Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Immunol ; 175(3): 458-67, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24215151

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by pronounced inflammation and leucocyte infiltration in affected joints. Despite significant therapeutic advances, a new targeted approach is needed. Our objective in this work was to investigate the anti-inflammatory effects of the Ras inhibitor farnesylthiosalicylic acid (FTS) on adjuvant-induced arthritis (AIA) in rats, an experimental model for RA. Following AIA induction in Lewis rats by intradermal injection of heat-killed Mycobacterium tuberculosis, rats were treated with either FTS or dexamethasone and assessed daily for paw swelling. Joints were imaged by magnetic resonance imaging and computerized tomography and analysed histologically. The anti-inflammatory effect of FTS was assessed by serum assay of multiple cytokines. After adjuvant injection rats demonstrated paw swelling, leucocyte infiltration, cytokine secretion and activation of Ras-effector pathways. Upon FTS treatment these changes reverted almost to normal. Histopathological analysis revealed that the synovial hyperplasia and leucocyte infiltration observed in the arthritic rats were alleviated by FTS. Periarticular bony erosions were averted. Efficacy of FTS treatment was also demonstrated by inhibition of CD4(+) and CD8(+) T cell proliferation and of interferon (IFN)-γ, tumour necrosis factor (TNF)-α, interleukin (IL)-6 and IL-17 release. The Ras effectors PI3K, protein kinase B (AKT), p38, and extracellular-regulated kinase (ERK) were significantly attenuated and forkhead box protein 3 (FoxP3) transcription factor, a marker of regulatory T cells, was significantly increased. Thus, FTS possesses significant anti-inflammatory and anti-arthritic properties and accordingly shows promise as a potential therapeutic agent for RA. Its effects are apparently mediated, at least in part, by a decrease in proinflammatory cytokines.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Experimental/metabolismo , Citocinas/metabolismo , Farnesol/análogos & derivados , Mediadores de Inflamación/metabolismo , Salicilatos/farmacología , Animales , Antiinflamatorios/administración & dosificación , Artritis Experimental/diagnóstico , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Modelos Animales de Enfermedad , Farnesol/administración & dosificación , Farnesol/farmacología , Articulaciones/efectos de los fármacos , Articulaciones/metabolismo , Articulaciones/patología , Masculino , Ratas , Salicilatos/administración & dosificación , Transducción de Señal , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo
2.
Science ; 242(4876): 268-70, 1988 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-2845579

RESUMEN

Sarafotoxins, a group of 21-residue cardiotoxic peptides from snake venom that induce coronary vasoconstriction, show high-affinity binding to rat atrial and brain membranes and activate the hydrolysis of phosphoinositides. Neither their binding nor their activity is affected by blockers or activators of known receptors and ion channels, suggesting that sarafotoxins act either directly on the phosphoinositide phosphodiesterase system or on a novel receptor. Their amino acid sequence shows a high degree of homology with that of endothelin, a recently described 21-residue vasoconstrictor peptide found in porcine aortic endothelium. This is remarkable, since endothelin is a natural compound of the mammalian vascular system while sarafotoxins are highly toxic components of snake venom.


Asunto(s)
Encéfalo/metabolismo , Miocardio/metabolismo , Fosfatidilinositoles/metabolismo , Venenos de Víboras/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Membrana Celular/metabolismo , Endotelinas , Activación Enzimática , Atrios Cardíacos/metabolismo , Hidrólisis , Inositol/metabolismo , Datos de Secuencia Molecular , Péptidos , Fosfatidilinositol Diacilglicerol-Liasa , Hidrolasas Diéster Fosfóricas/metabolismo , Ratas , Homología de Secuencia de Ácido Nucleico , Vasoconstricción
3.
Cell Death Differ ; 14(5): 895-906, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17096025

RESUMEN

Neurofibromatosis type 1 (NF1) is characterized by a high incidence of benign and malignant tumors attributed to loss of function of Nf1, which encodes neurofibromin, a tumor suppressor with Ras-GAP activity. Neurofibromin deficiency typically causes chronic activation of Ras, considered the major contributor to manifestation of NF1. Resistance to radio- and chemotherapy are typical of NF1-associated tumors, but the underlying mechanism is unknown. Here, we investigated interrelationships between neurofibromin expression, Ras activity, and sensitivity to apoptosis. Neurofibromin-deficient mouse embryonic fibroblasts (MEFs) and human NF1 tumor cells were more resistant than neurofibromin-expressing cells to apoptosis. Moreover, Nf1(-/-), Nf1(+/-), and Nf1(+/+) MEFs exhibited gene-dosage-related resistance to apoptosis. Resistance of the Nf1-deficient cells was mediated by two survival pathways: a Ras-dependent pathway, and a Ras-independent pathway promoted by the lack of an NF1-GRD-independent proapoptotic action of neurofibromin. Therefore, besides its Ras-dependent growth inhibition, neurofibromin can exert tumor suppression via a proapoptotic effect.


Asunto(s)
Apoptosis , Neurofibromina 1/metabolismo , Proteínas ras/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Transformada , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Farnesol/análogos & derivados , Farnesol/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Dosificación de Gen/efectos de los fármacos , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Ratones , Neurofibromina 1/deficiencia , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Salicilatos/farmacología , Virus 40 de los Simios , Serina-Treonina Quinasas TOR , Proteínas ras/antagonistas & inhibidores
4.
Oncogene ; 18(22): 3354-64, 1999 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-10362356

RESUMEN

We have examined fibroblast growth factor (FGF) receptor-1 mediated signal transduction in differentiation of endothelial cells (EC). The activated FGFR-1 couples to Ras through two adaptor proteins, FRS2 and Shc. In FGF-2 treated proliferating EC, FRS2 as well as Shc are tyrosine phosphorylated and interact with Grb2. In contrast, in FGF-2 treated differentiating cells, Shc, but not FRS2, is engaged in Grb2-interactions. Sustained MAP kinase activity has previously been implicated in differentiation. In FGF stimulated proliferating and differentiating endothelial cells, the MAP kinase Erk2 is activated in a sustained manner. Inhibition of MEK and MAP kinase activity by PD98059 treatment of cells, still allows EC tube formation. The FGFR-1 mediates activation of protein kinase C (PKC) through direct binding and activation of phospholipase C-gamma (PLC-gamma), and has also been shown to activate the cytoplasmic tyrosine kinase Src. Treatment of the cells with the PKC inhibitor bisindolylmaleimide does not prevent tube formation. In contrast, Src kinase activity is a prerequisite for EC differentiation, since treatment of the cells with PP1, a Src family specific inhibitor, abrogates tube formation. In differentiating EC, FGF-2 induces complex formation between Src and focal adhesion kinase (FAK). These data indicate that the Ras pathway is initiated via Shc or FRS2, dependent on the cellular program. Blocking the function of Src family kinases, attenuates differentiation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Endotelio Vascular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Genes ras , Dominios Homologos src , Animales , Becaplermina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , División Celular/fisiología , Línea Celular , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Flavonoides/farmacología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Proteína Adaptadora GRB2 , Indoles/farmacología , Isoenzimas/efectos de los fármacos , Isoenzimas/metabolismo , Maleimidas/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Fosfolipasa C gamma , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/efectos de los fármacos , Fosfolipasas de Tipo C/metabolismo , Proteínas ras/genética
5.
Oncogene ; 20(51): 7486-93, 2001 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-11709720

RESUMEN

Ras genes, frequently mutated in human tumors, promote malignant transformation. Ras transformation requires membrane anchorage, which is promoted by Ras farnesylcysteine carboxymethylester and by a second signal. Previously we showed that the farnesylcysteine mimetic, farnesylthiosalicylic acid (FTS) disrupts Ras membrane anchorage. To understand how this disruption contributes to inhibition of cell transformation we searched for new Ras-interacting proteins and identified galectin-1, a lectin implicated in human tumors, as a selective binding partner of oncogenic H-Ras(12V). The observed size of H-Ras(12V)-galectin-1 complex, which is equal to the sum of the molecular weights of Ras and galectin-1 indicates a direct binding interaction between the two proteins. FTS disrupted H-Ras(12V)-galectin-1 interactions. Overexpression of galectin-1 increased membrane-associated Ras, Ras-GTP, and active ERK resulting in cell transformation, which was blocked by dominant negative Ras. Galectin-1 antisense RNA inhibited transformation by H-Ras(12V) and abolished membrane anchorage of green fluorescent protein (GFP)-H-Ras(12V) but not of GFP-H-Ras wild-type (wt), GFP-K-Ras(12V), or GFP-N-Ras(13V). H-Ras(12V)-galectin-1 interactions establish an essential link between two proteins associated with cell transformation and human malignancies that can be exploited to selectively target oncogenic Ras proteins.


Asunto(s)
Membrana Celular/metabolismo , Genes ras/genética , Hemaglutininas/metabolismo , Proteínas ras/metabolismo , Animales , División Celular , Transformación Celular Neoplásica , ADN Complementario/metabolismo , Regulación hacia Abajo , Galectina 1 , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Mutación , Oligonucleótidos Antisentido/metabolismo , Plásmidos/metabolismo , Unión Proteica , Ratas , Transfección
6.
Oncogene ; 19(51): 5926-35, 2000 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-11127824

RESUMEN

Expression of oncogenic Ras in epithelial tumor cells is linked to the loss of transforming growth factor-beta (TGF-beta) anti-proliferative activity, and was proposed to involve inhibition of Smad2/3 nuclear translocation. Here we studied several epithelial cell lines expressing oncogenic N-RasK61 and show that TGF-beta-induced nuclear translocation of and transcriptional activation by Smad2/3 were unaffected. In contrast, oncogenic Ras mediated nuclearto-cytoplasmic mislocalization of p27KiP1 (p27) and of the cyclin-dependent kinase (CDK) CDK6, but not CDK2. Concomitantly, oncogenic Ras abrogated the ability of TGF-beta to release p27 from CDK6, to enhance its binding to CDK2 and to inhibit CDK2 activity. Inactivation of Ras by a specific antagonist restored the growth inhibitory response to TGF-beta with concurrent normalization of p27 and CDK6 localization. Therefore, the disruption of TGF-beta-mediated growth inhibition by oncogenic Ras appears to be due to lack of inhibition of CDK2, caused by the sequestration of p27 and CDK2 in different subcellular compartments and by the loss of TGF-beta-induced partner switching of p27 from CDK6 to CDK2.


Asunto(s)
Quinasas Ciclina-Dependientes , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Supresoras de Tumor , Proteínas ras/fisiología , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Quinasa 6 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Citoplasma/metabolismo , Expresión Génica , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Transactivadores/metabolismo , Transcripción Genética/fisiología , Factor de Crecimiento Transformador beta , Células Tumorales Cultivadas , Proteínas ras/biosíntesis , Proteínas ras/genética
7.
Oncogene ; 18(16): 2579-88, 1999 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-10353601

RESUMEN

Constitutively active Ras proteins, their regulatory components, and overexpressed tyrosine kinase receptors that activate Ras, are frequently associated with cell transformation in human tumors. This suggests that functional Ras antagonists may have anti-tumor activity. Studies in rodent fibroblasts have shown that S-trans, transfarnesylthiosalicylic acid (FTS) acts as a rather specific nontoxic Ras antagonist, dislodging Ras from its membrane anchorage domains and accelerating its degradation. FTS is not a farnesyltransferase inhibitor, and does not affect Ras maturation. Here we demonstrate that FTS also acts as a functional Ras antagonist in human pancreatic cell lines that express activated K-Ras (Panc-1 and MiaPaCa-2). In Panc-1 cells, FTS at a concentration of 25-100 microM reduced the amount of Ras in a dose-dependent manner and interfered with serum-dependent and epidermal growth factor-stimulated ERK activation, thus inhibiting both anchorage-dependent and anchorage-independent growth of Panc-1 cells in vitro. FTS also inhibited tumor growth in Panc-1 xenografted nude mice, apparently without systemic toxicity. Daily FTS treatment (5 mg/kg intraperitoneally) in mice with tumors (mean volume 0.07 cm3) markedly decreased tumor growth (after treatment for 18 days, tumor volume had increased by only 23+/-30-fold in the FTS-treated group and by 127+/-66-fold in controls). These findings suggest that FTS represents a new class of functional Ras antagonists with potential therapeutic value.


Asunto(s)
Antineoplásicos/uso terapéutico , Farnesol/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Salicilatos/uso terapéutico , Proteínas ras/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/toxicidad , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Farnesol/química , Farnesol/uso terapéutico , Farnesol/toxicidad , Humanos , Ratones , Ratones Desnudos , Salicilatos/química , Salicilatos/toxicidad , Células Tumorales Cultivadas
8.
Biochim Biophys Acta ; 801(3): 342-50, 1984 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-6487649

RESUMEN

Incorporation of unsaturated fatty acids into membrane fragments from rat brain cortex and medulla pons selectively increased the affinity of the muscarinic agonist, carbamylcholine. The affinity and number of binding sites for the labeled antagonist, N-[3H]methyl-4-piperidyl benzilate was unchanged. The effect on agonist binding was most prominent in the cortex, in which carbamylcholine IC50 values were decreased up to 5-fold. Selectivity of the effect was observed with fatty acids of chain length 18-20 carbons, unsaturation in position 11-12, and a cis conformation of the double bond being most effective. The effects of fatty acids on agonist binding were due primarily to alterations in the affinity constants for the binding reaction, with minor increases in the proportion of high-affinity sites. Transition metals selectively increased the percentage of high-affinity sites in the cortex, but in cis-vaccenic-acid-treated membranes more than additive effects of the metal were observed; both were reversed by GTP. GTP also reversed binding parameters in cis-vaccenic-acid-treated medulla membranes to control level. We conclude that the primary effect of the active fatty acids is to alter the thermodynamic properties of muscarinic agonist binding without markedly inducing interconversion.


Asunto(s)
Bencilatos , Carbacol/metabolismo , Corteza Cerebral/metabolismo , Ácidos Grasos/farmacología , Bulbo Raquídeo/metabolismo , Receptores Muscarínicos/metabolismo , Animales , Membrana Celular/metabolismo , Cobalto/farmacología , Ácidos Grasos/metabolismo , Guanosina Trifosfato/farmacología , Piperidinas/metabolismo , Ratas , Receptores Muscarínicos/efectos de los fármacos , Relación Estructura-Actividad
9.
Biochim Biophys Acta ; 1451(1): 187-95, 1999 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-10446400

RESUMEN

Farnesylthiosalicylic acid (FTS), a synthetic analog of the terminal prenylcysteine present in signaling proteins induces generation of superoxide ions, phospholipase C-driven hydrolysis of inositol lipids and calcium elevation in human neutrophils and DMSO-differentiated HL60 cells. These effects were ascribed to an interaction of the analog with elements responsible for recognition of specific prenylated proteins. The present study demonstrated that in addition to the release of intracellular calcium stores, FTS enhanced entry of Ca(2+) and Mn(2+) from the medium. The biphasic dependence of the influx on the concentration of FTS, as well as its insensitivity to inhibition by PMA and La(3+) suggest that the influx pathway activated by FTS is distinct from the previously described store-operated calcium channels of neutrophils. Consistent with the participation of a cellular membrane component in the interaction, FTS enhanced (45)Ca uptake in neutrophils and neutrophil cell membranes, but not in multilamellar vesicles. To establish specificity of the farnesyl moiety of FTS (C(15)), effects of three other analogs, geranylthiosalicylate, GTS (C(10)), geranylgeranylthiosalicylate, GGTS (C(20)), as well as the carboxymethyl ester FTS-Me on calcium homeostasis and superoxide production were investigated. GGTS dose-dependently elevated [Ca(2+)](i), induced quenching of the 360 nm Fura-2-calcium fluorescence by Mn(2+) and stimulated superoxide release, while GTS and FTS-Me were inactive. These results defined specific structural requirements for the functional interaction of prenylcysteine analogs with myeloid cells.


Asunto(s)
Calcio/metabolismo , Cisteína/análogos & derivados , Neutrófilos/efectos de los fármacos , Calcio/farmacología , Radioisótopos de Calcio , Medios de Cultivo/metabolismo , Cisteína/farmacología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Colorantes Fluorescentes , Fura-2 , Humanos , Liposomas , Manganeso/farmacología , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Tapsigargina/farmacología
10.
Biochim Biophys Acta ; 845(3): 428-35, 1985 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-4005300

RESUMEN

Binding properties of submaxillary gland muscarinic receptors and agonist-induced saliva secretion were studied in rats subjected to heat acclimation. The maximal binding capacity for the muscarinic antagonist N-[3H]methyl-4-piperidyl benzilate was increased from control value of 0.21 to 0.40 pmol/mg protein within 1-2 days of heat acclimation. The increase in the number of muscarinic receptors per gland (100%) was by far higher than the increase in tissue weight (20%), indicating higher density of receptors in the acinar cells of the treated rats. High levels of receptors coincided with the appearance of high-affinity binding sites for muscarinic agonists (oxotremorine, pilocarpine and carbamylcholine), and with reduced tissue sensitivity to pilocarpine. After 4-8 weeks of heat acclimation, the number of receptors as well as tissue response to pilocarpine returned to control levels. These results suggest a functional correlation between the transient upregulation muscarinic receptors in the submaxillary gland and the physiological activity in salivary secretion, and indicate that the high-affinity muscarinic receptors may attenuate saliva secretion during the initial phase of heat acclimation.


Asunto(s)
Aclimatación , Calor , Receptores Muscarínicos/fisiología , Glándula Submandibular/fisiología , Animales , Regulación de la Temperatura Corporal , Masculino , Parasimpaticomiméticos/metabolismo , Pilocarpina/farmacología , Ratas , Saliva/metabolismo , Glándula Submandibular/efectos de los fármacos
11.
Biochim Biophys Acta ; 1452(3): 228-42, 1999 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-10590312

RESUMEN

Ras proteins regulate cell growth, differentiation and apoptosis. Their activities depend on their anchorage to the inner surface of the plasma membrane, which is promoted by their common carboxy-terminal S-farnesylcysteine and either a stretch of lysine residues (K-Ras 4B) or S-palmitoyl moieties (H-Ras, N-Ras and K-Ras 4A). We previously demonstrated dislodgment of H-Ras from EJ cell membranes by S-trans,trans-farnesylthiosalicylic acid (FTS), and proposed that FTS disrupts the interactions between the S-prenyl moiety of Ras and the membrane anchorage domains. In support of this hypothesis, we now show that FTS, which is not a farnesyltransferase inhibitor, inhibits growth of NIH3T3 cells transformed by the non-palmitoylated K-Ras 4B(12V) or by its farnesylated, but unmethylated, K-Ras 4B(12) CVYM mutant. The growth-inhibitory effects of FTS followed the dislodgment and accelerated degradation of K-Ras 4B(12V), leading in turn to a decrease in its amount in the cells and inhibition of MAPK activity. FTS did not affect the rate of degradation of the K-Ras 4B, SVIM mutant which is not modified post-translationally, suggesting that only farnesylated Ras isoforms are substrates for facilitated degradation. The putative Ras-recognition sites (within domains in the cell membrane) appear to tolerate both C(15) and C(20) S-prenyl moeities, since geranylgeranyl thiosalicylic acid mimicked the growth-inhibitory effects of FTS in K-Ras 4B(12V)-transformed cells and FTS inhibited the growth of cells transformed by the geranylgeranylated K-Ras 4B(12V) CVIL isoform. The results suggest that FTS acts as a domain-targeted compound that disrupts Ras-membrane interactions. The fact that FTS can target K-Ras 4B(12V), which is insensitive to inhibition by farnesyltransfarase inhibitors, suggests that FTS may target Ras (and other prenylated proteins important for transformed cell growth) in an efficient manner that speaks well for its potential as an anticancer therapeutic agent.


Asunto(s)
Membrana Celular/efectos de los fármacos , Farnesol/análogos & derivados , Proteínas Proto-Oncogénicas p21(ras)/genética , Salicilatos/farmacología , Células 3T3 , Animales , División Celular/efectos de los fármacos , Línea Celular Transformada , Inhibidores Enzimáticos/farmacología , Farnesol/farmacología , Marcación de Gen , Genes ras/efectos de los fármacos , Ratones , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mutación , Isoformas de Proteínas/genética
12.
Biochim Biophys Acta ; 1226(3): 330-6, 1994 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-8054365

RESUMEN

The processing of ras and of other GTP-binding proteins includes a final reversible step in which the carboxy terminal prenylated cysteine is methylated by the enzyme prenylated protein methyltransferase (PPMTase). The significance of this modification and of the role of PPMTase in human tumors has yet to be fully elucidated. Here we characterize the PPMTase of human endometrial carcinomas (tumors in which the frequency of ras gene mutations is relatively high) and compare it to the PPMTase of the normal endometrium. Our results show that in both types of tissues the enzyme is bound to the membranes. It can utilize synthetic substrates such as N-acetyl-S-farnesyl-L-cysteine (Km = 18-20 microM) and is blocked by the PPMTase inhibitor S-farnesylthioacetic acid (Ki = 2 microM). In vitro methylation assays and [alpha-32P]GTP blot-overlay assays showed that the major endogenous PPMTase substrates are small GTP-binding proteins. Methylation of these proteins in vitro is blocked by farnesylthioacetic acid. The kinetic properties of PPMTase from the carcinomas and the normal tissues are very similar. However, levels of PPMTase activity (but not of its endogenous substrates) are higher in the carcinomatous endometrium than in the normal one. The elevated enzyme activity is restricted to the crude mitochondrial fraction (8.0 +/- 0.4 vs. 5.4 +/- 0.1 pmol N-acetyl farnesylcysteine methyl ester formed/min/mg protein by the carcinoma and by the normal endometrial preparations, respectively). As this fraction is enriched in plasma membranes, it appears that the elevated enzyme activity could be related to ras protein methylation; if so, selective methylation blockers might inhibit the growth of endometrial carcinomas.


Asunto(s)
Neoplasias Endometriales/enzimología , Proteína Metiltransferasas/metabolismo , Endometrio/enzimología , Femenino , Proteínas de Unión al GTP/metabolismo , Humanos , Cinética , Metilación , Proteína Metiltransferasas/antagonistas & inhibidores , Fracciones Subcelulares/enzimología , Especificidad por Sustrato
13.
Biochim Biophys Acta ; 1406(1): 40-50, 1998 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-9545527

RESUMEN

The carboxy terminal S-farnesylcysteine of Ras oncoproteins is required for their membrane anchorage and transforming activities. We showed previously that S-farnesylthiosalicylic acid (FTS) affects the membrane anchorage of activated H-Ras in EJ cells and inhibits their growth. We report here on structural elements in S-prenyl derivatives that specifically inhibit the growth of EJ cells, but not of untransformed Rat-1 cells. Inhibition of the Ras-dependent extracellular signal-regulated protein kinase (ERK), of DNA synthesis and of EJ cell growth were apparent after treatment with FTS or its 5-fluoro, 5-chloro and 4-fluoro derivatives or with the C20 S-geranylgeranyl derivative of thiosalicylic acid. The 4-Cl-FTS analogue was a weak inhibitor of EJ cell growth. The 3-Cl-FTS analogue and the FTS carboxyl methyl ester were inactive, as were the C10 S-geranyl derivative of thiosalicylic acid, farnesoic acid, N-acetyl-S-farnesyl-L-cysteine and S-farne-sylthiopropionic acid. The structural requirements for anti-Ras activity of S-prenyl analogues thus appear to be rather stringent. With regard to chain length, the C15 farnesyl group linked to a rigid backbone seems to be necessary and sufficient. A free carboxyl group in an appropriately rigid orientation, as in thiosalicylic acid, is also required. Halogenic substitutents on the benzene ring of the thiosalicylic acid are tolerated only at position 5 or 4. This information may facilitate the design of potent Ras antagonists and deepen our understanding of the mode of association of Ras with the plasma membrane.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Farnesol/análogos & derivados , Salicilatos/química , Salicilatos/farmacología , Proteínas ras/antagonistas & inhibidores , Animales , División Celular/efectos de los fármacos , Línea Celular , Cisteína/análogos & derivados , Cisteína/química , Relación Dosis-Respuesta a Droga , Farnesol/química , Farnesol/farmacología , Ratas , Transfección , Proteínas ras/química , Proteínas ras/genética
14.
J Mol Med (Berl) ; 77(11): 792-7, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10619439

RESUMEN

Merkel cell carcinoma (MCC) is a neuroendocrine malignancy showing poor response to a variety of therapeutic strategies. We evaluated the antitumor activity of S-trans, trans-farnesylthiosalicylic acid (FTS), a new inhibitor of Ras signal transduction, in a newly established SCID mouse xenotransplantation model for human MCC (seven animals per group). FTS injected intraperitoneally at 5 mg/kg per day for 2 weeks up-regulated the tumor suppressor p53 and induced tumor cell apoptosis in established MCCs growing subcutaneously in SCID mice. These effects led to a statistically significant inhibition of MCC growth (P<0.002). The mean tumor weights following FTS or control treatment were 0.32+/-0.15 g and 1.08+/-0.29 g, respectively. There was no evidence of FTS related toxicity at the effective dose used. Our findings stress the notion that FTS may qualify as a novel and rational treatment approach for MCC and possibly for other tumors that rely on tyrosine kinase signaling.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células de Merkel/tratamiento farmacológico , Farnesol/análogos & derivados , Salicilatos/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Células de Merkel/metabolismo , Carcinoma de Células de Merkel/patología , División Celular/efectos de los fármacos , Farnesol/administración & dosificación , Farnesol/farmacología , Farnesol/uso terapéutico , Femenino , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones SCID , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Salicilatos/administración & dosificación , Salicilatos/farmacología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Estadísticas no Paramétricas , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
15.
Cell Death Discov ; 1: 15047, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27551476

RESUMEN

Anaplastic Thyroid carcinoma is an extremely aggressive solid tumor that resists most treatments and is almost always fatal. Galectin-3 (Gal-3) is an important marker for thyroid carcinomas and a scaffold of the K-Ras protein. S-trans, transfarnesylthiosalicylic acid (FTS; Salirasib) is a Ras inhibitor that inhibits the active forms of Ras proteins. Modified citrus pectin (MCP) is a water-soluble citrus-fruit-derived polysaccharide fiber that specifically inhibits Gal-3. The aim of this study was to develop a novel drug combination designed to treat aggressive anaplastic thyroid carcinoma. Combined treatment with FTS and MCP inhibited anaplastic thyroid cells proliferation in vitro by inducing cell cycle arrest and increasing apoptosis rate. Immunoblot analysis revealed a significant decrease in Pan-Ras, K-Ras, Ras-GTP, p-ERK, p53, and Gal-3 expression levels and significant increase in p21 expression levels. In nude mice, treatment with FTS and MCP inhibited tumor growth. Levels of Gal-3, K-Ras-GTP, and p-ERK were significantly decreased. To conclude, our results suggest K-Ras and Gal-3 as potential targets in anaplastic thyroid tumors and herald a novel treatment for highly aggressive anaplastic thyroid carcinoma.

16.
Endocrinology ; 109(1): 235-9, 1981 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7238406

RESUMEN

The effects of L-T4 and betamethasone treatment of newborn mice on the development of the cholinergic muscarinic receptor in certain brain regions was studied using the potent labeled muscarinic antagonist [3H]4-N-methyl-piperidyl benzilate. Treatment with both L-T4 and betamethasone caused an accelerated accumulation of muscarinic receptors in the cortex 16 days post partum, with a subsequent reduction in level at 30 days. In the cerebellum and caudate putamen, only betamethasone caused a similar early accumulation of muscarinic receptors, while the later effect, namely a reduction in the level at 30 days, was seen with both hormones in these two regions as well as in the hippocampus. The results can explain some behavioral effects observed in other studies after treatment with these hormones.


Asunto(s)
Betametasona/farmacología , Encéfalo/crecimiento & desarrollo , Receptores Colinérgicos/fisiología , Receptores Muscarínicos/fisiología , Tiroxina/farmacología , Envejecimiento , Animales , Animales Recién Nacidos , Bencilatos/metabolismo , Encéfalo/efectos de los fármacos , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Cinética , Ratones , Parasimpatolíticos/metabolismo , Piperidinas/metabolismo , Receptores Muscarínicos/efectos de los fármacos
17.
Neurobiol Aging ; 8(2): 115-22, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3587488

RESUMEN

The objective of the present study was to determine the binding properties of muscarinic receptors in six brain regions in mature and old rats of both sexes by employing direct binding of [3H]-antagonist as well as of the labeled natural neurotransmitter, [3H]-acetylcholine [( 3H]-AcCh). In addition, age-related factors were evaluated in the modulation processes involved in agonist binding. The results indicate that as the rat ages the density of the muscarinic receptors is altered differently in the various brain regions: it is decreased in the cerebral cortex, hippocampus, striatum and olfactory bulb of both male and female rats, but is increased (58%) in the brain stem of senescent males while no significant change is observed for females. The use of the highly sensitive technique measuring direct binding of [3H]-AcCh facilitated the separate detection of age-related changes in the two classes (high- and low-affinity) of muscarinic agonist binding sites. In old female rats the density of high-affinity [3H]-AcCh binding sites was preserved in all tissues studied, indicating that the decreases in muscarinic receptor density observed with [3H]-antagonist represent a loss of low-affinity agonist binding sites. In contrast, [3H]-AcCh binding is decreased in the hypothalamus and increased in the brain stem of old male rats. These data imply sexual dimorphism of the aging process in central cholinergic mechanisms.


Asunto(s)
Acetilcolina/metabolismo , Bencilatos , Encéfalo/crecimiento & desarrollo , Oxotremorina/metabolismo , Parasimpatolíticos/metabolismo , Piperidinas/metabolismo , Receptores Muscarínicos/metabolismo , Envejecimiento , Animales , Encéfalo/metabolismo , Femenino , Masculino , Ratas , Ratas Endogámicas , Distribución Tisular , Tritio
18.
FEBS Lett ; 259(2): 233-6, 1990 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-2294014

RESUMEN

Evidence is presented for specific enzymatic methylation of 21-23 kDa membrane proteins in intact neuroblastoma N1E 115 cells, which is increased in dimethylsulfoxide-induced differentiated cells. Methylation of these proteins has characteristics typical of enzymatic reactions in which base labile volatile methyl groups are incorporated into proteins, consistent with the formation of protein carboxyl methylesters. However, these methylesters of the 21-23 kDa proteins are relatively stable compared to other protein carboxyl methylesters. The 3-fold increase in methylated 21-23 kDa proteins in the differentiated cells suggest biological significance in differentiation of the cell membranes.


Asunto(s)
Diferenciación Celular , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Células Tumorales Cultivadas/metabolismo , Animales , Línea Celular , Proteínas de la Membrana/aislamiento & purificación , Metilación , Ratones , Peso Molecular , Proteínas de Neoplasias/aislamiento & purificación , Neuroblastoma/patología , Células Tumorales Cultivadas/citología
19.
FEBS Lett ; 230(1-2): 167-70, 1988 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-2895019

RESUMEN

Binding of the labeled anticonvulsant drug [3H]dibenzocycloalkenimine [(3H]MK-801 to the N-methyl-D-aspartate (NMDA) receptor and its dissociation from the receptor at 25 degrees C are slow processes, both of which follow first order kinetics (t1/2 approximately equal to 70 and 180 min, respectively). Both reactions are markedly accelerated by glutamate and glycine (t1/2 approximately equal to 5-8 and 4 min, respectively), which allow bimolecular association kinetics of the labeled drug with the receptors whereas equilibrium binding of [3H]MK-801 (Kd 2-4 nM) is hardly affected by glutamate and glycine. The data suggest that MK-801 acts as a steric blocker of the NMDA receptor channel. The competitive antagonist D-(-)-2-amino-5-phosphovaleric acid (AP-5) freezes the receptor in a state which precludes either binding of [3H]MK-801 to the receptor channel or its dissociation from it. These findings have therapeutic implications.


Asunto(s)
Dibenzocicloheptenos/metabolismo , Receptores de Neurotransmisores/metabolismo , 2-Amino-5-fosfonovalerato , Animales , Anticonvulsivantes , Maleato de Dizocilpina , Glutamatos/farmacología , Ácido Glutámico , Glicina/farmacología , Cinética , Ratas , Receptores de N-Metil-D-Aspartato , Receptores de Neurotransmisores/efectos de los fármacos , Valina/análogos & derivados , Valina/farmacología
20.
FEBS Lett ; 241(1-2): 131-5, 1988 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-3197829

RESUMEN

Two distinct protein carboxyl methyltransferases (PCM) were identified in the electric organ of Torpedo ocellata. They were separated from each other in the active form by means of nondenaturing gel electrophoresis and by p-(chloromercuri)benzoate-agarose chromatography, and were individually identified by specific polyclonal antibodies. The existence of at least two distinct PCMs in eucaryotic cells raises the possibility that these enzymes are involved in distinct transmethylation reactions.


Asunto(s)
Órgano Eléctrico/enzimología , Isoenzimas/aislamiento & purificación , Proteína Metiltransferasas/aislamiento & purificación , Proteína O-Metiltransferasa/aislamiento & purificación , Animales , Electroforesis en Gel de Poliacrilamida , Sueros Inmunes , Immunoblotting , Isoenzimas/inmunología , Peso Molecular , Proteína O-Metiltransferasa/inmunología , Torpedo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA