Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1949: 307-324, 2019.
Article in English | MEDLINE | ID: mdl-30790264

ABSTRACT

For uncompromised in vitro assays for intermembrane lipid transfer and membrane fusion fluorescent membrane-spanning lipids have proved to be invaluable tools. These lipids in contrast to phosphoglycerolipids and sphingolipids are resistant to spontaneous as well as protein-mediated intermembrane transfer. Here I describe the synthesis of some homo-substituted fluorescent bipolar membrane-spanning lipids that bear a fluorescent tag either directly or via a phosphoethanolamine spacer to the lipid core. For the synthesis the lipid core of the bipolar membrane-spanning lipids, i.e., the tetraether lipid caldarchaeol, is prepared from cultures of the archaea Thermoplasma acidophilum.


Subject(s)
Cell Membrane/metabolism , Lipid Metabolism , Membrane Fusion , Membrane Lipids/metabolism , Archaea/metabolism , Cell Membrane/chemistry , Ethanolamines/chemistry , Ethanolamines/metabolism , Fluorescence Resonance Energy Transfer , Glyceryl Ethers/chemistry , Glyceryl Ethers/metabolism , Liposomes , Membrane Lipids/chemistry
2.
FEBS Lett ; 592(23): 3992-4006, 2018 12.
Article in English | MEDLINE | ID: mdl-30188569

ABSTRACT

The unveiling of ganglioside metabolism and biological function in the last decades depended on the extensive study of inherited human disease, studies with cultured cells, and specifically designed mouse models. Nonetheless, only few of the accomplishments made so far would have been possible without the use of labeled gangliosides, such as their radiolabeled and/or fluorescent analogs, as well as other modified gangliosides bearing cross-linking, affinity, or paramagnetic groups. Over the years, chemists and biochemists have made tremendous progress toward developing labeled gangliosides for their application in biological systems. These labeled ganglioside probes of the ganglio series have been successfully incorporated in cultured vertebrate cells and in artificial model membranes. In this Review, I provide an overview over the different methods, mostly semisynthetic procedures, to obtain these labeled ganglioside probes that have been used to elucidate the molecular basis of ganglioside-related biology as well as the physicochemical properties of gangliosides.


Subject(s)
Fluorescent Dyes/chemistry , Gangliosides/biosynthesis , Gangliosides/chemistry , Isotope Labeling , Animals , Carbohydrate Sequence , Cells, Cultured , Humans , Models, Animal , Research/trends
3.
Methods Mol Biol ; 1804: 323-356, 2018.
Article in English | MEDLINE | ID: mdl-29926417

ABSTRACT

Labeled gangliosides are invaluable tools to study their transport and metabolism within cells as well as to determine their distribution in membranes, and their interaction with membrane lipids and proteins. Here I describe established procedures to synthesize ganglioside derivatives with a fluorescent tag either attached to its sialooligosaccharide or ceramide portion. These procedures are chosen as to minimize the integrity of the ganglioside molecule and hence, to leave their native skeleton formally intact. The α-position of the stearic acid residue is favorable for the attachment both of hydrophilic and of lipophilic dyes. In some other cases, and starting from lyso-gangliosides, procedures are described by which a fluorescent tag bound to a short acyl chain replaces the native acyl chain of gangliosides.


Subject(s)
Fluorescent Dyes/chemistry , Gangliosides/chemical synthesis , Alkalies/chemistry , Amidohydrolases/metabolism , Esters/chemical synthesis , Esters/chemistry , Gangliosides/chemistry , Hydrolysis , Staining and Labeling , Stearic Acids/chemistry
4.
Methods Mol Biol ; 1804: 357-381, 2018.
Article in English | MEDLINE | ID: mdl-29926418

ABSTRACT

For the physicochemical studies aimed at elucidating the dynamic of gangliosides in membranes and their interaction with proteins and membrane lipids, photoactivatable and paramagnetic ganglioside derivatives have proved to be invaluable tools. Here, protocols for the synthesis of such ganglioside derivatives are described. These derivatives bear in their ceramide portion either a highly photoreactive (3-trifluoromethyl)phenyldiazirinyl- or a spin active doxyl-labeled acyl chain in place of their natural acyl chain.


Subject(s)
Gangliosides/chemical synthesis , Light , Spin Labels , Carbon Radioisotopes/chemistry , Esters/chemical synthesis , Gangliosides/chemistry , Proton Magnetic Resonance Spectroscopy
5.
Angew Chem Int Ed Engl ; 56(19): 5252-5257, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28378443

ABSTRACT

The main glycoforms of the hydrophobic lysosomal glycoprotein saposin D (SapD) were synthesized by native chemical ligation. An approach for the challenging solid-phase synthesis of the fragments was developed. Three SapD glycoforms were obtained following a general and robust refolding and purification protocol. A crystal structure of one glycoform confirmed its native structure and disulfide pattern. Functional assays revealed that the lipid-binding properties of three SapD glycoforms are highly affected by the single sugar moiety of SapD showing a dependency of the size and the type of N-glycan.


Subject(s)
Carbohydrates/chemistry , Saposins/chemical synthesis , Saposins/metabolism , Carbohydrate Conformation , Glycosylation , Humans , Hydrophobic and Hydrophilic Interactions , Particle Size , Saposins/chemistry
6.
J Lipid Res ; 58(3): 563-577, 2017 03.
Article in English | MEDLINE | ID: mdl-28126847

ABSTRACT

Glucosylceramide (GlcCer) is the primary storage lipid in the lysosomes of Gaucher patients and a secondary one in Niemann-Pick disease types A, B, and C. The regulatory roles of lipids on the hydrolysis of membrane bound GlcCer by lysosomal ß-glucocerebrosidase (GBA1) was probed using a detergent-free liposomal assay. The degradation rarely occurs at uncharged liposomal surfaces in the absence of saposin (Sap) C. However, anionic lipids stimulate GlcCer hydrolysis at low pH by up to 1,000-fold depending on the nature and position of the negative charges in their head groups while cationic lipids inhibit the degradation, thus showing the importance of electrostatic interactions between the polycationic GBA1 and the negatively charged vesicle surfaces at low pH. Ceramide, fatty acids, monoacylglycerol, and diacylglycerol also stimulate GlcCer hydrolysis while SM, sphingosine, and sphinganine play strong inhibitory roles, thereby explaining the secondary storage of GlcCer in Niemann-Pick diseases. Surprisingly, cholesterol stimulates GlcCer degradation in the presence of bis(monoacylglycero)phosphate (BMP). Sap C strongly stimulates GlcCer hydrolysis even in the absence of BMP and the regulatory roles of the intraendolysosomal lipids on its activity is discussed. Our data suggest that these strong modifiers of GlcCer hydrolysis affect the genotype-phenotype correlation in several cases of Gaucher patients independent of the types.


Subject(s)
Gaucher Disease/metabolism , Glucosylceramidase/genetics , Glucosylceramides/metabolism , Niemann-Pick Diseases/metabolism , Cholesterol/metabolism , Gaucher Disease/genetics , Gaucher Disease/pathology , Genetic Association Studies , Glucosylceramidase/metabolism , Humans , Hydrolysis , Lipid Metabolism/genetics , Lysophospholipids/metabolism , Lysosomes/enzymology , Monoglycerides/metabolism , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/pathology , Saposins/metabolism
7.
J Lipid Res ; 56(10): 1861-79, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269359

ABSTRACT

A Förster resonance energy transfer-based fusion and transfer assay was developed to study, in model membranes, protein-mediated membrane fusion and intermembrane lipid transfer of fluorescent sphingolipid analogs. For this assay, it became necessary to apply labeled reporter molecules that are resistant to spontaneous as well as protein-mediated intermembrane transfer. The novelty of this assay is the use of nonextractable fluorescent membrane-spanning bipolar lipids. Starting from the tetraether lipid caldarchaeol, we synthesized fluorescent analogs with fluorophores at both polar ends. In addition, we synthesized radioactive glycosylated caldarchaeols. These labeled lipids were shown to stretch through bilayer membranes rather than to loop within a single lipid layer of liposomes. More important, the membrane-spanning lipids (MSLs) in contrast to phosphoglycerides proved to be nonextractable by proteins. We could show that the GM2 activator protein (GM2AP) is promiscuous with respect to glycero- and sphingolipid transfer. Saposin (Sap) B also transferred sphingolipids albeit with kinetics different from GM2AP. In addition, we could unambiguously show that the recombinant activator protein Sap C x His6 induced membrane fusion rather than intermembrane lipid transfer. These findings showed that these novel MSLs, in contrast with fluorescent phosphoglycerolipids, are well suited for an uncompromised monitoring of membrane fusion and intermembrane lipid transfer.


Subject(s)
Glyceryl Ethers/metabolism , Membrane Fusion/physiology , Membrane Lipids/metabolism , Animals , Cells, Cultured , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Glyceryl Ethers/chemistry , Humans , Lipid Bilayers/metabolism , Liposomes/metabolism , Membrane Lipids/chemistry , Sphingolipids/chemistry , Sphingolipids/metabolism , Swine , Thermoplasma/metabolism
8.
J Lipid Res ; 56(9): 1747-61, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26175473

ABSTRACT

Ganglioside GM2 is the major lysosomal storage compound of Tay-Sachs disease. It also accumulates in Niemann-Pick disease types A and B with primary storage of SM and with cholesterol in type C. Reconstitution of GM2 catabolism with ß-hexosaminidase A and GM2 activator protein (GM2AP) at uncharged liposomal surfaces carrying GM2 as substrate generated only a physiologically irrelevant catabolic rate, even at pH 4.2. However, incorporation of anionic phospholipids into the GM2 carrying liposomes stimulated GM2 hydrolysis more than 10-fold, while the incorporation of plasma membrane stabilizing lipids (SM and cholesterol) generated a strong inhibition of GM2 hydrolysis, even in the presence of anionic phospholipids. Mobilization of membrane lipids by GM2AP was also inhibited in the presence of cholesterol or SM, as revealed by surface plasmon resonance studies. These lipids also reduced the interliposomal transfer rate of 2-NBD-GM1 by GM2AP, as observed in assays using Förster resonance energy transfer. Our data raise major concerns about the usage of recombinant His-tagged GM2AP compared with untagged protein. The former binds more strongly to anionic GM2-carrying liposomal surfaces, increases GM2 hydrolysis, and accelerates intermembrane transfer of 2-NBD-GM1, but does not mobilize membrane lipids.


Subject(s)
G(M2) Activator Protein/metabolism , G(M2) Ganglioside/metabolism , Liposomes/metabolism , Membrane Lipids/metabolism , Ceramides/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Fluorescence Resonance Energy Transfer , G(M2) Activator Protein/genetics , HEK293 Cells , Humans , Hydrolysis/drug effects , Lysophospholipids/administration & dosage , Membrane Lipids/genetics , Monoglycerides/administration & dosage , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/metabolism , Niemann-Pick Diseases/pathology , Sphingomyelins/metabolism , Surface Plasmon Resonance , Tay-Sachs Disease/genetics , Tay-Sachs Disease/metabolism , Tay-Sachs Disease/pathology , beta-Hexosaminidase alpha Chain/metabolism
9.
J Lipid Res ; 55(12): 2692-704, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25341943

ABSTRACT

Within recent years, ganglioside patterns have been increasingly analyzed by MS. However, internal standards for calibration are only available for gangliosides GM1, GM2, and GM3. For this reason, we prepared homologous internal standards bearing nonnatural fatty acids of the major mammalian brain gangliosides GM1, GD1a, GD1b, GT1b, and GQ1b, and of the tumor-associated gangliosides GM2 and GD2. The fatty acid moieties were incorporated after selective chemical or enzymatic deacylation of bovine brain gangliosides. For modification of the sphingoid bases, we developed a new synthetic method based on olefin cross metathesis. This method was used for the preparation of a lyso-GM1 and a lyso-GM2 standard. The total yield of this method was 8.7% for the synthesis of d17:1-lyso-GM1 from d20:1/18:0-GM1 in four steps. The title compounds are currently used as calibration substances for MS quantification and are also suitable for functional studies.


Subject(s)
Gangliosides/chemistry , Lipids/chemistry , Acylation , Amidohydrolases/metabolism , Animals , Brain/metabolism , Calibration , Cattle , Gangliosides/isolation & purification , Gangliosides/metabolism , Gangliosides/standards , Hydrolysis , Lipid Metabolism , Lipids/isolation & purification , Lipids/standards , Male , Molecular Structure , Neurons/chemistry , Reference Standards , Stereoisomerism , Testis/enzymology , Tissue Extracts/chemistry , beta-Galactosidase/metabolism
10.
Biochim Biophys Acta ; 1841(8): 1161-73, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24389251

ABSTRACT

The unraveling of sphingolipid metabolism and function in the last 40 years relied on the extensive study of inherited human disease and specifically-tailored mouse models. However, only few of the achievements made so far would have been possible without chemical biology tools, such as fluorescent and/or radio-labeled and other artificial substrates, (mechanism-based) enzyme inhibitors, cross-linking probes or artificial membrane models. In this review we provide an overview over chemical biology tools that have been used to gain more insight into the molecular basis of sphingolipid-related biology. Many of these tools are still of high relevance for the investigation of current sphingolipid-related questions, others may stimulate the tailoring of novel probes suitable to address recent and future issues in the field. This article is part of a Special Issue entitled Tools to study lipid functions.


Subject(s)
Proteins/metabolism , Sphingolipids/metabolism , Biological Transport , Carbohydrate Sequence , Cells, Cultured , Humans , Molecular Probes , Molecular Sequence Data , Protein Binding , Sphingolipids/chemistry
11.
Methods Enzymol ; 519: 1-38, 2013.
Article in English | MEDLINE | ID: mdl-23280106

ABSTRACT

Details of molecular membrane dynamics in living cells such as lipid-protein interactions or the incorporation of molecules into lipid "rafts" are often hidden to the observer because of the limited spatial resolution of conventional far-field optical microscopy. Fortunately, the superior spatial resolution of far-field stimulated-emission-depletion (STED) nanoscopy allows gaining new insights. Applying fluorescence correlation spectroscopy (FCS) in focal spots continuously tuned down to 30 nm in diameter distinguishes free from anomalous molecular diffusion due to transient binding, as for the diffusion of fluorescent phosphoglycero- and sphingolipid analogs in the plasma membrane of living cells. STED-FCS data recorded at different environmental conditions and on different lipid analogs reveal molecular details of the observed nanoscale trapping. Dependencies on the molecular structure of the lipids point to the distinct connectivity of the various lipids to initiate or assist cellular signaling events, but also outline strong differences to the characteristics of liquid-ordered and disordered phase separation in model membranes. STED-FCS is a highly sensitive and exceptional tool to study the membrane organization by introducing a new class of nanoscale biomolecular studies.


Subject(s)
Membrane Microdomains , Spectrometry, Fluorescence/methods , Diffusion , Fluorescent Dyes
12.
Biochim Biophys Acta ; 1818(7): 1777-84, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22450237

ABSTRACT

Several simplified membrane models featuring coexisting liquid disordered (Ld) and ordered (Lo) lipid phases have been developed to mimic the heterogeneous organization of cellular membranes, and thus, aid our understanding of the nature and functional role of ordered lipid-protein nanodomains, termed "rafts". In spite of their greatly reduced complexity, quantitative characterization of local lipid environments using model membranes is not trivial, and the parallels that can be drawn to cellular membranes are not always evident. Similarly, various fluorescently labeled lipid analogs have been used to study membrane organization and function in vitro, although the biological activity of these probes in relation to their native counterparts often remains uncharacterized. This is particularly true for raft-preferring lipids ("raft lipids", e.g. sphingolipids and sterols), whose domain preference is a strict function of their molecular architecture, and is thus susceptible to disruption by fluorescence labeling. Here, we analyze the phase partitioning of a multitude of fluorescent raft lipid analogs in synthetic Giant Unilamellar Vesicles (GUVs) and cell-derived Giant Plasma Membrane Vesicles (GPMVs). We observe complex partitioning behavior dependent on label size, polarity, charge and position, lipid headgroup, and membrane composition. Several of the raft lipid analogs partitioned into the ordered phase in GPMVs, in contrast to fully synthetic GUVs, in which most raft lipid analogs mis-partitioned to the disordered phase. This behavior correlates with the greatly enhanced order difference between coexisting phases in the synthetic system. In addition, not only partitioning, but also ligand binding of the lipids is perturbed upon labeling: while cholera toxin B binds unlabeled GM1 in the Lo phase, it binds fluorescently labeled GMI exclusively in the Ld phase. Fluorescence correlation spectroscopy (FCS) by stimulated emission depletion (STED) nanoscopy on intact cellular plasma membranes consistently reveals a constant level of confined diffusion for raft lipid analogs that vary greatly in their partitioning behavior, suggesting different physicochemical bases for these phenomena.


Subject(s)
Cell Membrane/chemistry , Lipid Bilayers/chemistry , Membrane Lipids/chemistry , Membrane Microdomains/chemistry , Cell Membrane/metabolism , Cholera Toxin/chemistry , Cholera Toxin/metabolism , Diffusion , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Ligands , Lipid Bilayers/metabolism , Membrane Lipids/metabolism , Membrane Microdomains/metabolism , Microscopy, Confocal , Nanotechnology , Protein Binding , Spectrometry, Fluorescence , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism
13.
J Lipid Res ; 51(7): 1747-60, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20179319

ABSTRACT

We examined the effect of Niemann-Pick disease type 2 (NPC2) protein and some late endosomal lipids [sphingomyelin, ceramide and bis(monoacylglycero)phosphate (BMP)] on cholesterol transfer and membrane fusion. Of all lipid-binding proteins tested, only NPC2 transferred cholesterol at a substantial rate, with no transfer of ceramide, GM3, galactosylceramide, sulfatide, phosphatidylethanolamine, or phosphatidylserine. Cholesterol transfer was greatly stimulated by BMP, little by ceramide, and strongly inhibited by sphingomyelin. Cholesterol and ceramide were also significantly transferred in the absence of protein. This spontaneous transfer of cholesterol was greatly enhanced by ceramide, slightly by BMP, and strongly inhibited by sphingomyelin. In our transfer assay, biotinylated donor liposomes were separated from fluorescent acceptor liposomes by streptavidin-coated magnetic beads. Thus, the loss of fluorescence indicated membrane fusion. Ceramide induced spontaneous fusion of lipid vesicles even at very low concentrations, while BMP and sphingomyelin did so at about 20 mol% and 10 mol% concentrations, respectively. In addition to transfer of cholesterol, NPC2 induced membrane fusion, although less than saposin-C. In this process, BMP and ceramide had a strong and mild stimulating effect, and sphingomyelin an inhibiting effect, respectively. Note that the effects of the lipids on cholesterol transfer mediated by NPC2 were similar to their effect on membrane fusion induced by NPC2 and saposin-C.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Endosomes/metabolism , Glycoproteins/metabolism , Membrane Fusion/physiology , Membrane Lipids/metabolism , Animals , Biological Transport/physiology , Carrier Proteins/genetics , Cattle , Ceramides/metabolism , Endosomes/ultrastructure , Glycoproteins/genetics , Humans , Hydrogen-Ion Concentration , Liposomes/chemistry , Liposomes/metabolism , Lysophospholipids/metabolism , Membrane Lipids/chemistry , Monoglycerides/metabolism , Niemann-Pick Diseases/metabolism , Niemann-Pick Diseases/physiopathology , Saposins/metabolism , Sphingomyelins/metabolism
14.
Nat Cell Biol ; 12(1): 11-8; sup pp 1-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023649

ABSTRACT

Incoming simian virus 40 (SV40) particles enter tight-fitting plasma membrane invaginations after binding to the carbohydrate moiety of GM1 gangliosides in the host cell plasma membrane through pentameric VP1 capsid proteins. This is followed by activation of cellular signalling pathways, endocytic internalization and transport of the virus via the endoplasmic reticulum to the nucleus. Here we show that the association of SV40 (as well as isolated pentameric VP1) with GM1 is itself sufficient to induce dramatic membrane curvature that leads to the formation of deep invaginations and tubules not only in the plasma membrane of cells, but also in giant unilamellar vesicles (GUVs). Unlike native GM1 molecules with long acyl chains, GM1 molecular species with short hydrocarbon chains failed to support such invagination, and endocytosis and infection did not occur. To conceptualize the experimental data, a physical model was derived based on energetic considerations. Taken together, our analysis indicates that SV40, other polyoma viruses and some bacterial toxins (Shiga and cholera) use glycosphingolipids and a common pentameric protein scaffold to induce plasma membrane curvature, thus directly promoting their endocytic uptake into cells.


Subject(s)
Endocytosis/physiology , G(M1) Ganglioside/chemistry , Simian virus 40/physiology , Animals , Caveolin 1/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , G(M1) Ganglioside/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , Receptors, Virus/physiology , Virus Replication
15.
J Biol Chem ; 284(14): 9372-81, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19224915

ABSTRACT

Arylsulfatase A (ASA) catalyzes the intralysosomal desulfation of 3-O-sulfogalactosylceramide (sulfatide) to galactosylceramide. The reaction requires saposin B (Sap B), a non-enzymatic proteinaceous cofactor which presents sulfatide to the catalytic site of ASA. The lack of either ASA or Sap B results in a block of sulfatide degradation, progressive intralysosomal accumulation of sulfatide, and the fatal lysosomal storage disease metachromatic leukodystrophy. We studied the coupled Sap B-ASA reaction in vitro using detergent-free micellar and liposomal assay systems and in vivo using cell culture models of metachromatic leukodystrophy. Under in vitro conditions, the reaction had a narrow pH optimum around pH 4.3 and was inhibited by mono- and divalent cations, phosphate and sulfite. Bis(monoacylglycero) phosphate and phosphatidic acid were activators of the reaction, underscoring a significant role of acidic phosphoglycerolipids in sphingolipid degradation. Desulfation was negligible when Sap B was substituted by Sap A, C, or D. Up to a molar ratio between Sap B and sulfatide of 1:5, an elevation of Sap B concentrations caused a sharp increase of sulfatide hydrolysis, indicating the requirement of unexpected high Sap B levels for maximum turnover. Feeding of ASA-deficient, sulfatide-storing primary mouse kidney cells with ASA caused partial clearance of sulfatide. Co-feeding of Sap B or its precursor prosaposin resulted in the lysosomal uptake of the cofactor but did not promote ASA-catalyzed sulfatide hydrolysis. This suggests that Sap B is not a limiting factor of the coupled Sap B-ASA reaction in mouse kidney cells even if sulfatide has accumulated to unphysiologically high levels.


Subject(s)
Cerebroside-Sulfatase/metabolism , Leukodystrophy, Metachromatic/enzymology , Models, Biological , Saposins/metabolism , Animals , Cells, Cultured , Cerebroside-Sulfatase/deficiency , Cerebroside-Sulfatase/genetics , Disulfides/metabolism , Enzyme Activation , Humans , Hydrolysis , Leukodystrophy, Metachromatic/genetics , Lipid Metabolism , Liposomes , Mice , Mice, Knockout , Substrate Specificity , Swine
16.
Nature ; 457(7233): 1159-62, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19098897

ABSTRACT

Cholesterol-mediated lipid interactions are thought to have a functional role in many membrane-associated processes such as signalling events. Although several experiments indicate their existence, lipid nanodomains ('rafts') remain controversial owing to the lack of suitable detection techniques in living cells. The controversy is reflected in their putative size of 5-200 nm, spanning the range between the extent of a protein complex and the resolution limit of optical microscopy. Here we demonstrate the ability of stimulated emission depletion (STED) far-field fluorescence nanoscopy to detect single diffusing (lipid) molecules in nanosized areas in the plasma membrane of living cells. Tuning of the probed area to spot sizes approximately 70-fold below the diffraction barrier reveals that unlike phosphoglycerolipids, sphingolipids and glycosylphosphatidylinositol-anchored proteins are transiently ( approximately 10-20 ms) trapped in cholesterol-mediated molecular complexes dwelling within <20-nm diameter areas. The non-invasive optical recording of molecular time traces and fluctuation data in tunable nanoscale domains is a powerful new approach to study the dynamics of biomolecules in living cells.


Subject(s)
Cell Membrane/metabolism , Membrane Lipids/analysis , Membrane Lipids/metabolism , Microscopy, Fluorescence/methods , Nanotechnology/methods , Cell Line , Cell Membrane/chemistry , Cell Survival , Cholesterol/analysis , Cholesterol/metabolism , Diffusion , Epithelial Cells/cytology , Ethanolamines/analysis , Ethanolamines/metabolism , Glycosylphosphatidylinositols/metabolism , Sphingomyelins/analysis , Sphingomyelins/metabolism , Time Factors
17.
Glycobiology ; 15(12): 1302-11, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16079415

ABSTRACT

The ganglioside-activator protein is an essential cofactor for the lysosomal degradation of ganglioside GM2 (GM2) by beta-hexosaminidase A. It mediates the interaction between the water-soluble exohydrolase and its membrane-embedded glycolipid substrate at the lipid-water interphase. Mutations in the gene encoding this glycoprotein result in a fatal neurological storage disorder, the AB variant of GM2-gangliosidosis. In order to efficiently and sensitively probe the glycolipid binding and membrane activity of this cofactor, we synthesized two new fluorescent glycosphingolipid (GSL) probes, 2-NBD-GM1 and 2-NBD-GM2. Both compounds were synthesized in a convergent and multistep synthesis starting from the respective gangliosides isolated from natural sources. The added functionality of 2-aminogangliosides allowed us to introduce the chromophore into the region between the polar head group and the hydrophobic anchor of the lipid. Both fluorescent glycolipids exhibited an extremely low off-rate in model membranes and displayed very efficient resonance energy transfer to rhodamine-dioleoyl phosphoglycerol ethanolamine (rhodamine-PE) as acceptor. The binding to GM2-activator protein (GM2AP) and the degrading enzyme was shown to be unaltered compared to their natural analogues. A novel fluorescence-resonance energy transfer (FRET) assay was developed to monitor in real time the protein-mediated intervesicular transfer of these lipids from donor to acceptor liposomes. The data obtained indicate that this rapid and robust system presented here should serve as a valuable tool to probe quantitatively and comprehensively the membrane activity of GM2AP and other sphingolipid activator proteins and facilitate further structure-function studies aimed at delineating independently the lipid- and the enzyme-binding mode of these essential cofactors.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , G(M1) Ganglioside/chemistry , G(M2) Activator Protein/chemistry , G(M2) Ganglioside/chemistry , Animals , Brain/pathology , Carbohydrate Sequence , Catalysis , Cattle , Chromatography, Thin Layer , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Gangliosides/chemistry , Gangliosidoses , Glycolipids/chemistry , Glycoproteins/chemistry , Humans , Lipids/chemistry , Models, Chemical , Molecular Sequence Data , Mutation , Spectrometry, Fluorescence , Sphingolipid Activator Proteins/chemistry , Sphingolipids/chemistry , Structure-Activity Relationship , Tay-Sachs Disease/metabolism , Time Factors , beta-N-Acetylhexosaminidases/chemistry
18.
J Lipid Res ; 44(9): 1772-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12777464

ABSTRACT

We studied the metabolism of radioactively labeled safingol (l-threo-dihydrosphingosine) in primary cultured neurons, B104 neuroblastoma cells, and Swiss 3T3 fibroblasts, and compared it to that of its natural stereoisomer d-erythro-dihydrosphingosine. Both sphingoid bases are used as biosynthetic precursors for complex sphingolipids, albeit to different rates. Whereas a considerable amount of the natural sphingoid base is also directed to the catabolic pathway (20-66%, cell type dependent), only a minor amount of the nonnatural safingol is subjected to catabolic cleavage, most of it being N-acylated to the respective stereochemical variant of dihydroceramide. Interestingly, N-acylation of safingol to l-threo-dihydroceramide is less sensitive to fumonisin B1 than the formation of the natural d-erythro-dihydroceramide. In addition, safingol-derived l-threo-dihydroceramide, unlike its physiologic counterpart, is not desaturated. Most of it either accumulates in the cells (up to 50%) or is used as a biosynthetic precursor of the respective dihydrosphingomyelin (up to 45%). About 5% is, however, glucosylated and channeled into the glycosphingolipid biosynthetic pathway. Our results demonstrate that, despite its nonnatural stereochemistry, safingol is recognized and metabolized preferentially by enzymes of the sphingolipid biosynthetic pathway. Furthermore, our data suggest that the cytotoxic potential of safingol is reduced rather than enhanced via its metabolic conversion.


Subject(s)
Antineoplastic Agents/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Animals , Biotransformation , Catalysis , Cells, Cultured , Ceramides/biosynthesis , Ceramides/metabolism , Humans , Mice , Neurons/cytology , Neurons/metabolism , Spectrometry, Mass, Electrospray Ionization , Sphingolipids/biosynthesis , Sphingolipids/metabolism
19.
Neurochem Res ; 27(7-8): 717-27, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12374206

ABSTRACT

Much discussion has centered on the biochemical mechanism by which ceramide is produced and functions as a signalling molecule in cells. To identify proteins involved in ceramide signalling, we synthesized a radioactively labelled ceramide analogue equipped with a photosensitive group: N-(p-trifluoromethyl-diazirinyl)phenyl-ethyl-2-[35S]-2-thioacetyl-D-erythro-C18-sphingosine ([35S]-TDS-ceramide). This compound was then employed in photo-affinity labelling experiments in primary cultured cerebellar neurons. Due to the hydrophobic nature of the compound, most of the cell-associated radioactivity was recovered in the lipid fraction while only about 0.1% of radioactivity was photocoupled to proteins. In order to improve protein labelling the cytosolic fraction of rapidly growing human neuroblastoma cells (SH-SY5Y) was isolated and subjected to ceramide affinity chromatography prior to photo-affinity labelling. Following electrophoresis proteins photocoupled to ceramide were identified by MALDI mass spectrometry in combination with tryptic digestion and turned out to be either cytoskeletal or stress proteins that are highly abundant in cytosol and contain at least one hydrophobic domain.


Subject(s)
Ceramides/metabolism , Nerve Tissue Proteins/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Mice , Photoaffinity Labels
20.
Mol Biol Cell ; 13(9): 3355-68, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12221139

ABSTRACT

In LAMP-2-deficient mice autophagic vacuoles accumulate in many tissues, including liver, pancreas, muscle, and heart. Here we extend the phenotype analysis using cultured hepatocytes. In LAMP-2-deficient hepatocytes the half-life of both early and late autophagic vacuoles was prolonged as evaluated by quantitative electron microscopy. However, an endocytic tracer reached the autophagic vacuoles, indicating delivery of endo/lysosomal constituents to autophagic vacuoles. Enzyme activity measurements showed that the trafficking of some lysosomal enzymes to lysosomes was impaired. Immunoprecipitation of metabolically labeled cathepsin D indicated reduced intracellular retention and processing in the knockout cells. The steady-state level of 300-kDa mannose 6-phosphate receptor was slightly lower in LAMP-2-deficient hepatocytes, whereas that of 46-kDa mannose 6-phosphate receptor was decreased to 30% of controls due to a shorter half-life. Less receptor was found in the Golgi region and in vesicles and tubules surrounding multivesicular endosomes, suggesting impaired recycling from endosomes to the Golgi. More receptor was found in autophagic vacuoles, which may explain its shorter half-life. Our data indicate that in hepatocytes LAMP-2 deficiency either directly or indirectly leads to impaired recycling of 46-kDa mannose 6-phosphate receptors and partial mistargeting of a subset of lysosomal enzymes. Autophagic vacuoles may accumulate due to impaired capacity for lysosomal degradation.


Subject(s)
Antigens, CD/physiology , Hepatocytes/metabolism , Lysosomes/metabolism , Animals , Antigens, CD/metabolism , Blotting, Western , Cathepsin D/metabolism , Cell Membrane , Cells, Cultured , Endocytosis , Endosomes , Genotype , Immunohistochemistry , Lipid Metabolism , Lysosomal Membrane Proteins , Mice , Microscopy, Electron , Microscopy, Fluorescence , Phenotype , Precipitin Tests , Protein Binding , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...