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











Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
Biochem J ; 359(Pt 1): 211-7, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11563985

RESUMO

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.


Assuntos
Regulação Enzimológica da Expressão Gênica , Genes ras/fisiologia , Isoenzimas/metabolismo , Neoplasias Mamárias Experimentais/enzimologia , Fosfolipase D/metabolismo , Proteína Quinase C/metabolismo , Animais , Células COS , Transformação Celular Neoplásica , Chlorocebus aethiops , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Humanos , Indóis/farmacologia , Luciferases/metabolismo , Maleimidas/farmacologia , Neoplasias Mamárias Experimentais/genética , Camundongos , Fosfolipase D/genética , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Transfecção
2.
J Biol Chem ; 276(46): 42834-42, 2001 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11551901

RESUMO

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.


Assuntos
Isoformas de Proteínas , Proteína Quinase C/química , Animais , Western Blotting , Mama/metabolismo , Células Cultivadas , Clonagem Molecular , Ciclina D1/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Flavonoides/farmacologia , Humanos , Isoenzimas/metabolismo , Luciferases/metabolismo , MAP Quinase Quinase 1 , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteína Quinase C-épsilon , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Transfecção
5.
Mol Cell Biol ; 19(12): 8052-65, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10567531

RESUMO

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.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ativação Transcricional , Proteínas ras/metabolismo , Células 3T3 , Animais , Transporte Biológico , Células COS , Fracionamento Celular , Membrana Celular/metabolismo , Citosol/metabolismo , Ativação Enzimática , Expressão Gênica , Variação Genética , Humanos , Líquido Intracelular , Isoenzimas , MAP Quinase Quinase 1 , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
6.
J Cell Biol ; 144(3): 413-25, 1999 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-9971737

RESUMO

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.


Assuntos
Actinas/metabolismo , Proteína Quinase C/metabolismo , Proteínas ras/metabolismo , Células 3T3 , Animais , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Isoenzimas , Maleimidas/farmacologia , Camundongos , Microscopia de Fluorescência , Mutação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Transdução de Sinais , Transfecção , Proteínas ras/genética
7.
Biochem Pharmacol ; 56(7): 861-9, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9774148

RESUMO

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.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Substituição de Aminoácidos/genética , Resistência a Múltiplos Medicamentos , Lactonas/farmacologia , Proteína Quinase C/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Ligação Competitiva/efeitos dos fármacos , Briostatinas , Resistencia a Medicamentos Antineoplásicos , Feminino , Células HeLa , Humanos , Células KB , Macrolídeos , Mutação Puntual
10.
EMBO J ; 17(14): 4046-55, 1998 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9670019

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Genes fos/genética , Isoenzimas/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteína Oncogênica p21(ras)/metabolismo , Proteína Quinase C/metabolismo , Ativação Transcricional/fisiologia , Animais , Linhagem Celular , Isoenzimas/genética , MAP Quinase Quinase 1 , Glândulas Mamárias Animais/citologia , Camundongos , Regiões Promotoras Genéticas/genética , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , RNA Antissenso , Transdução de Sinais/genética , Transfecção
11.
Ann Oncol ; 9 Suppl 5: S125-8, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9926251

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Doença de Hodgkin/tratamento farmacológico , Células de Reed-Sternberg/efeitos dos fármacos , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Genes bcl-2/efeitos dos fármacos , Genes ras/efeitos dos fármacos , Doença de Hodgkin/genética , Doença de Hodgkin/patologia , Humanos , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/metabolismo , Células de Reed-Sternberg/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/farmacologia
12.
J Med Chem ; 40(26): 4420-5, 1997 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-9435912

RESUMO

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].


Assuntos
Antineoplásicos/síntese química , Hidrazonas/síntese química , Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Citidina/metabolismo , DNA/biossíntese , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Hidrazonas/farmacologia , Hidroxiureia/farmacologia , Espectroscopia de Ressonância Magnética , Conformação Molecular , Estrutura Molecular , Ribonucleotídeo Redutases/antagonistas & inibidores , Ribonucleotídeo Redutases/metabolismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas
13.
J Biol Chem ; 271(48): 30505-9, 1996 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-8940018

RESUMO

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.


Assuntos
Cálcio/fisiologia , Receptores ErbB/fisiologia , Receptores de Fator de Crescimento Neural/fisiologia , Células 3T3 , Animais , Citosol/fisiologia , Isoenzimas/fisiologia , Camundongos , Fosfolipase C gama , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Transdução de Sinais , Fosfolipases Tipo C/fisiologia
14.
Adv Enzyme Regul ; 36: 385-407, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8869757

RESUMO

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.


Assuntos
Neoplasias/terapia , Fosfolipídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Antineoplásicos/farmacologia , Cálcio/metabolismo , Inibidores Enzimáticos/farmacologia , Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Inositol 1,4,5-Trifosfato/metabolismo , Estrutura Molecular , Fosfolipase D/antagonistas & inibidores , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/metabolismo , Trombina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores
15.
EMBO J ; 15(1): 73-82, 1996 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-8598208

RESUMO

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.


Assuntos
Transformação Celular Neoplásica , Receptores ErbB/fisiologia , Isoenzimas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator de Crescimento Neural/fisiologia , Fosfolipases Tipo C/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Primers do DNA/química , Ativação Enzimática , Humanos , Fosfatos de Inositol/metabolismo , Camundongos , Dados de Sequência Molecular , Fosfolipase C gama , Fosfotirosina/metabolismo , Ligação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Tirosina/química , Domínios de Homologia de src
16.
Cell Calcium ; 18(2): 120-34, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7585889

RESUMO

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.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Células 3T3 , Animais , Transporte Biológico , Bradicinina/farmacologia , Dexametasona/farmacologia , Fosfatos de Inositol/metabolismo , Manganês/metabolismo , Camundongos , Canais de Potássio/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Rubídio/metabolismo
17.
Anticancer Drug Des ; 10(5): 411-25, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7639930

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Fosfolipídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células 3T3/efeitos dos fármacos , Células 3T3/enzimologia , Células 3T3/fisiologia , Animais , Cálcio/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Sistema Livre de Células , Regulação da Expressão Gênica/efeitos dos fármacos , Genes fos , Inositol 1,4,5-Trifosfato/biossíntese , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Receptores de Trombina/efeitos dos fármacos , Receptores de Trombina/fisiologia , Transdução de Sinais/fisiologia , Trocadores de Sódio-Hidrogênio/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores
18.
FEBS Lett ; 352(1): 11-4, 1994 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-7925931

RESUMO

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.


Assuntos
Biopterinas/análogos & derivados , GMP Cíclico/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes fos/efeitos dos fármacos , Pteridinas/farmacologia , Células 3T3 , Animais , Sequência de Bases , Biopterinas/farmacologia , Cloranfenicol O-Acetiltransferase/genética , Genes fos/genética , Humanos , Camundongos , Dados de Sequência Molecular , Neopterina , Dibutirato de 12,13-Forbol/farmacologia , Regiões Promotoras Genéticas/genética , Proteína Quinase C/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
19.
Cell Signal ; 6(3): 285-97, 1994 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7917786

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Glândulas Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Sequências Reguladoras de Ácido Nucleico , Animais , Antineoplásicos/farmacologia , Sequência de Bases , Linhagem Celular , Células Cultivadas , Proteínas de Ligação a DNA , Dexametasona/farmacologia , Epitélio/metabolismo , Feminino , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas Nucleares , Gravidez , Proteína Quinase C/metabolismo , Fator de Resposta Sérica , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Transcrição/metabolismo , Transfecção
20.
Adv Enzyme Regul ; 34: 257-68, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7942278

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas ras/metabolismo , Animais , Briostatinas , Compartimento Celular , Linhagem Celular , Células Epiteliais , Regulação da Expressão Gênica/efeitos dos fármacos , Lactonas/farmacologia , Macrolídeos , Glândulas Mamárias Animais/citologia , Camundongos , Proteínas Recombinantes/biossíntese , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA