Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Med Rep ; 22(5): 3994-4002, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33000220

RESUMEN

Previous studies have shown that (GM3), a ganglioside, suppresses hepatoma cell motility and migration by inhibiting phosphorylation of EGFR and the activity of the PI3K/AKT signaling pathway. Therefore, the aim of the present study was to investigate whether the combined treatment of CD82 with gangliosides can exert a synergistic inhibitory effect on cell motility and migration. Epidermal growth factor receptor (EGFR) signaling was studied for its role in the mechanism through which CD82 and gangliosides synergistically inhibit the motility and migration of SW620 human colon adenocarcinoma cells. GM3 and/or GM2 treatment, and/or overexpression of CD82 was performed in SW620 cells. High-performance thin layer chromatography, reverse transcription-quantitative PCR, western blotting and flow cytometry assays were used to confirm the content changes of GM2, GM3 and CD82. In addition, the phosphorylation of EGFR, MAPK and Akt were evaluated by western blot analysis. SW620 cell motility was investigated using wound healing analysis and chemotaxis migration assay. The combination of GM3 and GM2 with CD82 was found to markedly suppress EGF-stimulated SW620 cell motility compared with the individual factors or combination of GM2 or GM3 with CD82 by inhibiting the phosphorylation of EGFR. The results suggested that CD82 in combination with either GM2 or GM3 can exert a synergistic inhibitory effect on cell motility and migration; however, the synergistic mechanisms elicited by GM2 or GM3 with CD82 differ.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Neoplasias del Colon/metabolismo , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Proteína Kangai-1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tirosina/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/patología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Receptores ErbB/metabolismo , Humanos , Proteína Kangai-1/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Transfección
2.
Biochim Biophys Acta ; 1863(7 Pt A): 1472-89, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27066976

RESUMEN

The definitive role of ganglioside GM2 in mediating tumor-induced growth and progression is still unknown. Here we report a novel role of ganglioside GM2 in mediating tumor cell migration and uncovered its mechanism. Data shows differential expression levels of GM2-synthase as well as GM2 in different human cancer cells. siRNA mediated knockdown of GM2-synthase in CCF52, A549 and SK-RC-26B cells resulted in significant inhibition of tumor cell migration as well as invasion in vitro without affecting cellular proliferation. Over-expression of GM2-synthase in low-GM2 expressing SK-RC-45 cells resulted in a consequent increase in migration thus confirming the potential role GM2 and its downstream partners play in tumor cell migration and motility. Further, treatment of SK-RC-45 cells with exogenous GM2 resulted in a dramatic increase in migratory and invasive capacity with no change in proliferative capacity, thereby confirming the role of GM2 in tumorigenesis specifically by mediating tumor migration and invasion. Gene expression profiling of GM2-synthase silenced cells revealed altered expression of several genes involved in cell migration primarily those controlling the integrin mediated signaling. GM2-synthase knockdown resulted in decreased phosphorylation of FAK, Src as well as Erk, while over-expression and/or exogenous GM2 treatment caused increased FAK and Erk phosphorylation respectively. Again, GM2 mediated invasion and Erk phosphorylation is blocked in integrin knockdown SK-RC-45 cells, thus confirming that GM2 mediated migration and phosphorylation of Erk is integrin dependent. Finally, confocal microscopy suggested co-localization while co-immunoprecipitation and surface plasmon resonance (SPR) confirmed direct interaction of membrane bound ganglioside, GM2 with the integrin receptor.


Asunto(s)
Movimiento Celular , Gangliósido G(M2)/metabolismo , Integrina beta1/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Gangliósido G(M2)/farmacología , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Inmunoprecipitación , Cinética , Microscopía Confocal , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/metabolismo , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/patología , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Resonancia por Plasmón de Superficie , Factores de Tiempo , Transfección , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
3.
Int J Biochem Cell Biol ; 45(11): 2349-58, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23968914

RESUMEN

The metastasis suppressor CD82/KAI-1, which is a member of the tetraspanin superfamily, has been proposed to exert its activity together with glycosphingolipids. However, the mechanism of CD82 inhibition has not been fully elucidated. The present study aimed to investigate the synergistic inhibition of cell migration by the tetraspanin CD82 and gangliosides and to correlate this inhibition with activation of epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor (HGFR/cMet) in Hepa1-6 cell lines, whose motility and migration is stimulated by epidermal growth factor (EGF) and hepatocyte growth factor (HGF) in vitro. We found that Hepa1-6 cells transfected with the CD82 gene exhibited decreased migration in response to EGF and HGF. EGF-stimulated phosphorylation of EGFR at Tyr1173 was inhibited in these cells, which contributed to the attenuation of EGFR. Ectopic expression of CD82 in Hepa1-6 cells inhibited HGF-stimulated tyrosine phosphorylation of cMet at Tyr1313 and Tyr1365 without affecting the expression of cMet. These inhibitory effects were enhanced when CD82 was introduced with Ganglioside GM3 alone or GM2/GM3. Reduction of CD82 expression by RNA interference together with depletion of glycosphingolipids with P4 significantly enhanced cell motility and increased the expression of EGFR and its phosphorylation at Tyr1173 in response to EGF. Increased cell motility and HGF-dependent activation of cMet at Tyr1313 and Tyr1365 resulted from decreased CD82 levels and increased GM3. Furthermore, CD82 expression selectively attenuated EGFR and cMet signalling via phosphatidylinositol 3-kinase/Akt but had no affect on the activity of the MAPK signalling pathway. These results suggest that the synergistic effects of CD82 and GM3 or GM2/GM3 on EGFR expression and phosphorylation and cMet activation are responsible for CD82 inhibition of EGF- and HGF-dependent cell motility and migration of Hepa1-6 cells.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Gangliósidos/farmacología , Proteína Kangai-1/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/farmacología , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
Mol Cell Biochem ; 382(1-2): 83-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23749170

RESUMEN

Ganglioside GM3 plays a well-documented and important role in the regulation of tumor cell proliferation, invasion, and metastasis by modulating tyrosine kinase growth factor receptors. However, the effect of GM3 on the hepatocyte growth factor receptor (HGFR, cMet) has not been fully delineated. In the current study, we investigated how GM3 affects cMet signaling and HGF-stimulated cell motility and migration using three hepatic cancer cell lines of mouse (Hca/A2, Hca/16A3, and Hepa1-6). Decreasing GM3 expression with the use of P4, a specific inhibitor for ganglioside synthesis inhibited the HGF-stimulated phosphorylation of cMet and activity of PI3K/Akt signaling pathway. In contrast, the increased expression of GM3 as a result of adding exogenous GM3 enhanced the HGF-stimulated phosphorylation of cMet and activity of PI3K/Akt signaling pathway. Furthermore, HGF-stimulated cell motility and migration in vitro were inhibited by reduced expression of GM3 and enhanced by increased expression of GM3. All the observations indicate that ganglioside GM3 promotes HGF-stimulated motility of murine hepatoma cell through enhanced phosphorylation of cMet at specific tyrosine sites and PI3K/Akt-mediated migration signaling.


Asunto(s)
Carcinoma Hepatocelular/patología , Movimiento Celular/efectos de los fármacos , Gangliósido G(M3)/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfotirosina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Carcinoma Hepatocelular/enzimología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Gangliósido G(M2)/farmacología , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Ratones , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 106(18): 7461-6, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19380734

RESUMEN

Epithelial-to-mesenchymal cell transition (EMT) is a basic process in embryonic development and cancer progression. The present study demonstrates involvement of glycosphingolipids (GSLs) in the EMT process by using normal murine mammary gland NMuMG, human normal bladder HCV29, and human mammary carcinoma MCF7 cells. Treatment of these cells with D-threo-1-(3',4'-ethylenedioxy)phenyl-2-palmitoylamino-3-pyrrolidino-1-propanol (EtDO-P4), the glucosylceramide (GlcCer) synthase inhibitor, which depletes all GSLs derived from GlcCer, (i) down-regulated expression of a major epithelial cell marker, E-cadherin; (ii) up-regulated expression of mesenchymal cell markers vimentin, fibronectin, and N-cadherin; (iii) enhanced haptotactic cell motility; and (iv) converted epithelial to fibroblastic morphology. These changes also were induced in these cell lines with TGF-beta, which is a well-documented EMT inducer. A close association between specific GSL changes and EMT processes induced by EtDO-P4 or TGF-beta is indicated by the following findings: (i) The enhanced cell motility of EtDO-P4-treated cells was abrogated by exogenous addition of GM2 or Gg4, but not GM1 or GM3, in all 3 cell lines. (ii) TGF-beta treatment caused changes in the GSL composition of cells. Notably, Gg4 or GM2 was depleted or reduced in NMuMG, and GM2 was reduced in HCV29. (iii) Exogenous addition of Gg4 inhibited TGF-beta-induced changes of morphology, motility, and levels of epithelial and mesenchymal markers. These observations indicate that specific GSLs play key roles in defining phenotypes associated with EMT and its reverse process (i.e., mesenchymal-to-epithelial transition).


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/fisiología , Glicoesfingolípidos/fisiología , Mesodermo/citología , Mesodermo/fisiología , Animales , Línea Celular , Movimiento Celular , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Gangliósido G(M2)/farmacología , Gangliósidos/farmacología , Glucosiltransferasas/antagonistas & inhibidores , Glicoesfingolípidos/metabolismo , Humanos , Mesodermo/efectos de los fármacos , Ratones , Propanolaminas/farmacología , Pirrolidinas/farmacología , Factor de Crecimiento Transformador beta/farmacología
6.
Clin Exp Immunol ; 130(3): 441-8, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12452834

RESUMEN

Dendritic cell (DC) development and function is critical in the initiation phase of any antigen-specific immune response against tumours. Impaired function of DC is one explanation as to how tumours escape immunosurveillance. In the presence of various soluble tumour-related factors DC precursors lose their ability to differentiate into mature DC and to activate T cells. Gangliosides are glycosphingolipids shed by tumours of neuroectodermal origin such as melanoma and neuroblastoma. In this investigation we address the question of whether gangliosides suppress the development and function of monocyte-derived DC in vitro. In the presence of gangliosides, the monocytic DC precursors showed increased adherence, cell spreading and a reduced number of dendrites. The expression of MHC class II molecules, co-stimulatory molecules and the GM-CSF receptor (CD116) on the ganglioside-treated DC was significantly reduced. Furthermore, the function of ganglioside-treated DC was impaired as observed in endocytosis, chemotactic and T cell proliferation assays. In contrast to monocytic DC precursors, mature DC were unaffected even when higher doses of gangliosides were added to the culture. With regard to their carbohydrate structure, five different gangliosides (GM2, GM3, GD2, GD3, GT1b), which are typically shed by melanoma and neuroblastoma, were tested for their ability to suppress DC development and function. Suppression was induced by GM2, but not by the other gangliosides. These data suggest that certain gangliosides impair DC precursors, implying a possible mechanism for tumour escape.


Asunto(s)
Gangliósidos/farmacología , Monocitos/citología , Tumores Neuroectodérmicos/inmunología , Escape del Tumor , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis de Leucocito/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Endocitosis/efectos de los fármacos , Citometría de Flujo , Gangliósido G(M2)/farmacología , Antígenos HLA-D/inmunología , Humanos , Inmunofenotipificación , Prueba de Cultivo Mixto de Linfocitos , Melanoma/inmunología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Neuroblastoma/inmunología
7.
Hypertension ; 38(5): 1030-7, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11711493

RESUMEN

Gangliosides, sialic acid-containing glycophospholipids, accumulate in atherosclerotic vessels and appear to regulate the proliferation of various cell types. Furthermore, vascular smooth muscle cell (VSMC) proliferation is associated with the development and progression of cardiovascular diseases. To demonstrate whether gangliosides are able to modulate the VSMC growth, the effect of gangliosides GM1, GM2, and GM3 on cell DNA synthesis and cell number has been examined. Moreover, we investigated possible intracellular mechanisms by which GM1 and GM2 elicit their mitogenic effects. Stimulation of VSMCs with GM1 and GM2 resulted in a dose-dependent increase in DNA synthesis and cell number, whereas GM3 caused a decrease in DNA synthesis. GM1 and GM2 (50 micromol/L) stimulate phosphorylation of extracellular signal-regulated kinases (ERKs) 1 and 2 and phosphorylation of the c-Jun N-terminal kinase (JNK), with a maximum at 15 minutes, but they do not have an effect on the phosphorylation of p38 mitogen-activated protein kinase (MAPK). GM3 (50 micromol/L), on the other hand, does not stimulate any of the 3 aforementioned MAPKs. Pretreatment of the cells with 20 micromol/L PD 098,059 caused a complete inhibition of ERK1/2 and JNK MAPK, whereas pretreatment with a Ras (farnesyl transferase) inhibitor did not abrogate the GM1- and GM2-induced ERK1/2 phosphorylation. Furthermore, GM1 and GM2 did not activate Raf-1 kinase. Interestingly, pretreatment of VSMCs with 100 nmol/L pertussis toxin resulted in a complete inhibition of the ERK1/2 phosphorylation. Finally, the GM1- and GM2-induced increase in cell number was significantly inhibited by PD 098,059. We may conclude that GM1 and GM2 stimulate ERK1/2 via a pertussis toxin-sensitive G(i)-coupled receptor through a Raf-1 kinase-independent pathway. Moreover, the GM1- and GM2-induced VSMC growth is ERK1/2 dependent.


Asunto(s)
Gangliósido G(M1)/farmacología , Gangliósido G(M2)/farmacología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/enzimología , Animales , División Celular/efectos de los fármacos , Células Cultivadas , ADN/biosíntesis , Gangliósido G(M3)/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Músculo Liso Vascular/efectos de los fármacos , Toxina del Pertussis , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ratas , Ratas Endogámicas WKY , Factores de Virulencia de Bordetella/farmacología
8.
J Lipid Res ; 42(8): 1197-202, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11483620

RESUMEN

Lysosphingolipids, which lack the fatty acid moiety of sphingolipids, are known to be accumulated in some variants of sphingolipid storage diseases. Here, we report that lysosphingolipids with naturally occurring stereochemical configurations induce apoptosis in mouse neuroblastoma Neuro2a cells. The intracellular dehydrogenase activity and [3H]thymidine incorporation of Neuro2a cells were strongly suppressed by the addition of lysosphingolipids in a dose-dependent manner, whereas the parental sphingolipids had no effect. Intranucleosomal DNA fragmentation, chromatin condensation, and phosphatidylserine externalization, which are typical features of apoptosis, were observed when the cells were cultured with 40-80 microM of lysosphingolipids for 24-48 h in the presence of 5% fetal calf serum. Activation of caspase-3-like enzyme occurred after addition of lysosphingolipids followed by incubation at 37 degrees C for 24 h. The addition of an inhibitor of caspases, ZVAD-fmk, to the Neuro2a cell culture completely inhibited the elevation of caspase-3 activity but not the DNA fragmentation. These results may indicate that a caspase-3 independent signaling pathway is involved in the lysosphingolipid-induced apoptosis and suggest that accumulation of lysosphingolipids, but not parental sphingolipids, triggers the apoptotic cascade in neuronal cells of patients with sphingolipidoses.


Asunto(s)
Apoptosis/efectos de los fármacos , Gangliósido G(M1)/análogos & derivados , Gangliósido G(M1)/farmacología , Gangliósido G(M2)/análogos & derivados , Gangliósido G(M2)/farmacología , Glicoesfingolípidos/química , Esfingosina/análogos & derivados , Amidohidrolasas/metabolismo , Animales , Caspasa 3 , Caspasas/metabolismo , Bovinos , Cromatina/ultraestructura , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Activación Enzimática , Gangliósido G(M1)/química , Gangliósido G(M1)/metabolismo , Gangliósido G(M2)/química , Gangliósido G(M2)/metabolismo , Glicoesfingolípidos/metabolismo , Humanos , Ratones , Neuroblastoma/patología , Oxidorreductasas/metabolismo , Fosfatidilserinas/metabolismo , Pseudomonas/enzimología , Psicosina/análogos & derivados , Transducción de Señal , Esfingolípidos/metabolismo , Esfingolípidos/farmacología , Esfingosina/farmacología , Células Tumorales Cultivadas
9.
Neurochem Res ; 26(4): 375-82, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11495348

RESUMEN

We previously reported that ciliary neurotrophic factor (CNTF) increased the serum-free cell survival of immortalized motor neuron-like cells (NSC-34), and addition of the exogenous ganglioside GalNAc beta4(Neu5Ac alpha3)Gal beta4GlcCer (GM2) facilitated cell survival together with CNTF. Moreover beta 1,4 N-acetylgalactosaminyltransferase (GM2 synthase) activity increased in NSC-34 cells cultured with CNTF. We now have examined whether CNTF-induced cell survival is associated with the collaboration between GM2 and the CNTF receptor (CNTF-R). Despite the presence of CNTF (50 ng/ml), anti-CNTF-R antibody caused cell death and prevented the up-regulation of GM2 synthase expression. The addition of GM2 (1 to 20 microM) abrogated the anti-CNTF-R antibody effect which shortened cell survival and blocked GM2 synthase activation. Use of [125I]CNTF showed the specificity of CNTF binding in NSC-34 cells in situ. GM2 produced a 5-fold increase in the CNTF binding affinity per cell but did not change the binding site number. The study by metabolic labeling with [1-(14)C]N-acetyl-D-galactosamine ([14C]GalNAc) showed that biosynthesized GM2 was involved in the immunoprecipitation of CNTF-R. These findings indicate that up-regulated GM2 synthesis induces functional conversion of CNTF-R to the activated state, in which it has affinity for CNTF. We conclude that GM2 is a bio-regulating molecule of CNTF-R in motor neurons.


Asunto(s)
Gangliósido G(M2)/farmacología , Neuronas/fisiología , Receptor de Factor Neurotrófico Ciliar/fisiología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Gangliósido G(M2)/metabolismo , Ratones , Neuronas/citología , Pruebas de Precipitina , Receptor de Factor Neurotrófico Ciliar/inmunología , Receptor de Factor Neurotrófico Ciliar/metabolismo
10.
J Clin Oncol ; 19(5): 1430-6, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11230488

RESUMEN

PURPOSE: High-dose interferon alfa-2b (IFNalpha2b) is the only established adjuvant therapy of resectable high-risk melanoma. GM2-KLH/QS-21 (GMK) is a chemically defined vaccine that is one of the best developed of a range of vaccine candidates for melanoma. A single-institution phase III trial conducted at Memorial Hospital served as the impetus for an intergroup adjuvant E1694/S9512/C509801 trial, which recently completed enrollment of 880 patients. To build on the apparent benefit of IFNalpha2b in resectable high-risk American Joint Committee on Cancer (AJCC) stage IIB or III melanoma, this phase II study was designed to evaluate the combination of GMK and IFNalpha2b. The E2696 trial was undertaken to evaluate the toxicity and other effects of the established adjuvant high-dose IFNalpha2b regimen in relation to immune responses to GMK and to evaluate the potential clinical and immunologic effects of the combined therapies. PATIENTS AND METHODS: This trial enrolled 107 patients with resectable high- or very high-risk melanoma (AJCC stages IIB, III, and IV). RESULTS: The results demonstrate that IFNalpha2b does not significantly inhibit immunoglobulin M or G serologic responses to the vaccine and that the combination of high-dose IFNalpha2b and GMK is well tolerated in this patient population. CONCLUSION: Cox analysis of the results of the combination with IFNalpha2b show improvement in the relapse-free survival of patients with very high-risk melanoma (including those with resectable M1 disease).


Asunto(s)
Antineoplásicos/farmacología , Vacunas contra el Cáncer/inmunología , Gangliósido G(M2)/uso terapéutico , Interferón-alfa/farmacología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Adulto , Formación de Anticuerpos , Antineoplásicos/efectos adversos , Antineoplásicos/inmunología , Quimioterapia Adyuvante , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Gangliósido G(M2)/farmacología , Humanos , Interferón alfa-2 , Interferón-alfa/efectos adversos , Interferón-alfa/inmunología , Masculino , Melanoma/tratamiento farmacológico , Persona de Mediana Edad , Recurrencia Local de Neoplasia , Proteínas Recombinantes , Factores de Riesgo , Neoplasias Cutáneas/tratamiento farmacológico
11.
Neuroreport ; 10(4): 703-6, 1999 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-10208534

RESUMEN

Some gangliosides have been regarded as autoantigens of immune-mediated neurological disorders such as Guillain-Barré syndrome (GBS), Miller Fisher syndrome and multifocal motor neuropathy. On the other hand, proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma), may be important in the pathogenesis of some neuroimmunological disorders. To clarify the interactions between immune cells and gangliosides, we investigated the effects of gangliosides on the production of proinflammatory cytokines in peripheral blood mononuclear cell (PBMC) cultures. We found that ganglioside GM2 markedly enhances the production of TNF-alpha and that TNF-alpha induction by coated GM2 is still more marked. These findings suggest that immune cells, especially monocytes/macrophages, cause inflammation upon encountering GM2.


Asunto(s)
Gangliósido G(M2)/farmacología , Monocitos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Células Cultivadas , Gangliósidos/farmacología , Humanos , Interferón gamma/biosíntesis , Interleucina-1/biosíntesis , Monocitos/efectos de los fármacos
12.
Neurochem Res ; 24(2): 281-6, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9972876

RESUMEN

We have examined whether ciliary neurotrophic factor (CNTF) can alter serum-free cell survival of immortalized motor neuron-like cells, which were established by fusing mouse neuroblastoma N18TG2 with mouse motor neurons. One of the cell lines, NSC-34 exhibited cell survival in the presence of CNTF. NSC-34 preserves the most characteristics of motor neurons, such as the formation of neuromuscular junctions on co-cultured myotube. GM2 ganglioside is characteristic of motor neurons, and expressed highly in NSC-34. When NSC-34 was cultured with exogenous GM2 ganglioside and CNTF, GM2 facilitated the cell survival effect of CNTF. In the addition, beta 1,4 N-acetylgalactosaminyltransferase (GM2 synthase) activity was enhanced up to 3.9-fold by culture in the presence of CNTF. GM2 might be a functional modulator of CNTF in motor neurons. It might be presented to cell surface by its enzyme activation, and become a signal of early stage, when CNTF rescues motor neurons.


Asunto(s)
Gangliósido G(M2)/farmacología , Neuronas Motoras/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Animales , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Factor Neurotrófico Ciliar , Sinergismo Farmacológico , Ratones , Neuronas Motoras/citología , Neuronas Motoras/enzimología , N-Acetilgalactosaminiltransferasas/metabolismo , Sialiltransferasas/metabolismo , Células Tumorales Cultivadas , Polipéptido N-Acetilgalactosaminiltransferasa
13.
Glycobiology ; 9(2): 101-14, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9949188

RESUMEN

We have previously shown that the extracellular matrix molecule tenascin-C inhibits fibronectin-mediated cell adhesion and neurite outgrowth by an interaction with a cellular RGD-independent receptor which interferes with the adhesion and neurite outgrowth promoting activities of the fibronectin receptor(s). Here we demonstrate that the inhibitory effect of tenascin-C on beta1integrin-dependent cell adhesion and neurite outgrowth is mediated by the interaction of the protein with membrane-associated disialogangliosides, which interferes with protein kinase C-related signaling pathways. First, in substratum mixtures with fibronectin, an RGD sequence-containing fragment of the molecule or synthetic peptide, tenascin-C inhibited cell adhesion and spreading by a disialoganglioside-dependent, sialidase-sensitive mechanism leading to an inhibition of protein kinase C. Second, the interaction of intact or trypsinized, i.e., cell surface glycoprotein-free, cells with immobilized tenascin-C was strongly inhibited by gangliosides or antibodies to gangliosides and tenascin-C. Third, preincubation of immobilized tenascin-C with soluble disialogangliosides resulted in a delayed cell detachment as a function of time. Similar to tenascin-C, immobilized antibody to GD2 (3F8) or sphingosine, a protein kinase C inhibitor, strongly inhibited RGD-dependent cell spreading. Finally, the degree of tenascin-C-induced inhibition of cell adhesion was proportional to the degree of disialoganglioside levels of expression by different cells suggesting the relevance of such mechanism in modulating integrin-mediated cell-matrix interactions during pattern formation or tumor progression.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Fibronectinas/farmacología , Gangliósidos/metabolismo , Integrina beta1/metabolismo , Neuritas/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Tenascina/farmacología , Animales , Células Cultivadas , Interacciones Farmacológicas , Gangliósido G(M1)/farmacología , Gangliósido G(M2)/farmacología , Gangliósidos/farmacología , Glicoesfingolípidos/farmacología , Humanos , Ratones , Oligopéptidos , Transducción de Señal
14.
Cell Immunol ; 183(2): 90-8, 1998 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9606992

RESUMEN

Monosialogangliosides, normal components of cell membranes, regulate cell development and differentiation in several organs. Our previous observation of dramatic premature thymic involution in cats with feline GM1 gangliosidosis, whose thymocytes have abnormally high cell surface gangliosides, suggested that excess GM1 ganglioside (GM1) could modulate thymocyte apoptosis in this disease (Cox et al., "Thymic Alterations in Feline GM1 Gangliosidosis," submitted). In these studies, we added exogenous GM1 to murine primary thymocyte cultures and demonstrated enhanced apoptosis in treated cells by DNA fragmentation, apoptotic body, and electrophoretic analyses. GM1-enhanced apoptosis was blocked by common apoptotic pathway inhibitors including aurintricarboxylic acid (inhibitor of endonuclease activity), actinomycin D (inhibitor of RNA transcription), and cycloheximide (inhibitor of protein synthesis). GM1 treatment primarily affected the immature CD4+ CD8+ subset, as shown by flow cytometric evaluation of fetal thymic organ culture and primary thymocyte cultures. Apoptosis also could be induced by GM2, GM3, and GT1b, whereas asialo-GM1 failed to do so, suggesting that the sialic acid moiety may play an important role in the induction of thymocyte apoptosis.


Asunto(s)
Apoptosis , Gangliósidos/farmacología , Linfocitos T/efectos de los fármacos , Timo/patología , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Células Cultivadas , Fragmentación del ADN , Femenino , Gangliósido G(M1)/farmacología , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico/farmacología , Timo/embriología
15.
Eur J Cell Biol ; 71(1): 79-88, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8884181

RESUMEN

Gangliosides appear to regulate proliferation of different cell types. In the present study, we investigated the effects of gangliosides GM1, GM2 and GM3 on platelet-derived growth factor (PDGF)-induced vascular smooth muscle cell (VSMC) growth. In addition, we examined the effects of gangliosides on the PDGF-BB-dependent signalling transduction pathway in rat aortic VSMC. GM2 and GM1 inhibit the PDGF-BB-dependent receptor tyrosine autophosphorylation, stimulation of the PLC-gamma 1, increase of inositol-1,4,5-trisphosphate (InsP3), elevation in cytosolic free Ca2+ ([Ca2+]i), expression of the immediate early growth response gene c-fos and cell proliferation with the following rank order of potency GM2 > GM1. Although GM3 did not influence the PDGF-BB-dependent receptor autophosphorylation and PLC-gamma 1 activation, it effectively inhibited the PDGF-BB-dependent InsP3 formation, [Ca2+]i and cell growth. Binding studies with 125I-PDGF-BB on VSMC in the presence and absence of 10 to 50 microM of each ganglioside revealed that GM1 and GM2 effectively inhibited the specific binding of PDGF-BB with an IC50 value of 20 microM for GM2 and 30 microM for GM1. GM3 had no significant effect on the specific 125I-PDGF-BB binding. These observations suggest that GM1 and GM2 may interact with PDGF-BB or its receptor resulting in a prevention of its binding. GM3 was able to suppress the PDGF-BB-dependent increase of InsP3 and [Ca2+]i downstream of the PDGF-BB-dependent receptor autophosphorylation and PLC-gamma 1 activity.


Asunto(s)
Gangliósido G(M1)/farmacología , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Músculo Liso Vascular/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Aorta/citología , Becaplermina , Calcio/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Fosfatos de Inositol/análisis , Isoenzimas/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Fosfolipasa C gamma , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-sis , ARN Mensajero/análisis , Ratas , Ratas Endogámicas WKY , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo
16.
Clin Immunol Immunopathol ; 79(2): 197-202, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8620627

RESUMEN

The effects of gangliosides on spontaneous immunoglobulin (Ig) production in human B cells were studied. Of the various gangliosides tested, including GM1, GM2, GM3, GD1a, GD1b, GD3, GT1b, and GQ1b, only GM2 inhibited Ig production, but not thymidine uptake, in human B cell lines. Moreover, the GM2-induced inhibition was blocked by anti-GM2 mAb, but not by control IgM. Of various cytokines, IL-10 and TNF-alpha each partially counteracted the GM2-induced inhibition, and addition of both IL-10 and TNF-alpha completely counteracted the inhibition. On the other hand, anti-IL-10 mAb plus anti-TNF-alpha mAb inhibited spontaneous Ig production. GM2 inhibited endogenous production of IL-10 and TNF-alpha without affecting the binding of IL-10 and TNF-alpha in B cell lines. GM2 also specifically inhibited spontaneous production of Ig, IL-10, and TNF-alpha in in vivo activated B cells obtained from normal donors. This inhibition was blocked by anti-GM2 mAb and was counteracted specifically by IL-10 plus TNF-alpha. Collectively, GM2 may inhibit spontaneous Ig production by inhibiting endogenous production of IL-10 and TNF-alpha in B cells.


Asunto(s)
Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Gangliósido G(M2)/farmacología , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/efectos de los fármacos , Inmunosupresores/farmacología , Linfocitos B/inmunología , Línea Celular , Citocinas/farmacología , Interacciones Farmacológicas , Humanos , Activación de Linfocitos/efectos de los fármacos
17.
Eur J Immunol ; 25(1): 302-5, 1995 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-7843249

RESUMEN

The effects of gangliosides on human IgE and IgG4 production were studied. Of the various gangliosides tested, only GM2 and GM3 inhibited the IgE and IgG4 production induced by interleukin (IL)-4 plus hydrocortisone (HC), or that induced by IL-13 plus HC, in human surface IgE- and IgG4-negative (sIgE-, sIgG4-) B cells without affecting the production of IgG1, IgG2, IgG3, IgM, IgA1 or IgA2. In contrast, GM1, GD1a, GD1b, GD3, GT1b and GQ1b were without effects. The GM2- and GM3-mediated inhibition was specific, since each was blocked by a corresponding antibody. Of the various factors tested. IL-6, IL-10, and tumor necrosis factor (TNF)-alpha enhanced the IgE and IgG4 production induced by IL-4 plus HC or by IL-13 plus HC, while IL-8 and transforming growth factor (TGF)-beta inhibited these responses. However, only TNF-alpha counteracted the GM2- and GM3-mediated inhibition of IgE and IgG4 production, while IL-6, IL-10, anti-IL-8 monoclonal antibody and anti-TGF-beta antibody failed to do so. Anti-TNF-alpha monoclonal antibody, but not control IgG1, not only inhibited IgE and IgG4 production in the absence of TNF-alpha but also blocked the counteraction of inhibition by TNF-alpha. In cultures containing IL-4 plus HC or IL-13 plus HC. GM2 and GM3 specifically inhibited TNF-alpha production without affecting TNF-alpha receptors, IL-6 production or IL-6 receptors. These results indicate that GM2 and GM3 inhibit IgE and IgG4 production by inhibiting endogenous TNF-alpha production.


Asunto(s)
Gangliósidos/fisiología , Inmunoglobulina E/biosíntesis , Inmunoglobulina G/biosíntesis , Linfocitos B/inmunología , Células Cultivadas , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Humanos , Hidrocortisona/farmacología , Interleucina-13/fisiología , Interleucina-4/fisiología , Tonsila Palatina/citología
18.
J Virol ; 68(1): 258-68, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8254737

RESUMEN

A virus-host cell-binding assay was developed and used to investigate specific binding between group A porcine rotavirus and MA-104 cells or porcine enterocytes. A variety of glycoconjugates and cellular components were screened for their ability to block rotavirus binding to cells. During these experiments a crude ganglioside mixture was observed to specifically block rotavirus binding. On the basis of these results, enterocytes were harvested from susceptible piglets and a polar lipid fraction was isolated by solvent extraction and partitioning. Throughout subsequent purification of this fraction by Sephadex partition, ion-exchange, silicic acid, and thin-layer chromatography, blocking activity behaved as a monosialoganglioside (GMX) that displayed a thin-layer chromatographic mobility between those of GM2 and GM3. The blocking activity of GMX was inhibited by treatment with neuraminidase and ceramide glycanase but not by treatment with protease or heat (100 degrees C). Further purification of GMX by high-pressure liquid chromatography resulted in the resolution of two monosialogangliosides, GMX and a band which comigrated with GM1 on thin-layer chromatography. These data suggest that a cell surface monosialoganglioside or family of monosialogangliosides may function as an in vivo relevant receptor for group A porcine rotavirus and that sialic acid is a required epitope for virus-binding activity.


Asunto(s)
Gangliósidos/metabolismo , Mucosa Intestinal/química , Receptores Virales/metabolismo , Rotavirus/metabolismo , Animales , Unión Competitiva , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Gangliósidos/aislamiento & purificación , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Neuraminidasa/farmacología , Receptores Virales/aislamiento & purificación , Porcinos
19.
J Neurochem ; 56(1): 105-12, 1991 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1702827

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Gangliósidos/farmacología , Glioma/metabolismo , Factores de Crecimiento Nervioso/farmacología , Animales , Sangre , Western Blotting , Secuencia de Carbohidratos , Gangliósido G(M1)/farmacología , Gangliósido G(M2)/farmacología , Gangliósido G(M3)/farmacología , Glucosamina/metabolismo , Glicoproteínas/metabolismo , Glicosilación , Cinética , Datos de Secuencia Molecular , Peso Molecular , Ratas , Tenascina , Células Tumorales Cultivadas , Tunicamicina/farmacología
20.
FEBS Lett ; 264(2): 249-52, 1990 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-2358071

RESUMEN

The only gangliosides in Burkitt's lymphoma EB-3 cells is GM3. Treatment of Burkitt's lymphoma EB-3 cells with gangliosides GM1 or GM3 results in their binding to and partial incorporation into the cell membrane. About 25% of cell-associated ganglioside GM1 can interact with the ricin. However, such an increase in the number of binding sites does not enhance but rather decreases the cytotoxic effect of ricin. A similar protective effect was observed when the cells were pretreated with ganglioside GM3. In contrast, the increase in ricin biding sites caused by pretreatment of the cells with neuraminidase was accompanied by increase in ricin cytotoxicity. These differences may be related to observed differences in the rate of ricin-endocytosis by native and ganglioside-treated cells.


Asunto(s)
Gangliósido G(M1)/farmacología , Gangliósido G(M2)/farmacología , Gangliósidos/farmacología , Ricina/metabolismo , Sitios de Unión , Linfoma de Burkitt , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Endocitosis , Gangliósido G(M1)/metabolismo , Gangliósido G(M2)/metabolismo , Liposomas/metabolismo , Neuraminidasa/metabolismo , Ricina/toxicidad , Temperatura , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA