Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Immunopathol Pharmacol ; 19(2): 339-56, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16831301

RESUMEN

The transition of prion protein from a mainly alpha-structured isoform (PrPC) to a beta sheet-containing protein (PrPSc) represents a major pathogenetic mechanism in prion diseases. To study the role of PrP structural conformation in prion-dependent neurodegeneration, we analysed the neurotoxicity of PrP in alpha and beta conformations, using a recombinant protein encompassing amino acids 90-231 of the human PrP (hPrP90-231). Using controlled thermal denaturation (53 degrees C, 1h) we converted hPrP90-231 in a structural isoform displaying PrPSc-related characteristics: high beta sheet content, increased aggregability and a slight increase in the resistance to protease K. In virtue of these structural changes, hPrP90-231 powerfully affected the survival of SH-SY5Y cells, inducing a caspase-3 and p38- dependent apoptosis. Conversely, in the native alpha-helix-rich conformation, hPrP90-231 did not show significant cell toxicity. The relationship between the structural state of hPrP90-231 and its neurotoxicity was demonstrated, inducing the thermal denaturation of the peptide in the presence of Congo red that prevented both the transition of hPrP90-231 into a beta-rich isoform and the acquisition of toxic properties. In conclusion, we report that the toxicity of hPrP90-231 is dependent on its three-dimensional structure, as is supposed to occur for the pathogen PrP during TSE.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas PrPC/química , Proteínas PrPC/farmacología , Amiloide/biosíntesis , Benzotiazoles , Caspasa 3 , Caspasa 7 , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dicroismo Circular , Endopeptidasa K/química , Colorantes Fluorescentes , Humanos , Hidrólisis , Immunoblotting , L-Lactato Deshidrogenasa/metabolismo , Microscopía Electrónica , Necrosis , Conformación Proteica , Desnaturalización Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad , Sales de Tetrazolio , Tiazoles/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Int J Immunopathol Pharmacol ; 18(1): 95-112, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15698515

RESUMEN

Peptides corresponding to three alpha helices present in the C-terminal region of the human prion protein have been synthesized and their structural autonomy analyzed by circular dichroism (CD) and NMR spectroscopy. The results obtained indicate that the protein fragment corresponding to the alpha 3-helix, in contrast to alpha 1 and alpha 2 peptides, shows a complete structural autonomy. The chemical shifts values found for NH and CHalpha resonance of the isolated alpha 3 peptide, formed by 30 aminoacid residues, were markedly and surprisingly similar to the corresponding values of the alpha 3-helix in the protein. The structural autonomy of the alpha 3-helix is profoundly determined by the presence of the conserved capping box and, in part, by the ionic bond formed between Glu200 and Lys204. On the basis of these observations a novel PrP consensus pattern, centered on the alpha 3-helix region, has been defined. The data indicate that this autonomous and highly conserved region of the PrPc likely plays a critical role in folding and stability. This gives an explanation of why many of pathogenic mutations occur in this part of the molecule, sharing relevant effects on the overall protein conformation. In particular the D202N capping mutation almost completely destabilizes the isolated alpha 3 peptide. While it is well known that the D202N substitution is associated with a GSS disease, the possible structural basis of this fatal pathology has never been investigated. We propose that a lower alpha 3-helical propensity leading to a major destabilization of the PrPc molecule initiates the pathogenic process associated with D202N capping mutation.


Asunto(s)
Mutación/fisiología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Priones/genética , Priones/metabolismo , Secuencia de Aminoácidos , Animales , Dicroismo Circular , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Fragmentos de Péptidos/síntesis química , Priones/síntesis química , Conformación Proteica
3.
Mol Endocrinol ; 15(10): 1838-52, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11579215

RESUMEN

The aim of this study was the characterization of the intracellular effectors of the antiproliferative activity of somatostatin in PC Cl3 thyroid cells. Somatostatin inhibited PC Cl3 cell proliferation through the activation of a membrane phosphotyrosine phosphatase. Conversely, PC Cl3 cells stably expressing the v-mos oncogene (PC mos) were completely insensitive to the somatostatin antiproliferative effects since somatostatin was unable to stimulate a phosphotyrosine phosphatase activity. In PC mos cells basal phosphotyrosine phosphatase activity was also reduced, suggesting that the expression of a specific phosphotyrosine phosphatase was impaired in these transformed cells. We suggested that this phosphotyrosine phosphatase could be r-PTP eta whose expression was abolished in the PC mos cells. To directly prove the involvement of r-PTP eta in somatostatin's effect, we stably transfected this phosphatase in PC mos cells. This new cell line (PC mos/PTP eta) recovered somatostatin's ability to inhibit cell proliferation, showing dose-dependence and time course similar to those observed in PC Cl3 cells. Conversely, the transfection of a catalytically inactive mutant of r-PTP eta did not restore the antiproliferative effects of somatostatin. PC mos/PTP eta cells showed a high basal phosphotyrosine phosphatase activity which, similarly to PC Cl3 cells, was further increased after somatostatin treatment. The specificity of the role of r-PTP eta in somatostatin receptor signal transduction was demonstrated by measuring its specific activity after somatostatin treatment in an immunocomplex assay. Somatostatin highly increased r-PTP eta activity in PCCl3 and PC mos/PTP eta (+300%, P < 0.01) but not in PCmos cells. Conversely, no differences in somatostatin-stimulated SHP-2 activity, (approximately +50%, P < 0.05), were observed among all the cell lines. The activation of r-PTP eta by somatostatin caused, acting downstream of MAPK kinase, an inhibition of insulin-induced ERK1/2 activation with the subsequent blockade of the phosphorylation, ubiquitination, and proteasome degradation of the cyclin-dependent kinase inhibitor p27(kip1). Ultimately, high levels of p27(kip1) lead to cell proliferation arrest. In conclusion, somatostatin inhibition of PC Cl3 cell proliferation requires the activation of r-PTP eta which, through the inhibition of MAPK activity, causes the stabilization of the cell cycle inhibitor p27(kip1).


Asunto(s)
División Celular/efectos de los fármacos , Proteínas Tirosina Fosfatasas/metabolismo , Somatostatina/farmacología , Glándula Tiroides/citología , Northern Blotting , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/fisiología , Expresión Génica , Insulina/farmacología , Cinética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Fosforilación , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , ARN Mensajero/análisis , Receptores de Somatostatina/fisiología , Transducción de Señal , Tirotropina/farmacología , Transfección , Proteínas Supresoras de Tumor/metabolismo , Vanadatos/farmacología
4.
Endocrinology ; 138(9): 3756-63, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9275062

RESUMEN

The effects of the stable expression of E1A and/or middle T oncogenes on the proliferative activity of PC Cl3 normal thyroid cells are reported. The proliferation of PC Cl3 cells is mainly regulated by insulin and TSH in a stimulatory way and by somatostatin in an inhibitory fashion. The transformed cell lines, named PC Py and PC E1A Py, show an autonomous pattern of proliferation. The blockade of phosphotyrosine phosphatase activity with vanadate increased the proliferation rate of PC Cl3 under basal and stimulated conditions and completely prevented the inhibitory activity of somatostatin, suggesting that in PC Cl3 cells, a tonic tyrosine phosphatase activity regulates basal and stimulated proliferation, and that a somatostatin-dependent increase in this activity may represent a cytostatic signal. Conversely, in both PC Py and PC E1A Py, vanadate did not modify basal and stimulated proliferation. We analyzed tyrosine phosphatase activity in the different cell lines basally and under conditions leading to the arrest of cell proliferation: confluence (contact inhibition), growth factor deprivation (starvation), and somatostatin treatment. Under basal conditions, tyrosine phosphatase activity was significantly lower in PC Py and PC E1APy cell lines than that in the normal cells. The inhibition of the proliferation induced by contact inhibition or somatostatin treatment was accompanied by an increase in tyrosine phosphatase activity only in PC Cl3 cells. The reduction in tyrosine phosphatase activity in PC E1APy cells correlated with a significant reduction in the expression of R-PTP eta, a tyrosine phosphatase cloned from PC Cl3 cells. Conversely, the expression of another receptor-like PTP, PTP mu, was unchanged. Thus, PTP eta may be a candidate to mediate inhibitory signals (i.e. activation of somatostatin receptors or cell to cell contact) on the proliferative activity of PC Cl3 cells, and the reduction of its expression in the transformed cell lines may lead to an alteration in the control of cell proliferation.


Asunto(s)
División Celular , Transformación Celular Neoplásica/genética , Oncogenes , Proteínas Tirosina Fosfatasas/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/enzimología , Animales , Northern Blotting , Línea Celular , Células Clonales , Medios de Cultivo , ADN/biosíntesis , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Expresión Génica , Sustancias de Crecimiento/administración & dosificación , Humanos , Insulina/farmacología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , Ratas , Somatostatina/farmacología , Vanadatos/farmacología
5.
Endocrinology ; 144(4): 1574-84, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12639942

RESUMEN

Somatostatin was reported to inhibit Kaposi's sarcoma (KS) cell (KS-Imm) xenografts through an antiangiogenic activity. Here, we show that somatostatin blocks growth of established KS-Imm tumors with the same efficacy as adriamycin, a clinically effective cytotoxic drug. Whereas KS-Imm cells do not express somatostatin receptors (SSTRs), endothelial cells express several SSTRs, in particular SSTR3. We investigated the molecular mechanisms and receptor specificity of somatostatin inhibition of angiogenesis. Somatostatin significantly inhibited angiogenesis in vivo in the matrigel sponge assay; this inhibition was mimicked by the SSTR3 agonist L-796778 and reversed by the SSTR3 antagonist BN81658, demonstrating involvement of SSTR3. In vitro experiments showed that somatostatin directly affected different endothelial cell line proliferation through a block of growth-factor-stimulated MAPK and endothelial nitric oxide (NO) synthase (eNOS) activities. BN81658 reversed somatostatin inhibition of cell proliferation, NO production, and MAPK activity, indicating that SSTR3 activation is required for the effects of somatostatin in vitro. Finally in vivo angiogenesis assays demonstrated that eNOS inhibition was a prerequisite for the antiangiogenic effects of somatostatin, because high concentrations of sodium nitroprusside, an NO donor, abolished the somatostatin effects. In conclusion, we demonstrate that somatostatin is a powerful antitumor agent in vivo that inhibits tumor angiogenesis through SSTR3-mediated inhibition of both eNOS and MAPK activities.


Asunto(s)
Hormonas/farmacología , Neovascularización Patológica/tratamiento farmacológico , Óxido Nítrico Sintasa/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/farmacología , Amidas/farmacología , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Activación Enzimática/efectos de los fármacos , Humanos , Técnicas In Vitro , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Nitrobencenos/farmacología , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inhibidores , Venas Umbilicales/citología
6.
J Mol Endocrinol ; 23(3): 299-306, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10601975

RESUMEN

The site of inhibition, by melatonin, of GnRH-dependent testosterone secretion was investigated in adult rat Leydig cells cultured in vitro. The various effects downstream of the binding of GnRH to its own receptor were isolated and mimicked by specific drugs. Testosterone secretion was then evaluated after 3 h stimulation with GnRH, thapsigargin (1 microM), phorbol-12-myristate-13-acetate (100 nM), arachidonic acid (20 microM), and ionomycin (1 microM) in the presence or absence of melatonin (215 nM). The effect of melatonin on the GnRH-induced changes in cytoplasmic calcium concentration ([Ca(2+)](i)) was also studied, using Fura-2 as fluorescent Ca(2+) indicator. Melatonin attenuated the increase in [Ca(2+)](i) and inhibited the testosterone secretion induced by GnRH, but not that induced by ionomycin. Both ionomycin and thapsigargin potentiated GnRH-induced testosterone secretion; however, ionomycin, but not thapsigargin, partially prevented the inhibitory effect of melatonin on cells stimulated with GnRH. The effect of melatonin was probably dependent on the binding of melatonin to its Gi-protein-coupled receptor, as the inhibitory effect on GnRH-induced secretion was supressed in cells pretreated with pertussis toxin in a concentration of 180 ng/ml for 20 h. Assay of 17-hydroxy-progesterone showed that, irrespective of the treatment, cells cultured with melatonin secreted greater amounts than controls. We conclude that melatonin reduces GnRH-induced testosterone secretion by 1) decreasing [Ca(2+)](i), through impairment of the GnRH-dependent release of Ca(2+) from intracellular stores and 2) blocking 17-20 desmolase enzymatic activity, an effect that occurs irrespective of changes in [Ca(2+)](i).


Asunto(s)
Calcio/metabolismo , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/farmacología , Células Intersticiales del Testículo/efectos de los fármacos , Melatonina/farmacología , Testosterona/metabolismo , 17-alfa-Hidroxiprogesterona/metabolismo , Animales , Ácido Araquidónico/farmacología , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Fura-2 , Ionomicina/farmacología , Células Intersticiales del Testículo/enzimología , Células Intersticiales del Testículo/metabolismo , Hormona Luteinizante/farmacología , Masculino , Melatonina/antagonistas & inhibidores , Toxina del Pertussis , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Estaurosporina/farmacología , Acetato de Tetradecanoilforbol/farmacología , Tapsigargina/antagonistas & inhibidores , Tapsigargina/farmacología , Factores de Virulencia de Bordetella/farmacología
7.
Neuroscience ; 74(1): 99-105, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8843080

RESUMEN

The aim of the present work was to investigate the mechanism by which the diffusible factor nitric oxide regulates GABAA receptor function in the brain. The effect of nitric oxide on GABAA receptor function has been studied in two different neuronal preparations: rat cerebral cortex microsacs and rat cerebellum granule cells in culture. In the first case, GABA-stimulated 36Cl-accumulation was studied as an index of GABAA receptor function. The maximal rate of GABA-stimulated 36Cl- accumulation (Vmax) was reduced by treatment of microsacs with nitric oxide chemical donors such as sodium nitroprusside (-26%) and S-nitroso-acetyl-penicillamine (-11%). The greater effect of the former agent is due to an additional interference by its breakdown products. The biochemical precursor L-arginine (1 mM) produced the same Vmax decrease as S-nitroso-acetyl-penicillamine. This effect was reversed by a nitric oxide synthase blocker and appears truly nitric oxide mediated. The action of nitric oxide in this system does not seem to imply cyclic GMP formation. GABAA receptor function was studied by whole-cell patch-clamp in rat cerebellum granule cells in culture. In this case, L-arginine (100 microM) profoundly reduced the Cl- current elicited by 10 microM GABA and its effect subsided following washing out. The effect of L-arginine was observed almost exclusively on the rapidly desensitizing component of the GABA-activated current. The action of L-arginine was blocked by a protein kinase G inhibitor and mimicked by its activators. Thus, it appears that this effect in these cells involves nitric oxide formation, cyclic GMP accumulation and protein kinase G-catalysed phosphorylation of GABAA receptor.


Asunto(s)
Cerebelo/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Óxido Nítrico/fisiología , Receptores de GABA-A/fisiología , Ácido gamma-Aminobutírico/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
8.
Eur J Endocrinol ; 141(4): 396-408, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10526255

RESUMEN

OBJECTIVE: Somatostatin is a powerful inhibitor of hormone secretion and cell proliferation. Treatment with somatostatin analogs in humans causes a reduction in size and secretory activity of some endocrine tumors, including somatotropic pituitary adenomas. Less studied are the effects of somatostatin agonists on non-functioning pituitary adenomas (NFPAs). In this study we characterized the effects of somatostatin and its analog lanreotide on the proliferation of NFPAs in vitro and the intracellular mechanisms involved. DESIGN: Twenty-three NFPA post-surgical specimens were analyzed for somatostatin receptor (SSTR) expression and 12 of them were cultured in vitro to study somatostatin's effects on cell proliferation, assessed by means of [(3)H]thymidine uptake, and the intracellular signaling. RESULTS: One or more SSTR subtypes were expressed in 90% of the adenomas tested. Somatostatin and lanreotide treatment inhibited phorbol myristate acetate (PMA)-induced cell proliferation. Vanadate pretreatment reversed somatostatin and lanreotide inhibition of PMA-induced DNA synthesis suggesting an involvement of tyrosine phosphatase in this effect. In the only adenoma tested, somatostatin directly induced a tyrosine phosphatase activity. Somatostatin and lanreotide caused also a significant inhibition of voltage-sensitive calcium channel activity induced by 40mmol/l K(+) depolarization in microfluorimetric analysis. CONCLUSIONS: These data show that somatostatin and lanreotide inhibit human NFPA cell proliferation in vitro, and suggest that activation of tyrosine phosphatases and inhibition of the activity of voltage-dependent calcium channels may represent intracellular signals mediating this effect.


Asunto(s)
Adenoma/tratamiento farmacológico , Péptidos Cíclicos/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Somatostatina/análogos & derivados , Somatostatina/farmacología , Adenoma/patología , Canales de Calcio/efectos de los fármacos , División Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Persona de Mediana Edad , Neoplasias Hipofisarias/patología , Células Tumorales Cultivadas
9.
Neurochem Int ; 41(1): 55-63, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11918972

RESUMEN

Prion diseases are fatal neurodegenerative disorders of the CNS of men and animals, characterized by spongiform degeneration of the CNS, astrogliosis and deposition of amyloid into the brain. The conversion of a cellular glycoprotein (the prion protein, PrP(C)) into an altered isoform (the prion scrapie, PrP(Sc)), which accumulates within the brain tissue by virtue of its resistance to the intracellular catabolism, is currently believed to represent the etiologic agent responsible for these diseases. Synthetic or recombinant polypeptides are commonly used to elucidate the mechanism of proteins involved in neurodegenerative diseases. Here we describe a procedure, which allows the synthesis and purification in its native folding, of the human prion protein fragment 90-231, corresponding to the protease resistant core of PrP(Sc). We synthesized the polypeptides 90-231 of both the wild type and the E200K mutant isoforms of PrP. Using a gluthatione S-transferase (GST) fusion protein approach, milligram amounts of polypeptides were obtained after expression in E. coli. The recovery of the purified fusion protein was monitored following the evaluation of the GST activity. The PrP fragment was released from the fusion protein immobilized on a glutathione-coupled agarose resin by direct cleavage with thrombin. The recombinant protein was identified by comassie stained acrylamide gel and by immunoblotting employing a monoclonal anti-PrP antibody. The peptide purified by gel filtration chromatography showed mainly an alpha-helix structure, as analysed by circular dichroism (CD) and an intact disulfide bridge. The same procedure was also successfully employed to synthesize and purify the E200K mutant PrP fragment.


Asunto(s)
Escherichia coli/genética , Priones/genética , Secuencia de Bases , Western Blotting , Cromatografía Liquida , Dicroismo Circular , Cartilla de ADN , Humanos , Espectrometría de Masas , Priones/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Espectrometría de Fluorescencia
10.
Ann N Y Acad Sci ; 1010: 610-22, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15033801

RESUMEN

Prion diseases are neurodegenerative disorders of the central nervous system of humans and animals, characterized by spongiform degeneration of the central nervous system, astrogliosis, and deposition of amyloid into the brain. The conversion of a cellular glycoprotein (prion protein, PrP(C)) into an altered isoform (PrP(Sc)) has been proposed to represent the causative event responsible for these diseases. The peptide corresponding to the residues 106-126 of PrP sequence (PrP106-126) is largely used to explore the neurotoxic mechanisms underlying the prion diseases. We investigated the intracellular signaling responsible for PrP106-126-dependent cell death in the SH-SY5Y human neuroblastoma cell line. In these cells, PrP106-126 treatment induced apoptotic cell death and the activation of caspase-3. The p38 MAP-kinase blockers (SB203580 and PD169316) prevented the apoptotic cell death evoked by PrP106-126 and Western blot analysis revealed that the exposure of the cells to the peptide induced p38 activation. However, whether the neuronal toxicity of PrP106-126 is caused by a soluble or fibrillar form of this peptide is still unknown. In this study, we correlated the structural state of this peptide with its neurotoxicity. We show that the two conserved glycines in position 114 and 119 prevent the peptide to assume a structured conformation, favoring its aggregation in amyloid fibrils. The substitution of both glycines with alanine residues (PrP106-126AA) generates a soluble nonamyloidogenic peptide, that retained its toxic properties when incubated with neuroblastoma cells. These data show that the amyloid aggregation is not necessary for the induction of the toxic effects of PrP106-126.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Apoptosis/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fragmentos de Péptidos/farmacología , Priones/farmacología , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Datos de Secuencia Molecular , Neuroblastoma/patología , Fragmentos de Péptidos/química , Priones/química , Proteínas Quinasas p38 Activadas por Mitógenos
11.
J Physiol Paris ; 94(3-4): 239-50, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11088001

RESUMEN

The mitogen activated protein (MAP) kinase cascade represents one of the major regulator of cell growth by hormones and growth factors. However, although the activation of this intracellular pathway has been often regarded as mediator of cell proliferation, in many cell types the increase in MAP kinase (also called extra-cellular signal regulated kinase: ERK) activity may result in cell growth arrest, depending on the length or the intensity of the stimulation. In this review we examine recent data concerning the effects of somatostatin on the MAP kinase cascade through one of its major receptor subtype, the somatostatin receptor 1 (SSTR1), stably expressed in CHO-K1 cells. Somatostatin inhibits the proliferative effects of basic FGF (bFGF) in CHO-SSTR1 cell line. However, in these cells, somatostatin robustly activates the MAP kinase and augments bFGF-induced stimulation of ERK. We show that the activation of ERK via SSTR1 is mediated by the betagamma subunit of a pertussis toxin-sensitive G-protein and requires both the small G protein Ras and the serine/threonine kinase Raf-1. Moreover the phosphatidyl inositol-3kinase and the cytosolic tyrosine kinase c-src participate in the signal transduction regulated by SSTRI to activate ERK, as well as it is involved the protein tyrosine phosphatase (PTP) SHP-2. Previous studies have suggested that somatostatin-stimulated PTP activity mediates the growth inhibitory actions of somatostatin, in CHO-SSTR1 cells. Thus, the activation of SHP-2 by SSTR1 may mediate the antiproliferative activity of somatostatin. SHP-2 may. in turn, regulate the activity of kinases upstream of ERK that require tyrosine dephosphorylation to be activated, such as c-src. Finally, the synergism between somatostatin and bFGF in the activation of ERK results in an increased expression of the cyclin-dependent kinase inhibitor p21cip/WAF1 as molecular effector of the antiproliferative activity of somatostatin.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Células CHO , División Celular/fisiología , Cricetinae , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6
12.
Int J Dev Neurosci ; 18(4-5): 481-92, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10817932

RESUMEN

Prion encephalopathies include fatal diseases of the central nervous system of men and animals characterized by nerve cell loss, glial proliferation and deposition of amyloid fibrils into the brain. During these diseases a cellular glycoprotein (the prion protein, PrP(C)) is converted, through a not yet completely clear mechanism, in an altered isoform (the prion scrapie, PrP(Sc)) that accumulates within the brain tissue by virtue of its resistance to the intracellular catabolism. PrP(Sc) is believed to be responsible for the neuronal loss that is observed in the prion disease. The PrP 106-126, a synthetic peptide that has been obtained from the amyloidogenic portion of the prion protein, represents a suitable model for studying the pathogenic role of the PrP(Sc), retaining, in vitro, some characteristics of the entire protein, such as the capability to aggregate in fibrils, and the neurotoxicity. In this work we present the results we have recently obtained regarding the action of the PrP 106-126 in different cellular models. We report that the PrP 106-126 induces proliferation of cortical astrocytes, as well as degeneration of primary cultures of cortical neurons or of neuroectodermal stable cell lines (GH(3) cells). In particular, these two opposite effects are mediated by the same attitude of the peptide to interact with the L-type calcium channels: in the astrocytes, the activity of these channels seems to be activated by PrP 106-126, while, in the cortical neurons and in the GH(3) cells, the same treatment causes a blockade of these channels causing a toxic effect.


Asunto(s)
Astrocitos/patología , Muerte Celular/fisiología , Gliosis/patología , Neuronas/patología , Fragmentos de Péptidos/toxicidad , Priones/toxicidad , Secuencia de Aminoácidos , Animales , Astrocitos/metabolismo , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Muerte Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Corteza Cerebral/citología , Síndrome de Creutzfeldt-Jakob/metabolismo , ADN/biosíntesis , Humanos , Etiquetado Corte-Fin in Situ , Datos de Secuencia Molecular , Neuronas/química , Neuronas/metabolismo , Nicardipino/farmacología , Fragmentos de Péptidos/química , Cloruro de Potasio/farmacología , Priones/química , Ratas , Timidina/farmacocinética , Tritio
13.
Naunyn Schmiedebergs Arch Pharmacol ; 349(6): 555-8, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7969504

RESUMEN

The existence of a pharmacological heterogeneity among the glutamate receptors sensitive to (RS)-alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) has been investigated in the adult rat central nervous system (CNS). AMPA stimulated [3H]noradrenaline ([3H]NA) release from hippocampal synaptosomes (pD2 = 4.58) and the production of cGMP in cerebellar slices (pD2 = 7.75). The AMPA effects in the two systems were tested against several glutamate receptor antagonists including 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX), 6,7-dinitro-quinoxaline-2,3-dione (DNQX), L-glutamate diethylester (GDEE), gamma-D-glutamyl-glycine (GDGG) and gamma-D-glutamyl-aminomethylsulphonate (GAMS). In both systems the AMPA effect was equally sensitive to CNQX or DNQX. However, while the AMPA-evoked increase of [3H]NA release from presynaptic terminals was not affected by GAMS, GDGG or GDEE, the postsynaptic cGMP response was prevented by GDGG and GDEE. It is concluded that rat hippocampus and cerebellum possess, respectively, presynaptic and postsynaptic AMPA-sensitive receptors involved in different functions and endowed with diverse pharmacological properties.


Asunto(s)
Sistema Nervioso Central/metabolismo , Receptores AMPA/antagonistas & inhibidores , Animales , Sistema Nervioso Central/efectos de los fármacos , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , GMP Cíclico/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Técnicas In Vitro , Masculino , Norepinefrina/administración & dosificación , Norepinefrina/metabolismo , Norepinefrina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
14.
Naunyn Schmiedebergs Arch Pharmacol ; 348(4): 347-51, 1993 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7904049

RESUMEN

The glutamatergic mossy fibre-->granule cell pathway has been investigated in rat cerebellar slices. Exposure to 35 mM KCl, a concentration of K+ known to elicit Ca(2+)-dependent releases of excitatory amino acids from cerebellar slices, raised cGMP levels. The cGMP response was decreased in a concentration-dependent manner by D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5) and by 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) indicating the involvement of ionotropic glutamate receptors of both the N-methyl-D-aspartate (NMDA) and the non-NMDA type. The K(+)-evoked production of cGMP was potently inhibited (EC50 = 1.21 nM) by 1-(2,5-dimethoxy-4-iodophenyl)-2-amino-propane (DOI), a selective 5-HT2 receptor agonist. The effect of DOI (0.01 microM) was antagonized by 0.03 microM of the 5-HT2 receptor antagonists ketanserin and methiothepin. At concentrations higher than 0.1 microM, both antagonists increased on their own the cGMP response elicited by high-K+. This effect was insensitive to tetrodotoxin. It had been previously shown that rat mossy fibre endings release glutamate upon depolarization and that such release can be inhibited by activation of 5-HT2 receptors sited on the mossy fibre endings. Altogether the available data suggest the following conclusions: a) the glutamate/aspartate endogenously released in cerebellar slices during K+ depolarization increase cGMP synthesis through the activation of both NMDA and non-NMDA receptors; b) a portion of the cGMP response can be prevented by 5-HT2 receptor activation and may reflect the activity of the mossy fibre--granule cell pathway.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Cerebelo/fisiología , Fibras Nerviosas/fisiología , Agonistas de Receptores de Serotonina/farmacología , Serotonina/fisiología , Transmisión Sináptica/fisiología , 2-Amino-5-fosfonovalerato/farmacología , 6-Ciano 7-nitroquinoxalina 2,3-diona , Anfetaminas/farmacología , Animales , Cerebelo/citología , AMP Cíclico/metabolismo , Gránulos Citoplasmáticos/fisiología , Técnicas In Vitro , Ketanserina/farmacología , Masculino , Metiotepina/farmacología , Potasio/farmacología , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/efectos de los fármacos , Antagonistas de la Serotonina , Tetrodotoxina/farmacología
15.
Life Sci ; 64(18): 1661-74, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10328526

RESUMEN

It is well-known that nucleotides, nucleosides and purine/pyrimidine bases enhance cell proliferation in vitro. Nevertheless, the molecular mechanisms involved in this mitogenic activity is still controversial, since these compounds are reported both to synergize with growth factor, and to act directly on purinergic receptor inducing per se a proliferative response. It was suggested that cell growth enhancement could be mediated by the A2 purinergic receptor activation. Here we report that a polydeoxyribonucleotide (PDRN) and adenosine are able to increase, the growth rate of human skin fibroblasts in primary cultures. The proliferative activity exerted by PDRN was significantly counteracted by the A2 antagonist 3, 7-Dimethyl-1-propargylxanthine (DMPX), but not by the A1 antagonist 8-cyclopentyl-1,3-dipropylxanthine (PD 116,948, DPCPX). Accordingly, the trophic action of PDRN was mimicked by the A2 agonist N6-[2-(3,5-Dimethoxyphenyl)-2-(methylphenyl)-ethyl]adenosine (DPMA), while the A1 agonist N6-Cyclopenthyladenosine (CPA) did not show any effect. In microfluorimetric studies, we observed that PDRN and adenosine increased the concentration of cytosolic calcium ions. The PDRN-evoked calcium rise was dose-dependent and DMPX sensitive. Taken together, our results suggest that PDRN may operate as a pro-drug providing the cultured cells with an effective amount of mitogenic deoxyribonucleotides, deoxyribonucleosides and bases; moreover, cell proliferation enhancement that has been induced by PDRN seems to be mediated, at least in part, by the activation of purinergic receptors of the A2 subtype.


Asunto(s)
División Celular/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , Receptores Purinérgicos P1/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Calcio/metabolismo , Citofotometría , Desoxirribonucleósidos/farmacología , Desoxirribonucleótidos/farmacología , Fibroblastos , Fura-2 , Humanos , Placenta/química , Sistemas de Mensajero Secundario , Teobromina/análogos & derivados , Teobromina/farmacología , Xantinas/farmacología
16.
Minerva Endocrinol ; 26(4): 273-6, 2001 Dec.
Artículo en Italiano | MEDLINE | ID: mdl-11782715

RESUMEN

Somatostatin and its analogs are active in the inhibition of the proliteration of sst receptor positive endocrine neoplasms, however their activity and mechanism in non-endocrine tumors is not clear. Somatostatin effectively inhibited the growth of a Kaposi's sarcoma xenograft in nude mice, yet in vitro the tumor cells did not express any known somatostatin receptors and were not growth inhibited by somatostatin. Histological examination revealed limited vascularization in the somatostatin treated tumors as compared to the controls. Somatostatin was a potent inhibitor of angiogenesis in an in vivo assay. In vitro, somatostatin inhibited endothelial cell growth and invasion. Migration of monocytes, important mediators of the angiogenic cascade, was also inhibited by somatostatin. Both cells types expressed somatostatin receptor mRNAs. These data demonstrate that somatostatin is a potent anti-tumor angiogenesis compound directly affecting both endothelial and monocytic cells. The debated function of somatostatin in tumor treatment and the design of therapeutic protocols should be reexamined considering these data.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antagonistas de Hormonas/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Sarcoma de Kaposi/tratamiento farmacológico , Sarcoma de Kaposi/patología , Somatostatina/análogos & derivados , Somatostatina/uso terapéutico , Animales , Humanos , Ratones , Trasplante de Neoplasias , Neovascularización Patológica/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Kaposi/diagnóstico
17.
Minerva Endocrinol ; 26(4): 277-83, 2001 Dec.
Artículo en Italiano | MEDLINE | ID: mdl-11782716

RESUMEN

BACKGROUND: This study was aimed to identify possible intracellular effectors of the somatostatin (SST) antiproliferative activity, in PCCl3 thyroid cells. METHODS: To prove the involvement of r-PTPeta in SST's effect, we studied th proliferative activity of subclones of PCCl3 cells that do or do not express this PTP. RESULTS: SST inhibited PCCl3 TSH+insulin-dependent cell proliferation through the induction of a phosphotyrosine phosphatase (PTP) activity, detected using the synthetic substrate pNPP (+150%, p<0.01). Conversely, PCCl3 cells stably expressing the v-mos oncogene (PCmos) were completely insensitive to SST antiproliferative effects due to the incapability of SST to increase PTP activity, that correlated with the abolishment of the expression of the receptor-like PTP, r-PTPeta. In the cells in which r-PTPeta was transfected (PCmos/ PTPeta) SST inhibited cell proliferation showing a dose-dependence similar to that observed in PCCl3 cells. Conversely, the transfection of a catalytically inactive mutant of r-PTPeta did not restore the responsivity to SST. Also in PCmos/PTPeta cells SST, treatment increased membrane PTP activity. CONCLUSIONS: SST inhibition of PCC13 cell proliferation requires the activation of r-PTPeta.


Asunto(s)
Antagonistas de Hormonas/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Somatostatina/farmacología , Glándula Tiroides/citología , Animales , División Celular , Línea Celular , ADN/biosíntesis , ADN/genética , Activación Enzimática/efectos de los fármacos , Isoenzimas/metabolismo , Ratas , Glándula Tiroides/efectos de los fármacos
18.
Cell Death Dis ; 2: e138, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21451573

RESUMEN

To define the mechanisms by which hPrP90-231 induces cell death, we analyzed its interaction with living cells and monitored its intracellular fate. Treatment of SH-SY5Y cells with fluorescein-5-isothiocyanate (FITC)-conjugated hPrP90-231 caused the accumulation of cytosolic aggregates of the prion protein fragment that increased in number and size in a time-dependent manner. The formation of large intracellular hPrP90-231 aggregates correlated with the activation of apoptosis. hPrP90-231 aggregates occurred within lysotracker-positive vesicles and induced the formation of activated cathepsin D (CD), indicating that hPrP90-231 is partitioned into the endosomal-lysosomal system structures, activating the proteolytic machinery. Remarkably, the inhibition of CD activity significantly reduced hPrP-90-231-dependent apoptosis. Internalized hPrP90-231 forms detergent-insoluble and SDS-stable aggregates, displaying partial resistance to proteolysis. By confocal microscopy analysis of lucifer yellow (LY) intracellular partition, we show that hPrP90-231 accumulation induces lysosome destabilization and loss of lysosomal membrane impermeability. In fact, although control cells evidenced a vesicular pattern of LY fluorescence (index of healthy lysosomes), hPrP90-231-treated cells showed diffuse cytosolic fluorescence, indicating LY diffusion through damaged lysosomes. In conclusion, these data indicate that exogenously added hPrP90-231 forms intralysosomal deposits having features of insoluble, protease-resistant aggregates and could trigger a lysosome-mediated apoptosis by inducing lysosome membrane permeabilization, followed by the release of hydrolytic enzymes.


Asunto(s)
Apoptosis , Endopeptidasa K/metabolismo , Lisosomas/metabolismo , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Muerte Celular , Línea Celular Tumoral , Citosol/química , Citosol/metabolismo , Humanos , Lisosomas/química , Proteínas PrPC/genética , Enfermedades por Prión/genética , Enfermedades por Prión/fisiopatología , Solubilidad
19.
Pharmacol Res ; 33(6): 297-305, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8971951

RESUMEN

This review systematically analyses recent knowledge of the biology of somatostatin receptors. Indeed, since the molecular cloning of five somatostatin receptors in 1992, a growing bulk of scientific data has been produced regarding the cell type localization, the physiological role and the biochemical intracellular pathways activated by the single somatostatin receptors. The aim of this review is to present all these data, also discussing the conflicting evidence that has been reported, to further simulate research efforts in the field.


Asunto(s)
Química Encefálica/fisiología , Receptores de Somatostatina/metabolismo , Transducción de Señal/fisiología , Animales , Clonación Molecular , Proteínas de Unión al GTP/metabolismo , Humanos , Ratas , Receptores de Somatostatina/química
20.
Neurochem Res ; 22(12): 1517-21, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9357019

RESUMEN

The effect of nitric oxide donors and L-arginine on the uptake of GABA was studied in synaptosomes purified from rat brain. The neurotransmitter uptake was significantly reduced by S-nitrosoacetylpenicillamine and by sodium nitroprusside, although in this case to a lesser extent. A slight inhibitory effect was found preincubating rat brain synaptosomes with 1 mM L-arginine as well. The S-nitrosoacetylpenicillamine effect gradually disappeared with decomposition of the substance by exposure to light. The nitric oxide effect appears to be mainly due to a decrease in the V for synaptosomal GABA uptake and seems to be related to a partial collapse of nerve endings ionic gradients. Functionally, it could result over time in a reduced availability of GABA at the synapses involved.


Asunto(s)
Encéfalo/efectos de los fármacos , Óxido Nítrico/farmacología , Sinaptosomas/efectos de los fármacos , Ácido gamma-Aminobutírico/efectos de los fármacos , Animales , Arginina/farmacología , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Masculino , Óxido Nítrico/metabolismo , Nitroarginina/farmacología , Nitroprusiato/farmacología , Penicilamina/análogos & derivados , Penicilamina/farmacología , Ratas , Ratas Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina , Sinaptosomas/metabolismo , Ácido gamma-Aminobutírico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA