Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Biochem J ; 440(1): 127-35, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21793801

ABSTRACT

S49 mouse lymphoma cells undergo apoptosis in response to the ALP (alkyl-lysophospholipid) edelfosine (1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine), FasL (Fas ligand) and DNA damage. S49 cells made resistant to ALP (S49(AR)) are defective in sphingomyelin synthesis and ALP uptake, and also have acquired resistance to FasL and DNA damage. However, these cells can be re-sensitized following prolonged culturing in the absence of ALP. The resistant cells show sustained ERK (extracellular-signal-regulated kinase)/Akt activity, consistent with enhanced survival signalling. In search of a common mediator of the observed cross-resistance, we found that S49(AR) cells lacked the PtdIns(3,4,5)P(3) phosphatase SHIP-1 [SH2 (Src homology 2)-domain-containing inositol phosphatase 1], a known regulator of the Akt survival pathway. Re-sensitization of the S49(AR) cells restored SHIP-1 expression as well as phosphoinositide and sphingomyelin levels. Knockdown of SHIP-1 mimicked the S49(AR) phenotype in terms of apoptosis cross-resistance, sphingomyelin deficiency and altered phosphoinositide levels. Collectively, the results of the present study suggest that SHIP-1 collaborates with sphingomyelin synthase to regulate lymphoma cell death irrespective of the nature of the apoptotic stimulus.


Subject(s)
Phospholipid Ethers/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fas Ligand Protein/metabolism , Inositol Polyphosphate 5-Phosphatases , Lymphoma/pathology , Mice , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Proto-Oncogene Proteins c-akt/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism
2.
PLoS One ; 6(4): e19170, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559477

ABSTRACT

FRET-based sensors for cyclic Adenosine Mono Phosphate (cAMP) have revolutionized the way in which this important intracellular messenger is studied. The currently prevailing sensors consist of the cAMP-binding protein Epac1, sandwiched between suitable donor- and acceptor fluorescent proteins (FPs). Through a conformational change in Epac1, alterations in cellular cAMP levels lead to a change in FRET that is most commonly detected by either Fluorescence Lifetime Imaging (FLIM) or by Sensitized Emission (SE), e.g., by simple ratio-imaging. We recently reported a range of different Epac-based cAMP sensors with high dynamic range and signal-to-noise ratio. We showed that constructs with cyan FP as donor are optimal for readout by SE, whereas other constructs with green FP donors appeared much more suited for FLIM detection. In this study, we present a new cAMP sensor, termed (T)Epac(VV), which employs mTurquoise as donor. Spectrally very similar to CFP, mTurquoise has about doubled quantum efficiency and unlike CFP, its fluorescence decay is strictly single-exponential. We show that (T)Epac(VV) appears optimal for detection both by FLIM and SE, that it has outstanding FRET span and signal-to-noise ratio, and improved photostability. Hence, (T)Epac(VV) should become the cAMP sensor of choice for new experiments, both for FLIM and ratiometric detection.


Subject(s)
Biosensing Techniques , Cyclic AMP/chemistry , Fluorescence Resonance Energy Transfer/methods , Animals , Cell Line , Cloning, Molecular , Equipment Design , Fluorescence , Fluorescence Resonance Energy Transfer/instrumentation , Fluorescent Dyes/chemistry , Genetic Techniques , HeLa Cells , Humans , Mice , Microscopy, Confocal/methods , Signal Processing, Computer-Assisted
3.
Biochem J ; 425(1): 225-34, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-19824885

ABSTRACT

The ALP (alkyl-lysophospholipid) edelfosine (1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine) induces apoptosis in S49 mouse lymphoma cells. A variant cell line, S49AR, made resistant to ALP, was found previously to be impaired in ALP uptake via lipid-raft-mediated endocytosis. In the present paper, we report that these cells display cross-resistance to Fas/CD95 ligation [FasL (Fas ligand)], and can be gradually resensitized by prolonged culturing in the absence of ALP. Fas and ALP activate distinct apoptotic pathways, since ALP-induced apoptosis was not abrogated by dominant-negative FADD (Fas-associated protein with death domain), cFLIP(L) [cellular FLICE (FADD-like interleukin 1beta-converting enzyme)-inhibitory protein long form] or the caspase 8 inhibitor Z-IETD-FMK (benzyloxycarbonyl-Ile-Glu-Thr-Asp-fluoromethylketone). ALP-resistant cells showed decreased Fas expression, at both the mRNA and protein levels, in a proteasome-dependent fashion. The proteasome inhibitor MG132 partially restored Fas expression and resensitized the cells to FasL, but not to ALP. Resistant cells completely lacked SM (sphingomyelin) synthesis, which seems to be a unique feature of the S49 cell system, having very low SM levels in parental cells. Lack of SM synthesis did not affect cell growth in serum-containing medium, but retarded growth under serum-free (SM-free) conditions. SM deficiency determined in part the resistance to ALP and FasL. Exogenous short-chain (C12-) SM partially restored cell-surface expression of Fas in lipid rafts and FasL sensitivity, but did not affect Fas mRNA levels or ALP sensitivity. We conclude that the acquired resistance of S49 cells to ALP is associated with down-regulated SM synthesis and Fas gene transcription and that SM in lipid rafts stabilizes Fas expression at the cell surface.


Subject(s)
Drug Resistance, Neoplasm , Lysophospholipids/pharmacology , Sphingomyelins/metabolism , fas Receptor/metabolism , Animals , Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Fas Ligand Protein/pharmacology , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Leupeptins/pharmacology , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Microscopy, Confocal , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sphingomyelins/deficiency , Transfection , fas Receptor/genetics
4.
Biochem Pharmacol ; 74(10): 1456-65, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17803969

ABSTRACT

Perifosine is a member of the class of synthetic alkylphospholipids (APLs) and is being evaluated as anti-cancer agent in several clinical trials. These single-chain APLs accumulate in cellular membranes and disturb lipid-dependent signal transduction, ultimately causing apoptosis in a variety of tumor cells. The APL prototype edelfosine was previously found to be endocytosed by S49 mouse lymphoma cells via lipid rafts. An edelfosine-resistant cell variant, S49(AR), was found to be cross-resistant to other APLs, including perifosine. This resistance was due to defective synthesis of the raft constituent sphingomyelin, which abrogated APL cellular uptake. Sensitivity of S49 cells to edelfosine was higher than perifosine, which correlated with a relatively higher uptake. Human KB epidermal carcinoma cells were much more sensitive to APLs than S49 cells. Their much higher APL uptake was highly dependent on intracellular ATP and ambient temperature, and was blocked by chlorpromazine, independent of canonical endocytic pathways. We found no prominent role of lipid rafts for APL uptake in these KB cells; contrary to S49(AR) cells, perifosine-resistant KBr cells display normal sphingomyelin synthesis, whereas APL uptake by the responsive KB cells was insensitive to treatment with methyl-beta-cyclodextrin, a cholesterol-sequestrator and inhibitor of raft-mediated endocytosis. In conclusion, different mechanisms determine APL uptake and consequent apoptotic toxicity in lymphoma versus carcinoma cells. In the latter cells, APL uptake is mainly determined by a raft- and endocytosis-independent process, but metabolic energy-dependent process, possibly by a lipid transporter.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Drug Resistance, Neoplasm , Lymphoma/metabolism , Membrane Microdomains/metabolism , Phospholipid Ethers/pharmacology , Phosphorylcholine/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Cell Line, Tumor , Humans , KB Cells , Mice , Phospholipids/metabolism , Phosphorylcholine/pharmacology
5.
Mol Cancer Ther ; 6(8): 2337-45, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17699729

ABSTRACT

Single-chain alkylphospholipids, unlike conventional chemotherapeutic drugs, act on cell membranes to induce apoptosis in tumor cells. We tested four different alkylphospholipids, i.e., edelfosine, perifosine, erucylphosphocholine, and compound D-21805, as inducers of apoptosis in the mouse lymphoma cell line S49. We compared their mechanism of cellular entry and their potency to induce apoptosis through inhibition of de novo biosynthesis of phosphatidylcholine at the endoplasmic reticulum. Alkylphospholipid potency closely correlated with the degree of phosphatidylcholine synthesis inhibition in the order edelfosine > D-21805 > erucylphosphocholine > perifosine. In all cases, exogenous lysophosphatidylcholine, an alternative source for cellular phosphatidylcholine production, could partly rescue cells from alkylphospholipid-induced apoptosis, suggesting that phosphatidylcholine biosynthesis is a direct target for apoptosis induction. Cellular uptake of each alkylphospholipid was dependent on lipid rafts because pretreatment of cells with the raft-disrupting agents, methyl-beta-cyclodextrin, filipin, or bacterial sphingomyelinase, reduced alkylphospholipid uptake and/or apoptosis induction and alleviated the inhibition of phosphatidylcholine synthesis. Uptake of all alkylphospholipids was inhibited by small interfering RNA (siRNA)-mediated blockage of sphingomyelin synthase (SMS1), which was previously shown to block raft-dependent endocytosis. Similar to edelfosine, perifosine accumulated in (isolated) lipid rafts independent on raft sphingomyelin content per se. However, perifosine was more susceptible than edelfosine to back-extraction by fatty acid-free serum albumin, suggesting a more peripheral location in the cell due to less effective internalization. Overall, our results suggest that lipid rafts are critical membrane portals for cellular entry of alkylphospholipids depending on SMS1 activity and, therefore, are potential targets for alkylphospholipid anticancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lymphoma/pathology , Membrane Microdomains/drug effects , Phospholipids/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Endocytosis/drug effects , HeLa Cells , Humans , Mice , Phosphatidylcholines/biosynthesis , Phospholipid Ethers/chemistry , Phospholipid Ethers/pharmacology , Phospholipids/chemistry , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Phosphorylcholine/pharmacology , Sphingomyelins/biosynthesis , Time Factors
6.
Biochem J ; 401(2): 541-9, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17049047

ABSTRACT

The ALP (alkyl-lysophospholipid) edelfosine (1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine; Et-18-OCH3) induces apoptosis in S49 mouse lymphoma cells. To this end, ALP is internalized by lipid raft-dependent endocytosis and inhibits phosphatidylcholine synthesis. A variant cell-line, S49AR, which is resistant to ALP, was shown previously to be unable to internalize ALP via this lipid raft pathway. The reason for this uptake failure is not understood. In the present study, we show that S49AR cells are unable to synthesize SM (sphingomyelin) due to down-regulated SMS1 (SM synthase 1) expression. In parental S49 cells, resistance to ALP could be mimicked by small interfering RNA-induced SMS1 suppression, resulting in SM deficiency and blockage of raft-dependent internalization of ALP and induction of apoptosis. Similar results were obtained by treatment of the cells with myriocin/ISP-1, an inhibitor of general sphingolipid synthesis, or with U18666A, a cholesterol homoeostasis perturbing agent. U18666A is known to inhibit Niemann-Pick C1 protein-dependent vesicular transport of cholesterol from endosomal compartments to the trans-Golgi network and the plasma membrane. U18666A reduced cholesterol partitioning in detergent-resistant lipid rafts and inhibited SM synthesis in S49 cells, causing ALP resistance similar to that observed in S49AR cells. The results are explained by the strong physical interaction between (newly synthesized) SM and available cholesterol at the Golgi, where they facilitate lipid raft formation. We propose that ALP internalization by lipid-raft-dependent endocytosis represents the retrograde route of a constitutive SMS1- and lipid-raft-dependent membrane vesicular recycling process.


Subject(s)
Apoptosis/drug effects , Membrane Microdomains/chemistry , Phospholipid Ethers/pharmacology , Transferases (Other Substituted Phosphate Groups)/biosynthesis , Androstenes/pharmacology , Animals , Bridged-Ring Compounds/pharmacology , Cholesterol/analysis , Cholesterol/metabolism , Down-Regulation , Endocytosis/drug effects , Fatty Acids, Monounsaturated/pharmacology , Gene Expression/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Norbornanes , Phospholipid Ethers/metabolism , RNA, Small Interfering/pharmacology , Sphingomyelins/analysis , Sphingomyelins/metabolism , Thiocarbamates , Thiones/pharmacology , Transferases (Other Substituted Phosphate Groups)/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...