Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros











Base de datos
Tipo de estudio
Intervalo de año de publicación
1.
J Biol Chem ; 276(46): 42834-42, 2001 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-11551901

RESUMEN

Transcriptional activation of the cyclin D1 by oncogenic Ras appears to be mediated by several pathways leading to the activation of multiple transcription factors which interact with distinct elements of the cyclin D1 promoter. The present investigations revealed that cyclin D1 induction by transforming Ha-Ras is MEK- and Rac-dependent and requires the PKC isotypes epsilon, lambda, and zeta, but not cPKC-alpha. This conclusion is based on observations indicating that cyclin D1 induction by transforming Ha-Ras was depressed in a dose-dependent manner by PD98059, a selective inhibitor of the mitogen-activated kinase kinase MEK-1, demonstrating that Ha-Ras employs extracellular signal-regulated kinases (ERKs) for signal transmission to the cyclin D1 promoter. Evidence is presented that PKC isotypes epsilon and zeta, but not lambda are required for the Ras-mediated activation of ERKs. Expression of kinase-defective, dominant negative (DN) mutants of nPKC-epsilon or aPKC-zeta inhibit ERK activation by constitutively active Raf-1. Phosphorylation within the TEY motif and subsequent activation of ERKs by constitutively active MEK-1 was significantly inhibited by DN aPKC-zeta, indicating that aPKC-zeta functions downstream of MEK-1 in the pathway leading to cyclin D1 induction. In contrast, TEY phosphorylation induced by constitutively active MEK-1 was not effected by nPKC-epsilon, suggesting another position for this kinase within the cascade investigated. Transformation by oncogenic Ras requires activation of several Ras effector pathways which may be PKC-dependent and converge on the cyclin D1 promoter. Therefore, we investigated a role for PKC isotypes in the Ras-Rac-mediated transcriptional regulation of cyclin D1. We have been able to reveal that cyclin D1 induction by oncogenic Ha-Ras is Rac-dependent and requires the PKC isotypes epsilon, lambda, and zeta, but not cPKC-alpha. Evidence is presented that aPKC-lambda acts upstream of Rac, between Ras and Rac, whereas the PKC isotypes epsilon and zeta act downstream of Rac and are required for the activation of ERKs.


Asunto(s)
Isoformas de Proteínas , Proteína Quinasa C/química , Animales , Western Blotting , Mama/metabolismo , Células Cultivadas , Clonación Molecular , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Células Epiteliales/metabolismo , Flavonoides/farmacología , Humanos , Isoenzimas/metabolismo , Luciferasas/metabolismo , MAP Quinasa Quinasa 1 , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteína Quinasa C/metabolismo , Proteína Quinasa C-alfa , Proteína Quinasa C-epsilon , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Activación Transcripcional , Transfección
2.
Biochem J ; 359(Pt 1): 211-7, 2001 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11563985

RESUMEN

The activation of phospholipase D (PLD) by transforming Ras is well documented. Although two distinct PLD isoforms, PLD1 and PLD2, have been cloned from mammalian cells, it has remained unclear whether both isoenzymes are activated by Ras and, if this is the case, whether they are stimulated by a common mechanism. In the present study we show that expression of transforming Ras in HC11 mouse mammary epithelial cells enhanced the activity of endogenous PLD. Co-expression of Ras with either PLD1b or PLD2 resulted in elevated activities of both PLD isoenzymes in HC11 cells, indicating that transforming Ras was capable of activating both PLD isoforms in vivo. Ras-induced activation of PLD was resistant to the protein kinase C (PKC) inhibitor GF109203X, which preferentially affects conventional- and novel-type PKCs, but sensitive to Ro-31-8220, which inhibits atypical PKCs more effectively. Co-transfection of atypical PKC-iota with either PLD1b or PLD2 led to a selective activation of PLD2 by PKC-iota, whereas PLD1b was not affected. PLD1b, however, was found to be a potent activator of PKC-iota, whereas PLD2 was less effective in this respect. The data suggest that PKC-iota acts upstream of PLD2 and that PLD1b is implicated in the activation of PKC-iota. The data are discussed as indicating a putative signalling cascade comprising Ras-->PLD1b-->PKC-iota-->PLD2. Evidence for the implication of this pathway in the transcriptional regulation of cyclin D1 is also presented.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Genes ras/fisiología , Isoenzimas/metabolismo , Neoplasias Mamarias Experimentales/enzimología , Fosfolipasa D/metabolismo , Proteína Quinasa C/metabolismo , Animales , Células COS , Transformación Celular Neoplásica , Chlorocebus aethiops , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Indoles/farmacología , Luciferasas/metabolismo , Maleimidas/farmacología , Neoplasias Mamarias Experimentales/genética , Ratones , Fosfolipasa D/genética , Unión Proteica , Proteína Quinasa C/antagonistas & inhibidores , Transfección
5.
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
6.
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
8.
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
10.
EMBO J ; 17(14): 4046-55, 1998 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9670019

RESUMEN

The implication of protein kinase C (PKC) isoforms cPKC-alpha, nPKC-epsilon, aPKC-lambda and aPKC-zeta in the transcriptional activation of a c-fos promoter-driven CAT-reporter construct by transforming Ha-Ras has been investigated. This was achieved by employing antisense constructs encoding RNA directed against isoform-specific 5' sequences of the corresponding mRNA, and expression of PKC mutants representing either kinase-defective, dominant negative, or constitutively active forms of the PKC isoforms. The data indicate that in HC11 mouse mammary epithelial cells, transforming Ha-Ras requires the activities of the three PKC isozymes: aPKC-lambda, nPKC-epsilon and aPKC-zeta, not, however, of cPKC-alpha, for the transcriptional activation of c-fos. Co-expression of oncogenic Ha-Ras with combinations of kinase-defective, dominant negative and constitutively active mutants of the various PKC isozymes are in agreement with a tentative model suggesting that, in the signaling pathway from Ha-Ras to the c-fos promoter, aPKC-lambda acts upstream whereas aPKC-zeta functions downstream of nPKC-epsilon.


Asunto(s)
Células Epiteliales/metabolismo , Genes fos/genética , Isoenzimas/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteína Oncogénica p21(ras)/metabolismo , Proteína Quinasa C/metabolismo , Activación Transcripcional/fisiología , Animales , Línea Celular , Isoenzimas/genética , MAP Quinasa Quinasa 1 , Glándulas Mamarias Animales/citología , Ratones , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/fisiología , ARN sin Sentido , Transducción de Señal/genética , Transfección
11.
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
12.
J Biol Chem ; 272(7): 4072-8, 1997 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-9020116

RESUMEN

Phosphorylation of myristoylated alanine-rich protein kinase C substrate (MARCKS) in intact cells has been employed as an indicator for activation of protein kinase C (PKC). Specific PKC isoenzymes responsible for MARCKS phosphorylation under physiological conditions, however, remained to be identified. In our present study using stably transfected NIH 3T3 cell clones we demonstrate that expression of constitutively active mutants of either conventional cPKC-alpha or novel nPKC-epsilon increased phosphorylation of endogenous MARCKS in the absence of phorbol 12,13-dibutyrate in intact mouse fibroblasts, implicating that each of these PKC isoforms itself is sufficient to induce enhanced MARCKS phosphorylation. Similarly, ectopic expression of a constitutively active mutant of PKC-theta significantly increased MARCKS phosphorylation compared to vector controls, identifying PKC-theta as a MARCKS kinase. The PKC-specific inhibitor GF 109203X (bisindolylmaleimide I) reduced MARCKS phosphorylation in intact cells at a similar dose-response as enzymatic activity of recombinant isoenzymes cPKC-alpha, nPKC-epsilon, and nPKC-theta in vitro. Consistently, phorbol 12,13-dibutyrate-dependent MARCKS phosphorylation was significantly reduced in cell lines expressing dominant negative mutants of either PKC-alpha K368R or (dominant negative) PKC-epsilon K436R. The fact, that the constitutively active PKC-lambda A119E mutant did not alter the MARCKS phosphorylation underscores the assumption that atypical PKC isoforms are not involved in this process. We conclude that under physiological conditions, conventional cPKC-alpha and novel nPKC-epsilon, but not atypical aPKC-lambda are responsible for MARCKS phosphorylation in intact NIH 3T3 fibroblasts.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Isoenzimas/metabolismo , Proteínas de la Membrana , Proteína Quinasa C/metabolismo , Proteínas/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Maleimidas/farmacología , Ratones , Datos de Secuencia Molecular , Sustrato de la Proteína Quinasa C Rico en Alanina Miristoilada , Forbol 12,13-Dibutirato/farmacología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores
13.
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
14.
J Biol Chem ; 271(48): 30505-9, 1996 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-8940018

RESUMEN

Stimulation by epidermal growth factor (EGF) of NIH3T3 cells overexpressing the EGF receptor (EGFR) results in a release of Ca2+ from internal stores. Ca2+ release is followed by an influx of extracellular calcium which can be recorded by the influx of the calcium surrogate Mn2+. Both Ca2+ release and Mn2+/Ca2+ influx are inhibited by expression of the dominant negative Asn17-Ras mutant and abrogated by microinjected neutralizing anti-Ras antibody Y13-259, whereas microinjection of the anti-Ras antibody Y13-238 which does not interact with the effector binding domain of Ras is without any effect on the EGF-induced Ca2+ transient. Neither Asn17-Ha-Ras nor the Y13-259 antibody interferes with the thapsigargin-induced Mn2+/Ca2+ influx. The nerve growth factor receptor (Trk)-mediated Ca2+ transient was found to be unaffected by the dominant negative Ras mutant or microinjected neutralizing anti-Ras antibodies. Substitution of the phospholipase Cgamma1 (PLCgamma1) binding site of the EGFR by the PLCgamma binding domain of Trk renders the EGFR-induced Ca2+ influx insensitive to the expression of Asn17-Ha-Ras, whereas the Ca2+ signal induced by Trk carrying the PLC binding site of EGFR is Ras-dependent and abrogated by the dominant negative Ras mutant. It is concluded that the Ca2+ transient induced by the activated EGFR, not, however, the Ca2+ transient elicited by the activated NGFR/Trk, is a Ras-mediated phenomenon and that the role of Ras in regulating EGFR-induced Ca2+ influx depends on the structure of the PLCgamma binding domain.


Asunto(s)
Calcio/fisiología , Receptores ErbB/fisiología , Receptores de Factor de Crecimiento Nervioso/fisiología , Células 3T3 , Animales , Citosol/fisiología , Isoenzimas/fisiología , Ratones , Fosfolipasa C gamma , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Transducción de Señal , Fosfolipasas de Tipo C/fisiología
15.
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
16.
EMBO J ; 15(1): 73-82, 1996 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-8598208

RESUMEN

The exchange of nerve growth factor receptor/Trk and epidermal growth factor receptor (EGFR) phospholipase C gamma (PLC gamma) binding sites resulted in the transfer of their distinct affinities for this Src homology 2 domain-containing protein. Relative to wild-type EGFR, the PLC gamma affinity increase of the EGFR switch mutant EGFR.X enhanced its inositol trisphosphate (IP3) and calcium signals and resulted in a more sustained mitogen-activated protein (MAP) kinase activation and accelerated receptor dephosphorylation. In parallel, EGFR.X exhibited a significantly decreased mitogenic and transforming potential in NIH 3T3 cells. Conversely, the transfer of the EGFR PLC gamma binding site into the Trk cytoplasmic domain context impaired the IP3/calcium signal and attenuated the MAP kinase activation and receptor dephosphorylation, but resulted in an enhancement of the ETR.X exchange mutant mitogenic and oncogenic capacity. Our findings establish the significance of PLC gamma affinity for signal definition, the role of this receptor tyrosine kinase substrate as a negative feedback regulator and the importance of this regulatory function for mitogenesis and its disturbance in oncogenic aberrations.


Asunto(s)
Transformación Celular Neoplásica , Receptores ErbB/fisiología , Isoenzimas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Fosfolipasas de Tipo C/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Cartilla de ADN/química , Activación Enzimática , Humanos , Fosfatos de Inositol/metabolismo , Ratones , Datos de Secuencia Molecular , Fosfolipasa C gamma , Fosfotirosina/metabolismo , Unión Proteica , Transducción de Señal , Relación Estructura-Actividad , Tirosina/química , Dominios Homologos src
17.
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
18.
Anticancer Drug Des ; 10(5): 411-25, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7639930

RESUMEN

The interference of several new hexadecylphosphocholine analogues with mitogenic signal transduction was investigated in NIH3T3 fibroblasts by studying the effects of these agents on thrombin-induced inositol 1,4,5-trisphosphate (Ins(1,4,5)P3) formation and the subsequent Ca2+ release, on protein kinase C (PKC) in cell-free extracts, on the PKC-mediated activation of the Na+/H+ antiporter and on c-fos induction. The compounds investigated include hexadecylphosphocholine (HePC), octadecyl-[2-(N-methyl-piperidinio)-ethyl]-phosphate (D20133), octadecyl-(N,N-dimethyl-piperidinio-4-yl)-phosphate (D21266); octadecyl-[2-(trimethyl-arsonio)-ethyl]-phosphate (D21805) and hexadecylphospho-L-serine (HePS). The data indicate that (i) all compounds inhibit the thrombin-induced progression of growth-arrested NIH3T3 cells into S phase with similar IC50 values; (ii) the common denominator of all compounds is a reduction of Ins(1,4,5)P3 formation, resulting in an attenuation of Ca2+ release; (iii) the direct interaction with PKC does not significantly contribute to the antitumor activity of these agents; (iv) the new HePC congeners D21266, D21133 and D21805 affect the same targets as HePC, i.e. PKC and phosphatidylinositol 4,5-bisphosphate-specific phospholipase C (PLC). The lower toxicities of these compounds cannot be explained by a less pronounced inhibition of PKC or PLC, respectively.


Asunto(s)
Antineoplásicos/farmacología , Fosfolípidos/farmacología , Transducción de Señal/efectos de los fármacos , Células 3T3/efectos de los fármacos , Células 3T3/enzimología , Células 3T3/fisiología , Animales , Calcio/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Sistema Libre de Células , Regulación de la Expresión Génica/efectos de los fármacos , Genes fos , Inositol 1,4,5-Trifosfato/biosíntesis , Ratones , Regiones Promotoras Genéticas/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Receptores de Trombina/efectos de los fármacos , Receptores de Trombina/fisiología , Transducción de Señal/fisiología , Intercambiadores de Sodio-Hidrógeno/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA