Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 90(4): 917-25, 2004 Feb 23.
Article in English | MEDLINE | ID: mdl-14970874

ABSTRACT

Anticancer drugs generally have intracellular targets, implicating transport over the plasma membrane. For amphiphilic agents, such as the anthracycline doxorubicin, this occurs by passive diffusion. We investigated whether exogenous membrane-permeable lipid analogues improve this drug influx. Combinations of drugs and lipid analogues were coadministered to cultured endothelial cells and various tumour cell lines, and subsequent drug accumulation in cells was quantified. We identified N-hexanoyl-sphingomyelin (SM) as a potent enhancer of drug uptake. Low micromolar amounts of this short-chain sphingolipid, being not toxic itself, enhanced the uptake of doxorubicin up to 300% and decreased its EC(50) toxicity values seven- to 14-fold. N-hexanoyl SM acts at the level of the plasma membrane, but was found not incorporated in (isolated) lipid rafts, and artificial disruption or elimination of raft constituents did not affect its drug uptake-enhancing effect. Further, any mechanistic role of the endocytic machinery, membrane leakage or ABC-transporter-mediated efflux could be excluded. Finally, a correlation was established between the degree of drug lipophilicity, as defined by partitioning in a two-phase octanol-water system, and the susceptibility of the drug towards the uptake-enhancing effect of the sphingolipid. A clear optimum was found for amphiphilic drugs, such as doxorubicin, epirubicin and topotecan, indicating that N-hexanoyl-SM might act by modulating the average degree of plasma membrane lipophilicity, in turn facilitating transbilayer drug diffusion. The concept of short-chain sphingolipids as amphiphilic drug potentiators provides novel opportunities for improving drug delivery technologies.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/pharmacokinetics , Sphingomyelins/pharmacology , Adenocarcinoma , Animals , Breast Neoplasms/pathology , Cell Death , Cell Membrane , Diffusion , Drug Delivery Systems , Drug Interactions , Drug Synergism , Endothelial Cells , Fibroblasts , Humans , Mice , Mice, Knockout , Tumor Cells, Cultured
2.
Circ Res ; 89(11): 957-68, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11717151

ABSTRACT

Sphingolipids have emerged as a new class of lipid mediators. In response to various extracellular stimuli, sphingolipid turnover can be stimulated in vascular cells and cardiac myocytes. Subsequent generation of sphingolipid molecules such as ceramide, sphingosine, and sphingosine-1-phosphate, is followed by regulation of ion fluxes and activation of various signaling pathways leading to smooth muscle cell proliferation, endothelial cell differentiation or apoptotic cell death, cell contraction, retraction, or migration. The importance of sphingolipids in cardiovascular signaling is illustrated by recent observations implicating them in physiological processes such as vasculogenesis as well as in frequent pathological conditions, including atherosclerosis and its complications.


Subject(s)
Heart Diseases/etiology , Myocardium/metabolism , Sphingolipids/physiology , Animals , Apoptosis , Cell Division , Coronary Artery Disease/etiology , Humans , Ion Transport , Myocardial Contraction , Myocardial Reperfusion Injury/etiology , Myocardium/cytology , Neovascularization, Physiologic , Radiation Injuries/etiology , Signal Transduction , Sphingolipids/chemistry
3.
Int J Cancer ; 94(2): 157-65, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11668492

ABSTRACT

Multidrug-resistant (MDR) cancer cells have been shown to have an accumulation of glucosylceramide (GlcCer). In this study, we aim at localizing, at subcellular level, where these lipids accumulate. Neutral lipids and phospholipid containing organelles have been identified using confocal fluorescence microscopy and microspectrofluorometry by monitoring the emission of the fluorescent probe Nile-red. Data from confocal fluorescence microscopy analysis shows accumulation of neutral lipids in cytoplasmic droplets of MDR human carcinoma MCF7R cells. Microspectrofluorometric measurements show an increase of the gold-yellow emission intensity in MCF7R cells, corresponding to neutral lipids. Similar observations were made in human MDR vincristine-HL60 and doxorubicin-KB selected cells. Total cellular glucosylceramide (GlcCer) measurements using [(3)H]-palmitic acid and thin layer chromatography show a significant increase of GlcCer in MCF7R cells. Moreover, MCF7R cells treated with fluorescent GlcCer-bodipy exhibit an accumulation of this lipid in cytoplasmic droplets. Treatment of MCF7R cells with 1-phenyl-2-palmitoylamino-3-morpholino-1-propanolol (PPMP), a potent inhibitor of GlcCer synthase, attenuates the Nile-red fluorescence emission emanating from these structures and reverses MDR. Moreover, Golgi compartments stained with fluorescent PPMP-bodipy, show an increase in the Golgi compartments density. Treatment of MCF7R cells with cyclosporine A (CSA), tamoxifen (TMX) and 3'-azido-3'deoxythymidine (AZT) leads to the same effect observed in the presence of PPMP. Treatment of MCF7 and MCF7R with the beta-glucosidase inhibitor conduritol beta-epoxide (CBE) significantly increases resistance to daunorubicin only in MCF7R cells. These data demonstrate also that: (i) CSA, an inhibitor of MDR, has an additional target in addition to P-glycoprotein; and (ii) TMX (used in breast cancer treatment and prevention) and AZT (used in the treatment of HIV) could have side effects by disturbing lipid metabolism and inhibiting many cellular functions required in normal cells.


Subject(s)
Cytoplasm/metabolism , Glucosylceramides/metabolism , Neoplasms/metabolism , Cell Nucleus/metabolism , Daunorubicin/pharmacokinetics , Daunorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Glucosylceramides/analysis , Humans , Inositol/analogs & derivatives , Inositol/pharmacology , Microscopy, Confocal , Neoplasms/drug therapy , Tumor Cells, Cultured
4.
J Membr Biol ; 181(3): 153-62, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11420602

ABSTRACT

Administration of most chemotherapeutic agents eventually results in the onset of apoptosis, despite the agents' variety in structure and molecular targets. Ceramide, the central molecule in cellular glycosphingolipid metabolism, has recently been identified as an important mediator of this process. Indeed, one of the events elicited by application of many cytotoxic drugs is an accumulation of this lipid. Treatment failure in cancer chemotherapy is largely attributable to multidrug resistance, in which tumor cells are typically cross-resistant to multiple chemotherapeutic agents. Different cellular mechanisms underlying this phenomenon have been described. Of these the drug efflux pump activity of P-glycoprotein and the multidrug resistance-associated proteins are the most extensively studied examples. Recently, an increased cellular capacity for ceramide glycosylation has been recognized as a novel multidrug resistance mechanism. Indeed, virtually all multidrug-resistant cells exhibit a deviating sphingolipid composition, most typically, increased levels of glucosylceramide. On the other hand, several direct molecular interactions between sphingolipids and drug efflux proteins have been described. Therefore, in addition to a role in the multidrug resistance phenotype by which ceramide accumulation and, thus, the onset of apoptosis are prevented, an indirect role for sphingolipids might be envisaged, by which the activity of these efflux proteins is modulated. In this review, we present an overview of the current understanding of the interesting relations that exist between sphingolipid metabolism and multidrug resistance.


Subject(s)
Drug Resistance, Multiple , Sphingolipids/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Apoptosis/drug effects , Ceramides/metabolism , Drug-Related Side Effects and Adverse Reactions , Glucosyltransferases/metabolism , Humans , Signal Transduction , Sphingolipids/chemistry
5.
Biochem J ; 355(Pt 3): 859-68, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11311151

ABSTRACT

Sphingomyelinases hydrolyse sphingomyelin to ceramide, a process involved in signal-transduction routes leading to apoptosis and various other cellular responses. In the present study, we investigated the sphingomyelinase content of caveolae, invaginated plasma-membrane microdomains that contain a variety of signalling molecules. These structures are highly enriched in sphingomyelin as well as in ceramide, which suggests that metabolism of these lipids might, to some extent, occur locally. By cell fractionation, we demonstrate that, in addition to a previously reported minute amount of acidic sphingomyelinase activity, a substantial amount of neutral sphingomyelinase activity resides in caveolae of human skin fibroblasts. This caveolar neutral sphingomyelinase activity was also detected in Niemann-Pick disease type A fibroblasts, which are completely devoid of functional acidic sphingomyelinase. Neutral (but not acidic) sphingomyelinase activity was specifically inhibited by a peptide that corresponds to the scaffolding domain of caveolin, which suggests a direct molecular interaction between the two proteins. In addition, this finding implies a cytosolic orientation of the caveolar neutral sphingomyelinase. Interestingly, stimulation of fibroblasts with tumour necrosis factor alpha (TNFalpha) resulted in a partial shift of its p55 receptor to caveolin-enriched membrane fractions and the appearance of caveolin-sensitive neutral sphingomyelinase activity in the non-caveolar fractions. These results suggest that (part of) the presently identified caveolar neutral sphingomyelinase activity is involved in TNFalpha signalling.


Subject(s)
Caveolins/pharmacology , Signal Transduction/physiology , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Caveolae/metabolism , Caveolins/chemistry , Cells, Cultured , Cholesterol/deficiency , Cholesterol/metabolism , Enzyme Activation/drug effects , Humans , Niemann-Pick Diseases/pathology , Protein Structure, Tertiary , Sphingolipids/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors
6.
FEBS Lett ; 474(1): 107-10, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10828460

ABSTRACT

Regulation of capacitative Ca(2+) entry was studied in two different multidrug resistance (MDR) protein (MRP1) overexpressing cell lines, HT29(col) and GLC4/ADR. MRP1 overexpression was accompanied by a decreased response to thapsigargin. Moreover, inhibition of capacitative Ca(2+) entry by D, L-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) was abolished in MRP1 overexpressing cells. Both PDMP and the MRP1 inhibitor MK571 greatly reduced InsP(3)-mediated (45)Ca(2+) release from intracellular stores in HT29 cells. Again, these effects were virtually abolished in HT29(col) cells. Our results point to a modulatory role of MRP1 on intracellular calcium concentration ([Ca(2+)](i)) homeostasis which may contribute to the MDR phenotype.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Calcium/metabolism , Gene Expression , Homeostasis , ATP-Binding Cassette Transporters/antagonists & inhibitors , Adenocarcinoma , Calcium Radioisotopes , Colonic Neoplasms , Humans , Inositol 1,4,5-Trisphosphate/pharmacology , Morpholines/pharmacology , Multidrug Resistance-Associated Proteins , Propionates/pharmacology , Quinolines/pharmacology , Thapsigargin/pharmacology , Tumor Cells, Cultured
7.
Int J Cancer ; 87(2): 172-8, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10861470

ABSTRACT

We have obtained a novel multidrug resistant cell line, derived from HT29 G(+) human colon carcinoma cells, by selection with gradually increasing concentrations of the anti-mitotic, microtubule-disrupting agent colchicine. This HT29(col) cell line displayed a 25-fold increase in colchicine resistance and exhibited cross-resistance to doxorubicin, VP16, vincristine and taxol. Immunoblotting, combined with RT-PCR showed that the multidrug resistance phenotype was conferred by specific overexpression of the multidrug resistance protein 1. Confocal scanning laser microscopy revealed that multidrug resistance protein 1 specifically localized in the plasma membrane of HT29(col) cells. In a functional assay, using the fluorescent multidrug resistance protein 1 substrate 5-carboxyfluorescein, an increased efflux activity of HT29(col) cells was measured, as compared to the wild-type HT29 G(+) cells. MK571, a specific inhibitor of multidrug resistance protein 1, blocked the 5-carboxyfluorescein efflux, but only partially reversed resistance to colchicine, indicating that additional multidrug resistance mechanisms operate in HT29(col) cells. In conclusion, these results show for the first time overexpression of a functional multidrug resistance protein 1 under colchicine pressure, indicating that colchicine is not a P-glycoprotein-specific substrate. Colchicine-induced overexpression of multidrug resistance protein 1 is accompanied by a changed sphingolipid composition, i.e., enhanced levels of glucosylceramide and galactosylceramide. In addition, ceramide, a lipid messenger molecule involved in apoptosis-related signal transduction processes, was much more abundant in HT29(col) cells, which is indicative of a stress response.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Colonic Neoplasms/metabolism , Sphingolipids/biosynthesis , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Ceramides/biosynthesis , Colchicine/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Resistance, Multiple , Etoposide/pharmacology , Fluoresceins/pharmacology , Galactosylceramides/biosynthesis , Glucosylceramides/biosynthesis , HT29 Cells , Humans , Immunoblotting , Immunohistochemistry , Microscopy, Confocal , Microtubules/drug effects , Mitosis/drug effects , Multidrug Resistance-Associated Proteins , Paclitaxel/pharmacology , Propionates/pharmacology , Quinolines/pharmacology , Signal Transduction , Vincristine/pharmacology
8.
J Biol Chem ; 275(36): 28316-25, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10871611

ABSTRACT

The sequence similarity with bacterial neutral sphingomyelinase resulted in the isolation of putative mammalian counterparts and, subsequently, identification of similar molecules in a number of other eukaryotic organisms. Based on sequence similarities and previous characterization of the mammalian enzymes, we have chemically modified specific residues and performed site-directed mutagenesis in order to identify critical catalytic residues and determinants for membrane localization. Modification of histidine residues and the substrate protection experiments demonstrated the presence of reactive histidine residues within the active site. Site directed mutagenesis suggested an essential role in catalysis for two histidine residues (His-136 and His-272), which are conserved in all sequences. Mutations of two additional histidines (His-138 and His-151), conserved only in eukaryotes, resulted in reduced neutral sphingomyelinase activity. In addition to sphingomyelin, the enzyme also hydrolyzed lysophosphatidylcholine. Exposure to an oxidizing environment or modification of cysteine residues using several specific compounds also inactivated the enzyme. Site-directed mutagenesis of eight cysteine residues and gel-shift analysis demonstrated that these residues did not participate in the catalytic reaction and suggested the involvement of cysteines in the formation/breakage of disulfide bonds, which could underlie the reversible inactivation by the oxidizing compounds. Cellular localization studies of a series of deletion mutants, expressed as green fluorescent protein fusion proteins, demonstrated that the transmembrane region contains determinants for the endoplasmic reticulum localization.


Subject(s)
Sphingomyelin Phosphodiesterase/chemistry , Sphingomyelin Phosphodiesterase/metabolism , Type C Phospholipases/chemistry , Type C Phospholipases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Catalysis , Consensus Sequence , Conserved Sequence , DNA Primers , Histidine , Humans , Kinetics , Mammals , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Alignment , Sequence Homology, Amino Acid
9.
Clin Cancer Res ; 6(3): 942-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741719

ABSTRACT

In this study, we show that an inhibitor of glycosphin-golipid biosynthesis, D,L-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), increases the chemosensitivity of neuroblastoma tumor cells for Taxol and vincristine. At noneffective low doses of Taxol or vincristine, the addition of a noneffective dose of PDMP resulted in 70% cytotoxicity, indicating synergy. Such an effect was not observed for etoposide (VP16). PDMP caused an early (6 h) increase in ceramide (Cer) levels, but the excess Cer was metabolically removed in the long-term (96 h). However, upon incubation with PDMP in combination with Taxol, but not with etoposide, Cer levels remained elevated at 96 h. These results suggest that neuroblastoma cells are normally able to metabolically remove excess Cer, but lose this capacity upon exposure to microtubule modulating anticancer agents (Taxol or vincristine). In addition, PDMP treatment resulted in a decreased efflux of [14C]Taxol and [3H]vincristine from neuroblastoma cells, similar to treatment with PSC833 or MK571, suggesting an effect of PDMP on the transporter proteins P-glycoprotein and/or multidrug resistance protein. PDMP did not further reduce [14C]Taxol or [3H]vincristine efflux in PSC833-treated cells, although it did further diminish cell survival under these conditions. We conclude that a combined administration of nontoxic concentrations of PDMP and either Taxol or vincristine results in highly sensitized neuroblastoma cells. This appears to involve a sustained elevation of Cer levels, possibly in concert with increased drug accumulation.


Subject(s)
Antineoplastic Agents/pharmacology , Morpholines/pharmacology , Neuroblastoma/prevention & control , Paclitaxel/pharmacology , Vincristine/pharmacology , Animals , Cell Survival/drug effects , Ceramides/metabolism , Cyclosporins/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Neuroblastoma/pathology , Paclitaxel/metabolism , Tumor Cells, Cultured , Vincristine/metabolism
10.
Biochem Biophys Res Commun ; 266(2): 492-6, 1999 Dec 20.
Article in English | MEDLINE | ID: mdl-10600530

ABSTRACT

In the present study we show that neutral hexanoyl-(glyco)sphingolipids inhibit P-glycoprotein (Pgp) activity in human ovarian 2780AD cells. By contrast, hexanoylceramide and the gangliosides GM(3) and GM(2) had no effect on Pgp activity, whereas sphingosine had a stimulating effect. In the case of hexanoylglucosylceramide, inhibition of Pgp activity by was reflected by a regained doxorubicin sensitivity of cells, which were grown in medium supplemented with the lipid. Our results lead to the conclusion that a direct transmodulation of Pgp activity by glycolipids occurs, depending on lipid headgroup structure, which can result in reduced resistance to the chemotherapeutic agent doxorubicin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Glucosylceramides/pharmacology , Sphingolipids/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Doxorubicin/pharmacology , Drug Resistance, Multiple , Female , Humans , Ovarian Neoplasms , Rhodamine 123/metabolism , Sphingosine/pharmacology , Tumor Cells, Cultured
11.
Eur J Biochem ; 258(1): 233-42, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9851714

ABSTRACT

Previous work suggested that glucosylceramide (GlcCer) plays a role in the regulation of cell differentiation of HT29 human colon tumor cells. In the present study, we investigated the role of GlcCer in the cellular release of carcinoembryonic antigen (CEA), a marker for cell differentiation. This was done by modulating the intracellular level of the glycolipid, according to two different approaches. The cells were treated with D,L-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), which resulted in a specific lowering of the cellular GlcCer pool. Alternatively, by exogenous addition of a short-chain analog of the lipid, hexanoyl(C6)-GlcCer, the cellular pool was enhanced. The results demonstrate that PDMP causes an increase in the release of CEA, while exogenous C6-GlcCer suppresses its release. Furthermore, the enhanced release of CEA in the presence of PDMP, could be completely reversed upon exogenous addition of C6-GlcCer. Control experiments reveal that a potential interference of the well-known modulator of cell physiology, ceramide (Cer), can be excluded. Long-term depletion of GlcCer resulted in a change in a morphological feature of differentiation of the cells, i.e. an increase in apical membrane surface with microvilli brush borders, accompanied by an enhanced expression of the cytoskeletal protein villin. These results, together with the observations on modulation of the differentiation marker CEA by GlcCer, provide support for the conclusion that GlcCer interferes with the differentiation of HT29 cells.


Subject(s)
Carcinoembryonic Antigen/metabolism , Glucosylceramides/physiology , Cell Differentiation , Glucosylceramides/antagonists & inhibitors , Glucosylceramides/biosynthesis , HT29 Cells , Humans
12.
Biochem Biophys Res Commun ; 247(3): 802-8, 1998 Jun 29.
Article in English | MEDLINE | ID: mdl-9647774

ABSTRACT

Recently, (glyco)sphingolipids (SL) like ceramide (Cer) and glucosylceramide (GlcCer) have been shown to be involved in signaling pathways leading to differentiation and apoptosis in several cell types, including the colon adenocarcinoma cell line HT29. Intracellular levels of Cer can be modulated by ligands such as interferon-gamma (IFN gamma). In the present study we show that IFN gamma, depending on its concentration, has both differentiation- and apoptosis-inducing effects on HT29 cells. Since both phenomena have been related to SL-mediated signaling in other cell types, we next examined whether IFN gamma was able to induce changes in the SL levels of HT29 cells. Remarkably, no significant changes in these levels could be revealed, implying that SL are not involved in IFN gamma-induced differentiation and/or apoptosis of HT29 cells. This observation provides evidence for the hypothesis that SL-mediated signaling pathways might be more cell type specific than is generally assumed.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Glucosylceramides/metabolism , Interferon-gamma/pharmacology , Signal Transduction/physiology , Carcinoembryonic Antigen/metabolism , Ceramides/metabolism , DNA Fragmentation/drug effects , Glycosphingolipids/metabolism , HT29 Cells , Histones/metabolism , Humans
13.
Biochem J ; 331 ( Pt 2): 563-9, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9531498

ABSTRACT

Ceramide (Cer) has been implicated in the regulation of apoptosis. In this study, we elevated cellular Cer levels in human colon-carcinoma (HT29(rev)) cells by incubating the cells in the presence of bacterial sphingomyelinase (bSMase) or, alternatively, in the presence of C2-Cer, a short-chain analogue of the sphingolipid. bSMase treatment did not induce apoptosis in these cells, as revealed by a lack of both DNA fragmentation and cleavage of poly(ADP-ribose)polymerase. In contrast, apoptosis did occur upon addition of C2-Cer. These findings led us to study whether differences in the metabolic fate of the excess of Cer, as generated by both treatments, contributed to the observed difference in apoptosis-inducing capacity. C2-Cer was rapidly taken up by HT29(rev) cells and accumulated due to the absence of substantial metabolic conversion. Upon addition of bSMase, hydrolysis of sphingomyelin resulted in a reduction of that pool to 20% compared with control values, accompanied by a multi-fold increase in Cer level. In spite of the continuous presence of active bSMase, the Cer increase turned out to be transient. Cer levels reached their maximum 1-2 h after addition of bSMase, followed by a significant decrease. Excessive Cer was mainly turned over via cerebrosides into complex glycolipids, including gangliosides. In the presence of glucosylceramide synthase- and/or ceramidase inhibitors, this conversion was significantly blocked and bSMase-generated Cer accumulated in the cells. However, even under these conditions apoptosis did not occur. In conclusion, the inability of bSMase to induce apoptosis of HT29(rev) cells does not appear to be due to rapid metabolic conversion of excessive Cer. Since apoptosis is induced upon addition of C2-Cer, we therefore propose that the intracellular target involved in the propagation of the apoptotic signal is reached by C2-Cer, but not by bSMase-generated Cer.


Subject(s)
Apoptosis , Ceramides/metabolism , Ceramides/pharmacology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Fragmentation , Humans , Hydrolysis , Poly(ADP-ribose) Polymerases/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...