Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 241
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34948386

ABSTRACT

The recent identification of plasma membrane (Ca2+)-ATPase (PMCA)-Neuroplastin (Np) complexes has renewed attention on cell regulation of cytosolic calcium extrusion, which is of particular relevance in neurons. Here, we tested the hypothesis that PMCA-Neuroplastin complexes exist in specific ganglioside-containing rafts, which could affect calcium homeostasis. We analyzed the abundance of all four PMCA paralogs (PMCA1-4) and Neuroplastin isoforms (Np65 and Np55) in lipid rafts and bulk membrane fractions from GM2/GD2 synthase-deficient mouse brains. In these fractions, we found altered distribution of Np65/Np55 and selected PMCA isoforms, namely PMCA1 and 2. Cell surface staining and confocal microscopy identified GM1 as the main complex ganglioside co-localizing with Neuroplastin in cultured hippocampal neurons. Furthermore, blocking GM1 with a specific antibody resulted in delayed calcium restoration of electrically evoked calcium transients in the soma of hippocampal neurons. The content and composition of all ganglioside species were unchanged in Neuroplastin-deficient mouse brains. Therefore, we conclude that altered composition or disorganization of ganglioside-containing rafts results in changed regulation of calcium signals in neurons. We propose that GM1 could be a key sphingolipid for ensuring proper location of the PMCA-Neuroplastin complexes into rafts in order to participate in the regulation of neuronal calcium homeostasis.


Subject(s)
G(M1) Ganglioside/metabolism , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Brain/metabolism , Cells, Cultured , G(M1) Ganglioside/analysis , Male , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Neurons/metabolism , Plasma Membrane Calcium-Transporting ATPases/analysis
2.
Histochem Cell Biol ; 154(5): 565-578, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33079236

ABSTRACT

Lysosomal storage diseases are the most common cause of neurodegeneration in children. They are characterised at the cellular level by the accumulation of storage material within lysosomes. There are very limited therapeutic options, and the search for novel therapies has been hampered as few good small animal models are available. Here, we describe the use of light sheet microscopy to assess lipid storage in drug and morpholino induced zebrafish models of two diseases of cholesterol homeostasis with lysosomal dysfunction: First, Niemann-Pick type C disease (NPC), caused by mutations in the lysosomal transmembrane protein NPC1, characterised by intralysosomal accumulation of cholesterol and several other lipids. Second, Smith-Lemli-Opitz syndrome (SLOS), caused by mutations in 7-dehydrocholesterol reductase, which catalyses the last step of cholesterol biosynthesis and is characterised by intralysosomal accumulation of dietary cholesterol. This is the first description of a zebrafish SLOS model. We find that zebrafish accurately model lysosomal storage and disease-specific phenotypes in both diseases. Increased cholesterol and ganglioside GM1 were observed in sections taken from NPC model fish, and decreased cholesterol in SLOS model fish, but these are of limited value as resolution is poor, and accurate anatomical comparisons difficult. Using light sheet microscopy, we were able to observe lipid changes in much greater detail and identified an unexpected accumulation of ganglioside GM1 in SLOS model fish. Our data demonstrate, for the first time in zebrafish, the immense potential that light sheet microscopy has in aiding the resolution of studies involving lysosomal and lipid disorders.


Subject(s)
Cholesterol/analysis , Disease Models, Animal , G(M1) Ganglioside/analysis , Niemann-Pick Disease, Type C/diagnosis , Smith-Lemli-Opitz Syndrome/diagnosis , Zebrafish , Animals , Cholesterol/metabolism , G(M1) Ganglioside/metabolism , Lysosomes/metabolism , Microscopy, Fluorescence , Niemann-Pick Disease, Type C/metabolism , Smith-Lemli-Opitz Syndrome/metabolism
3.
Glycoconj J ; 37(3): 329-343, 2020 06.
Article in English | MEDLINE | ID: mdl-32198666

ABSTRACT

It has been recently reported by our group that GM1-oligosaccharide added to neuroblastoma cells or administered to mouse experimental model mimics the neurotrophic and neuroprotective properties of GM1 ganglioside. In addition to this, differently from GM1, GM1-oligosaccharide is not taken up by the cells, remaining solubilized into the extracellular environment interacting with cell surface proteins. Those characteristics make GM1-oligosaccharide a good tool to study the properties of the endogenous GM1, avoiding to interfere with the ganglioside natural metabolic pathway. In this study, we show that GM1-oligosaccharide administered to mice cerebellar granule neurons by interacting with cell surface induces TrkA-MAP kinase pathway activation enhancing neuron clustering, arborization and networking. Accordingly, in the presence of GM1-oligosaccharide, neurons show a higher phosphorylation rate of FAK and Src proteins, the intracellular key regulators of neuronal motility. Moreover, treated cells express increased level of specific neuronal markers, suggesting an advanced stage of maturation compared to controls. In parallel, we found that in the presence of GM1-oligosaccharide, neurons accelerate the expression of complex gangliosides and reduce the level of the simplest ones, displaying the typical ganglioside pattern of mature neurons. Our data confirms the specific role of GM1 in neuronal differentiation and maturation, determined by its oligosaccharide portion. GM1-oligosacchairide interaction with cell surface receptors triggers the activation of intracellular biochemical pathways responsible for neuronal migration, dendrites emission and axon growth.


Subject(s)
Cell Differentiation/drug effects , G(M1) Ganglioside/pharmacology , Gangliosides/metabolism , Neurons/drug effects , Animals , Cell Differentiation/physiology , Cell Movement/drug effects , Cells, Cultured , Cerebellum/cytology , Female , G(M1) Ganglioside/analysis , G(M1) Ganglioside/metabolism , Lipid Metabolism/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Proteins/genetics , Proteins/metabolism , Receptor, trkA/metabolism
4.
Proc Natl Acad Sci U S A ; 113(20): 5592-7, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27143722

ABSTRACT

The cancer stem cells (CSCs) of glioblastoma multiforme (GBM), a grade IV astrocytoma, have been enriched by the expressed marker CD133. However, recent studies have shown that CD133(-) cells also possess tumor-initiating potential. By analysis of gangliosides on various cells, we show that ganglioside D3 (GD3) is overexpressed on eight neurospheres and tumor cells; in combination with CD133, the sorted cells exhibit a higher expression of stemness genes and self-renewal potential; and as few as six cells will form neurospheres and 20-30 cells will grow tumor in mice. Furthermore, GD3 synthase (GD3S) is increased in neurospheres and human GBM tissues, but not in normal brain tissues, and suppression of GD3S results in decreased GBM stem cell (GSC)-associated properties. In addition, a GD3 antibody is shown to induce complement-dependent cytotoxicity against cells expressing GD3 and inhibition of GBM tumor growth in vivo. Our results demonstrate that GD3 and GD3S are highly expressed in GSCs, play a key role in glioblastoma tumorigenicity, and are potential therapeutic targets against GBM.


Subject(s)
Brain Neoplasms/pathology , Gangliosides/physiology , Glioblastoma/pathology , Neoplastic Stem Cells/chemistry , Sialyltransferases/physiology , AC133 Antigen/analysis , Animals , Cell Line, Tumor , G(M1) Ganglioside/analysis , Gangliosides/analysis , Glioblastoma/chemistry , Glioblastoma/etiology , Humans , Mice , Proto-Oncogene Proteins c-met/metabolism , Sialyltransferases/analysis
5.
Anal Chem ; 90(15): 8873-8880, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29972017

ABSTRACT

The use of exosomes for diagnostic and disease monitoring purposes is becoming particularly appealing in biomedical research because of the possibility to study directly in biological fluids some of the features related to the organs from which exosomes originate. A paradigmatic example are brain-derived exosomes that can be found in plasma and used as a direct read-out of the status of the central nervous system (CNS). Inspired by recent remarkable development of plasmonic biosensors, we have designed a surface plasmon resonance imaging (SPRi) assay that, taking advantage of the fact that exosome size perfectly fits within the surface plasmon wave depth, allows the detection of multiple exosome subpopulations of neural origin directly in blood. By use of an array of antibodies, exosomes derived from neurons and oligodendrocytes were isolated and detected with good sensitivity. Subsequently, by injecting a second antibody on the immobilized vesicles, we were able to quantify the amount of CD81 and GM1, membrane components of exosomes, on each subpopulation. In this way, we have been able to demonstrate that they are not homogeneously expressed but exhibit a variable abundance according to the exosome cellular origin. These results confirm the extreme variability of exosome composition and demonstrate how SPRi can provide an effective tool for their characterization. Besides, our work paves the road toward more precise clinical studies on the use of exosomes as potential biomarkers of neurodegenerative diseases.


Subject(s)
Brain/cytology , Exosomes/chemistry , Neurons/chemistry , Oligodendroglia/chemistry , Plasma/chemistry , Surface Plasmon Resonance/methods , Adult , Antibodies, Immobilized/chemistry , Female , G(M1) Ganglioside/analysis , Humans , Male , Tetraspanin 28/analysis
6.
Cell Physiol Biochem ; 51(4): 1544-1565, 2018.
Article in English | MEDLINE | ID: mdl-30497064

ABSTRACT

BACKGROUND/AIMS: Red blood cells (RBC) have been shown to exhibit stable submicrometric lipid domains enriched in cholesterol (chol), sphingomyelin (SM), phosphatidylcholine (PC) or ganglioside GM1, which represent the four main lipid classes of their outer plasma membrane leaflet. However, whether those lipid domains co-exist at the RBC surface or are spatially related and whether and how they are subjected to reorganization upon RBC deformation are not known. METHODS: Using fluorescence and/or confocal microscopy and well-validated probes, we compared these four lipid-enriched domains for their abundance, curvature association, lipid order, temperature dependence, spatial dissociation and sensitivity to RBC mechanical stimulation. RESULTS: Our data suggest that three populations of lipid domains with decreasing abundance coexist at the RBC surface: (i) chol-enriched ones, associated with RBC high curvature areas; (ii) GM1/PC/chol-enriched ones, present in low curvature areas; and (iii) SM/PC/chol-enriched ones, also found in low curvature areas. Whereas chol-enriched domains gather in increased curvature areas upon RBC deformation, low curvature-associated lipid domains increase in abundance either upon calcium influx during RBC deformation (GM1/PC/chol-enriched domains) or upon secondary calcium efflux during RBC shape restoration (SM/PC/chol-enriched domains). Hence, abrogation of these two domain populations is accompanied by a strong impairment of the intracellular calcium balance. CONCLUSION: Lipid domains could contribute to calcium influx and efflux by controlling the membrane distribution and/or the activity of the mechano-activated ion channel Piezo1 and the calcium pump PMCA. Whether this results from lipid domain biophysical properties, the strength of their anchorage to the underlying cytoskeleton and/or their correspondence with inner plasma membrane leaflet lipids remains to be demonstrated.


Subject(s)
Cholesterol/analysis , Erythrocyte Membrane/metabolism , Erythrocytes/cytology , G(M1) Ganglioside/analysis , Membrane Microdomains/metabolism , Phosphatidylcholines/analysis , Biomechanical Phenomena , Cell Shape , Cholesterol/metabolism , Erythrocyte Membrane/chemistry , Erythrocyte Membrane/ultrastructure , Erythrocytes/chemistry , Erythrocytes/metabolism , Erythrocytes/ultrastructure , G(M1) Ganglioside/metabolism , Humans , Ion Channels/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/ultrastructure , Phosphatidylcholines/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism
7.
Acta Odontol Scand ; 72(8): 694-700, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24655314

ABSTRACT

OBJECTIVE: To determine and compare the presence and in situ localization of the glycosphingolipid ganglioside GM1 in human salivary glands using the biomarkers for GM1: cholera toxin and antibodies against GM1. MATERIALS AND METHODS: Immunohistochemical analyses were performed on sections of adult human submandibular, parotid and palatinal glands using cholera toxin sub-unit B and two polyclonal antibodies against ganglioside GM1 as biomarkers. RESULTS: Immunofluorescence microscopy showed that the toxin and antibodies were co-localized in some acini but not in others. The cholera toxin mainly reacted with the cell membranes of the mucous acini in the submandibular gland, while incubation with the antibody against GM1 gave rise to a staining of the cytoplasm. The cytoplasm in some secretory acinar cells in the parotid gland was stained by the cholera toxin, whereas only small spots on the plasma membranes reacted with anti-GM1. The plasma membranes in the parotid excretory ducts appeared to react to anti-GM1, but not to cholera toxin. CONCLUSIONS: Cholera toxin induces the expression of ion channels and carriers in the small intestine and increases the production of secretory mucins. Although their mutual immunohistochemical localization may differ, both cholera toxin and ganglioside GM1 are present in the mucin-producing acini from salivary glands. This could point to a relationship between ganglioside expression and production of salivary mucins.


Subject(s)
Cholera Toxin , G(M1) Ganglioside/analysis , Salivary Glands/chemistry , Adult , Antibodies , Biomarkers , Cadaver , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cytoplasm/chemistry , Cytoplasm/ultrastructure , Humans , Immunohistochemistry , Microscopy, Fluorescence , Mucins/chemistry , Parotid Gland/chemistry , Parotid Gland/cytology , Salivary Ducts/chemistry , Salivary Ducts/cytology , Salivary Glands/cytology , Salivary Glands, Minor/chemistry , Salivary Glands, Minor/cytology , Serous Membrane/chemistry , Serous Membrane/cytology , Submandibular Gland/chemistry , Submandibular Gland/cytology
8.
Biochim Biophys Acta ; 1820(9): 1437-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22206893

ABSTRACT

BACKGROUND: Mono-, di- and trisialo gangliosides are major glycosphingolipids in the brain of higher vertebrates involved in lipid raft assembly. In contrast, the fish brain is abundant in polisialo-gangliosides, whose function is implicated in the modulation of repulsive and attractive intercellular interactions during embryonic development and a temperature adaptation process. The histological distribution of gangliosides is usually studied in rodent and mammalian brains, but to date it has not been described in the case of fish brain. METHODS: Gangliosides were extracted from adult brains of trout, carp and zebrafish and separated by TLC. High-affinity anti-ganglioside (GM1, GD1a, GD1b, GT1b) IgG antibodies were used for immunohistochemistry. RESULTS: In trout and carp brains GM1 and GT1b are expressed in the same neuronal cell bodies from the telencephalon to the spinal cord. In zebrafish brain GM1 was not detected, whereas GT1b is a general neuropil staining. GD1a is specific for unmyelinated parallel fibers in carp and zebrafish brains as well as parallel fibers in the molecular layer of all cerebellar divisions. In trout brain GD1b is found in parallel fibers of the cerebellum, but not in the tectum mesencephali. GD1b is expressed in zebrafish neuronal cell bodies. CONCLUSIONS: Each studied species has a different expression of complex gangliosides. GT1b is widely present, whereas GD1a and GD1b appear in a specific group of unmyelinated fibers and could be used as their specific marker. GENERAL SIGNIFICANCE: This is the first report on mono-, di- and trisialo ganglioside (GM1, GD1a, GD1b and GT1b) distribution in the brain of adult Actinopterygian fishes. This article is part of a Special Issue entitled Glycoproteomics.


Subject(s)
Brain/metabolism , Fishes/metabolism , Gangliosides/metabolism , Animals , Antibody Affinity , Brain Chemistry , Carps/metabolism , G(M1) Ganglioside/analysis , G(M1) Ganglioside/isolation & purification , G(M1) Ganglioside/metabolism , Gangliosides/analysis , Gangliosides/immunology , Gangliosides/isolation & purification , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Tissue Distribution , Trout/metabolism , Zebrafish/metabolism
9.
Carbohydr Polym ; 312: 120795, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37059535

ABSTRACT

Glycosphingolipids (GSLs) in human milk regulate the immune system, support intestinal maturation, and prevent gut pathogens. The structural complexity and low abundance of GSLs limits their systematic analysis. Here, we coupled the use of monosialoganglioside 1-2-amino-N-(2-aminoethyl) benzamide (GM1-AEAB) derivatives as internal standards with HILIC-MS/MS to qualitatively and quantitatively compare GSLs in human, bovine, and goat milk. One neutral glycosphingolipid (GB) and 33 gangliosides were found in human milk, of which 22 were newly detected and three were fucosylated. Five GB and 26 gangliosides were identified in bovine milk, of which 21 were newly discovered. Four GB and 33 gangliosides were detected in goat milk, 23 of them newly reported. GM1 was the main GSL in human milk; whereas disialoganglioside 3 (GD3) and monosialogangloside 3 (GM3) were dominant in bovine and goat milk, respectively; N-acetylneuraminic acid (Neu5Ac) was detected in >88 % of GSLs in bovine and goat milk. N-hydroxyacetylneuraminic acid (Neu5Gc)-modified GSLs were 3.5 times more abundant in goat than in bovine milk; whereas GSLs modified with both Neu5Ac and Neu5Gc were 3 times more abundant in bovine than in goat milk. Given the health benefits of different GSLs, these results will facilitate the development of custom-designed human milk-based infant formula.


Subject(s)
Glycosphingolipids , Tandem Mass Spectrometry , Humans , Animals , Glycosphingolipids/chemistry , G(M1) Ganglioside/analysis , Gangliosides/analysis , Gangliosides/chemistry , Milk, Human/chemistry , Goats
10.
Glycobiology ; 22(12): 1721-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22833314

ABSTRACT

A 6-cm fresh proximal ileum surgical specimen from a blood group A(1)Le(a-b+) secretor individual was used for stepwise isolation of epithelial cells from villus tip to crypt bottom by gentle washing with ethylenediaminetetraacetic acid-containing buffer. Acid and non-acid sphingolipids were prepared from the epithelial cell fractions and the non-epithelial intestinal residue. Molecular information on the sphingolipid composition was obtained without further isolation of individual species by applying thin-layer chromatography using chemical and biological (monoclonal antibodies, cholera toxin, Escherichia coli) detection reagents, mass spectrometry and proton NMR spectroscopy of derivatized glycolipids. In this way, the structure of major and minor saccharides, ceramide components and their relative amounts were obtained. Epithelial cells and non-epithelial residue were distinctly different in their sphingolipid composition. Sphingomyelin was the major single component in both compartments. Characteristic for epithelial cells was the dominance of monoglycosylceramides, sulphatides and blood group fucolipids (mainly Le(b) hexaglycosylceramides and ALe(b) heptaglycosylceramides). The non-epithelial residue had about five times less glycolipids mainly mono-, di-, tri- and tetra-glycosylceramides and gangliosides, including the GM1 ganglioside. The ceramides were more hydroxylated (1-2 additional hydroxyls) in epithelial cell glycolipids compared with the non-epithelial residue. Combined with a separate detailed study on the glycoproteins of the same epithelial cell preparation, this human intestinal sample is the only epithelial cell preparation where both protein- and lipid-linked saccharides are characterized in detail.


Subject(s)
Ileum/chemistry , Intestinal Mucosa/chemistry , Sphingomyelins/analysis , Ceramides/analysis , Epithelial Cells/chemistry , Female , G(M1) Ganglioside/analysis , Glycolipids/analysis , Humans , Ileum/cytology , Intestinal Mucosa/cytology , Microvilli/chemistry , Middle Aged
11.
J Muscle Res Cell Motil ; 33(5): 341-50, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22968393

ABSTRACT

The neurotrophic factor-like activity of monosialoganglioside (GM1) has been shown to activate tyrosine kinase receptors (Trk). Targets of neuronal innervation play a vital role in regulating the survival and differentiation of innervating neurotrophin-responsive neurons. Both GM1 and target skeletal muscle (SKM) cells are essential for the maintenance of the function of neurons. However, much less is known about the effects of GM1 or/and target SKM cells on the expression of Trk receptors in dorsal root ganglion (DRG) neurons. Here we have tested what extent to the expression of TrkA, TrkB, and TrkC receptors in primary cultured of DRG neurons in absence or presence of GM1 or/and SKM cells. In this experiment, we found that: (1) GM1 promoted expression of TrkA and TrkB but not TrkC in primary cultured DRG neurons; (2) target SKM cells promoted expression of TrkC but not TrkA and TrkB in neuromuscular cocultures without GM1 treatment; and (3) GM1 and target SKM cells had additional effects on expression of these three Trk receptors. The results of the present study offered new clues for a better understanding of the association of GM1 and target SKM on the expression of Trk receptors.


Subject(s)
G(M1) Ganglioside/pharmacology , Ganglia, Spinal/enzymology , Muscle, Skeletal/physiology , Neurons/enzymology , Receptor, trkA/biosynthesis , Receptor, trkB/biosynthesis , Receptor, trkC/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , G(M1) Ganglioside/analysis , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
12.
Hum Mol Genet ; 17(11): 1556-68, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18270209

ABSTRACT

Mammalian sialidase Neu4, ubiquitously expressed in human tissues, is located in the lysosomal and mitochondrial lumen and has broad substrate specificity against sialylated glycoconjugates. To investigate whether Neu4 is involved in ganglioside catabolism, we transfected beta-hexosaminidase-deficient neuroglia cells from a Tay-Sachs patient with a Neu4-expressing plasmid and demonstrated the correction of storage due to the clearance of accumulated GM2 ganglioside. To further clarify the biological role of Neu4, we have generated a stable loss-of-function phenotype in cultured HeLa cells and in mice with targeted disruption of the Neu4 gene. The silenced HeLa cells showed reduced activity against gangliosides and had large heterogeneous lysosomes containing lamellar structures. Neu4(-/-) mice were viable, fertile and lacked gross morphological abnormalities, but showed a marked vacuolization and lysosomal storage in lung and spleen cells. Lysosomal storage bodies were also present in cultured macrophages preloaded with gangliosides. Thin-layer chromatography showed increased relative level of GD1a ganglioside and a markedly decreased level of GM1 ganglioside in brain of Neu4(-/-) mice suggesting that Neu4 may be important for desialylation of brain gangliosides and consistent with the in situ hybridization data. Increased levels of cholesterol, ceramide and polyunsaturated fatty acids were also detected in the lungs and spleen of Neu4(-/-) mice by high-resolution NMR spectroscopy. Together, our data suggest that Neu4 is a functional component of the ganglioside-metabolizing system, contributing to the postnatal development of the brain and other vital organs.


Subject(s)
Gangliosides/metabolism , Lysosomes/metabolism , Neuraminidase/genetics , Neuraminidase/physiology , Animals , Behavior, Animal , Brain/enzymology , Brain/physiology , Brain/ultrastructure , Catalysis , G(M1) Ganglioside/analysis , G(M1) Ganglioside/metabolism , G(M2) Ganglioside/analysis , G(M2) Ganglioside/metabolism , Gangliosides/analysis , HeLa Cells , Humans , Lung/enzymology , Lung/ultrastructure , Mice , Mice, Knockout , Neuraminidase/metabolism , RNA Interference , Spleen/enzymology , Spleen/ultrastructure , Tissue Distribution , beta-N-Acetylhexosaminidases/genetics
13.
Glycoconj J ; 26(8): 929-33, 2009 Nov.
Article in English | MEDLINE | ID: mdl-18415015

ABSTRACT

After the discovery of glycosphingolipid (GSL) glycan detaching enzymes, Rhodococcal endoglycoceramidase (EGCase) and leech ceramide glycanase (CGase), the method for enzymatically releasing glycans from GSLs has become the method of choice for preparing intact ceramide-free oligosaccharide chains from GSLs. This paper describes (1) the preparation of the intact oligosaccharides from GM1 (II(3)NeuAcGgOse(4)Cer) and GbOse(4)Cer as examples to show the use of CGase to prepare intact glycan chains from GSLs, and (2) the specificity and detergent requirements of Rhodococcal EGCases for the release of glycan chains from different GSLs.


Subject(s)
Glycosphingolipids/metabolism , Oligosaccharides/analysis , Animals , Carbohydrate Sequence , Chromatography, Thin Layer , Detergents/pharmacology , G(M1) Ganglioside/analysis , Globosides/analysis , Globosides/chemistry , Glycoside Hydrolases/metabolism , Glycosphingolipids/chemistry , Hydrolysis/drug effects , Leeches , Molecular Sequence Data , Oligosaccharides/chemistry , Rhodococcus/enzymology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity/drug effects
14.
Sci Rep ; 9(1): 17684, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31776384

ABSTRACT

GM1 ganglioside, a monosialic glycosphingolipid and a crucial component of plasma membranes, accumulates in lysosomal storage disorders, primarily in GM1 gangliosidosis. The development of biomarkers for simplifying diagnosis, monitoring disease progression and evaluating drug therapies is an important objective in research into neurodegenerative lysosomal disorders. With this in mind, we established fluorescent imaging and flow-cytometric methods to track changes in GM1 ganglioside levels in patients with GM1 gangliosidosis and in control cells. We also evaluated GM1 ganglioside content in patients' cells treated with the commercially available Miglustat, a substrate inhibitor potentially suitable for the treatment of late-onset GM1 gangliosidosis. The flow-cytometric method proved to be sensitive, unbiased, and rapid in determining variations in GM1 ganglioside content in human lymphocytes derived from small amounts of fresh blood. We detected a strong correlation between GM1 ganglioside content and the clinical severity of GM1 gangliosidosis. We confirm the ability of Miglustat to act as a substrate reduction agent in the patients' treated cells. As well as being suitable for diagnosing and managing patients with GM1 gangliosidosis this method could be useful in the diagnosis and management of other lysosomal diseases, such as galactosialidosis, Type C Niemann-Pick, and any other disease with pathologic variations of GM1 ganglioside.


Subject(s)
G(M1) Ganglioside/analysis , G(M1) Ganglioside/metabolism , Gangliosidosis, GM1/classification , Gangliosidosis, GM1/diagnosis , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Biomarkers/analysis , Biomarkers/metabolism , Cells, Cultured , Disease Progression , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry/methods , Gangliosidosis, GM1/blood , Gangliosidosis, GM1/pathology , Glycoside Hydrolase Inhibitors/pharmacology , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Optical Imaging/methods , Phenotype , Severity of Illness Index
15.
Anal Chem ; 80(19): 7631-4, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18717573

ABSTRACT

We report a system that allows the simultaneous aspiration of one or more cells into each of five capillaries for electrophoresis analysis. A glass wafer was etched to create an array of 1-nL wells. The glass was treated with poly(2-hydroxyethyl methacrylate) to control cell adherence. A suspension of formalin-fixed cells was placed on the surface, and cells were allowed to settle. The concentration of cells and the settling time were chosen so that there was, on average, one cell per well. Next, an array of five capillaries was placed so that the tip of each capillary was in contact with a single well. A pulse of vacuum was applied to the distal end of the capillaries to aspirate the content of each well into a capillary. Next, the tips of the capillaries were placed in running buffer and potential was applied. The cells lysed upon contact with the running buffer, and fluorescent components were detected at the distal end of the capillaries by laser-induced fluorescence. The electrophoretic separation efficiency was outstanding, generating over 750,000 theoretical plates (1,800,000 plates/m). In this example, AtT-20 cells were used that had been treated with TMR-G(M1). The cells were allowed to metabolize this substrate into a series of products before the cells were fixed. The number of cells found in each well was estimated visually under the microscope and was described by a Poisson distribution with mean of 0.98 cell/well. This system provides an approach to high-throughput chemical cytometry.


Subject(s)
Electrophoresis, Capillary/methods , Flow Cytometry/methods , Gangliosides/metabolism , Animals , Cell Line , Ceramides/analysis , Ceramides/metabolism , Electrophoresis, Capillary/instrumentation , Flow Cytometry/instrumentation , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , G(M1) Ganglioside/analysis , G(M1) Ganglioside/metabolism , G(M2) Ganglioside/analysis , G(M2) Ganglioside/metabolism , Gangliosides/analysis , Nanotechnology/instrumentation , Nanotechnology/methods , Rhodamines/chemistry , Rhodamines/metabolism
16.
Cytometry A ; 73(10): 918-25, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18727060

ABSTRACT

Membrane rafts are involved in a broad variety of biological processes. Their protein composition under growth factor stimulation, anti-inflammatory or proinflammatory microenvironments, or in the course of pathogenic infections still remains to be determined. However, current techniques aimed at the identification of particular proteins on membrane rafts are not devoid of pitfalls. Membrane rafts were obtained by detergent-free based differential centrifugation from Jurkat T cells and J774 macrophages. Membrane rafts were labeled with fluorochrome-labeled antibodies directed against different cell membrane molecules, and with fluorochrome-labeled cholera toxin B that targets GM1 and analyzed by flow cytometry. CD3, CD11a, and GM1 were shown to be differentially expressed on Jurkat T cell-derived membrane rafts, indicating heterogeneity in membrane rafts composition. On the other hand, it was shown in J774 cell-derived membrane rafts that most but not all CD14 is present in the GM1-containing membrane fragments, thus confirming the heterogeneity of membrane rafts composition in other cell lines. The method described here allows the fluorometric assessment of the relative expression of more than one membrane raft component at a time, and at a single vesicle level in a fast and sensitive manner. This method seems to be a suitable approach to evaluate the molecular heterogeneity in membrane rafts composition.


Subject(s)
Flow Cytometry/methods , Membrane Microdomains/chemistry , Animals , CD11a Antigen/analysis , CD3 Complex/analysis , Cell Fractionation , Cell Separation/methods , G(M1) Ganglioside/analysis , Humans , Jurkat Cells , Membrane Microdomains/ultrastructure , Mice , Microscopy, Electron
17.
J Microsc ; 232(2): 225-34, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19017221

ABSTRACT

The localization of asialo-GM1 in ordered membrane raft domains in HeLa cells has been examined using a combination of membrane fractionation and fluorescence imaging. The glycolipid is enriched in Triton X-100 insoluble membrane fractions that contain high concentrations of cholesterol and caveolin-1 but is also found in detergent soluble membrane fractions. Near-field fluorescence microscopy shows that a fraction of the asialo-GM1 is localized in small nanoscale clusters that have an upper limit for the average diameter of approximately 90 nm and are partially colocalized with caveolae membrane domains. In addition to clusters, a diffuse, non-clustered population of asialo-GM1 is observed and is hypothesized to correspond to glycolipid isolated in detergent soluble membrane fractions.


Subject(s)
Cell Membrane/chemistry , G(M1) Ganglioside/analysis , Microscopy, Fluorescence/methods
18.
Biochem J ; 407(1): 31-40, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17608628

ABSTRACT

In order to search for novel components of lipid membrane microdomains involved in neural signalling pathways, mAbs (monoclonal antibodies) were raised against the detergent-insoluble membrane fraction of PC12 (pheochromocytoma) cells. Among the 22 hybrid clones, mAb PR#1 specifically detected a fucoganglioside Fuc(Gal)-GM1 [a-fucosyl(a-galactosyl)-GM1], a ganglioside homologous with GM1a (II3NeuAc,GgOse4Cer), as a novel member of microdomain components with biological functions. In the presence of mAb PR#1 in the culture medium, the outgrowth of neurites was induced in PC12 cells in a dose-dependent manner, with no effects on cell proliferation, suggesting that Fuc(Gal)-GM1 is preferentially involved in PC12 cell neuritogenesis. Effects through Fuc(Gal)-GM1 were different from those through GM1a during differentiation, e.g. under PR#1 treatment on Fuc(Gal)-GM1, round cell bodies with thinner cell processes were induced, whereas treatment with CTB (cholera toxin B subunit), a specific probe for GM1a, produced flattened cell bodies with thicker pro-cesses. Molecular analysis demonstrated that the PR#1-Fuc(Gal)-GM1 pathway was associated with Fyn and Yes of the Src family of kinases, although Src itself was not involved. No association was found with TrkA (tropomyosin receptor kinase A) and ERKs (extracellular-signal-regulated kinases), which are responsible for GM1a-induced differentiation. From these findings, it is suggested that a fucoganglioside Fuc(Gal)-GM1 provides a functional platform distinct from that of GM1a for signal transduction in PC12 cell differentiation.


Subject(s)
G(M1) Ganglioside/analogs & derivatives , Gangliosides/metabolism , Membrane Lipids/metabolism , Neurites/metabolism , Animals , Antibodies, Monoclonal/metabolism , G(M1) Ganglioside/analysis , G(M1) Ganglioside/metabolism , Immunohistochemistry , Membrane Lipids/chemistry , Mice , Mice, Inbred BALB C , Neurites/ultrastructure , PC12 Cells , Rats , Signal Transduction , Spectrometry, Mass, Fast Atom Bombardment , Time Factors , src-Family Kinases/metabolism
19.
Int J Biol Macromol ; 114: 830-835, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29626601

ABSTRACT

Mounting evidence indicates soluble Aß42 oligomers as the most toxic species causing neuronal death which leads to the onset and progression of Alzheimer disease (AD). Recently, it has been found that neurotoxic Aß42 oligomers grow from monomeric species or arise following secondary nucleation by preformed mature fibrils. Thus, the use of natural compounds such as polyphenols to hinder the growth or to remodel Aß42 fibrils is one of the most promising strategies for AD treatment. In our previous study, we showed that 1, 2, 4-trihydroxynaphthalene-2-O-ß-d-glucopyranoside (THNG) inhibits Aß42 aggregation during the early steps of the aggregation process, inhibits its conformational change to a ß-sheet-rich structure, decreases its polymerization, inhibits its fibrillogenisis and reduces oxidative stress and aggregate cytotoxicity. Here, we used different spectroscopic and cell culture methods to check the effect of THNG on fibrils disaggregation. We showed that THNG binds to mature Aß42 fibrils, rearrange their secondary structure, and remodels them into non-amyloid, less toxic, species by inhibiting their interaction with the plasma membrane. Our findings reveal that THNG is a good agent to remodel amyloid fibrils and could be used as a starting molecular scaffold to design new anti-AD drugs.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/drug effects , Lawsonia Plant/chemistry , Peptide Fragments/chemistry , Amyloid/toxicity , Amyloid beta-Peptides/toxicity , Cell Line, Tumor , Circular Dichroism , Drug Evaluation, Preclinical , Fluorescence Resonance Energy Transfer , G(M1) Ganglioside/analysis , Humans , Membrane Microdomains , Microscopy, Electron , Molecular Structure , Neuroblastoma/pathology , Peptide Fragments/toxicity , Plant Leaves/chemistry
20.
J Clin Invest ; 113(8): 1176-87, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15085197

ABSTRACT

Systemic lupus erythematosus (SLE) is characterized by abnormalities in T lymphocyte receptor-mediated signal transduction pathways. Our previous studies have established that lymphocyte-specific protein tyrosine kinase (LCK) is reduced in T lymphocytes from patients with SLE and that this reduction is associated with disease activity and parallels an increase in LCK ubiquitination independent of T cell activation. This study investigated the expression of molecules that regulate LCK homeostasis, such as CD45, C-terminal Src kinase (CSK), and c-Cbl, in lipid raft domains from SLE T cells and investigated the localization of these proteins during T cell receptor (TCR) triggering. Our results indicate that the expression of raft-associated ganglioside, GM1, is increased in T cells from SLE patients and LCK may be differentially regulated due to an alteration in the association of CD45 with lipid raft domains. CD45 tyrosine phosphatase, which regulates LCK activity, was differentially expressed and its localization into lipid rafts was increased in T cells from patients with SLE. Furthermore, T cells allowed to "rest" in vitro showed a reversal of the changes in LCK, CD45, and GM1 expression. The results also revealed that alterations in the level of GM1 expression and lipid raft occupancy cannot be induced by serum factors from patients with SLE but indicated that cell-cell contact, activating aberrant proximal signaling pathways, may be important in influencing abnormalities in T cell signaling and, therefore, function in patients with SLE.


Subject(s)
G(M1) Ganglioside/analysis , Lupus Erythematosus, Systemic/immunology , Membrane Microdomains/physiology , Signal Transduction , T-Lymphocytes/physiology , Adolescent , Adult , Aged , Cholera Toxin/metabolism , Humans , Leukocyte Common Antigens/analysis , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/analysis , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL