Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Biochem Soc Trans ; 33(Pt 5): 1170-3, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16246074

ABSTRACT

Over the past several years, studies of sphingolipid functions in the baker's yeast Saccharomyces cerevisiae have revealed that the sphingoid LCBs (long-chain bases), dihydrosphingosine and PHS (phytosphingosine), are important signalling molecules or second messengers under heat stress and during non-stressed conditions. LCBs are now recognized as regulators of AGC-type protein kinase (where AGC stands for protein kinases A, G and C) Pkh1 and Pkh2, which are homologues of mammalian phosphoinositide-dependent protein kinase 1. LCBs were previously shown to activate Pkh1 and Pkh2, which then activate the downstream protein kinase Pkc1. We have recently demonstrated that PHS stimulates Pkh1 to activate additional downstream kinases including Ypk1, Ypk2 and Sch9. We have also found that PHS acts downstream of Pkh1 and partially activates Ypk1, Ypk2 and Sch9. These kinases control a wide range of cellular processes including growth, cell wall integrity, stress resistance, endocytosis and aging. As we learn more about the cellular processes controlled by Ypk1, Ypk2 and Sch9, we will have a far greater appreciation of LCBs as second messengers.


Subject(s)
Saccharomyces cerevisiae/physiology , Signal Transduction/physiology , Sphingolipids/physiology , Animals , Hot Temperature , Mammals , Protein Kinases/metabolism , Sphingosine/analogs & derivatives , Sphingosine/physiology
2.
Anal Biochem ; 298(2): 283-92, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11700984

ABSTRACT

The molecular species of dihydrosphingosines and phytosphingosines and their 1-phosphates with carbon chain lengths from 16 to 20 have been tagged with the fluorescent amino group reagent, 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate. All these derivatives could be resolved by reversed phase HPLC on a C18 column. A convenient one-pot method is described whereby lipid extracts from strains of Saccharomyces cerevisiae containing carbon chain length homologs of sphingolipid long chain bases and their phosphorylated derivatives were directly reacted with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate, ester lipids were deacylated, and the reaction mixtures were subjected to liquid chromatography. Five molecular species of both sphingolipid long chain bases and their phosphorylated derivatives are for the first time separated and analyzed. The procedure is quite sensitive, requiring only approximately 10(8) wild-type cells.


Subject(s)
Aminoquinolines/analysis , Carbamates/analysis , Chromatography, High Pressure Liquid/methods , Phosphates/analysis , Saccharomyces cerevisiae/chemistry , Sphingolipids/analysis , Reproducibility of Results , Sensitivity and Specificity , Sphingosine/analogs & derivatives , Sphingosine/analysis
3.
Genetics ; 158(4): 1397-411, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11514435

ABSTRACT

css1 mutants display a novel defect in Schizosaccharomyces pombe cell wall formation. The mutant cells are temperature-sensitive and accumulate large deposits of material that stain with calcofluor and aniline blue in their periplasmic space. Biochemical analyses of this material indicate that it consists of alpha- and beta-glucans in the same ratio as found in cell walls of wild-type S. pombe. Strikingly, the glucan deposits in css1 mutant cells do not affect their overall morphology. The cells remain rod shaped, and the thickness of their walls is unaltered. Css1p is an essential protein related to mammalian neutral sphingomyelinase and is responsible for the inositolphosphosphingolipid-phospholipase C activity observed in S. pombe membranes. Furthermore, expression of css1(+) can compensate for loss of ISC1, the enzyme responsible for this activity in Saccharomyces cerevisiae membranes. Css1p localizes to the entire plasma membrane and secretory pathway; a C-terminal fragment of Css1p, predicted to encode a single membrane-spanning segment, is sufficient to direct membrane localization of the heterologous protein, GFP. Our results predict the existence of an enzyme(s) or process(es) essential for the coordination of S. pombe cell wall formation and division that is, in turn, regulated by a sphingolipid metabolite.


Subject(s)
Glucans/metabolism , Schizosaccharomyces/metabolism , Sphingomyelin Phosphodiesterase/chemistry , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelin Phosphodiesterase/physiology , Amino Acid Sequence , Cell Division , Cell Membrane/enzymology , Cell Wall/metabolism , Cloning, Molecular , Epitopes , Gene Deletion , Immunoblotting , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Molecular Sequence Data , Mutation , Plasmids/metabolism , Polysaccharides/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sphingolipids/metabolism , Subcellular Fractions , Temperature , Time Factors , Type C Phospholipases/metabolism
4.
Curr Genet ; 40(4): 221-33, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11795842

ABSTRACT

Sphingolipid long-chain base phosphates (LCBPs) regulate cell proliferation, movement and differentiation in higher eukaryotes. To study the function of LCBPs in Saccharomyces cerevisiae, we inactivated LCBP breakdown pathways. Elimination of both the Dpll lyase and the Lcb3 phosphatase pathways by gene deletion was lethal, indicating that these enzymes regulate LCBP levels to prevent accumulation. Lethality was prevented by eliminating the major LCB kinase. Lcb4p, which synthesizes LCBPs, but not by eliminating the minor LCB kinase, Lcb5p. These data imply that death results from an accumulation of LCBPs made by the Lcb4p kinase. By regulating Lcb4 kinase activity, we found that cell death correlates with LCBP accumulation and that C18 dihydrosphingosine-l-P (DHS-P) and C20 DHS-P are most likely the killing molecules. LCB levels were found to be most elevated in a strain lacking Lcb4 kinase, Dpll lyase and Lcb3 phosphatase activity. Analysis of mutant strains suggests that the C18 and C20 species of LCBPs are preferentially degraded by the Lcb3 phosphate phosphatase, while the Dpll lyase prefers C16 DHS-P as a substrate. These and other data indicate the existence of an unknown mechanism(s) for regulating LCB levels. Our results demonstrate that LCBPs may be used in some circumstances to regulate yeast cell growth.


Subject(s)
Phosphates/physiology , Saccharomyces cerevisiae/physiology , Sphingolipids/physiology , Cell Survival , Gene Deletion , Genotype , Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/physiology , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/genetics , Sphingosine/metabolism
5.
Curr Genet ; 38(4): 191-201, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11126778

ABSTRACT

Strains of Saccharomyces cerevisiae termed sphingolipid compensatory (SLC) do not grow at low pH when the cells lack sphingolipids. To begin to understand why sphingolipids are required for growth at low pH, we isolated derivatives of SLC strains, termed low pH resistant (LprR), carrying the LPR suppressor gene that allows growth at pH 4.1 when cells lack sphingolipids. Suppression is due to mutation of a single nuclear gene. The LPR suppressor gene functions, at least in part, by enhancing the ability of cells lacking sphingolipids to generate a net efflux of protons in suspension fluid with a pH range of 4.0-6.0. The LPR suppressor gene also enables cells lacking sphingolipids to maintain their intracellular pH near neutrality when the pH of the suspension fluid is low, unlike cells lacking the suppressor gene, which cannot maintain their intracellular pH in the face of a low external pH. These results demonstrate that some functions(s) of sphingolipids necessary for growth at low pH can be bypassed by a suppressor mutation. Attempts to clone the LPR suppressor gene were not successful, but they led to the isolation of the CWP2 gene, which encodes a major mannoprotein component of the outer cell wall. It was isolated because an increased copy number has the unusual property of increasing the frequency at which LprR strains arise. As we show here, part of the reason for this effect is that the CWP2 gene is essential for generating a net efflux of protons and for controlling intracellular pH in LprR strains that lack sphingolipids. These results suggest new cellular functions for the Cwp2 protein.


Subject(s)
Genes, Suppressor , Membrane Glycoproteins/genetics , Mutation , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Sphingomyelins/metabolism , Base Sequence , Cloning, Molecular , DNA Primers , Hydrogen-Ion Concentration , Nuclear Magnetic Resonance, Biomolecular , Phenotype , Saccharomyces cerevisiae/growth & development
6.
Proc Natl Acad Sci U S A ; 97(17): 9531-6, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10931938

ABSTRACT

We have previously identified a Saccharomyces cerevisiae mutant that is markedly more resistant than wild-type to Dahlia merckii antimicrobial peptide 1 (DmAMP1), an antifungal plant defensin isolated from seeds of dahlia (Dahlia merckii). A complementation approach was followed that consisted of the introduction of a genomic library of DmAMP1-sensitive wild-type yeast into the DmAMP1-resistant yeast mutant and screening for restored sensitivity to DmAMP1. The gene determining sensitivity of S. cerevisiae to DmAMP1 was identified as IPT1, a gene encoding an enzyme involved in the last step of the synthesis of the sphingolipid mannose-(inositol-phosphate)(2)-ceramide. Strains with a nonfunctional IPT1 allele lacked mannose-(inositol-phosphate)(2)-ceramide in their plasma membranes, bound significantly less DmAMP1 compared with wild-type strains, and were highly resistant to DmAMP1-mediated membrane permeabilization. All of these phenotypic deviations could be restored by reintroduction of a functional IPT1 gene. Our data support a model in which membrane patches containing sphingolipids act as binding sites for DmAMP1 or, alternatively, are required to anchor membrane or cell wall-associated proteins, which themselves interact with DmAMP1.


Subject(s)
Antifungal Agents/pharmacology , Asteraceae/chemistry , Defensins , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Plant Proteins/pharmacology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/drug effects , Sphingolipids/biosynthesis , Alleles , Antifungal Agents/metabolism , Binding Sites , Cell Division/drug effects , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cloning, Molecular , Genes, Fungal/genetics , Genetic Complementation Test , Microbial Sensitivity Tests , Mutation/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Plant Proteins/metabolism , Protein Binding , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Sphingolipids/metabolism
8.
Biochim Biophys Acta ; 1438(3): 305-21, 1999 Jun 10.
Article in English | MEDLINE | ID: mdl-10366774

ABSTRACT

Our knowledge of sphingolipid metabolism and function in Saccharomyces cerevisiae is growing rapidly. Here we discuss the current status of sphingolipid metabolism including recent evidence suggesting that exogenous sphingoid long-chain bases must first be phosphorylated and then dephosphorylated before incorporation into ceramide. Phenotypes of strains defective in sphingolipid metabolism are discussed because they provide hints about the undiscovered functions of sphingolipids and are one of the major reasons for studying this model eukaryote. The long-chain base phosphates, dihydrosphingosine-1-phosphate and phytosphingosine-1-phosphate, have been hypothesized to play roles in heat stress resistance, perhaps acting as signaling molecules. We evaluate the data supporting this hypothesis and suggest future experiments needed to verify it. Finally, we discuss recent clues that may help to reveal how sphingolipid synthesis and total cellular sphingolipid content are regulated.


Subject(s)
Saccharomyces cerevisiae/metabolism , Sphingolipids/metabolism , Ceramides/metabolism , Hot Temperature , Phenotype , Saccharomyces cerevisiae/genetics , Sphingolipids/biosynthesis , Sphingosine/analogs & derivatives , Sphingosine/metabolism
9.
J Bacteriol ; 181(4): 1134-40, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973338

ABSTRACT

Sphingolipid long-chain bases and their phosphorylated derivatives, for example, sphingosine-1-phosphate in mammals, have been implicated as signaling molecules. The possibility that Saccharomyces cerevisiae cells also use long-chain-base phosphates to regulate cellular processes has only recently begun to be examined. Here we present a simple and sensitive procedure for analyzing and quantifying long-chain-base phosphates in S. cerevisiae cells. Our data show for the first time that phytosphingosine-1-phosphate (PHS-1-P) is present at a low but detectable level in cells grown on a fermentable carbon source at 25 degreesC, while dihydrosphingosine-1-phosphate (DHS-1-P) is only barely detectable. Shifting cells to 37 degreesC causes transient eight- and fivefold increases in levels of PHS-1-P and DHS-1-P, respectively, which peak after about 10 min. The amounts of both compounds return to the unstressed levels by 20 min after the temperature shift. These data are consistent with PHS-1-P and DHS-1-P being signaling molecules. Cells unable to break down long-chain-base phosphates, due to deletion of DPL1 and LCB3, show a 500-fold increase in PHS-1-P and DHS-1-P levels, grow slowly, and survive a 44 degreesC heat stress 10-fold better than parental cells. These and other data for dpl1 or lcb3 single-mutant strains suggest that DHS-1-P and/or PHS-1-P act as signals for resistance to heat stress. Our procedure should expedite experiments to determine how the synthesis and breakdown of these compounds is regulated and how the compounds mediate resistance to elevated temperature.


Subject(s)
Phospholipids/chemistry , Saccharomyces cerevisiae/chemistry , Sphingolipids/chemistry , Sphingosine/analogs & derivatives , Heat Stress Disorders , Models, Biological , Mutation , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sphingosine/analysis
10.
Biochim Biophys Acta ; 1426(2): 347-57, 1999 Jan 06.
Article in English | MEDLINE | ID: mdl-9878820

ABSTRACT

Many advances in our understanding of fungal sphingolipids have been made in recent years. This review focuses on the types of sphingolipids that have been found in fungi and upon the genes in Saccharomyces cerevisiae, the common baker's yeast, that are necessary for sphingolipid metabolism. While only a small number of fungi have been examined, most contain sphingolipids composed of ceramide derivatized at carbon-1 with inositol phosphate. Further additions include mannose and then other carbohydrates. The second major class of fungal sphingolipids is the glycosylceramides, having either glucose or galactose attached to ceramide rather than inositol phosphate. The glycosylceramides sometimes contain additional carbohydrates. Knowledge of the genome sequence has expedited identification of S. cerevisiae genes necessary for sphingolipid metabolism. At least one gene is known for most steps in S. cerevisiae sphingolipid metabolism, but more are likely to be identified so that the 13 known genes are likely to grow in number. The AUR1 gene is necessary for addition of inositol phosphate to ceramide and has been identified as a target of several potent antifungal compounds. This essential step in yeast sphingolipid synthesis, which is not found in humans, appears to be an excellent target for the development of more effective antifungal compounds, both for human and for agricultural use.


Subject(s)
Saccharomyces cerevisiae/metabolism , Sphingolipids/biosynthesis , Amino Acid Sequence , Glycosphingolipids/biosynthesis , Hexosyltransferases/metabolism , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Saccharomyces cerevisiae/enzymology , Sequence Homology, Amino Acid
11.
J Biol Chem ; 273(31): 19437-42, 1998 Jul 31.
Article in English | MEDLINE | ID: mdl-9677363

ABSTRACT

Sphingolipid long chain bases (LCBs) and phosphorylated derivatives, particularly sphingosine 1-phosphate, are putative signaling molecules. To help elucidate the physiological roles of LCB phosphates, we identified two Saccharomyces cerevisiae genes, LCB4 (YOR171c) and LCB5 (YLR260w), which encode LCB kinase activity. This conclusion is based upon the synthesis of LCB kinase activity in Escherichia coli expressing either LCB gene. LCB4 encodes most (97%) Saccharomyces LCB kinase activity, with the remainder requiring LCB5. Log phase lcb4-deleted yeast cells make no LCB phosphates, showing that the Lcb4 kinase synthesizes all detectable LCB phosphates under these growth conditions. The Lcb4 and Lcb5 proteins are paralogs with 53% amino acid identity but are not related to any known protein, thus revealing a new class of lipid kinase. Two-thirds of the Lcb4 and one-third of the Lcb5 kinase activity are in the membrane fraction of yeast cells, a puzzling finding in that neither protein contains a membrane-localization signal. Both enzymes can use phytosphingosine, dihydrosphingosine, or sphingosine as substrate. LCB4 and LCB5 should be useful for probing the functions of LCB phosphates in S. cerevisiae. Potential mammalian cDNA homologs of the LCB kinase genes may prove useful in helping to understand the function of sphingosine 1-phosphate in mammals.


Subject(s)
Fungal Proteins/chemistry , Lysophospholipids , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases/chemistry , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Sphingolipids/metabolism , Amino Acid Sequence , Genes, Fungal/genetics , Kinetics , Membrane Proteins/chemistry , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Recombinant Proteins/chemistry , Sequence Alignment , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Substrate Specificity
12.
J Biol Chem ; 273(18): 11062-8, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9556590

ABSTRACT

The Saccharomyces cerevisiae gene SYR2, necessary for growth inhibition by the cyclic lipodepsipeptide syringomycin E, is shown to be required for 4-hydroxylation of long chain bases in sphingolipid biosynthesis. Four lines of support for this conclusion are presented: (a) the predicted Syr2p shows sequence similarity to diiron-binding membrane enzymes involved in oxygen-dependent modifications of hydrocarbon substrates, (b) yeast strains carrying a disrupted SYR2 allele produced sphingoid long chain bases lacking the 4-hydroxyl group present in wild type strains, (c) 4-hydroxylase activity was increased in microsomes prepared from a SYR2 overexpression strain, and (d) the syringomycin E resistance phenotype of a syr2 mutant strain was suppressed when grown under conditions in which exogenous 4-hydroxysphingoid long chain bases were incorporated into sphingolipids. The syr2 strain produced wild type levels of sphingolipids, substantial levels of hydroxylated very long chain fatty acids, and the full complement of normal yeast sphingolipid head groups. These results show that the SYR2 gene is required for the 4-hydroxylation reaction of sphingolipid long chain bases, that this hydroxylation is not essential for growth, and that the 4-hydroxyl group of sphingolipids is necessary for syringomycin E action on yeast.


Subject(s)
Fungal Proteins/metabolism , Mixed Function Oxygenases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Sphingolipids/metabolism , Amino Acid Sequence , Fungal Proteins/chemistry , Fungal Proteins/genetics , Hydroxylation , Iron/metabolism , Molecular Sequence Data , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid
13.
J Biol Chem ; 273(13): 7235-43, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9516416

ABSTRACT

Sphingolipid-related metabolites have been implicated as potential signaling molecules in many studies with mammalian cells as well as in some studies with yeast. Our previous work showed that sphingolipid-deficient strains of Saccharomyces cerevisiae are unable to resist a heat shock, indicating that sphingolipids are necessary for surviving heat stress. Recent evidence suggests that one role for the sphingolipid intermediate ceramide may be to act as a second messenger to signal accumulation of the thermoprotectant trehalose. We examine here the mechanism for generating the severalfold increase in ceramide observed during heat shock. As judged by compositional analysis and mass spectrometry, the major ceramides produced during heat shock are similar to those found in complex sphingolipids, a mixture of N-hydroxyhexacosanoyl C18 and C20 phytosphingosines. Since the most studied mechanism for ceramide generation in animal cells is via a phospholipase C-type sphingomyelin hydrolysis, we examined S. cerevisiae for an analogous enzyme. Using [3H]phytosphingosine and [3H]inositol-labeled yeast sphingolipids, a novel membrane-associated phospholipase C-type activity that generated ceramide from inositol-P-ceramide, mannosylinositol-P-ceramide, and mannose(inositol-P)2-ceramide was demonstrated. The sphingolipid head groups were concomitantly liberated with the expected stoichiometry. However, other data demonstrate that the ceramide generated during heat shock is not likely to be derived by breakdown of complex sphingolipids. For example, the water-soluble fraction of heat-shocked cells showed no increase in any of the sphingolipid head groups, which is inconsistent with complex sphingolipid hydrolysis. Rather, we find that de novo ceramide synthesis involving ceramide synthase appears to be responsible for heat-induced ceramide elevation. In support of this hypothesis, we find that the potent ceramide synthase inhibitor, australifungin, completely inhibits both the heat-induced increase in incorporation of [3H]sphinganine into ceramide as well as the heat-induced increase in ceramide as measured by mass. Thus, heat-induced ceramide most likely arises by temperature activation of the enzymes that generate ceramide precursors, activation of ceramide synthase itself, or both.


Subject(s)
Ceramides/biosynthesis , Hot Temperature , Saccharomyces cerevisiae/metabolism , Antifungal Agents/pharmacology , Chromatography, High Pressure Liquid , Hydrolysis , Kinetics , Saccharomyces cerevisiae/drug effects , Sphingolipids/metabolism , Tetrahydronaphthalenes/pharmacology , Type C Phospholipases/metabolism
14.
J Biol Chem ; 273(5): 2829-34, 1998 Jan 30.
Article in English | MEDLINE | ID: mdl-9446592

ABSTRACT

Sphingoid long chain bases have many effects on cells including inhibition or stimulation of growth. The physiological significance of these effects is unknown in most cases. To begin to understand how these compounds inhibit growth, we have studied Saccharomyces cerevisiae cells. Growth of tryptophan (Trp-) auxotrophs was more strongly inhibited by phytosphingosine (PHS) than was growth of Trp+ strains, suggesting that PHS diminishes tryptophan uptake and starves cells for this amino acid. This hypothesis is supported by data showing that growth inhibition is relieved by increasing concentrations of tryptophan in the culture medium and by multiple copies of the TAT2 gene, encoding a high affinity tryptophan transporter. Measurement of tryptophan uptake shows that it is inhibited by PHS. Finally, PHS treatment induces the general control response, indicating starvation for amino acids. Multiple copies of TAT2 do not protect cells against two other cationic lipids, stearylamine, and sphingosine, indicating that the effect of PHS on tryptophan utilization is specific. Other data demonstrate that PHS reduces uptake of leucine, histidine, and proline by specific transporters. Our data suggest that PHS targets proteins in the amino acid transporter family but not other distantly related membrane transporters, including those necessary for uptake of adenine and uracil.


Subject(s)
Amino Acids/metabolism , Carrier Proteins/drug effects , Escherichia coli Proteins , Membrane Transport Proteins/drug effects , Saccharomyces cerevisiae/drug effects , Sphingosine/analogs & derivatives , Amino Acid Transport Systems , Amino Acids, Cyclic/metabolism , Biological Transport/drug effects , Carrier Proteins/metabolism , Leucine/metabolism , Membrane Transport Proteins/metabolism , Proline/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sphingosine/pharmacology
15.
J Biol Chem ; 272(47): 29620-5, 1997 Nov 21.
Article in English | MEDLINE | ID: mdl-9368028

ABSTRACT

Knowledge of the Saccharomyces cerevisiae genes and proteins necessary for sphingolipid biosynthesis is far from complete. Such information should expedite studies of pathway regulation and sphingolipid functions. Using the Aur1 protein sequence, recently identified as necessary for synthesis of the sphingolipid inositol-P-ceramide (IPC), we show that a homolog (open reading frame YDR072c), termed Ipt1 (inositolphosphotransferase 1) is necessary for synthesis of mannose-(inositol-P)2-ceramide (M(IP)2C), the most abundant and complex sphingolipid in S. cerevisiae. This conclusion is based upon analysis of an ipt1-deletion strain, which fails to accumulate M(IP)2C and instead accumulates increased amounts of the precursor mannose-inositol-P-ceramide. The mutant also fails to incorporate radioactive precursors into M(IP)2C, and membranes prepared from it do not incorporate [3H-inositol]phosphatidylinositol into M(IP)2C, indicating a lack of M(IP)2C synthase activity (putatively phosphatidylinositol:mannose-inositol-P-ceramide phosphoinositol transferase). M(IP)2C synthase activity is inhibited in the micromolar range by aureobasidin A, but drug sensitivity is over 1000-fold lower than reported for IPC synthase activity. An ipt1-deletion mutant has no severe phenotypic effects but is slightly more resistant to growth inhibition by calcium ions. Identification of the IPT1 gene should be helpful in determining the function of the M(IP)2C sphingolipid and in determining the catalytic mechanism of IPC and M(IP)2C synthases.


Subject(s)
Depsipeptides , Glycosphingolipids/biosynthesis , Hexosyltransferases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Catalysis , Drug Resistance, Microbial , Fungal Proteins/metabolism , Hexosyltransferases/metabolism , Molecular Sequence Data , Open Reading Frames , Peptides, Cyclic/metabolism , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Saccharomyces cerevisiae/metabolism
16.
J Biol Chem ; 272(48): 30196-200, 1997 Nov 28.
Article in English | MEDLINE | ID: mdl-9374502

ABSTRACT

The ability of organisms to quickly respond to stresses requires the activation of many intracellular signal transduction pathways. The sphingolipid intermediate ceramide is thought to be particularly important for activating and coordinating signaling pathways during mammalian stress responses. Here we present the first evidence that ceramide and other sphingolipid intermediates are signaling molecules in the Saccharomyces cerevisiae heat stress response. Our data show a 2-3-fold transient increase in the concentration of C18-dihydrosphingosine and C18-phytosphingosine, more than a 100-fold transient increase in C20-dihydrosphingosine and C20-phytosphingosine, and a more stable 2-fold increase in ceramide containing C18-phytosphingosine and a 5-fold increase in ceramide containing C20-phytosphingosine following heat stress. Treatment of cells with dihydrosphingosine activates transcription of the TPS2 gene encoding a subunit of trehalose synthase and causes trehalose, a known thermoprotectant, to accumulate. Dihydrosphingosine induces expression of a STRE-LacZ reporter gene, showing that the global stress response element, STRE, found in many yeast promoter sequences can be activated by sphingolipid signals. The TPS2 promoter contains four STREs that may mediate dihydrosphingosine responsiveness. Using genetic and other approaches it should be possible to identify sphingolipid signaling pathways in S. cerevisiae and quantify the importance of each during heat stress.


Subject(s)
Depsipeptides , Glucosyltransferases/genetics , Heat-Shock Proteins/genetics , Hot Temperature , Multienzyme Complexes/genetics , Phosphoric Monoester Hydrolases/genetics , Saccharomyces cerevisiae/metabolism , Sphingolipids/metabolism , Fungal Proteins/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Peptides, Cyclic/pharmacology , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Signal Transduction , Transcription, Genetic , Trehalose/metabolism
17.
J Biol Chem ; 272(26): 16110-7, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9195906

ABSTRACT

To identify genes necessary for sphingolipid synthesis in Saccharomyces cerevisiae we developed a procedure to enrich for mutants unable to incorporate exogenous long chain base into sphingolipids. We show here that a mutant strain, AG84-3, isolated by using the enrichment procedure, makes sphingolipids from endogenously synthesized but not from exogenously supplied long chain base. A gene termed LCB3 (YJL134W, GenBank designation X87371x21), which complements the long chain base utilization defect of strain AG84-3, was isolated from a genomic DNA library. The gene is predicted to encode a protein with multiple membrane-spanning domains and a COOH-terminal glycosylphosphatidylinositiol cleavage/attachment site. Deletion of the lcb3 gene in a wild type genetic background reduces the rate of exogenous long chain base incorporation into sphingolipids and makes the host strain more resistant to growth inhibition by long chain bases. Only one protein in current data bases, the S. cerevisiae open-reading frame YKR053C, whose function is unknown, shows homology to the Lcb3 protein. The two proteins are not, however, functional homologs because deletion of the YKR053C open reading frame does not impair long chain base utilization or enhance resistance of cells to growth inhibition by long chain bases. Based upon these data we hypothesize that the Lcb3 protein is a plasma membrane transporter capable of transporting sphingoid long chain bases into cells. It is the first candidate for such a transporter and the first member of what appears to be a new class of membrane-bound proteins.


Subject(s)
Genes, Fungal , Membrane Proteins/genetics , Saccharomyces cerevisiae/genetics , Sphingolipids/metabolism , Alleles , Amino Acid Sequence , Membrane Proteins/physiology , Molecular Sequence Data , Sphingosine/analogs & derivatives , Sphingosine/metabolism
18.
J Biol Chem ; 272(15): 9809-17, 1997 Apr 11.
Article in English | MEDLINE | ID: mdl-9092515

ABSTRACT

We have identified a Saccharomyces cerevisiae gene necessary for the step in sphingolipid synthesis in which inositol phosphate is added to ceramide to form inositol-P-ceramide, a reaction catalyzed by phosphatidylinositol:ceramide phosphoinositol transferase (IPC synthase). This step should be an effective target for antifungal drugs. A key element in our experiments was the development of a procedure for isolating mutants defective in steps in sphingolipid synthesis downstream from the first step including a mutant defective in IPC synthase. An IPC synthase defect is supported by data showing a failure of the mutant strain to incorporate radioactive inositol or N-acetylsphinganine into sphingolipids and, by using an improved assay, a demonstration that the mutant strain lacks enzyme activity. Furthermore, the mutant accumulates ceramide when fed exogenous phytosphingosine as expected for a strain lacking IPC synthase activity. Ceramide accumulation is accompanied by cell death, suggesting the presence of a ceramide-activated death response in yeast. A gene, AUR1 (YKL004w), that complements the IPC synthase defect and restores enzyme activity and sphingolipid synthesis was isolated. Mutations in AUR1 had been shown previously to give resistance to the antifungal drug aureobasidin A, leading us to predict that the drug should inhibit IPC synthase activity. Our data show that the drug is a potent inhibitor of IPC synthase with an IC50 of about 0.2 nM. Fungal pathogens are an increasing threat to human health. Now that IPC synthase has been shown to be the target for aureobasidin A, it should be possible to develop high throughput screens to identify new inhibitors of IPC synthase to combat fungal diseases.


Subject(s)
Antifungal Agents/pharmacology , Depsipeptides , Drug Resistance, Microbial/genetics , Fungal Proteins/genetics , Hexosyltransferases/metabolism , Sphingolipids/biosynthesis , Amino Acid Sequence , Ceramides/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Molecular Sequence Data , Peptides, Cyclic/pharmacology , Saccharomyces cerevisiae , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
19.
J Bacteriol ; 179(5): 1513-20, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9045807

ABSTRACT

Sphingolipids are normally necessary for growth of Saccharomyces cerevisiae cells, but mutant strains that bypass the need for sphingolipids have been identified. Such bypass mutants fail to grow under stressful conditions, including low pH (pH 4.1), when they lack sphingolipids. To begin to understand why sphingolipids seem to be necessary for coping with low-pH stress, we screened a genomic library and selected a suppressor gene, CWP2 (cell wall protein 2), that when present in multiple copies partially compensates for the lack of sphingolipids and enhances survival at low pH. To explain these results, we present evidence that sphingolipids are required for a normal rate of transport of glycosylphosphatidylinositol (GPI)-anchored proteins, including Cwp2 and Gas1/Gpg1, from the endoplasmic reticulum (ER) to the Golgi apparatus. The effect of sphingolipids is specific for transport of GPI-anchored proteins because no effect on the rate of transport of carboxypeptidase Y, a non-GPI-anchored protein, was observed. Since the Gasl protein accumulated in the ER with a GPI anchor in cells lacking sphingolipids, we conclude that sphingolipids are not necessary for anchor attachment. Therefore, sphingolipids must be necessary for a step in formation of COPII vesicles or for their transport to the Golgi apparatus. Our data identify the Cwp2 protein as a vital component in protecting cells from the stress of low pH.


Subject(s)
Fungal Proteins/metabolism , Glycosylphosphatidylinositols/metabolism , Membrane Glycoproteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Sphingolipids/physiology , Biological Transport , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Genes, Fungal , Genes, Suppressor , Golgi Apparatus/metabolism , Hydrogen-Ion Concentration , Membrane Glycoproteins/genetics , Protein Precursors/metabolism , Saccharomyces cerevisiae/genetics
20.
J Bacteriol ; 178(21): 6223-6, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8892822

ABSTRACT

Hyphal forms of the human pathogen Candida albicans have been found to contain substantial quantities of phosphosphingolipids. These lipids were fractionated into three classes by normal-phase high-performance liquid chromatography. The first class contained equimolar amounts of phosphorus, inositol, phytosphingosines, and fatty acids; their composition and chromatographic behavior suggest that these compounds are inositolphosphorylceramides. The second class contained equimolar amounts of phosphorus, mannosylinositol, phytosphingosines, and fatty acids; their composition and chromatographic behavior indicate that these compounds are mannosylinositolphosphorylceramides. The third class of compounds contained phosphorus, mannosylinositol, inositol, phytosphingosines, and fatty acids in a molar ratio of 2:1:1:1:1; their composition and chromatographic behavior indicate that these compounds are mannosyldiinositolphosphorylceramides. Molecular species in each class differ in the composition of long chain bases and fatty acids; the most abundant long chain bases were C18 and C20 phytosphingosines, and the most abundant fatty acids were hydroxy and nonhydroxy C24-26. The array of sphingolipids in C. albicans is similar to that of Saccharomyces cerevisiae. Sphingolipids have been shown to be essential in S. cerevisiae, thus these lipids, which are not present in animals, offer a potentially unique target for antifungal chemotherapy against C. albicans.


Subject(s)
Candida albicans/metabolism , Ceramides/chemistry , Alkalies , Fatty Acids/chemistry , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...