g(m1)-ganglioside has been researched along with Glioma* in 22 studies
22 other study(ies) available for g(m1)-ganglioside and Glioma
Article | Year |
---|---|
Penetration of the blood-brain barrier and the anti-tumour effect of a novel PLGA-lysoGM1/DOX micelle drug delivery system.
Effective treatment of glioma and other central nervous system (CNS) diseases is hindered by the presence of the blood-brain barrier (BBB). A novel nano-delivery vehicle system composed of PLGA-lysoGM1/DOX micelles was developed to cross the BBB for CNS treatment. We have shown that doxorubicin (DOX) as a model drug encapsulated in PLGA-lysoGM1 micelles can achieve up to 3.8% loading efficiency and 61.6% encapsulation efficiency by the orthogonal test design. Our in vitro experiments demonstrated that PLGA-lysoGM1/DOX micelles had a slow and sustainable drug release under physiological conditions and exhibited a high cellular uptake through the macropinocytosis and the autophagy/lysosomal pathways. In vivo experimental studies in zebrafish and mice confirmed that PLGA-lysoGM1/DOX micelles could cross the BBB and be specifically accumulated in the brain. Moreover, an excellent anti-glioma effect was observed in intracranial glioma-bearing rats. Therefore, PLGA-lysoGM1/DOX micelles not only effectively can cross the BBB, but our results also suggest that they have great potential for anti-glioma therapy and other central nervous system diseases. Topics: Animals; Blood-Brain Barrier; Brain Neoplasms; Doxorubicin; Drug Delivery Systems; G(M1) Ganglioside; Glioma; Humans; Male; Mice; Micelles; Polylactic Acid-Polyglycolic Acid Copolymer; Zebrafish | 2020 |
Penetration of blood-brain barrier and antitumor activity and nerve repair in glioma by doxorubicin-loaded monosialoganglioside micelles system.
For the treatment of glioma and other central nervous system diseases, one of the biggest challenges is that most therapeutic drugs cannot be delivered to the brain tumor tissue due to the blood-brain barrier (BBB). The goal of this study was to construct a nanodelivery vehicle system with capabilities to overcome the BBB for central nervous system administration. Doxorubicin as a model drug encapsulated in ganglioside GM1 micelles was able to achieve up to 9.33% loading efficiency and 97.05% encapsulation efficiency by orthogonal experimental design. The in vitro study demonstrated a slow and sustainable drug release in physiological conditions. In the cellular uptake studies, mixed micelles could effectively transport into both human umbilical vein endothelial cells and C6 cells. Furthermore, biodistribution imaging of mice showed that the DiR/GM1 mixed micelles were accumulated sustainably and distributed centrally in the brain. Experiments on zebrafish confirmed that drug-loaded GM1 micelles can overcome the BBB and enter the brain. Among all the treatment groups, the median survival time of C6-bearing rats after administering DOX/GM1 micelles was significantly prolonged. In conclusion, the ganglioside nanomicelles developed in this work can not only penetrate BBB effectively but also repair nerves and kill tumor cells at the same time. Topics: Animals; Animals, Genetically Modified; Antibiotics, Antineoplastic; Blood-Brain Barrier; Brain Neoplasms; Doxorubicin; Drug Delivery Systems; G(M1) Ganglioside; Gangliosides; Glioma; Human Umbilical Vein Endothelial Cells; Humans; Male; Micelles; Nerve Regeneration; Rats, Wistar; Tissue Distribution; Zebrafish | 2017 |
C6 cells express a sodium-calcium exchanger/GM1 complex in the nuclear envelope but have no exchanger in the plasma membrane: comparison to astrocytes.
Previous work demonstrated the presence of an isoform of Na(+)/Ca(2+) exchanger in the nuclear envelope of neurons and NG108-15 cells that is tightly associated with GM1 ganglioside and potentiated by the latter. This contrasted with the Na(+)/Ca(2+) exchanger(s) in the plasma membrane, which were suggested to associate more loosely with GM1. To study these aspects of Na(+)/Ca(2+) exchanger expression in nonneuronal neural cells, we have examined nuclear and plasma membrane exchanger patterns in astrocytes and C6 cells, a glia-derived line. We find both cell types contain the tightly associated exchanger/GM1 complex in the nuclear envelope but, surprisingly, only astrocytes possess Na(+)/Ca(2+) exchanger activity in the plasma membrane. This is the first reported example of a cell (C6) with Na(+)/Ca(2+) exchangers in the nuclear envelope but not in the plasma membrane. RT-PCR established the presence of the NCX1 subtype in C6 cells and both NCX1 and NCX2 in astrocytes. Comparison was made with NG108-15 cells, which have Na(+)/Ca(2+) exchangers in both nuclear and plasma membranes, and Jurkat cells, which have no Na(+)/Ca(2+) exchanger in either membrane. Culturing of C6 cells in the presence dibutyryl-cAMP caused upregulation of a high molecular weight isoform of the exchanger together with GM1 in the nuclear envelope, resulting in significant elevation of Na(+)/Ca(2+) exchanger activity in the latter. Application of exogenous GM1 to nuclei from non-treated cells also potentiated exchanger activity, although to a lesser degree. The Na(+)/Ca(2+) exchanger/GM1 complex occurs in the inner membrane of the nuclear envelope, suggesting a functional role in transferring Ca(2+) between nucleoplasm and the envelope lumen. Topics: Animals; Astrocytes; Cell Line, Tumor; Cell Membrane; Cells, Cultured; Central Nervous System Neoplasms; G(M1) Ganglioside; Glioma; Neuroglia; Nuclear Envelope; Protein Isoforms; Rats; Sodium-Calcium Exchanger; Tissue Distribution | 2004 |
Vaccination with MCP-1 cDNA transfectant on human malignant glioma in nude mice induces migration of monocytes and NK cells to the tumor.
Recently, studies on vaccination with tumor cells genetically engineered to produce monocyte chemoattractant protein-1 (MCP-1) have provided some encouragement. These studies have shown infiltration of certain types of lymphocytes at the tumor site. However, natural killer (NK) cells have not yet been assessed. We obtained a human malignant glioma cell line producing human MCP-1 constitutively by transfection of MCP-1 cDNA. We then test the effect of vaccination with the MCP-1 transfectant on nude mice. Although vaccination with MCP-1 transfectant did not reduce the tumor in our study, it was associated with the infiltration of large numbers of NK cells and monocytes at the tumor site. The site of vaccination also showed large numbers of monocytes. NK cells were detected with anti-asialo GM1 antibody, and monocytes were detected immunohistochemically with F4/80. We assumed that infiltrating monocytes at the site of vaccination could promote the infiltration of monocytes and NK cells into the tumor site without T-cell mediated transduction because the host lacked T-cell function. Topics: Animals; Cancer Vaccines; Cell Movement; Chemokine CCL2; G(M1) Ganglioside; Glioma; Humans; Killer Cells, Natural; Male; Mice; Mice, Inbred BALB C; Mice, Nude; Monocytes; Transfection; Vaccination | 2001 |
Expression of the GM1-species, [NeuN]-GM1, in a case of human glioma.
Altered glycosylation is a common feature in tumors of various kind and particular interest has been focused on the expression of tumor-associated gangliosides. We have previously identified some human glioma-associated gangliosides and in this study yet another, not previously described, ganglioside has been isolated. The ganglioside was prepared from human glioma tissue taken at autopsy. The new ganglioside bound cholera-toxin B-subunit and its structure was confirmed by fast atom bombardment-mass spectrometry to be NeuN-GM1 (II3NeuNH2-GgOse4Cer). In the dissected tumor specimen, the concentration of NeuN-GM1 was 0.1 micromol/g wet weight and accounted for approximately 20% of the monosialoganglioside fraction. Normal human brain tissue specimens (n = 10) did not contain detectable (>0.5 nmol/g wet weight of tissue) amounts of NeuN-GM1, indicating that this ganglioside might be associated with human glioma. However, none of the 17 other tumour specimens reveal any detectable amounts of this ganglioside. In conclusion, NeuN GM1 is a glioma-associated ganglioside but its exceptional expression limits its relevance as a molecule involved in general tumor biology. Topics: Brain; Brain Neoplasms; Carbohydrate Sequence; Chromatography, Thin Layer; Enzyme-Linked Immunosorbent Assay; G(M1) Ganglioside; Glioma; Humans; Molecular Sequence Data; Spectrometry, Mass, Fast Atom Bombardment | 1999 |
GM1 activates the MAP kinase cascade through a novel wortmannin-sensitive step upstream from c-Raf.
Topics: Androstadienes; Calcium-Calmodulin-Dependent Protein Kinases; Enzyme Activation; G(M1) Ganglioside; Glioma; Humans; Phosphodiesterase Inhibitors; Phosphoinositide-3 Kinase Inhibitors; Proto-Oncogene Proteins c-raf; Signal Transduction; Tumor Cells, Cultured; Wortmannin | 1998 |
Ganglioside GM1 activates the mitogen-activated protein kinase Erk2 and p70 S6 kinase in U-1242 MG human glioma cells.
Gangliosides are implicated in the regulation of cellular proliferation as evidenced by differences in ganglioside composition associated with malignant transformation and density of cells in culture, as well as their inhibitory effects when added to cells growing in culture. Exogenously added gangliosides have a bimodal effect on proliferation in U-1242 MG glioma cells, inhibiting DNA synthesis in growing cells and stimulating it in quiescent cells. We investigated the mechanisms involved in stimulation of DNA synthesis using [3H]thymidine incorporation and immune complex kinase assays to identify responsible signal transduction pathways. Treatment of quiescent U-1242 MG cells with GM1 caused activation of the mitogen-activated protein (MAP) kinase isoform Erk2. Pretreatment with the specific MAP kinase kinase inhibitor PD98059 prevented the GM1-stimulated Erk2 activation and GM1-stimulated DNA synthesis. GM1 treatment stimulated another distinct signaling pathway leading to activation of p70 S6 kinase (p70s6k), and this was prevented by pretreatment with rapamycin. Rapamycin also inhibited GM1-stimulated DNA synthesis. Activation of both pathways and stimulation of DNA synthesis were inhibited by forskolin treatment; however, GM1 had no effect on cyclic AMP levels. Platelet-derived growth factor also activated both Erk2 and p70s6k but did not cause DNA synthesis, suggesting that GM1 may stimulate additional cascades, which also contribute to GM1-mediated DNA synthesis. Topics: Calcium-Calmodulin-Dependent Protein Kinases; Colforsin; Cyclic AMP; DNA; Enzyme Activation; Enzyme Inhibitors; Flavonoids; G(M1) Ganglioside; Glioma; Humans; Immunosuppressive Agents; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase Kinases; Mitogens; Platelet-Derived Growth Factor; Polyenes; Protein Kinase Inhibitors; Protein Serine-Threonine Kinases; Ribosomal Protein S6 Kinases; Signal Transduction; Sirolimus; Tumor Cells, Cultured | 1997 |
Opioid receptor and calcium channel regulation of adenylyl cyclase, modulated by GM1, in NG108-15 cells: competitive interactions.
GM1 ganglioside was previously shown to function as a specific regulator of excitatory opioid activity in dorsal root ganglion neurons and F11 hybrid cells, as seen in its facilitation of opioid-induced activation of adenylyl cyclase and its ability to dramatically reduce the threshold opioid concentration required to prolong the action potential duration. The elevated levels of GM1 resulting from chronic opioid exposure of F11 cells were postulated to cause the ensuing opioid excitatory supersensitivity. We now show that GM1 promotes opioid (DADLE)-induced activation of adenylyl cyclase in NG108-15 cells which possess the delta-type of receptor. In keeping with previous studies of other systems, this can be envisioned as conformational interaction of GM1 with the receptor that results in uncoupling of the receptor from Gi and facilitated coupling to Gs. This would also account for the observation that DADLE-induced attenuation of forskolin-stimulated adenylyl cyclase was reversed by GM1, provided the cells were not pretreated with pertussis toxin. When the cells were so pretreated, GM1 evoked an unexpected attenuation of forskolin-stimulated adenylyl cyclase attributed to GM1-promoted influx of calcium which was postulated to inhibit a calcium-sensitive form of adenylyl cyclase. This is concordant with several studies showing GM1 to be a potent modulator of calcium flux. Pertussis toxin in these experiments exerted dual effects, one being to promote interaction of the delta-opioid receptor with Gs through inactivation of Gi, and the other to enhance the GM1-promoted influx of calcium by inactivation of Go; the latter is postulated to function as constitutive inhibitor of the relevant calcium channel. NG108-15 cells thus provide an interesting example of competitive interaction between two GM1-regulated systems involving enhancement of both opioid receptor excitatory activity and calcium influx. Topics: Adenylate Cyclase Toxin; Adenylyl Cyclases; Aminopterin; Binding, Competitive; Calcium Channels; Cholera Toxin; Cyclic AMP; Enkephalin, Leucine-2-Alanine; Fluorescent Dyes; G(M1) Ganglioside; Glioma; Hybrid Cells; Hypoxanthine; Neuroblastoma; Pertussis Toxin; Receptors, Opioid; Thymidine; Tumor Cells, Cultured; Virulence Factors, Bordetella | 1997 |
Fuc-GM1 ganglioside mimics the receptor function of GM1 for cholera toxin.
The ability of Fuc-GM1 ganglioside to mimic the receptor function of GM1 for cholera toxin (CT) has been investigated. For this purpose, rat glioma C6 cultured cells were enriched with Fuc-GM1 and the responsiveness to CT was compared with that of cells enriched with GM1 ganglioside. Fuc-GM1 was taken up by cells as rapidly and to the same extent as GM1. When comparable amounts of ganglioside were associated, the cells enriched with Fuc-GM1 bound the same amount of 125I-CT as did cells enriched with GM1. Under conditions in which GM1- and Fuc-GM1-enriched cells bound comparable amounts of CT, the Fuc-GM1-treated cells accumulated virtually the same amount of cyclic AMP as did GM1-treated cells, and activation of adenylate cyclase was also similar. The lag time preceding the CT-induced cAMP accumulation was the same in Fuc-GM1- and GM1-enriched cells. High-sensitivity isothermal titration calorimetry (ITC) experiments showed that the association constants of CT with Fuc-GM1 or GM1 ganglioside were comparable (4 x 10(7) M-1 and 1.9 x 10(7) M-1, respectively, at 25 degrees C). Also, the association constants of the B-subunit pentamer with Fuc-GM1 or GM1 ganglioside were comparable (about 3 x 10(7) M-1 and 7 x 10(7) M-1, respectively, at 25 degrees C). Topics: Adenylyl Cyclases; Animals; Calorimetry; Cattle; Cell Survival; Cholera Toxin; Cyclic AMP; G(M1) Ganglioside; Glioma; Kinetics; Rats; Receptors, Cell Surface; Receptors, Immunologic; Swine; Tritium; Tumor Cells, Cultured | 1992 |
Intoxication of cultured cells by cholera toxin: evidence for different pathways when bound to ganglioside GM1 or neoganglioproteins.
We previously reported that when the oligosaccharide of ganglioside GM1 is covalently attached to cell surface proteins of GM1-deficient rat glioma C6 cells, the cells bind large amounts of cholera toxin (CT) but their cAMP response to CT is not enhanced [Pacuszka, T., & Fishman, P. H. (1990) J. Biol. Chem. 265, 7673-7668]. We now report that when such cells were exposed to CT in the presence of chloroquine, an acidotropic agent, they accumulated cAMP. This raised the possibility that CT bound to cell surface "neoganglioproteins" may be entering the cells through a different pathway from that of CT-bound GM1. To further explore this phenomenon, we covalently attached GM1 oligosaccharide to human transferrin (Tf). The modified protein (GM1OS-Tf) bound with high affinity to Tf receptors on HeLa cells and increased the binding of CT to the cells. The bound CT, however, was unable to activate adenylyl cyclase as measured by cyclic AMP accumulation. By contrast, treatment of HeLa cells with GM1 increased both CT binding and stimulation of cyclic AMP accumulation. Control cells and cells treated with either GM1 or GM1OS-Tf were exposed to CT in the presence of chloroquine. Whereas chloroquine had little or no effect on the response of control or GM1-treated cells to CT, it made the cells treated with GM1OS-Tf responsive to the toxin. Our results indicate that CT bound to its natural receptor GM1 enters the cells through a pathway different from that of toxin bound to neoganglioproteins. Topics: Animals; Chloroquine; Cholera Toxin; Cyclic AMP; Endocytosis; G(M1) Ganglioside; Glioma; HeLa Cells; Humans; Rats; Receptors, Transferrin; Transferrin; Tumor Cells, Cultured | 1992 |
Nerve growth factor mediates monosialoganglioside-induced release of fibronectin and J1/tenascin from C6 glioma cells.
C6 rat glioma cells incubated in serum-free medium with D-[14C]glucosamine secrete, on stimulation with nerve growth factor (NGF) or monosialogangliosides (MSGs), several glycoproteins (Gps), the most prominent of which are a 270-, 220-, and 69-kDa Gp. Several growth factors, hormones, phorbol ester, and disialo- and trisialogangliosides did not stimulate secretion. Western blot analysis of the conditioned medium from C6 cells stimulated with NGF or MSG identified one distinct band of approximately 220 kDa for fibronectin and J1/tenascin, which comigrated. Antiserum to NGF prevented NGF-stimulated release and also blocked MSG-evoked release. The 220-kDa band was labeled after pulse labeling with [35S]methionine in the presence of NGF, and by a 15-min chase period radioactively labeled J1/tenascin could be immunoprecipitated. Tunicamycin drastically inhibited almost completely release of the 220-kDa Gp labeled by D-[14C]glucosamine or [35S]methionine. These results extend the range of neurotrophic properties attributed to NGF to cells of glial origin and suggest that NGF regulates secretion of extracellular matrix proteins. MSG stimulation of fibronectin and J1/tenascin secretion may be mediated by NGF or an NGF-like molecule also secreted by the C6 glioma cells. Topics: Animals; Blood; Blotting, Western; Carbohydrate Sequence; Cell Adhesion Molecules, Neuronal; Extracellular Matrix Proteins; Fibronectins; G(M1) Ganglioside; G(M2) Ganglioside; G(M3) Ganglioside; Gangliosides; Glioma; Glucosamine; Glycoproteins; Glycosylation; Kinetics; Molecular Sequence Data; Molecular Weight; Nerve Growth Factors; Rats; Tenascin; Tumor Cells, Cultured; Tunicamycin | 1991 |
Effects of GM1 and 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (NeuAc2en) on neuroblastoma (Neuro 2a) and human glioma cells (U1242 MG).
Several lines of evidence suggest that gangliosides may play a role in the regulation of growth in many cell types. Here we describe the effects on growth of two different cell lines by the addition of two different chemicals which have been reported to elevate the cellular ganglioside content through different mechanisms. Growth of neuroblastoma (Neuro 2a) cells in medium containing fetal bovine serum was inhibited in a dose-dependent fashion by both exogenous GM1 ganglioside and NeuAc2en, an inhibitor of sialidase activity. In contrast, growth of glioma cells (U-1242 MG) was not affected by exogenous GM1 or NeuAc2en in the presence of as little as 1% calf serum. However, NeuAc2en inhibited growth of U-1242 MG cells stimulated by platelet-derived growth factor in serum-free medium. These results demonstrate that the growth inhibitory effects of ganglioside on U-1242 MG but not Neuro 2a cells can be counteracted by serum, suggesting that the mechanisms through which gangliosides affect cell growth may be different for different growth factors and cell types. Topics: Animals; Azides; Brain Neoplasms; Cell Division; Culture Media; Culture Media, Serum-Free; Depression, Chemical; G(M1) Ganglioside; Glioma; Humans; Mice; Neuraminidase; Neuroblastoma; Platelet-Derived Growth Factor; Sialic Acids; Tumor Cells, Cultured | 1991 |
Metabolism of cholesterol, phosphatidylethanolamine and stearylamine analogues of GM1 ganglioside by rat glioma C6 cells.
Tritium-labeled neoglycolipids consisting of the oligosaccharide of ganglioside GM1 attached to cholesterol (GM1OSNH-X-CHOL), phosphatidylethanolamine (GM1OS-PE) and stearylamine (GM1OSNHC18) were synthesized and their uptake and metabolism by GM1-deficient rat glioma C6 cells were determined. When the neoglycolipids were added to serum-free culture medium, all three were rapidly taken up by the cells and initially inserted into the plasma membrane based on their resistance to trypsin and their ability to bind cholera toxin. With time, the neoglycolipids underwent internalization as the ratio of cell-associated radioactivity to cell surface toxin binding increased; this process was slow for GM1OSNH-X-CHOL and GM1OS-PE and rapid for GM1OSNHC18. Analysis of lipids extracted from the cells indicated that the neoglycolipids also underwent metabolism to GD1aOS-based analogues. In addition, GM1OSNH-X-CHOL and GM1OSNHC18 were degraded to their GM2OS-based analogues, whereas GM2OS-PE was not detected. In contrast, large amounts of 3H were recovered in the medium from cells treated with GM1OS-PE and the label was associated with material that behaved neither as an oligosaccharide or a neoglycolipid. In the presence of monensin or chloroquine, metabolism of the three neoglycolipids was inhibited. Thus, GM1OS-based neoglycolipids were taken up by the cells, internalized and sorted both to the Golgi apparatus (sialylated to GD1aOS-based analogues) and to lysosomes (hydrolyzed to GM2OS-based analogues). The rate and extent of these processes, however, were strongly influenced by the nature of lipid moiety. Topics: Amines; Animals; Carbohydrate Sequence; Chloroquine; Cholesterol; G(M1) Ganglioside; Glioma; Molecular Sequence Data; Monensin; Phosphatidylethanolamines; Rats; Tumor Cells, Cultured | 1991 |
Cautionary note on the use of the B subunit of cholera toxin as a ganglioside GM1 probe: detection of cholera toxin A subunit in B subunit preparations by a sensitive adenylate cyclase assay.
The use of the B subunit of cholera toxin, a protein that binds specifically to ganglioside GM1, has provided a new paradigm for studying physiological functions of ganglioside GM1. The B subunit inhibited the growth of rat glioma C6 cells that had been pretreated with ganglioside GM1. In some preparations of the B subunit, the inhibition was independent of adenylate cyclase activation and was due to the binding of the B subunit to ganglioside GM1 inserted onto the cell surface. However, in other preparations of the B subunit, there was an additional inhibitory effect due to small contaminations with the A subunit, which caused increases in intracellular cyclic adenosine monophosphate (cAMP) levels and concomitant growth inhibition. This vanishingly small contamination with the A subunit could not be detected by conventional protein sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis but could be measured utilizing a sensitive adenylate cyclase activation assay. Thus caution must be used to ensure that any biological effects of the B subunit are not due to contaminating A subunit and are due solely to the binding of the B subunit to ganglioside GM1 exposed on the cell surface. This is especially important in cyclic nucleotide-sensitive systems. Topics: Adenylyl Cyclases; Animals; Cholera Toxin; Cyclic AMP; DNA, Neoplasm; G(M1) Ganglioside; Glioma; Macromolecular Substances; Molecular Probes; Rats; Tumor Cells, Cultured | 1990 |
Generation of cell surface neoganglioproteins. GM1-neoganglioproteins are non-functional receptors for cholera toxin.
GM1 (II3Neu5Ac-GgOse4Cer)-oligosaccharide was prepared from the ganglioside by ozonolysis and alkaline fragmentation, reductively aminated and coupled to the heterobifunctional cross-linker succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate. The resulting derivative reacted with free sulfhydryl groups and readily cross-linked to cell surface components on rat glioma C6 cells which are GM1-deficient. Attachment of the GM1-oligosaccharide derivative, which was monitored by increased binding of 125I-cholera toxin to the cells, was both time- and concentration-dependent. Prior treatment of the cells with dithiothreitol enhanced the attachment by generating additional free sulfhydryl groups. The affinity of cholera toxin for cells treated with the GM1-oligosaccharide derivative or with GM1 was similar. The nature of the newly generated toxin receptors was determined by Western blotting. Membranes from derivatized cells were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and the resolved components were electrophoretically transferred to a nitrocellulose sheet which was overlain with 125I-cholera toxin. The toxin bound to a wide variety of membrane proteins, most of which were trypsin-sensitive. No such binding was observed using membranes from control cells. Although the GM1-neoganglioproteins newly generated on the surface of rat glioma C6 cells readily bound cholera toxin, the cells did not become more responsive to the toxin as measured by increased production of cyclic AMP or activation of adenylate cyclase. In contrast, cells exposed to GM1 became highly responsive to the toxin. Thus, neoganglioproteins on the cell surface appear to behave as nonfunctional receptors for cholera toxin. Topics: Adenylyl Cyclases; Animals; Brain; Carbohydrate Sequence; Cattle; Cell Line; Cholera Toxin; Cyclic AMP; Enzyme Activation; G(M1) Ganglioside; Gangliosides; Glioma; Glycoproteins; Kinetics; Molecular Sequence Data; Oligosaccharides; Rats; Receptors, Cell Surface; Receptors, Immunologic | 1990 |
Coordinate regulation of ganglioside glycosyltransferases in differentiating NG108-15 neuroblastoma x glioma cells.
The enzymatic basis for ganglioside regulation during differentiation of NG108-15 mouse neuroblastoma x rat glioma hybrid cells was studied. This cell line contains four gangliosides that lie along the same biosynthetic pathway: GM3, GM2, GM1, and GD1a. Chemically induced neuronal differentiation of NG108-15 cells led to an 80% drop in the steady-state level of their major ganglioside, GM3, a sixfold increase in the level of a minor ganglioside, GM2 (which became the predominant ganglioside of differentiated cells); and relatively little change in the levels of GM1 and GD1a, which lie further along the same biosynthetic pathway. The enzymatic basis for this selective change in ganglioside expression was investigated by measuring the activity of two glycosyltransferases involved in ganglioside biosynthesis. UDP-N-acetylgalactosamine: GM3 N-acetylgalactosaminyltransferase (GM2-synthetase) activity increased fivefold during butyrate-induced differentiation, whereas UDP-galactose: GM2 galactosyltransferase (GM1-synthetase) activity decreased to 10% of its control level. Coordinate regulation of these two glycosyltransferases appears to be primarily responsible for the selective increase of GM2 expression during NG108-15 differentiation. Topics: Alprostadil; Animals; Bucladesine; Cell Differentiation; G(M1) Ganglioside; G(M2) Ganglioside; G(M3) Ganglioside; Galactosyltransferases; Ganglioside Galactosyltransferase; Gangliosides; Glioma; Hybrid Cells; Kinetics; Mass Spectrometry; Mice; N-Acetylgalactosaminyltransferases; Neuroblastoma; Polypeptide N-acetylgalactosaminyltransferase; Rats; Theophylline; Tumor Cells, Cultured | 1989 |
Insertion of ganglioside GM1 into rat glioma C6 cells renders them susceptible to growth inhibition by the B subunit of cholera toxin.
The B subunit of cholera toxin does not affect the growth of rat glioma C6 cells which are deficient of its receptor, ganglioside GM1. Insertion of ganglioside GM1 into the plasma membrane of C6 cells renders them susceptible to inhibition of DNA synthesis by the B subunit. Exposure of C6 cells to butyrate induces an elevation of ganglioside GM1 as measured by an increase in binding of iodinated cholera toxin and also results in an inhibition of DNA synthesis by the B subunit. The extent of inhibition of DNA synthesis correlated with the binding of B subunit and was independent of adenylate cyclase activation or increases in intracellular cAMP levels. Topics: Adenylyl Cyclases; Animals; Butyrates; Cell Division; Cholera Toxin; Cyclic AMP; DNA; Dose-Response Relationship, Drug; G(M1) Ganglioside; Glioma; Rats; Tumor Cells, Cultured | 1988 |
Ganglioside potentiation of NGF-independent trophic agents on sensory ganglia.
The ability of gangliosides to potentiate nerve growth factor (NGF)-independent trophic agents was determined by examining the capacity of an exogenous mixture of bovine brain gangliosides (BBG) and the monosialoganglioside GM1 to enhance the neuritogenic action of conditioned media (CM). CM were prepared with cultures of C6 glioma cells, neonatal rat astroglial cells, rat L6 myoblasts and chick embryonic skeletal muscle. Chick embryonic (9 day) dorsal root ganglia (DRG) were cultured on collagen-coated surfaces. The nutrient media with serum added or serum-free N1 medium were supplemented with 50% of one of the CM with or without BBG (150 micrograms/ml) or GM1 (150 micrograms/ml). The neuritogenic responses of DRG 48 h in vitro were evaluated microscopically on the basis of neurite length and number. The neurite promoting action of the factor(s) present in the various CM was potentiated by BBG or GM1 and resulted in increased neurite length and number. Topics: Animals; Cattle; Cell Line; Cells, Cultured; Chick Embryo; Culture Media; Dendrites; G(M1) Ganglioside; Ganglia, Spinal; Gangliosides; Glioma; Growth Substances; Muscles; Neuroglia; Neurons; Neurons, Afferent; Rats | 1988 |
Fluorescent derivatives of ganglioside GM1 function as receptors for cholera toxin.
A fluorescent derivative of ganglioside GM1 was prepared by oxidation of the sialic acid residue with sodium periodate and reaction of the resulting aldehyde with Lucifer yellow CH. The biological activity of the fluorescent derivative was compared with that of native GM1 using GM1-deficient rat glioma C6 cells. When the cells were exposed to either native or fluorescent GM1, their ability to bind 125I-labeled cholera toxin was increased to a similar extent. This increase in binding was directly proportional to the amount of ganglioside added to the medium. The affinity of the toxin for cells treated with either native or fluorescent GM1 also was similar. More importantly, the fluorescent GM1 was as effective as native GM1 in enhancing the responsiveness of the cells to cholera toxin. Thus, the ganglioside-treated cells exhibited a 9-fold increase in toxin-stimulated cyclic AMP production over cells not exposed to GM1. There was a similar increase in iodotoxin binding and toxin-stimulated cyclic AMP accumulation in cells treated with other GM1 derivatives containing rhodaminyl or dinitrophenyl groups. On the basis of these results, it is clear that these modified gangliosides retain the ability to function as receptors for cholera toxin. Consequently, fluorescent gangliosides are likely to be useful as probes for investigating the dynamics and function of these membrane components. Topics: Animals; Cell Line; Cholera Toxin; Chromatography, Thin Layer; Cyclic AMP; Fluorescent Dyes; G(M1) Ganglioside; Glioma; Isoquinolines; Kinetics; Rats; Receptors, Cell Surface; Receptors, Immunologic | 1985 |
Ecto-5'-nucleotidase regeneration after chemical modification of the plasma membrane.
Treatment of the C6 glioblastoma cell with trinitrobenzenesulfonic acid (TNBS) resulted in the selective inactivation of ecto-5'-nucleotidase under conditions which maintained cell viability. Cells respond to ecto-enzyme inactivation by replacing 80% of lost activity within 24 hrs. A lag time of 4-6 hrs before ecto-5'-nucleotidase replacement began and its complete blockage by cycloheximide indicated that the source of replaced enzyme was de novo synthesis and not an intracellular pool. Release of 5'-nucleotidase activity into culture medium in the form of membraneous vesicles slowed during the active recovery period and then steadily increased with time as the plasma membrane enzyme level approached normal. TNBS did not exert a direct inhibitory action upon the exfoliative process as release of vesicular GM1 and protein were little affected. Decrease in exfoliated 5'-nucleotidase activity may be due to a selective conservation of the enzyme in the exfoliative process. Topics: 5'-Nucleotidase; Adenosine Triphosphatases; Animals; Cell Line; Cell Membrane; Cholera Toxin; Cycloheximide; Fibroblasts; G(M1) Ganglioside; Glioma; Humans; Male; Nitrobenzenes; Nucleotidases; Rats; Trinitrobenzenesulfonic Acid | 1983 |
Uptake and metabolism of exogenous gangliosides by cultured cells: effect of choleragen on the turnover of GM1.
When added to the culture medium, 3H-labeled GM1 (tritiated predominantly in the terminal galactose residue) was taken up by murine NCTC 2071 and rat glioma C6 cells, both of which are GM1-deficient. Upon incubating the labeled cells in fresh medium, the cell-associated GM1 was metabolized by the cells with a half-life of 1 to 2 days. Some of the GM1 was converted to GD1a but the bulk of the label appeared in the medium as degradation products. When GM1 labeled in the sialic acid or lipid portion of the molecule was utilized, GM2 also was detected with time in the cells and only a small fraction of the radioactivity was detected in the medium. The rat glioma C6 cells appeared unable to degrade the GM2 that they accumulated; this was demonstrated directly by incubating the cells with labeled GM2. The uptake and subsequent metabolism of GM1 was observed over a wide range of GM1 concentrations (10(-8) to 10(-4) M). The GM1-treated cells initially bound more iodinated choleragen than did untreated cells; but with time, binding capacity decreased. When GM1-treated cells were transferred to fresh medium in the presence of excess choleragen, the amount of cell-associated GM1 remained relatively constant for several days; the conversion of GM1 to GD1a also was blocked. Although labeled GM3 and GD1b also were taken up by the cells, choleragen had no effect on their subsequent metabolism. Choleragenoid, the binding subunit of choleragen, also inhibited GM1 metabolism without activating adenylate cyclase. These results indicate that exogenous gangliosides taken up by cultured cells are metabolized and that choleragen, which binds with high affinity to GM1, specifically prevents the metabolism of this ganglioside. Topics: Animals; Cattle; Cell Count; Cell Division; Cell Line; Cholera Toxin; Chromatography, Thin Layer; G(M1) Ganglioside; Gangliosides; Glioma; Mice; Rats | 1983 |
Differences in the beta-adrenergic responsiveness between high and low passage rat glioma C6 cells.
Responsiveness to catecholamines was studied in two different strains of rat glioma C6 cells. The C6 cells of low passage possessed a high capacity to accumulate cyclic AMP in response to (-)-isoproterenol. Cholera toxin was also able to stimulate cyclic AMP accumulation in these cells. High passage C6 cells were unresponsive to (-)-isoproterenol or to cholera toxin except in the presence of a high concentration of phosphodiesterase inhibitor. The affinity of beta-adrenergic receptors on both strains for (-) [3H] dihydroalprenolol was similar; however, C6 low passage possessed several times the number of beta-adrenergic receptors found in C6 high passage. This difference correlated with the difference found in (-)-isoproterenol-stimulated adenylate cyclase between C6 low passage and high passage. The sodium fluoride-stimulated adenylate cyclase was similar in both strains. Cyclic AMP phosphodiesterase activity was 2-3 times higher in homogenates of C6 high passage than in low passage. In intact cells, the rate of breakdown of cyclic AMP was 5-times faster in C6 high passage than in low passage. Thus, differences in beta-adrenergic receptor number and phosphodiesterase activity explain in part the lack of responsiveness of C6 high passage. Our studies indicate that continuous subculturing of rat glioma C6 cells led to complex alterations in the beta-adrenergic receptor-adenylate cyclase system. Topics: Adenylyl Cyclases; Animals; Cell Line; Cell Membrane; Cholera Toxin; Cyclic AMP; Dihydroalprenolol; G(M1) Ganglioside; Glioma; Isoproterenol; Kinetics; Neoplasms, Experimental; Rats; Receptors, Adrenergic; Receptors, Adrenergic, beta | 1981 |