fumonisin B1, a classical inhibitor of ceramide synthases, but differed in the efficiency to inhibit biosynthesis of short-chain versus long-chain ceramides. In vitro kinetic studies revealed that FTY720 is a competitive inhibitor of ceramide synthase 2 toward dihydrosphingosine with an apparent K(i) of 2.15 microm. FTY720-induced up-regulation of DHS1P level was mediated by sphingosine kinase (SphK) 1, but not SphK2, as confirmed by experiments using SphK1/2 silencing with small interfering RNA. Our data demonstrate for the first time the ability of FTY720 to inhibit ceramide synthases and modulate the intracellular balance of signaling sphingolipids. These findings open a novel direction for therapeutic applications of FTY720 that focuses on inhibition of ceramide biosynthesis, ceramide-dependent signaling, and the up-regulation of DHS1P generation in cells.


Subject(s)
Endothelium, Vascular/drug effects , Immunosuppressive Agents/pharmacology , Lung/drug effects , Oxidoreductases/antagonists & inhibitors , Propylene Glycols/pharmacology , Pulmonary Artery/drug effects , Sphingosine/analogs & derivatives , Cells, Cultured , Ceramides/metabolism , Chromatography, Liquid , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Fingolimod Hydrochloride , Humans , Lung/cytology , Lung/enzymology , Lysophospholipids/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Sphingosine/metabolism , Sphingosine/pharmacology , Tandem Mass Spectrometry , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...