Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Oncogene ; 33(45): 5225-37, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24336328

RESUMO

Since their discovery in the late 1970s, protein kinase C (PKC) isozymes represent one of the most extensively studied signaling kinases. PKCs signal through multiple pathways and control the expression of genes relevant for cell cycle progression, tumorigenesis and metastatic dissemination. Despite the vast amount of information concerning the mechanisms that control PKC activation and function in cellular models, the relevance of individual PKC isozymes in the progression of human cancer is still a matter of controversy. Although the expression of PKC isozymes is altered in multiple cancer types, the causal relationship between such changes and the initiation and progression of the disease remains poorly defined. Animal models developed in the last years helped to better understand the involvement of individual PKCs in various cancer types and in the context of specific oncogenic alterations. Unraveling the enormous complexity in the mechanisms by which PKC isozymes have an impact on tumorigenesis and metastasis is key for reassessing their potential as pharmacological targets for cancer treatment.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Proteína Quinase C/genética , Animais , Progressão da Doença , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias/enzimologia , Neoplasias/patologia , Proteína Quinase C/metabolismo , Transdução de Sinais/genética
2.
Oncogene ; 31(20): 2593-600, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21996750

RESUMO

Protein kinase C (PKC)ɛ, a member of the novel PKC family, has key roles in mitogenesis and survival in normal and cancer cells. PKCɛ is frequently overexpressed in epithelial cancers, particularly in lung cancer. Using a short-hairpin RNA approach, here we established that PKCɛ is required for non-small cell lung carcinoma (NSCLC) growth in vitro as well as tumor growth when inoculated into athymic mice. Moreover, sustained delivery of a PKCɛ-selective inhibitor peptide, ɛV1-2, reduced xenograft growth in mice. Both RNA interference depletion and pharmacological inhibition of PKCɛ caused a marked elevation in the number of apoptotic cells in NSCLC tumors. PKCɛ-depleted NSCLC cells show elevated expression of pro-apoptotic proteins of the Bcl-2 family, caspase recruitment domain-containing proteins and tumor necrosis factor ligands/receptor superfamily members. Moreover, a Gene Set Enrichment Analysis revealed that a vast majority of the genes changed in PKCɛ-depleted cells were also deregulated in human NSCLC. Our results strongly suggest that PKCɛ is required for NSCLC cell survival and maintenance of NSCLC tumor growth. Therefore, PKCɛ may represent an attractive therapeutic target for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/enzimologia , Neoplasias Pulmonares/enzimologia , Proteína Quinase C-épsilon/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular , Genes bcl-2 , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fragmentos de Peptídeos/farmacologia , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptores do Fator de Necrose Tumoral/metabolismo
3.
Biochem Soc Trans ; 34(Pt 5): 855-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17052214

RESUMO

We have established a novel role for the second messenger DAG (diacylglycerol), a product of PtdIns(4,5)P2 hydrolysis by PLC (phospholipase C). In addition to its well-known function as a protein kinase C activator, DAG produced by stimulation of the epidermal growth factor receptor causes the redistribution of the Rac-GAP (GTPase-activating protein) beta2-chimaerin to the plasma membrane, where it associates with the active form of Rac1 and promotes the inactivation of this small G-protein. This represents the first example of a Rac-GAP regulated directly by DAG in response to the activation of a tyrosine kinase receptor, and suggests a previously unappreciated role for this lipid as a negative modulator of Rac signalling.


Assuntos
Diglicerídeos/fisiologia , Sistemas do Segundo Mensageiro , Proteínas rac de Ligação ao GTP/fisiologia , Animais , Receptores ErbB/fisiologia , Homeostase , Proteínas de Neoplasias/fisiologia , Fosfolipase C gama/metabolismo , Fosfolipases/metabolismo , Proteína Quinase C/fisiologia , Receptores de Droga/fisiologia , Transdução de Sinais
4.
Cell Growth Differ ; 12(12): 603-11, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11751455

RESUMO

Rho family GTPases Rac and Cdc42 are pivotal regulators of apoptosis in multiple cell types. However, little is known about the mechanism by which these GTPases are regulated in response to apoptotic stimuli. Here, we demonstrate that TIAM1, a Rac-specific guanine nucleotide exchange factor, is cleaved by caspases during apoptosis. TIAM1 cleavage occurs in multiple cell lines in response to diverse apoptotic stimuli such as ceramide, Fas, and serum deprivation. Processing occurs at residue 993 of TIAM1 and removes the NH(2)-terminal of TIAM's two pleckstrin homology domains, leaving a stable fragment containing the Dbl homology and COOH-terminal pleckstrin homology domains. This leads to functional inactivation of TIAM1, as determined by failure of the cleavage product to stimulate GTP loading of Rac in vivo. Furthermore, this product is defective in signaling to two independent Rac effectors, c-Jun NH(2)-terminal kinase and serum response factor. Finally, we demonstrate that in cells treated with ceramide, cleavage of TIAM1 coincided with the inactivation of endogenous Rac. These results reveal a novel mechanism for regulating guanine nucleotide exchange factor activity and GTPase-mediated signaling pathways.


Assuntos
Apoptose , Caspases/metabolismo , Proteínas/metabolismo , Células 3T3 , Animais , Sítios de Ligação , Proteínas Sanguíneas/química , Células COS , Linhagem Celular , Membrana Celular/metabolismo , Ceramidas/metabolismo , Ceramidas/farmacologia , Ativação Enzimática , GTP Fosfo-Hidrolases/metabolismo , Fatores de Troca do Nucleotídeo Guanina , Humanos , Immunoblotting , Proteínas Quinases JNK Ativadas por Mitógeno , Células Jurkat , Luciferases/metabolismo , Camundongos , Microscopia de Fluorescência , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas de Neoplasias , Células PC12 , Fosfoproteínas/química , Testes de Precipitina , Estrutura Terciária de Proteína , Ratos , Fator de Resposta Sérica/metabolismo , Transdução de Sinais , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Fatores de Tempo , Células Tumorais Cultivadas , Proteínas rac de Ligação ao GTP/metabolismo
5.
Cell Growth Differ ; 12(10): 517-24, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11682463

RESUMO

Mouse melanoma B16 F1 cells cultured in RPMI 1640 supplemented with the melanin precursors tyrosine and phenylalanine display increased melanin levels and elevated migration while down-regulating protein kinase C (PKC)zeta to low levels. Although control experiments rule out a direct role by melanin, PKCzeta down-regulation is shown to be a critical determinant of cell migration. Transfection of high-motility cells with either wild-type PKCzeta or its regulatory domain suppresses migration. Known to bind to the regulatory domain of PKCzeta, the proapoptotic protein prostate apoptosis response-4 (Par-4) coimmunoprecipitates with PKCzeta as a 47-kDa protein. Transfection of Par-4 (or its leucine zipper element) further suppresses migration of low-motility cells (which express high levels of PKCzeta), whereas high-motility cells (which express low levels of PKCzeta) are unaffected by Par-4 overexpression. It is proposed that in nonmetastatic cells, the PKCzeta Par-4 complex provides a brake on migration that is released by melanin precursors that initiate PKCzeta down-regulation. Elevation of PKCzeta in melanoma cells, or preventing its down-regulation through the dietary restriction of tyrosine and phenylalanine, may therefore control metastatic behavior.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Proteína Quinase C/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Western Blotting , Proteínas de Transporte/metabolismo , Adesão Celular , Linhagem Celular , Movimento Celular , Separação Celular , Células Cultivadas , DNA Complementar/metabolismo , Regulação para Baixo , Melaninas/biossíntese , Melanoma Experimental , Camundongos , Mutagênese Sítio-Dirigida , Metástase Neoplásica , Fenilalanina/metabolismo , Testes de Precipitina , Relação Estrutura-Atividade , Transfecção , Células Tumorais Cultivadas , Tirosina/metabolismo
6.
Curr Pharm Des ; 7(17): 1725-44, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11562308

RESUMO

Recent years have seen extensive growth in the understanding of the role(s) of the various PKC isozymes and novel receptors for the phorbol ester tumor promoters. The PKC family of serine-threonine kinases is an important regulator of signaling cascades that control cell proliferation and death, and therefore represent targets for cancer therapy. While past interests have focused on PKC-selective inhibitors, more recently, intensive research has been underway for selective activators and inhibitors for each individual PKC isozyme. In the past few years a large number of PKC activators and inhibitors with potential as anticancer agents have been developed. A number of these compounds are already in Phase II clinical testing. As a new generation of cancer chemotherapeutic agents are designed, developed and put through a series of rigorous clinical trials, we can anticipate achieving exquisite control over PKC-mediated regulatory pathways, leading ultimately to a greater understanding of different cancers.


Assuntos
Antineoplásicos/metabolismo , Proteínas de Caenorhabditis elegans , Forbóis/metabolismo , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Receptores de Droga/metabolismo , Animais , Antineoplásicos/química , Sítios de Ligação , Proteínas de Transporte , Desenho de Fármacos , Humanos , Forbóis/química , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/fisiologia , Receptores de Droga/química
7.
Arch Biochem Biophys ; 388(1): 74-80, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11361144

RESUMO

In this study we have evaluated the specificity of different PKC isozymes for the phosphorylation of the catalytic alpha1 subunit of rat renal Na+,K+-ATPase (alpha1 Na+,K+-ATPase). Using in vitro phosphotransferase assays we found that classical PKCs (cPKCs) alpha, betaI, and gamma efficiently phosphorylate alpha1 Na+,K+-ATPase. However, alpha1 Na+,K+-ATPase was a poor substrate for the novel PKCs (nPKCs) delta and epsilon. Two-dimensional phosphopeptide mapping revealed a similar pattern of phosphorylation by all cPKCs. The functional significance of this finding was evaluated by measuring Na+,K+-ATPase activity (assessed by 86Rb+ uptake) in COS-7 cells expressing the rat alpha1 Na+,K+-ATPase. 1-oleoyl-2-acetoyl-sn-glycerol (OAG), a nonselective PKC activator, inhibited Na+,K+-ATPase activity in this system. On the other hand, 12-deoxyphorbol-13-phenylacetate (DPP), which preferentially activates nPKCepsilon, did not affect 86Rb+ uptake. These results indicate a differential pattern of phosphorylation and regulation of rat renal Na+,K+-ATPase activity by PKC isoforms and suggest an important role for cPKCs in the physiological regulation of the pump.


Assuntos
Proteína Quinase C/química , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Células COS , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Irritantes/farmacologia , Isoenzimas , Ouabaína/farmacologia , Mapeamento de Peptídeos , Ésteres de Forbol/farmacologia , Fosforilação , Isoformas de Proteínas , Proteína Quinase C/metabolismo , Transporte Proteico , Ratos , Rubídio/farmacologia , Sódio/farmacologia , Fatores de Tempo , Transfecção
8.
Bioorg Med Chem Lett ; 11(8): 955-9, 2001 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-11327599

RESUMO

In view of its role in tumor promotion and signal transduction, protein kinase C (PKC) has proven to be an exciting target for cancer therapy. With the aid of molecular modeling, we rationally designed and stereoselectively synthesized a new class of rigidified pyrrolidone-based PKC activators. Pyrrolidone 15 was found to exhibit reasonable affinity for PKCdelta, with lower affinity for the other isozymes tested. Pyrrolidone 2 causes the dose-dependent induction of apoptosis in LNCaP prostate cancer cells. This apoptotic effect could be markedly potentiated by the use of LNCaP cells overexpressing the PKCalpha or delta isozymes.


Assuntos
Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Ativadores de Enzimas/síntese química , Isoenzimas/efeitos dos fármacos , Neoplasias da Próstata/fisiopatologia , Proteína Quinase C/efeitos dos fármacos , Pirrolidinonas/síntese química , Antineoplásicos/farmacologia , Apoptose/fisiologia , Relação Dose-Resposta a Droga , Desenho de Fármacos , Ativadores de Enzimas/farmacologia , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Próstata/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteína Quinase C-delta , Pirrolidinonas/farmacologia , Estereoisomerismo , Células Tumorais Cultivadas
9.
J Med Chem ; 44(7): 1028-34, 2001 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-11297449

RESUMO

N-Benzyladriamycin-14-valerate (AD 198) is a semisynthetic anthracycline with experimental antitumor activity superior to that of doxorubicin (DOX). AD 198, unlike DOX, only weakly binds DNA, is a poor inhibitor of topoisomerase II, and circumvents anthracycline-resistance mechanisms, suggesting a unique mechanism of action for this novel analogue. The phorbol ester receptors, protein kinase C (PKC) and beta2-chimaerin, were recently identified as selective targets for AD 198 in vitro. In vitro, AD 198 competes with [3H]PDBu for binding to a peptide containing the isolated C1b domain of PKC-delta (deltaC1b domain). In the present study molecular modeling is used to investigate the interaction of AD 198 with the deltaC1b domain. Three models are identified wherein AD 198 binds into the groove formed between amino acid residues 6-13 and 21-27 of the deltaC1b domain in a manner similar to that reported for phorbol-13-acetate and other ligands of the C1 domain. Two of the identified models are consistent with previous experimental data demonstrating the importance of the 14-valerate side chain of AD 198 in binding to the C1 domain as well as current data demonstrating that translocation of PKC-alpha to the membrane requires the 14-valerate substituent. In this regard, the carbonyl of the 14-valerate participates in hydrogen bonding to the deltaC1b while the acyl chain is positioned for stabilization of the membrane-bound protein-ligand complex in a manner analogous to the acyl chains of the phorbol esters. These studies provide a structural basis for the interaction of AD 198 with the deltaC1b domain and a starting point for the rational design of potential new drugs targeting PKC and other proteins with C1 domains.


Assuntos
Antibióticos Antineoplásicos/química , Proteínas de Caenorhabditis elegans , Doxorrubicina/química , Isoenzimas/química , Proteína Quinase C/química , Receptores de Droga/química , Proteínas de Transporte , Doxorrubicina/análogos & derivados , Modelos Moleculares , Ligação Proteica , Proteína Quinase C-delta
10.
J Biol Chem ; 276(18): 15537-46, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11297525

RESUMO

In this study, we examined the mitogen-activated protein kinase (MAPK) cascade in micrometastatic cell lines generated from rib bone marrow (RBM) of patients undergoing resection of esophagogastric malignancies. The molecular mechanism(s) involved in esophagogastric MAPK activation have not previously been investigated. Constitutive activation of both ERK1 and -2 isoforms was evident in each of the five RBM cell lines. Elk-1, a transcription factor activated by the ERK1/2 pathway was also found to be constitutively activated. Cell lines generated from metastases of involved lymph nodes (OC2) and ascites (OC1) of patients with esophageal cancer do not display, however, hyperphosphorylation of ERK1/2. Constitutive RBM ERK1/2 activation is protein kinase C and phosphatidylinositol 3-kinase dependent. Surprisingly, constitutive ERK1/2 activation is MEK-independent. Pharmacological inhibition of MEK with two specific inhibitors, PD 98059 and U0126, were both ineffective in blocking ERK activation. Similarly, the use of a dominant negative MEK mutant was without effect. Interestingly, experiments overexpressing two different dominant negative Pak1 mutants significantly reduced RBM ERK1/2 activation, albeit not to the same extent for all cell lines. We also examined the role of three different phosphatases, PAC1, MKP-1, and -2. While RBM ERK1/2 activation was found to be PAC1- and MKP-2-independent, surprisingly, MKP-1 was down-regulated in all five RBM cell lines. In conclusion, we provide evidence for the first time for a MEK-independent constitutive ERK1/2 activation pathway in esophagogastric RBM cell lines. These findings have important implications for drug treatment strategies which currently target MEK in other forms of cancer.


Assuntos
Neoplasias da Medula Óssea/secundário , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Costelas/enzimologia , Sequência de Bases , Neoplasias da Medula Óssea/enzimologia , Neoplasias da Medula Óssea/patologia , Primers do DNA , Ativação Enzimática , Neoplasias Esofágicas/patologia , Esôfago , Proteína Quinase 3 Ativada por Mitógeno , Estômago , Neoplasias Gástricas/patologia
11.
J Biol Chem ; 276(21): 18303-12, 2001 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-11278894

RESUMO

The novel phorbol ester receptor beta2-chimaerin is a Rac-GAP protein possessing a single copy of the C1 domain, a 50-amino acid motif initially identified in protein kinase C (PKC) isozymes that is involved in phorbol ester and diacylglycerol binding. We have previously shown that, like PKCs, beta2-chimaerin binds phorbol esters with high affinity in a phospholipid-dependent manner (Caloca, M. J., Fernandez, M. N., Lewin, N. E., Ching, D., Modali, R., Blumberg, P. M., and Kazanietz, M. G. (1997) J. Biol. Chem. 272, 26488-26496). In this paper we report that like PKC isozymes, beta2-chimaerin is translocated by phorbol esters from the cytosolic to particulate fraction. Phorbol esters also induce translocation of alpha1 (n)- and beta1-chimaerins, suggesting common regulatory mechanisms for all chimaerin isoforms. The subcellular redistribution of beta2-chimaerin by phorbol esters is entirely dependent on the C1 domain, as revealed by deletional analysis and site-directed mutagenesis. Interestingly, beta2-chimaerin translocates to the Golgi apparatus after phorbol ester treatment, as revealed by co-staining with the Golgi marker BODIPY-TR-ceramide. Structure relationship analysis of translocation using a series of PKC ligands revealed substantial differences between translocation of beta2-chimaerin and PKCalpha. Strikingly, the mezerein analog thymeleatoxin is not able to translocate beta2-chimaerin, although it very efficiently translocates PKCalpha. Phorbol esters also promote the association of beta2-chimaerin with Rac in cells. These data suggest that chimaerins can be positionally regulated by phorbol esters and that each phorbol ester receptor class has distinct pharmacological properties and targeting mechanisms. The identification of selective ligands for each phorbol ester receptor class represents an important step in dissecting their specific cellular functions.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas de Neoplasias/metabolismo , Ésteres de Forbol/metabolismo , Transdução de Sinais , Animais , Células COS , Proteínas de Transporte , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Receptores de Droga/metabolismo
12.
Biochem Pharmacol ; 60(10): 1417-24, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11020443

RESUMO

The phorbol ester tumor promoters and related analogs are widely used as potent activators of protein kinase C (PKC). The phorbol esters mimic the action of the lipid second messenger diacylglycerol (DAG). The aim of this commentary is to highlight a series of important and controversial concepts in the pharmacology and regulation of phorbol ester receptors. First, phorbol ester analogs have marked differences in their biological properties. This may be related to a differential regulation of PKC isozymes by distinct analogs. Moreover, it seems that marked differences exist in the ligand recognition properties of the C1 domains, the phorbol ester/DAG binding sites in PKC isozymes. Second, an emerging theme that we discuss here is that phorbol esters also target receptors unrelated to PKC isozymes, a concept that has been largely ignored. These novel receptors lacking kinase activity include chimaerins (a family of Rac-GTPase-activating proteins), RasGRP (a Ras exchange factor), and Unc-13/Munc-13 (a family of proteins involved in exocytosis). Unlike the classical and novel PKCs, these "non-kinase" phorbol ester receptors possess a single copy of the C1 domain. Interestingly, each receptor class has unique pharmacological properties and biochemical regulation. Lastly, it is well established that phorbol esters and related analogs can translocate each receptor to different intracellular compartments. The differential pharmacological properties of the phorbol ester receptors can be exploited to generate specific agonists and antagonists that will be helpful tools to dissect their cellular function.


Assuntos
Proteínas de Caenorhabditis elegans , Carcinógenos/farmacologia , Diglicerídeos/metabolismo , Ésteres de Forbol/farmacologia , Proteína Quinase C/metabolismo , Receptores de Droga/metabolismo , Animais , Transporte Biológico , Carcinógenos/toxicidade , Proteínas de Transporte , Sequência Conservada , Diglicerídeos/química , Ativação Enzimática , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Mimetismo Molecular , Ésteres de Forbol/toxicidade , Conformação Proteica , Proteína Quinase C/química , Proteína Quinase C/efeitos dos fármacos , Receptores de Droga/química , Receptores de Droga/efeitos dos fármacos , Sistemas do Segundo Mensageiro
13.
FEBS Lett ; 483(1): 27-32, 2000 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-11033350

RESUMO

The C1 domains are 50 amino acid sequences present in protein kinase C (PKC) isozymes that are responsible for binding of phorbol esters and the lipid second messenger diacylglycerol (DAG). We found that bacterially expressed C1b domain of PKCdelta induces germinal vesicle breakdown (GVBD) when microinjected into Xenopus laevis oocytes. Injection of the C1b domain of PKCdelta significantly enhanced insulin- but not progesterone-induced maturation. Interestingly, the PKCdelta C1b domain markedly synergized with normal Ras protein to induce oocyte maturation when both proteins were co-injected in oocytes. Our results demonstrate that the purified C1b domain of PKCdelta is sufficient to promote meiotic maturation of X. laevis oocytes probably through activation of components of the insulin/Ras signaling pathway.


Assuntos
Isoenzimas/farmacologia , Meiose/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Proteína Quinase C/farmacologia , Substituição de Aminoácidos , Animais , Sítios de Ligação/genética , Relação Dose-Resposta a Droga , Feminino , Insulina/farmacologia , Isoenzimas/genética , Isoenzimas/metabolismo , Microinjeções , Mutação , Oócitos/crescimento & desenvolvimento , Ésteres de Forbol/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/farmacologia , Progesterona/farmacologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Proteínas Recombinantes/farmacologia , Fatores de Tempo , Xenopus , Proteínas ras/farmacologia , Domínios de Homologia de src/genética
14.
Mol Carcinog ; 28(1): 5-11, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10820483

RESUMO

In addition to the well-characterized interaction with classical and novel protein kinase C (PKC) isozymes, the phorbol ester tumor promoters bind to other receptors lacking kinase activity. Among these novel phorbol ester receptors, two families of proteins may play a role in the regulation of cell growth and malignant transformation: chimaerins and ras guanyl-releasing protein (ras-GRP). These proteins possess a single copy of the C1 domain that is involved in binding of phorbol esters and the lipid second messenger diacylglycerol. Four isoforms of chimaerins (alpha1-, alpha2-, beta1-, and beta2-chimaerins) have been isolated to-date, all of them possessing GTPase-activating protein activity for Rac, a small GTP-binding protein that controls actin cytoskeleton organization, cell-cycle progression, adhesion, and migration. Ras-GRP is a guanine nucleotide exchange factor for ras and promotes malignant transformation in fibroblasts in a phorbol ester-dependent manner. The C1 domain in Ras-GRP may, therefore, have a dominant role in Ras-GRP activation and is essential for phorbol ester-dependent activation of downstream effectors of ras, i.e., the mitogen-activated protein kinase cascade. Thus, a novel concept emerges in which phorbol esters may exert cellular responses through pathways not involving phorbol ester-responsive PKC isozymes. The discovery of "nonPKC" phorbol ester receptors adds an additional level of complexity to the understanding of phorbol ester effects and the molecular mechanisms of carcinogenesis.


Assuntos
Carcinógenos , Transformação Celular Neoplásica , Fatores de Troca do Nucleotídeo Guanina , Ésteres de Forbol , Proteína Quinase C/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Carcinógenos/toxicidade , Transformação Celular Neoplásica/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Humanos , Isoenzimas/fisiologia , Ésteres de Forbol/toxicidade , Transdução de Sinais
15.
J Biol Chem ; 275(11): 7574-82, 2000 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-10713064

RESUMO

Phorbol esters, the activators of protein kinase C (PKC), induce apoptosis in androgen-sensitive LNCaP prostate cancer cells. The role of individual PKC isozymes as mediators of this effect has not been thoroughly examined to date. To study the involvement of the novel isozyme PKCdelta, we used a replication-deficient adenovirus (PKCdeltaAdV), which allowed for a tightly controlled expression of PKCdelta in LNCaP cells. A significant reduction in cell number was observed after infection of LNCaP cells with PKCdeltaAdV. Overexpression of PKCdelta markedly enhanced the apoptotic effect of phorbol 12-myristate 13-acetate in LNCaP cells. PKCdelta-mediated apoptosis was substantially reduced by the pan-caspase inhibitor z-VAD and by Bcl-2 overexpression. Importantly, and contrary to other cell types, PKCdelta-mediated apoptosis does not involve its proteolytic cleavage by caspase-3, suggesting that allosteric activation of PKCdelta is sufficient to trigger apoptosis in LNCaP cells. In addition, phorbol ester-induced apoptosis was blocked by a kinase-deficient mutant of PKCdelta, supporting the concept that PKCdelta plays an important role in the regulation of apoptotic cell death in LNCaP prostate cancer cells.


Assuntos
Apoptose , Isoenzimas/metabolismo , Neoplasias da Próstata/enzimologia , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Ativação Enzimática , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Masculino , Mutação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteína Quinase C-delta , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas
17.
J Biol Chem ; 275(15): 11418-24, 2000 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-10753958

RESUMO

Pulse treatment of U-937 promonocytic cells with cadmium chloride (2 h at 200 microM) provoked apoptosis and induced a rapid phosphorylation of p38 mitogen-activated protein kinase (p38(MAPK)) as well as a late phosphorylation of extracellular signal-regulated protein kinases (ERK1/2). However, although the p38(MAPK)-specific inhibitor SB203580 attenuated apoptosis, the process was not affected by the ERK-specific inhibitor PD98059. The attenuation of the cadmium-provoked apoptosis by SB203580 was a highly specific effect. In fact, the kinase inhibitor did not prevent the generation of apoptosis by heat shock and camptothecin, nor the generation of necrosis by cadmium treatment of glutathione-depleted cells, nor the cadmium-provoked activation of the stress response. The generation of apoptosis was preceded by intracellular H(2)O(2) accumulation and was accompanied by the disruption of mitochondrial transmembrane potential, both of which were inhibited by SB203580. On the other hand, the antioxidant agent butylated hydroxyanisole-inhibited apoptosis but did not prevent p38(MAPK) phosphorylation. In a similar manner, p38(MAPK) phosphorylation was not affected by the caspase inhibitors Z-VAD and DEVD-CHO, which nevertheless prevented apoptosis. These results indicate that p38(MAPK) activation is an early and specific regulatory event for the cadmium-provoked apoptosis in promonocytic cells.


Assuntos
Apoptose/efeitos dos fármacos , Cádmio/toxicidade , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Caspases/metabolismo , Ativação Enzimática , Flavonoides/farmacologia , Humanos , Imidazóis/farmacologia , Potenciais da Membrana , Mitocôndrias/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Necrose , Estresse Oxidativo , Piridinas/farmacologia , Células U937 , Proteínas Quinases p38 Ativadas por Mitógeno
18.
Endocrinology ; 141(1): 146-52, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10614633

RESUMO

Several reports have indicated that protein kinase C (PKC) is an important regulator of proliferation in thyroid cells. Unlike TSH, the mitogenic effects of phorbol esters are accompanied by de-differentiation. The role of individual PKC isoforms in thyroid cell proliferation and differentiation has not been examined. Recent studies have implicated the atypical PKCzeta, a phorbol ester-unresponsive isozyme, in cell proliferation, death, and survival. We overexpressed PKCzeta in Wistar rat thyroid (WRT) cells and determined that PKCzeta conferred TSH-independent DNA synthesis and cell proliferation. Cells overexpressing PKCzeta show higher levels of phosphorylated p42/p44 MAPK compared with vector-transfected cells. Experiments using a luciferase reporter for Elk-1 revealed that PKCzeta overexpressing cells exhibit higher basal Elk-1 transcriptional activity than vector-transfected control cells. Interestingly, stimulation of Elk-1 transcriptional activity by MEK1, a p42/p44 MAPK kinase, was significantly enhanced in cells overexpressing PKCzeta. Strikingly, TSH retained the ability to stimulate Tg expression in cells expressing PKCzeta. These results suggest that PKCzeta stimulates TSH-independent mitogenesis through a p42/p44 MAPK-dependent pathway. Unlike overexpression of Ras or phorbol ester treatment, PKC overexpression does not impair thyroglobulin (Tg) expression.


Assuntos
Proteínas de Ligação a DNA , Proteína Quinase C/fisiologia , Glândula Tireoide/citologia , Tireotropina/fisiologia , Animais , Western Blotting , Divisão Celular , Células Cultivadas , DNA/biossíntese , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Vetores Genéticos/genética , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase C/biossíntese , Proteína Quinase C/genética , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Wistar , Fatores de Transcrição/genética , Proteínas Elk-1 do Domínio ets
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...