Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Cell Biol ; 144(3): 413-25, 1999 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-9971737

RESUMEN

Expression of transforming Ha-Ras L61 in NIH3T3 cells causes profound morphological alterations which include a disassembly of actin stress fibers. The Ras-induced dissolution of actin stress fibers is blocked by the specific PKC inhibitor GF109203X at concentrations which inhibit the activity of the atypical aPKC isotypes lambda and zeta, whereas lower concentrations of the inhibitor which block conventional and novel PKC isotypes are ineffective. Coexpression of transforming Ha-Ras L61 with kinase-defective, dominant-negative (DN) mutants of aPKC-lambda and aPKC-zeta, as well as antisense constructs encoding RNA-directed against isotype-specific 5' sequences of the corresponding mRNA, abrogates the Ha-Ras-induced reorganization of the actin cytoskeleton. Expression of a kinase-defective, DN mutant of cPKC-alpha was unable to counteract Ras with regard to the dissolution of actin stress fibers. Transfection of cells with constructs encoding constitutively active (CA) mutants of atypical aPKC-lambda and aPKC-zeta lead to a disassembly of stress fibers independent of oncogenic Ha-Ras. Coexpression of (DN) Rac-1 N17 and addition of the phosphatidylinositol 3'-kinase (PI3K) inhibitors wortmannin and LY294002 are in agreement with a tentative model suggesting that, in the signaling pathway from Ha-Ras to the cytoskeleton aPKC-lambda acts upstream of PI3K and Rac-1, whereas aPKC-zeta functions downstream of PI3K and Rac-1. This model is supported by studies demonstrating that cotransfection with plasmids encoding L61Ras and either aPKC-lambda or aPKC-zeta results in a stimulation of the kinase activity of both enzymes. Furthermore, the Ras-mediated activation of PKC-zeta was abrogated by coexpression of DN Rac-1 N17.


Asunto(s)
Actinas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas ras/metabolismo , Células 3T3 , Animales , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Isoenzimas , Maleimidas/farmacología , Ratones , Microscopía Fluorescente , Mutación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Transducción de Señal , Transfección , Proteínas ras/genética
2.
Mol Cell Biol ; 19(12): 8052-65, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10567531

RESUMEN

Expression of constructs encoding fusion proteins of ERK1 and ERK2 containing a C-terminal farnesylation motif (CAAX) is predominantly localized at the cell membrane and was activated by coexpression of constitutively active Ha-RasL61 and epidermal growth factor. Both fusion proteins significantly inhibit the transcriptional activation of a c-fos-chloramphenicol acetyltransferase reporter induced by RasL61, constitutively active MEK1, or constitutively active RafBXB. The corresponding SAAX chimeras or overexpression of the wild-type ERKs did not interfere with the transcriptional activation of c-fos. The inhibition of the Ras-mediated c-fos induction by ERK2-CAAX can in part be rescued by coexpression of a wild-type ERK2 but not by wild-type ERK1. We find that ERK1-CAAX acts in the same fashion, indicating that mitogen-activated protein kinase (MAPK)-CAAX chimeras interact in an isotype-specific manner. It is demonstrated that both ERK1-CAAX and ERK2-CAAX associate with the corresponding endogenous ERKs, which explains the isotype-specific inhibitory effects of the ERK-CAAX chimeras. Evidence is presented that expression of ERK-CAAX fusion proteins inhibits the nuclear translocation of the corresponding endogenous ERKs. Disruption of MAPK translocation by membrane targeting provides additional, independent proof that nuclear translocation of ERKs is essential for the transcriptional activation of c-fos.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Activación Transcripcional , Proteínas ras/metabolismo , Células 3T3 , Animales , Transporte Biológico , Células COS , Fraccionamiento Celular , Membrana Celular/metabolismo , Citosol/metabolismo , Activación Enzimática , Expresión Génica , Variación Genética , Humanos , Líquido Intracelular , Isoenzimas , MAP Quinasa Quinasa 1 , Ratones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
3.
Mol Cell Biol ; 8(10): 4212-6, 1988 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-3141785

RESUMEN

An elevation of the intracellular pH and a rise in the cytoplasmic Ca2+ concentration are considered important mitogenic signals which are observed after stimulation by various growth factors. In a preceding report it was demonstrated that the expression of Ha-ras or v-mos in cells transfected with Ha-ras or v-mos, respectively, leads to an activation of the Na+/H+ antiporter and a concomitant rise in intracellular pH (W. Doppler, R. Jaggi, and B. Groner, Gene 54:145-151, 1987). This report describes the effect of the Ha-ras and v-mos oncogenes on intracellular Ca2+ release. The expression of Ha-ras in NIH 3T3 cells carrying a glucocorticoid-inducible transforming Ha-ras gene caused a desensitization of the Ca2+-mobilizing system to serum growth factors. The induction of p21ras in cells carrying the corresponding glucocorticoid-inducible proto-oncogene did not affect the Ca2+ response to growth factors. Conditions leading to the expression of the transforming Ha-ras gene but not those causing the induction of the normal Ha-ras gene yielded an increase in phosphatidylinositol turnover and a concomitant rise in inositol phosphates. Results similar to those obtained with the transforming Ha-ras gene were seen after the expression of v-mos. The data are consistent with a mechanism in which expression of the transforming Ha-ras gene leads to a release of Ca2+ from intracellular stores via elevated levels of inositol trisphosphate.


Asunto(s)
Calcio/metabolismo , Genes ras , Sustancias de Crecimiento/farmacología , Fosfatos de Inositol/metabolismo , Oncogenes , Fosfatos de Azúcar/metabolismo , Animales , Línea Celular , Transformación Celular Neoplásica/fisiopatología , Dexametasona/farmacología , Proteínas de Unión al GTP/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Sustancias de Crecimiento/sangre , Ratones , Fosfatidilinositoles/metabolismo
4.
Cancer Res ; 48(9): 2454-7, 1988 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-3356009

RESUMEN

5-N,N-Dimethylamiloride protects Ehrlich ascites tumor cells against the antiproliferative effect of nitrogen mustard. The drug reduces the frequency of DNA interstrand cross-links introduced by nitrogen mustard. Cells with a defective choline carrier are not protected against nitrogen mustard by dimethylamiloride. As nitrogen mustard is taken up by the choline carrier, it is concluded that the recently reported inhibition of the choline transport system by amiloride and its dimethyl derivatives (W. Doppler et al., Biochem. Pharmacol., 36: 1645-1649, 1987) is responsible for the protection against the alkylating agent.


Asunto(s)
Amilorida/análogos & derivados , Carcinoma de Ehrlich/patología , Mecloretamina/farmacología , Amilorida/farmacología , Animales , Transporte Biológico/efectos de los fármacos , División Celular/efectos de los fármacos , Colina/farmacocinética , Colina/farmacología , ADN de Neoplasias/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos
5.
Cancer Res ; 51(3): 807-12, 1991 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-1846318

RESUMEN

Hexadecylphosphocholine (HePC) inhibits protein kinase C (PKC) from NIH3T3 cells in cell-free extracts with a 50% inhibitory concentration of about 7 microM. Inhibition is competitive with regard to phosphatidylserine with a Ki of 0.59 microM. In order to determine whether HePC affects PKC in intact cells, the bombesin or tetradecanoylphorbolacetate-induced, PKC-mediated activation of the Na+/H(+)-antiporter was determined. It is demonstrated that HePC causes a drastic inhibition of this enzyme indicating a similar sensitivity of PKC to HePC in intact cells compared to cell-free extracts. In addition to the effects on PKC, treatment of NIH3T3 cells with HePC depresses the bombesin-induced formation of inositol 1,4,5-trisphosphate and the concomitant mobilization of intracellular Ca2+. Dose-response curves for the inhibition of inositol 1,4,5-trisphosphate formation and Ca2+ mobilization reveal 50% inhibitory concentrations of 2 or 5 microM, respectively. Polyphosphorylated phosphoinositides accumulate in HePC-treated cells indicating that the depression of inositol 1,4,5-trisphosphate generation is not caused by an inhibition of phosphoinositide kinases. Addition of bombesin to HePC-treated cells in the presence of LiCl revealed no evidence for an accelerated rate of inositol 1,4,5-trisphosphate turnover by the phospholipid analogue. It is concluded that HePC inhibits phosphoinositidase C in intact cells. The data strongly suggest that the growth-inhibitory effect of HePC is at least in part explained by the interference with mitogenic signal transduction.


Asunto(s)
Proteínas Portadoras/metabolismo , Fosfatos de Inositol/biosíntesis , Fosforilcolina/análogos & derivados , Proteína Quinasa C/antagonistas & inhibidores , Bombesina/antagonistas & inhibidores , Bombesina/farmacología , División Celular/efectos de los fármacos , Litio/farmacología , Fosforilcolina/farmacología , Intercambiadores de Sodio-Hidrógeno , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología
6.
Cancer Res ; 51(21): 5821-5, 1991 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-1718584

RESUMEN

The dihydropyridine derivative B859-35 inhibits phospholipid- and calcium-dependent protein kinase C (PKC) in cell-free extracts from NIH3T3 cells. Inhibition is competitive with regard to phosphatidylserine. At 1 microM phosphatidylserine, half-maximal inhibition (IC50) is obtained at approximately 2.5 microM B859-35. 12-O-Tetradecanoylphorbol-13-acetate (TPA)-dependent activation of the Na+/H+ antiporter was used to determine whether the enzyme is also affected in intact cells. The activity of the antiporter was monitored by following the dimethylamiloride-sensitive cytosolic alkalinization. It is demonstrated that B859-35 depresses the TPA-induced alkalinization with an IC50 of 5 microM, indicating that PKC in intact cells and the enzyme in cell-free extracts are equally sensitive to the drug. TPA-induced expression of the c-fos gene was used as an additional marker for intracellular PKC activity. Activation of c-fos expression was determined by measuring chloramphenicol acetyltransferase (CAT) activity in cells transfected with a c-fosCAT construct in which the CAT gene is expressed under the control of the endogenous human c-fos promoter. The studies revealed that 2.5 microM B859-35, a concentration equivalent to the IC50 in cell-free extracts, significantly depresses TPA-induced c-fosCAT expression. B859-35 inhibited cellular proliferation of NIH3T3 cells with an IC50 of approximately 5 microM. This is close to the IC50 for the anti-PKC activity of B859-35. It is suggested that the inhibition of PKC contributes to the growth inhibition following exposure to B859-35.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , División Celular/efectos de los fármacos , Dihidropiridinas/farmacología , Genes fos/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Acetato de Tetradecanoilforbol/farmacología , 1-Metil-3-Isobutilxantina/farmacología , Células 3T3 , Animales , Secuencia de Bases , Proteínas Portadoras/metabolismo , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Elementos de Facilitación Genéticos , Expresión Génica/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Cinética , Ratones , Datos de Secuencia Molecular , Fosfatidilserinas/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Intercambiadores de Sodio-Hidrógeno , Transcripción Genética/efectos de los fármacos , Transfección
7.
Cell Signal ; 6(3): 285-97, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7917786

RESUMEN

The mechanism by which transforming Ha-ras induces c-fos expression in HC11 mouse mammary epithelial cells was investigated with regard to controversial data concerning the role of protein kinase C (PKC) and the required promoter elements of the fos gene. HC11 cells carrying a glucocorticoid-inducible Ha-ras (val12) construct were transfected with a chloramphenicol acetyltransferase (CAT) reporter gene under the control of a human fos promoter which includes the serum response element (SRE), the adjacent c-fos AP-1 site (FAP) and the cAMP response element (CRE). Induction of the Ha-ras gene by dexamethasone lead to a transactivation of expression of the transfected fos promoter construct which was inhibited by the PKC inhibitor BM41440 and abrogated in PKC-'depleted' cells. A similar transactivation was observed when the fos promoter construct was co-transfected with a constitutively active ras expression vector. Again, this effect was depressed by the PKC inhibitor and abolished in PKC-'depleted' cells. 'PKC-depletion' was achieved by long-term exposure to 12-O-tetradecanoylphorbol-13-acetate. This procedure was shown to deplete cells of PKC alpha and to reduce significantly PKC epsilon. Long-term exposure to bryostatin 1 selectively depletes PKC alpha. Depletion of PKC alpha by bryostatin 1 does not reduce the transcriptional activation of the SRE-FAP-TK-CAT (TK: thymidine kinase) construct by Ha-ras. In order to delineate the promoter elements mediating the transcriptional activation, constructs which lack the FAP and the CRE sites but contain an intact SRE were co-transfected with the ras construct. Elimination of the FAP and CRE sequences did not affect the transcriptional activation by Ha-ras (val12). It is concluded that in HC11 cells, transforming Ha-ras activates c-fos expression in a PKC-dependent manner, presumably implying PKC epsilon, and that the SRE is sufficient to mediate transcriptional activation.


Asunto(s)
Regulación de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Antineoplásicos/farmacología , Secuencia de Bases , Línea Celular , Células Cultivadas , Proteínas de Unión al ADN , Dexametasona/farmacología , Epitelio/metabolismo , Femenino , Glándulas Mamarias Animales/citología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Nucleares , Embarazo , Proteína Quinasa C/metabolismo , Factor de Respuesta Sérica , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/metabolismo , Transfección
8.
Cell Calcium ; 18(2): 120-34, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7585889

RESUMEN

The effect of an induction of transforming Ha-ras on Ca2+ influx into NIH3T3 cells was studied employing Fura-2 quenching by Mn2+. The expression of transforming p21Ha-ras caused a significant increase in Mn2+ influx which was blocked by Cd2+, La3+, niguldipine and the Ca(2+)-channel blocker SK&F96365. This effect was specific for transforming Ha-ras and was not seen after overexpression of the Ha-ras proto-oncogene or v-mos. In addition to the enhanced Mn2+ influx, transforming p21Ha-ras elicited an increased efflux of the K(+)-congener 86Rb+ which was inhibitable by Ca(2+)-channel blockers and charybdotoxin, a selective inhibitor of high and intermediate conductance Ca(2+)-dependent K+ channels. Charybdotoxin did not reduce the increase in Mn2+ influx by ras, demonstrating that the activation of Ca(2+)-dependent K+ channels was not required for the sustained Mn2+/Ca2+ influx in the presence of transforming Ha-ras. In ras-expressing cells, the bradykinin-induced Mn2+ influx and charybdotoxin sensitive 86Rb+ efflux were markedly potentiated. The increase in the inositol- 1,4,5-trisphosphate and inositol-1,3,4,5-tetrakisphosphate levels by ras is not sufficient to explain the elevated Mn2+ influx. The mitogenic response to an expression of transforming Ha-ras was inhibited by the Ca(2+)-channel blockers not, however, by charybdotoxin. These data suggest the existence of an agonist-independent activation of a receptor- or second messenger-operated Ca2+ channel by transforming Ha-ras which is necessary for the mitogenic response to the activation of the oncogene.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Transformación Celular Neoplásica/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células 3T3 , Animales , Transporte Biológico , Bradiquinina/farmacología , Dexametasona/farmacología , Fosfatos de Inositol/metabolismo , Manganeso/metabolismo , Ratones , Canales de Potasio/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Rubidio/metabolismo
9.
FEBS Lett ; 352(1): 11-4, 1994 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-7925931

RESUMEN

We have previously shown that neopterin enhances hydrogen peroxide and chloramine T activity in a luminol-dependent chemiluminescence assay and strengthens toxicity of these agents against bacteria at slightly alkaline pH (pH 7.5), while 7,8-dihydroneopterin was shown to be a scavenger independent of the pH value. Besides various oxidants, phenolic antioxidants were shown to specifically induce expression of the c-fos and c-jun mRNAs. Using an inducible cfosCAT reporter transactivation system we studied the function of the pteridine derivatives on c-fos transactivation. For the first time, we demonstrate that neopterin and 7,8-dihydroneopterin, particularly together with cyclic guanosine monophosphate, induce c-fos gene expression. In humans, interferon-gamma induces the release of neopterin and 7,8-dihydroneopterin and also the synthesis of nitric oxide radical which in turn stimulate the formation of cGMP. Thus, in certain situations all three substances, namely neopterin, 7,8-dihydroneopterin and cGMP, may be present locally and even in the circulation at the same time. Based on our findings this constellation would significantly enhance the risk of c-fos gene expression and therefore promote tumour growth and development.


Asunto(s)
Biopterinas/análogos & derivados , GMP Cíclico/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes fos/efectos de los fármacos , Pteridinas/farmacología , Células 3T3 , Animales , Secuencia de Bases , Biopterinas/farmacología , Cloranfenicol O-Acetiltransferasa/genética , Genes fos/genética , Humanos , Ratones , Datos de Secuencia Molecular , Neopterin , Forbol 12,13-Dibutirato/farmacología , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos
10.
J Med Chem ; 40(26): 4420-5, 1997 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-9435912

RESUMEN

A series of N-heteroaryl hydrazones derived from aryl N-heteroaryl or bis-N-heteroaryl methanones was prepared in search for potential novel antitumor agents. The stereochemistry of these compounds was established by means of NMR spectroscopy. Antiproliferative activity was determined in a panel of human tumor cell lines (CCRF-CEM, Burkitt's lymphoma, HeLa, ZR-75-1, HT-29, and MEXF 276L) in vitro. Generally, the new compounds were found to be more potent (IC50 = 0.011-0.436 microM) than the ribonucleotide reductase inhibitor hydroxyurea (IC50 = 140 microM). Most of the compounds exhibited the highest activity against Burkitt's lymphoma with an IC50 of 0.011-0.035 microM. [14C]Cytidine incorporation into DNA was quantitated for selected hydrazones (Z-A, E-1, Z-3, Z-4, E-5, Z-5, E-13, E-18, Z-19, Z-24, and E-26) as a measure of the inhibition of ribonucleotide reductase in Burkitt's lymphoma cells. The E-configurated compounds were found to inhibit [14C]cytidine incorporation to a greater extent (IC50 = 0.67-5.05 microM) than the Z-isomers (IC50 = 7.20 to > 10 microM). Principal component analysis of the IC50 values obtained for inhibition of cell proliferation revealed that the cell lines tested can be grouped into three main families showing different sensitivities toward the compounds in our series [(i) CCRF-CEM, Burkitt's lymphoma, and Hela; (ii) HT-29; and (iii) MEXF 276 L].


Asunto(s)
Antineoplásicos/síntesis química , Hidrazonas/síntesis química , Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Citidina/metabolismo , ADN/biosíntesis , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Hidrazonas/farmacología , Hidroxiurea/farmacología , Espectroscopía de Resonancia Magnética , Conformación Molecular , Estructura Molecular , Ribonucleótido Reductasas/antagonistas & inhibidores , Ribonucleótido Reductasas/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
11.
Biochem Pharmacol ; 56(7): 861-9, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9774148

RESUMEN

Bryostatin 1 is a new antitumor agent which modulates the enzyme activity of protein kinase C (PKC, phospholipid-Ca2+-dependent ATP:protein transferase, EC 2.7.1.37). Several reports have suggested that the pumping activity of the multidrug resistance gene 1 (MDR1)-encoded multidrug transporter P-glycoprotein (PGP) is enhanced by a PKC-mediated phosphorylation. It was shown here that bryostatin 1 was a potent modulator of multidrug resistance in two cell lines over-expressing a mutant MDR1-encoded PGP, namely KB-C1 cells and HeLa cells transfected with an MDR1-V185 construct (HeLa-MDR1-V185) in which glycine at position 185 (G185) was substituted for valine (V185). Bryostatin 1 is not able to reverse the resistance of cells over-expressing the wild-type form (G185) of PGP, namely CCRF-ADR5000 cells and HeLa cells transfected with a MDR1-G185 construct (HeLa-MDR1-G185). Treatment of HeLa-MDR1-V185 cells with bryostatin 1 was accompanied by an increase in the intracellular accumulation of rhodamine 123, whereas no such effect could be observed in HeLa-MDR1-G185 cells. HeLa-MDR1-V185 cells expressed the PKC isoforms alpha, delta and zeta. Down-modulation of PKC alpha and delta by 12-O-tetradecanoylphorbol-13-acetate (TPA) did not affect the drug accumulation by bryostatin 1. Bryostatin 1 depleted PKC alpha completely and PKC delta partially. In HeLa-MDR1-V185 cells, short-term exposure to bryostatin 1, which led to a PKC activation, was as efficient in modulating the pumping activity of PGP as long-term exposure leading to PKC depletion. Bryostatin 1 competed with azidopine for binding to PGP in cells expressing the MDR1-V185 and MDR1-G185 forms of PGP. It is concluded that bryostatin 1: i) interacts with both the mutated MDR1-V185 and the wild-type MDR1-G185; ii) reverses multidrug resistance and inhibits drug efflux only in PGP-V185 mutants; and iii) that this effect is not due to an interference of PKC with PGP. For gene therapy, it is important to reverse the specific resistance of a mutant in the presence of a wild-type transporter and vice versa. Our results show that it is possible to reverse a specific mutant PGP.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Sustitución de Aminoácidos/genética , Resistencia a Múltiples Medicamentos , Lactonas/farmacología , Proteína Quinasa C/fisiología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Unión Competitiva/efectos de los fármacos , Brioestatinas , Resistencia a Antineoplásicos , Femenino , Células HeLa , Humanos , Células KB , Macrólidos , Mutación Puntual
12.
J Cancer Res Clin Oncol ; 115(5): 435-8, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2808481

RESUMEN

In view of various reports describing differences in histone acetylation between normal rat liver and hepatomas, the behaviour of histone acetyltransferase (EC 2.3.1.48) activity was elucidated in normal rat liver and in a spectrum of well-characterized rat hepatomas of slow, intermediate and rapid growth rates. In all tumours the acetyltransferase specific activity, expressed as nmol h-1 mg total protein-1, was higher than in the corresponding normal livers and the rise correlated positively with the proliferation rates of the tumors. No difference is observed if acetyltransferase activity is expressed per milligram of histone. This is explained by elevated ratios of histones and of DNA to total protein in the hepatomas compared to the ratios in normal liver. Electrophoretic analysis of [3H]acetate-labeled histones revealed similar patterns in hepatoma and normal liver. The extent of histone H4 acetylation, as indicated by the frequency distribution of non-, mono-, di-, tri-, and tetraacetylated H4-species, was found to be identical in hepatomas and normal liver. The histone protein and acetate labeling patterns were near normal in the slowly growing hepatomas.


Asunto(s)
Acetiltransferasas/análisis , Neoplasias Hepáticas Experimentales/enzimología , Proteínas de Saccharomyces cerevisiae , Acetilación , Animales , Histona Acetiltransferasas , Histonas/análisis , Histonas/metabolismo , Hígado/enzimología , Ratas , Ratas Endogámicas ACI , Ratas Endogámicas BUF
13.
Adv Enzyme Regul ; 34: 257-68, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-7942278

RESUMEN

HC-11 mouse mammary epithelial cells stably transfected with a glucocorticoid-inducible Ha-ras construct encoding a transforming (val12) p21Ha-ras were cotransfected with a c-fos-CAT construct containing the human c-fos promoter up to position -711 and the CAT reporter gene. Expression of Ha-ras by dexamethasone leads to a transcriptional activation of the fos-CAT construct which was found to be sensitive to the PKC inhibitor ilmofosine (BM41440) and abrogated by PKC depletion following long-term exposure to TPA. The responsiveness to Ha-ras is retained if only the portion of the fos promoter covering the serum response element (SRE) and the adjacent fos AP-1 (FAP) site are put in front of a CAT gene linked to a thymidine kinase (TK) promoter. Further depletion of the FAP-site does not affect the inducibility by Ha-ras. Transcriptional activation of the SRE-FAP-TK-CAT as well as the SRE-TK-CAT constructs by Ha-ras is sensitive to the PKC-inhibitor ilmofosine (BM41440) and blocked by long-term exposure to TPA. Long-term exposure to TPA depletes cells of PKC alpha and significantly reduces the PKC epsilon levels. Long-term exposure in bryostatin 1 selectively depletes PKC alpha. Depletion of PKC alpha by bryostatin 1 does not reduce the transcriptional activation of the SRE-FAP-TK-CAT-construct by Ha-ras. It is concluded that (i) transforming Ha-ras induces c-fos in HC-11 cells via PKC (presumably epsilon), (ii) the signal is mediated to the serum response element (SRE) of the fos promoter and (iii) the fos AP-1 (FAP) site is not required for this mechanism.


Asunto(s)
Regulación de la Expresión Génica , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas ras/metabolismo , Animales , Brioestatinas , Compartimento Celular , Línea Celular , Células Epiteliales , Regulación de la Expresión Génica/efectos de los fármacos , Lactonas/farmacología , Macrólidos , Glándulas Mamarias Animales/citología , Ratones , Proteínas Recombinantes/biosíntesis , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética , Transfección
14.
Adv Enzyme Regul ; 36: 385-407, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8869757

RESUMEN

Mitogenic signalling mechanisms emerged as novel targets for tumor chemotherapy. Current strategies for pharmacological interventions are briefly discussed. Phospholipid analogues are treated in greater detail. It is shown here that this new class of antitumor agents acts as inhibitors of mitogenic signal transduction. The common target of all phospholipid analogues studied so far is the phosphatidylinositol (PI)-specific phospholipase C (PLC). This results in an attenuated formation of inositol-1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). The reduction in IP3-levels leads to a depressed release of Ca2+ from internal stores, and the reduced formation of DAG interferes with the growth factor-induced activation of protein-kinase C (PKC). In addition to the effect on PI-specific PLC, most phospholipid analogues inhibit PKC directly by interacting with the regulatory domain of the enzyme. This effect, however, is not observed with all phospholipid analogues. Some potent growth inhibitory representatives from this group like hexadecylphosphoserine or hexadecylphosphonoserine do not affect PKC in cell-free extracts. It is concluded, therefore, that the direct inhibition of PKC is not required for the growth-inhibitory activity of these agents. The ability of phospholipid analogues to interact with PKC was also not found to be correlated the occurrence of unwanted side effects. Phospholipid analogues have also been found to act as inhibitors of phospholipase D (PLD). However, in this case the correlation to the growth inhibitory potency of various phospholipid analogues was less clear, so that the contribution of the PLD inhibition to the growth inhibitory effect of these agents still remains to be established. The inhibition of the thrombin-induced rise in cytosolic free Ca2+ by phospholipid analogues is reversible by washing the cells in phospholipid-free medium. These findings suggest that phospholipid analogues do not cause persistent membrane damage and may act as cytostatic rather than cytotoxic agents.


Asunto(s)
Neoplasias/terapia , Fosfolípidos/farmacología , Transducción de Señal/efectos de los fármacos , Antineoplásicos/farmacología , Calcio/metabolismo , Inhibidores Enzimáticos/farmacología , Inositol 1,4,5-Trifosfato/antagonistas & inhibidores , Inositol 1,4,5-Trifosfato/metabolismo , Estructura Molecular , Fosfolipasa D/antagonistas & inhibidores , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Fosfoserina/análogos & derivados , Fosfoserina/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Trombina/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores
20.
Ann Oncol ; 9 Suppl 5: S125-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9926251

RESUMEN

Concepts for the treatment of Hodgkin's lymphomas based on novel insights of the molecular mechanisms responsible for the maintenance of the transformed phenotype of Reed-Sternberg cells, their proliferation and sensitivity to radiation and anti-tumor agents are discussed. The potentials of some recently developed new signal transduction inhibitors for the treatment of Hodgkin's lymphomas are discussed in greater detail and comprise agents directed against Janus kinase 2 (JAK 2); Signal Transducers and Activators of Transcription (STAT factors); agents directed against SH 2-domains: the fes/fps oncogene, Ras; protein kinase C (PKC) isotypes and means of inducing radiation or drug-induced apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Enfermedad de Hodgkin/tratamiento farmacológico , Células de Reed-Sternberg/efectos de los fármacos , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Genes bcl-2/efectos de los fármacos , Genes ras/efectos de los fármacos , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Humanos , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , Células de Reed-Sternberg/fisiología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA