Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Phytother Res ; 37(5): 1839-1849, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36512326

RESUMO

Salidroside, a prominent active ingredient in traditional Chinese medicines, is garnering increased attention because of its unique pharmacological effects against ischemic heart disease via MAPK signaling, which plays a critical role in regulating the evolution of ventricular hypertrophy. However, the function of Salidroside on myocardial hypertrophy has not yet been elucidated. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with Salidroside (100 mg kg-1  day-1 ) by oral gavage for 3 weeks starting 1 week after surgery. Four weeks after TAC surgery, the mice were subjected to echocardiography and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes were used to validate the protective effects of Salidroside in response to Angiotensin II (Ang II, 1 µM) stimulation. Here, we proved that Salidroside dramatically inhibited hypertrophic reactions generated by pressure overload and isoproterenol (ISO) injection. Salidroside prevented the activation of the TAK1-JNK/p38 axis. Salidroside pretreatment of TAK1-inhibited cardiomyocytes shows no additional attenuation of Ang II-induced cardiomyocytes hypertrophy and signaling pathway activation. The overexpression of constitutively active TAK1 removed the protective effects of Salidroside on myocardial hypertrophy. TAC-induced increase of TLR4 protein expression was reduced considerably in the Salidroside treated mice. Transient transfection of small interfering RNA targeting TLR4 (siTLR4) in cardiomyocytes did not further decrease the activation of the TAK1/JNK-p38 axis. In conclusion, Salidroside functioned as a TLR4 inhibitor and displayed anti-hypertrophic action via the TAK1/JNK-p38 pathway.


Assuntos
Estenose da Valva Aórtica , Cardiomegalia , Receptor 4 Toll-Like , Animais , Camundongos , Ratos , Estenose da Valva Aórtica/metabolismo , Cardiomegalia/tratamento farmacológico , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Células Cultivadas , Modelos Animais de Doenças , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , MAP Quinase Quinase Quinases/farmacologia , Camundongos Endogâmicos C57BL , Miócitos Cardíacos , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
2.
J Pharmacol Sci ; 143(3): 133-140, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32253104

RESUMO

Endogenous noradrenaline (NA) has multiple bioactive functions and, in the central nervous system (CNS), has been implicated in modulating neuroinflammation via ß-adrenergic receptors (ß-ARs). Microglia, resident macrophages in the CNS, have a central role in the brain immune system and have been reported to be activated by NA. However, intracellular signaling mechanisms of the AR-mediated proinflammatory responses of microglia are not fully understood. Using a rapid and stable in vitro reporter assay system to evaluate IL-1ß production in microglial BV2 cells, we found that NA and the ß-AR agonist isoproterenol upregulated the IL-1ß reporter activity. This effect was suppressed by ß-AR antagonists. We further examined the involvement of EPAC (exchange protein directly activated by cAMP) and TPL2 (tumor progression locus 2, MAP3K8) and found that inhibitors for EPAC and TPL2 reduced AR agonist-induced IL-1ß reporter activity. These inhibitors also suppressed NA-induced endogenous Il1b mRNA expression and IL-1ß protein production. Our results suggest that EPAC and TPL2 are involved in ß-AR-mediated IL-1ß production in microglial cells, and extend our understanding of its intracellular signaling mechanism.


Assuntos
Acetilcisteína/análogos & derivados , Eritromicina/análogos & derivados , Interleucina-1beta/metabolismo , MAP Quinase Quinase Quinases/farmacologia , Microglia/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , Acetilcisteína/farmacologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Eritromicina/farmacologia , Expressão Gênica/efeitos dos fármacos , Interleucina-1beta/genética , Isoproterenol/farmacologia , MAP Quinase Quinase Quinases/fisiologia , Camundongos , Norepinefrina/farmacologia , Norepinefrina/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Adrenérgicos beta , Transdução de Sinais , Regulação para Cima/efeitos dos fármacos
3.
Clin Exp Dermatol ; 39(3): 376-84, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24635082

RESUMO

BACKGROUND: Metastatic melanoma is a disease with high mortality and limited therapeutic options. MicroRNAs (miRNAs) can be used to classify melanoma stage. METHODS: Expression of the miRNA miR-125b and serine/threonine kinase mixed lineage kinase (MLK)3 was assessed in primary malignant melanoma tissues and several melanoma cell lines by quantitative reverse transcription PCR. The effect of MLK3 and miR-125b on cell proliferation was evaluated by MTS assay, and cell invasion was evaluated by Transwell invasion assays. Targeting of MLK3 by miR-125b was evaluated using luciferase reporter assay and western blotting. RESULTS: We found significantly increased levels of MLK3 in metastatic primary malignant melanomas and melanoma cell lines, with levels being especially high in metastatic lines. To investigate the functional significance of MLK3, we used knockdown MLK3, which was found to suppress cell growth and invasion. Using bioinformatics, we identified MLK3 as one potential target of miR-125b. miRNA transfection and luciferase assay confirmed that MLK3 was regulated by miR-125b at both the transcriptional and translational levels. Cell proliferation and cell invasion was inhibited by overexpression of miR-125b. CONCLUSIONS: MLK3 is upregulated in metastatic melanoma, and regulates cell proliferation and invasion in melanoma cells. MLK3 is a direct target of miR-125b.


Assuntos
Proliferação de Células/efeitos dos fármacos , MAP Quinase Quinase Quinases/fisiologia , Melanoma/metabolismo , MicroRNAs/metabolismo , Neoplasias Cutâneas/metabolismo , Western Blotting , Linhagem Celular Tumoral/metabolismo , Humanos , MAP Quinase Quinase Quinases/farmacologia , MicroRNAs/farmacologia , Invasividade Neoplásica
4.
Br J Cancer ; 107(1): 129-36, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22644295

RESUMO

BACKGROUND: Tumour cell metastasis involves cell adhesion and invasion, processes that depend on signal transduction, which can be influenced by the tumour microenvironment. N-6 polyunsaturated fatty acids, found both in the diet and in response to inflammatory responses, are important components of this microenvironment. METHODS: We used short hairpin RNA (shRNA) knockdown of TGF-ß-activated kinase-1 (TAK1) in human tumour cells to examine its involvement in fatty acid-stimulated cell adhesion and invasion in vitro. An in vivo model of metastasis was developed in which cells, stably expressing firefly luciferase and either a control shRNA or a TAK1-specific shRNA, were injected into the mammary fat pads of mice fed diets, rich in n-6 polyunsaturated fatty acids. Tumour growth and spontaneous metastasis were monitored with in vivo and in situ imaging of bioluminescence. RESULTS: Arachidonic acid activated TAK1 and downstream kinases in MDA-MB-435 breast cancer cells and led to increased adhesion and invasion. Knockdown of TAK1 blocked this activation and inhibited both cell adhesion and invasion in vitro. Tumour growth at the site of injection was not affected by TAK1 knockdown, but both the incidence and extent of metastasis to the lung were significantly reduced in mice injected with TAK1 knockdown cells compared with mice carrying control tumour cells. CONCLUSION: These data demonstrate the importance of TAK1 signalling in tumour metastasis in vivo and suggest an opportunity for antimetastatic therapies.


Assuntos
Adesão Celular/efeitos dos fármacos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Invasividade Neoplásica/prevenção & controle , Metástase Neoplásica/prevenção & controle , Animais , Ácido Araquidônico , Neoplasias da Mama , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , MAP Quinase Quinase Quinases/biossíntese , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/farmacologia , Camundongos , Transplante de Neoplasias , RNA Interferente Pequeno/farmacologia
5.
Vaccine ; 27(41): 5589-98, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19646407

RESUMO

Improving vaccine immunogenicity remains a major challenge in the fight against developing country diseases like malaria and AIDS. We describe a novel strategy to identify new DNA vaccine adjuvants. We have screened components of the Toll-like receptor signalling pathways for their ability to activate pro-inflammatory target genes in transient transfection assays and assessed in vivo adjuvant activity by expressing the activators from the DNA backbone of vaccines. We find that a robust increase in the immune response necessitates co-expression of two activators. Accordingly, the combination of tak1 and tram elicits synergistic reporter activation in transient transfection assays. In a mouse model this combination, but not the individual molecules, induced approximately twofold increases in CD8+ T-cell immune responses. These results indicate that optimal immunogenicity may require activation of distinct innate immune signalling pathways. Thus this strategy offers a novel route to the discovery of a new generation of adjuvants.


Assuntos
Adjuvantes Imunológicos/farmacologia , MAP Quinase Quinase Quinases/farmacologia , Receptores de Interleucina/metabolismo , Vacinas de DNA/imunologia , Adjuvantes Imunológicos/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Feminino , Perfilação da Expressão Gênica , Humanos , Interferon gama/metabolismo , MAP Quinase Quinase Quinases/genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Interleucina/genética
6.
ChemMedChem ; 3(2): 336-44, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18081134

RESUMO

Receptor rearrangement upon ligand binding (induced fit) is a major stumbling block in docking and virtual screening. Even though numerous studies have stressed the importance of including protein flexibility in ligand docking, currently available methods provide only a partial solution to the problem. Most of these methods, being computer intensive, are often impractical to use in actual drug discovery settings. We had earlier shown that ligand-induced receptor side-chain conformational changes could be modeled statistically using data on known receptor-ligand complexes. In this paper, we show that a similar approach can be used to model more complex changes like backbone flips and loop movements. We have used p38 MAPK as a test case and have shown that a few simple structural features of ligands are sufficient to predict the induced variation in receptor conformations. Rigorous validation, both by internal resampling methods and on an external test set, corroborates this finding and demonstrates the robustness of the models. We have also compared our results with those from an earlier molecular dynamics simulation study on DFG loop conformations of p38 MAPK, and found that the results matched in the two cases. Our statistical approach enables one to predict the final ligand-induced conformation of the active site of a protein, based on a few ligand properties, prior to docking the ligand. We can do this without having to trace the step-by-step process by which this state is arrived at (as in molecular dynamics simulations), thereby drastically reducing computational effort.


Assuntos
Algoritmos , Simulação por Computador , Desenho de Fármacos , MAP Quinase Quinase Quinases/farmacologia , Proteínas Proto-Oncogênicas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Cristalografia por Raios X , Ligantes , MAP Quinase Quinase Quinases/química , Modelos Químicos , Ligação Proteica , Conformação Proteica , Proteínas Proto-Oncogênicas/química , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Relação Estrutura-Atividade , Proteínas Quinases p38 Ativadas por Mitógeno/química
7.
Am J Physiol Renal Physiol ; 292(5): F1471-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17299140

RESUMO

We have previously demonstrated that transforming growth factor-beta(1) (TGF-beta(1)) rapidly activates the mitogen-activated protein kinase kinase 3 (MKK3)-p38 MAPK signaling cascade, leading to the induction of type I collagen synthesis in mouse glomerular mesangial cells (Wang L, Ma R, Flavell RA, Choi ME. J Biol Chem 277: 47257-47262, 2002). In the present study, we investigated the functional role of upstream TGF-beta-activated kinase 1 (TAK1) and TAK1-binding protein 1 (TAB1) in the TGF-beta(1) signaling cascade. Rapid activation of endogenous TAK1 activity by TGF-beta(1) was observed in mouse mesangial cells. Transient overexpression of TAK1 with TAB1 enhanced the activation of MKK3 and p38 MAPK with or without TGF-beta(1) stimulation, whereas a dominant-negative mutant of TAK1 (TAK1DN) suppressed TGF-beta(1)-induced activation of MKK3 and p38 MAPK. Moreover, constitutive expression of TAK1DN reduced steady-state protein levels of MKK3 and p38 MAPK as well as MKK3 phosphorylation. Increased p38alpha MAPK activity by ectopic expression of either TAB1 or wild-type p38alpha MAPK resulted in enhanced TGF-beta(1)-induced type I collagen expression. In contrast, constitutive expression of TAK1DN inhibited collagen induction. Taken together, our data indicate that TAK1 and TAB1 play a pivotal role as upstream signal transducers activating the MKK3-p38 MAPK signaling cascade that leads to the induction of type I collagen expression by TGF-beta(1). In addition, our findings also suggest that TAK1 has a novel function in regulation of the steady-state protein levels of MKK3 and p38 MAPK.


Assuntos
Colágeno Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , MAP Quinase Quinase 3/metabolismo , MAP Quinase Quinase Quinases/fisiologia , Fator de Crescimento Transformador beta1/farmacologia , Animais , Células Cultivadas , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Genes Dominantes , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Isoenzimas/genética , Isoenzimas/metabolismo , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/farmacologia , Camundongos , Mutação , Transdução de Sinais/fisiologia , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Oncogene ; 24(21): 3459-71, 2005 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-15735667

RESUMO

Mutations in BRAF, a component of extracellular signal-regulated kinases 1 and 2 (ERK) cascade, are frequent in melanoma. It is important to understand how BRAF mutations contribute to malignant traits including anchorage- and growth factor-independence. We have previously shown that efficient activation of ERK in normal human epidermal melanocytes (NHEM) requires both adhesion to the extracellular matrix and growth factors. Mutant V599E BRAF is sufficient to promote ERK activation independent of adhesion and growth factors. Here, we analysed regulation of G1 cell cycle events in NHEM and human melanoma cells. We show that S phase entry in NHEM requires both adhesion and growth factor signaling through the MEK-ERK pathway. This control correlates with induction of cyclin D1 and downregulation of p27Kip1, two key G1 cell cycle events. In melanoma cells expressing V599E BRAF, cyclin D1 was constitutively expressed independent of adhesion but dependent upon MEK activation and nuclear accumulation of ERK. Reduction of cyclin D1 levels by RNA interference inhibited S phase entry in melanoma cells. Importantly, expression of V599E BRAF in NHEM was sufficient to promote cyclin D1 promoter activity in the absence of adhesion. Additionally, p27Kip1 levels were downregulated in V599E BRAF-expressing melanoma cells and active BRAF was sufficient to downregulate p27Kip1 in serum-starved NHEM. Thus, adhesion-growth factor cooperation, leading to efficient activation of ERK, regulates cyclin D1 and p27Kip1 levels in human melanocytes and mutant BRAF overrides adhesion-growth factor control of these two G1 cell cycle proteins in melanomas. These findings provide important insight into how BRAF mutations contribute to aberrant human melanocyte proliferation.


Assuntos
Adesão Celular , Proteínas de Ciclo Celular/biossíntese , Ciclina D1/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , MAP Quinase Quinase Quinases/farmacologia , Melanoma/genética , Melanoma/patologia , Proteínas Proto-Oncogênicas B-raf/farmacologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/biossíntese , Ciclo Celular/genética , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/metabolismo , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Análise Mutacional de DNA , Regulação para Baixo , Genes Supressores de Tumor , Humanos , Melanócitos , Regiões Promotoras Genéticas , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo
9.
Int J Oncol ; 26(3): 737-44, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703831

RESUMO

The extracellular signal-regulated kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway plays a critical role in the anticancer action in vitro. ERK1/2 activation or phosphorylation is responsible for increased cyclooxygenase-2 (COX-2) protein expression in some cancer cells treated with selective COX-2 inhibitor NS398. We determined the effect of NS398 on ERK signaling and the synergistic effect of combined treatment with NS398 and a specific MEK inhibitor U0126 on three human endometrial cancer cell lines: Ishikawa, HEC-1A and AN3CA cells. Results showed that NS398 and U0126 individually, and especially the combination of both exhibited profound anti-proliferation of all three cell lines in a time- and concentration-dependent manner by [3-(4, 5)-dimethylthiazol-z-yl]-2, 5-diphenyl tetrazolium bromide (MTT) assay. The phosphorylated ERK1/2 was up-regulated in HEC-1A and AN3CA cells, but the COX-2 protein expression was unchanged in the three cancer cell lines treated with NS398 alone. However, both phosphorylated ERK1/2 and COX-2 protein expression were concentration-dependently decreased in all three cell types by combined treatment with NS398 and U0126 assessed by western blot analysis. Simultaneously, the combination of NS398 and U0126 resulted in 2-fold increase in apoptosis of all three lines over that by the individual alone, and enhanced G0/G1 phase arrest of Ishikawa and HEC-1A cells induced by U0126 treatment determined by flow cytometry. The synergistic and complementary effects of combining NS398 and U0126 were found to be associated with activation of caspase-3, alterations of Bcl-2 family proteins and cell cycle regulatory proteins detected by western blot analysis. Taken together, these findings correlate with blocking MEK-ERK signaling cascade and down-regulating COX-2 protein expression in endometrial cancer cells with combination treatment of NS398 and U0126, suggesting that the combinatory use of NS398 and specific MEK inhibitors may be valuable for chemotherapy or chemoprevention of human endometrial cancer.


Assuntos
Butadienos/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Neoplasias do Endométrio/patologia , Inibidores Enzimáticos/farmacologia , Nitrilas/farmacologia , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Western Blotting , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , Feminino , Humanos , MAP Quinase Quinase Quinases/farmacologia , Transdução de Sinais , Células Tumorais Cultivadas
10.
Int J Oncol ; 26(3): 751-6, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15703833

RESUMO

I-2PP2A/SET, the translocation breakpoint-encoded protein expressed in acute undifferentiated leukemia, was identified as an inhibitor of protein phosphatase 2A (PP2A). Induction of exogenous I-2PP2A/SET at a ratio of 1:1 to the endogenous protein resulted in suppression of cell proliferation. In contrast, siRNA-mediated depletion of I-2PP2A/SET resulted in enhanced cell proliferation. Depletion of I-2PP2A/SET was accompanied with a decrease in the number of cells in G1 and an increase in cells in S phase. To examine the mode of action by which I-2PP2A/SET suppresses cell proliferation, we determined the effect of over-expressed I-2PP2A/SET on ERK activation. I-2PP2A/SET suppressed activation of ERK following EGF stimulation but did not affect activation levels of stress kinases, JNK and p38. By contrast, knocking down I-2PP2A/SET by siRNA resulted in enhancement of ERK and MEK activations, suggesting that I-2PP2A/SET negatively regulates MEK/ERK. These data suggest that I-2PP2A/SET negatively regulates cell growth by inhibiting the G1/S transition and inhibiting the MEK/ERK pathway stimulated by external stimuli. These data demonstrate that I-2PP2A/SET potentially functions as a tumor suppressor.


Assuntos
Proliferação de Células , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/farmacologia , MAP Quinase Quinase Quinases/farmacologia , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/farmacologia , Ciclo Celular , Proteínas de Ligação a DNA , Perfilação da Expressão Gênica , Células HeLa , Chaperonas de Histonas , Humanos , Proteína Fosfatase 2
11.
Int J Oncol ; 25(6): 1671-6, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15547704

RESUMO

Testicular germ cell tumours (TGCT) are the most common solid tumour among young males. Whereas in 1970s, only 5% of patients with a metastatic testicular tumours survived their disease, these days 80% of patients treated by modern cisdiamminedichloroplatinum (cisplatin, CDDP)-based chemotherapy are cured. Although data are accumulating on the effect of the mitogen-activated protein kinase (MAPK) family on the CDDP-induced apoptosis in tumour cells, the mechanisms by which CDDP initiates apoptosis in TGCT are not completely understood. Applying Western blot and phosphorylated kinase-specific ELISA analyses, flow cytometry, blocking experiments, and morphological methods we sought here to define the MAPK pathway(s) involved in the CDDP-induced apoptosis in the human TGCT cell line NCCIT. Our experiments showed that within hours of CDDP application only the extracellular signal-regulated kinase (ERK) was dually phosphorylated and caspase-3 became active. Functional assays using chemical inhibitors demonstrated that the phosphorylation of ERK was mediated by reactive oxygen species in an Raf-1-independent manner and required the activation of caspase-3. Thus, our data suggest that CDDP mediates its apoptosis-inducing effect on the human malignant testicular germ cells, at least partially, through activation of the MEK-ERK signaling pathway in a ROS-dependent, Raf-1-independent manner.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cisplatino/farmacologia , Neoplasias Embrionárias de Células Germinativas/patologia , Espécies Reativas de Oxigênio/efeitos adversos , Neoplasias Testiculares/patologia , Western Blotting , Ensaio de Imunoadsorção Enzimática , Humanos , MAP Quinase Quinase Quinases/farmacologia , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Fosforilação , Células Tumorais Cultivadas
12.
Prostate ; 59(2): 214-25, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15042621

RESUMO

BACKGROUND: The androgen receptor (AR) is a ligand-activated transcription factor that mediates the biological responses of androgens in the prostate gland. This study focuses on the chemopreventive agents, resveratrol and genistein, on AR-mediated transcription in prostate cancer cells. RESULTS: We found that resveratrol and genistein activated AR-driven gene expression at low concentrations, whereas they repressed the AR-dependent reporter gene activity at high concentrations. We determined that resveratrol and genistein induced AR-driven gene expression by activating the Raf-MEK-ERK kinase pathway. The ERK1 kinase phosphorylated the AR on multiple sites in vitro, but this phosphorylation event did not contribute to the resveratrol-induced AR transactivation. CONCLUSIONS: In vitro and in vivo studies have indicated that resveratrol and genistein are promising chemopreventive agents. Given the clear evidence that AR pathways are involved in the development and progression of prostate cancer, these data showed that the ability to modulate AR function would contribute the observed chemopreventive activity of resveratrol and genistein.


Assuntos
Anticarcinógenos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genisteína/farmacologia , MAP Quinase Quinase Quinase 1 , Neoplasias da Próstata/genética , Neoplasias da Próstata/prevenção & controle , Receptores Androgênicos/efeitos dos fármacos , Receptores Androgênicos/fisiologia , Estilbenos/farmacologia , Sequência de Aminoácidos , Relação Dose-Resposta a Droga , Humanos , MAP Quinase Quinase Quinases/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Dados de Sequência Molecular , Fosforilação , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-raf/farmacologia , Resveratrol , Transdução de Sinais , Transfecção
13.
Br J Cancer ; 90(2): 283-8, 2004 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-14735164

RESUMO

The Raf-MEK-ERK signalling pathway controls fundamental cellular processes including proliferation, differentiation and survival. It remains enigmatic how this pathway can reliably convert a myriad of extracellular stimuli in specific biological responses. Recent results have shown that the Raf family isoforms A-Raf, B-Raf and Raf-1 have different physiological functions. Here we review how Raf isozyme diversity contributes to the specification of functional diversity, in particular regarding the role of Raf isozymes in cancer.


Assuntos
MAP Quinase Quinase Quinase 1 , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/farmacologia , Neoplasias/fisiopatologia , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/farmacologia , Transdução de Sinais , Diferenciação Celular , Sobrevivência Celular , Humanos , Isoenzimas
14.
J Neurochem ; 87(1): 238-47, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12969270

RESUMO

Inflammatory cell signaling leading to transcriptional activation is primarily mediated by signal transduction via mitogen-activated protein kinase (MAPK) and NFkappaB pathways. A common upstream kinase that signals the activation of these pathways is TGFbeta-activated kinase 1 (TAK1), which itself becomes activated in response to cytokines and upon engagement of a class of cell surface receptors involved in innate immunity, that is Toll-like receptors (TLRs) by bacterial and viral pathogens. This study directly tests the role of TAK1 in the induction of inducible nitric oxide (NO) synthase (iNOS) in glial cells, which represent immune-regulatory cells of the CNS, by transient transfection assays. Transfection of C-6 glia, primary astrocytes and a rat microglial cell line with TAK1 (but not its inactive form) along with its activator protein, TAK1-binding protein 1 (TAB1) resulted in a marked stimulation of a co-transfected rat iNOS promoter-reporter construct (iNOS-Luc). TAK1-induced iNOS-Luc activity was substantially inhibited by pharmacological inhibitors of the known downstream kinases, p38 MAPK and JNK (SB203580 and SP620125), and was almost completely blocked by co-expression of a phosphorylation mutant of IkappaB. TAK1/TAB1 also induced the production of NO and the expression of iNOS in microglial cells in a p38 MAPK-, JNK- and NFkappaB-dependent manner. The results of these studies provide evidence for an important role for TAK1-mediated intracellular signaling, via p38 MAPK, JNK and NFkappaB, in the transcriptional activation of iNOS in glial cells.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , MAP Quinase Quinase Quinases/metabolismo , Neuroglia/enzimologia , Óxido Nítrico Sintase/genética , Animais , Astrócitos/citologia , Astrócitos/enzimologia , Proteínas de Transporte/metabolismo , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Microglia/citologia , Microglia/enzimologia , NF-kappa B/metabolismo , Neuroglia/citologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Regiões Promotoras Genéticas , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ativação Transcricional/efeitos dos fármacos , Transfecção
15.
J Biol Chem ; 278(45): 44377-84, 2003 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-12947087

RESUMO

The cytokine transforming growth factor beta (TGF-beta) plays an important role in preventing tumor formation by blocking cell cycle progression. Accordingly, many cancers demonstrate mutations in TGF-beta signaling components or enhanced expression of inhibitors of the TGF-beta pathway such as Smad7. In this report we show that the oncoprotein HER2/Neu is able to collaborate with the ETS transcription factor ER81 to activate Smad7 transcription in breast, endometrial, and ovarian cancer cell lines. ER81 binds to two ETS sites within the Smad7 promoter, and mutation of one of these ETS sites greatly decreases Smad7 induction by HER2/Neu and ER81. Furthermore, we show that Smad7 activation involves the processing of signals from HER2/Neu to ER81 via the ERK mitogen-activated protein kinase pathway. Thus, we have uncovered a novel mechanism by which oncogenic HER2/Neu, in collaboration with ER81, can induce carcinogenesis through Smad7 up-regulation. Moreover, we show that TAK1, a TGF-beta-activated protein kinase, stimulates ER81 via the p38 mitogen-activated protein kinase pathway and thereby induces the Smad7 promoter. This suggests that attenuation of TGF-beta signaling by activating Smad7 transcription may proceed not only through TGF-beta receptor-regulated Smad proteins but also through an independent pathway involving ER81 and TAK1.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , MAP Quinase Quinase Quinases/fisiologia , Receptor ErbB-2/fisiologia , Transativadores/genética , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Sítios de Ligação , Neoplasias da Mama , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/farmacologia , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , MAP Quinase Quinase Quinases/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Ovarianas , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas/genética , Receptor ErbB-2/genética , Transdução de Sinais , Proteína Smad7 , Fator de Transcrição AP-1/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/farmacologia , Transfecção , Células Tumorais Cultivadas
16.
J Virol ; 76(9): 4567-79, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11932422

RESUMO

The Epstein-Barr virus-encoded latent infection membrane protein 1 (LMP1) is a pleiotropic protein, the activities of which include effects on cell transformation and phenotype, growth, and survival. The ability of LMP1 to mediate at least some of these phenomena could be attributed to the activation of the transcription factor NF-kappaB. LMP1 promotes NF-kappaB activation through the recruitment of the adapter protein TRAF2 and the formation of a dynamic multiprotein complex that includes the NF-kappaB kinase, the IkappaB kinases, and their downstream targets, IkappaBs and p105. In this study, we have identified the oncogenic kinase Tpl-2/Cot as a novel component of LMP1-induced NF-kappaB signaling. We show that Tpl-2 is expressed in primary biopsies from patients with nasopharyngeal carcinoma and Hodgkin's disease, where LMP1 is also found. Inducible expression of LMP1 promotes the activation of Tpl-2, and a catalytically inactive Tpl-2 mutant suppresses LMP1-induced NF-kappaB signaling. In colocalization and coimmunoprecipitation experiments, Tpl-2 and TRAF2 were found to interact with Tpl-2 functioning downstream of TRAF2. Consistent with this observation, catalytically inactive Tpl-2 also blocked CD40-mediated NF-kappaB activation, which largely depends on TRAF2. The ability of Tpl-2 to influence LMP1-induced NF-kappaB occurs through modulation of both IkappaBalpha and p105 functions. Furthermore, Tpl-2 was found to influence the expression of angiogenic mediators, such as COX-2 in LMP1-transfected cells. These data identify Tpl-2 as a component of LMP1 signaling downstream of TRAF2 and as a modulator of LMP1-mediated effects.


Assuntos
Proteínas de Transporte/metabolismo , MAP Quinase Quinase Quinases/metabolismo , NF-kappa B/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Ribossômicas , Transdução de Sinais , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Carcinoma/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Ciclo-Oxigenase 2 , Proteínas do Citoesqueleto , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Doença de Hodgkin/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Isoenzimas/metabolismo , Proteínas com Domínio LIM , MAP Quinase Quinase Quinases/farmacologia , Proteínas de Membrana , Camundongos , Neoplasias Nasofaríngeas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator 2 Associado a Receptor de TNF , Transfecção
17.
Hepatology ; 34(5): 953-63, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11679966

RESUMO

After liver injury, hepatic stellate cells (HSCs) undergo a process of activation with expression of smooth muscle alpha-actin (alpha-SMA), an increased proliferation rate, and a dramatic increase in synthesis of type I collagen. The intracellular signaling mechanisms of activation and perpetuation of the activated phenotype in HSCs are largely unknown. In this study the role of the stress-activated protein kinases, c-Jun N-terminal kinase (JNK) and p38, were evaluated in primary cultures of rat HSCs. The effect of JNK was assessed by using an adenovirus expressing a dominant negative form of transforming growth factor beta (TGF-beta)-activated kinase 1 (TAK1) (Ad5dnTAK1) and a new selective pharmacologic inhibitor SP600125. The effect of p38 was assessed with the selective pharmacologic inhibitor SB203580. These kinases were inhibited starting either in quiescent HSCs (culture day 1) or in activated HSCs (culture day 5). Although blocking TAK1/JNK and p38 decreased the expression of alpha-SMA protein in early stages of HSC activation, no effect was observed when TAK1/JNK or p38 were inhibited in activated HSCs. JNK inhibition increased and p38 inhibition decreased collagen alpha1(I) mRNA level as measured by RNase protection assays, with maximal effects observed in early stages of HSC activation. Furthermore, TAK1/JNK inhibition decreased HSC proliferation, whereas p38 inhibition led to an increased proliferation rate of HSCs, independently of its activation status. These results show novel roles for the TAK1/JNK pathway and p38 during HSC activation in culture. Despite similar activators of TAK1/JNK and p38, their functions in HSCs are distinct and opposed.


Assuntos
Fígado/efeitos dos fármacos , MAP Quinase Quinase Quinases/farmacologia , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Actinas/antagonistas & inibidores , Animais , Antracenos/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/genética , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases JNK Ativadas por Mitógeno , Fígado/citologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Liso/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Quinases p38 Ativadas por Mitógeno
18.
Am J Physiol Gastrointest Liver Physiol ; 281(5): G1279-89, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11668037

RESUMO

Transforming growth factor-beta (TGF-beta)-activated kinase 1 (TAK1), a serine/threonine kinase, is reported to function in the signaling pathways of TGF-beta, interleukin 1, and ceramide. However, the physiological role of TAK1 in vivo is largely unknown. To assess the function of TAK1 in vivo, dominant-negative TAK1 (dnTAK1) was expressed in the rat liver by adenoviral gene transfer. dnTAK1 expression abrogated c-Jun NH(2)-terminal kinase and c-Jun but not nuclear factor (NF)-kappaB or SMAD activation after partial hepatectomy (PH). Expression of dnTAK1 or TAM-67, a dominant-negative c-Jun, induced G(0) exit in quiescent liver and accelerated cell cycle progression after PH. Finally, dnTAK1 and TAM-67 induced c-myc expression in the liver before and after PH, suggesting that G(0) exit induced by dnTAK1 and TAM-67 is mediated by c-myc induction.


Assuntos
Regulação da Expressão Gênica , Genes Dominantes , Genes myc/fisiologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/fisiologia , Fase de Repouso do Ciclo Celular/fisiologia , Animais , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Proteínas de Ligação a DNA/fisiologia , Ativação Enzimática/efeitos dos fármacos , Genes jun/efeitos dos fármacos , Genes jun/fisiologia , Genes myc/efeitos dos fármacos , Hepatectomia/métodos , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase Quinases/farmacologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Ratos , Ratos Sprague-Dawley , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Proteína Smad3 , Proteína Smad4 , Transativadores/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno
19.
Ann Hematol ; 80(5): 284-91, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11446731

RESUMO

Thrombopoietin (TPO) plays a critical role not only in proliferation and differentiation of megakaryocytes but also in erythroid differentiation. We have investigated whether the different pathway of mitogen-activated protein kinase (MAPK) after TPO stimulation may discriminate megakaryocyte and erythroid differentiation. In this study, we have used human CD34+ hematopoietic progenitor cells (HPCs) from cord blood (CB) in serum-free liquid culture supplemented with TPO, to compare the respective effects of specific inhibitors of MAPK kinase (MEK) (PD98059) and p38 MAP kinase (p38) (SB203580) on megakaryocyte and erythroid development. PD98059, but not SB203580, significantly suppressed TPO-induced megakaryocyte differentiation when examined by the expression of CD41 and polyploidy assay. In the presence of SB203580, CD34+/CD36+ erythroid progenitors clearly decreased, whereas they increased when cultured with PD98059. These results indicate that activation of extracellular-signal-regulated kinase (ERK) is required for TPO-induced megakaryocyte differentiation and that p38 is required for TPO-induced erythroid differentiation.


Assuntos
Células Precursoras Eritroides/citologia , Megacariócitos/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Trombopoetina/farmacologia , Antígenos CD34/sangue , Diferenciação Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células Precursoras Eritroides/imunologia , Humanos , Recém-Nascido , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/farmacologia , Megacariócitos/imunologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/biossíntese , Complexo Glicoproteico GPIb-IX de Plaquetas/biossíntese , Ploidias , Proteínas Quinases p38 Ativadas por Mitógeno
20.
J Neurosci ; 21(14): 4949-57, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11438570

RESUMO

Superior cervical ganglion (SCG) sympathetic neurons die by apoptosis when deprived of nerve growth factor (NGF). It has been shown previously that the induction of apoptosis in these neurons at NGF withdrawal requires both the activity of the small GTP-binding protein Cdc42 and the activation of the c-Jun N-terminal kinase (JNK) pathway. The mixed lineage kinase 3 (MLK3) belongs to a family of mitogen-activated protein (MAP) kinase kinase kinases. MLK3 contains a Cdc42/Rac interactive-binding (CRIB) domain and activates both the JNK and the p38 MAP kinase pathways. In this study the role of MLK3 in the induction of apoptosis in sympathetic neurons has been investigated. Overexpression of an active MLK3 induces activation of the JNK pathway and apoptosis in SCG neurons. In addition, overexpression of kinase dead mutants of MLK3 blocks apoptosis as well as c-Jun phosphorylation induced by NGF deprivation. More importantly, MLK3 activity seems to increase by 5 hr after NGF withdrawal in both differentiated PC12 cells and SCG neurons. We also show that MLK3 lies downstream of Cdc42 in the neuronal death pathway. Regulation of MLK3 in neurons seems to be dependent on MLK3 activity and possibly on an additional cellular component, but not on its binding to Cdc42. These results suggest that MLK3, or a closely related kinase, is a physiological element of NGF withdrawal-induced activation of the Cdc42-c-Jun pathway and neuronal death. MLK3 therefore could be an interesting therapeutic target in a number of neurodegenerative diseases involving neuronal apoptosis.


Assuntos
Apoptose/fisiologia , MAP Quinase Quinase Quinases/metabolismo , Neurônios/enzimologia , Motivos de Aminoácidos/fisiologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Expressão Gênica , Genes Dominantes , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/farmacologia , Microinjeções , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênese Sítio-Dirigida , Fator de Crescimento Neural/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Gânglio Cervical Superior , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína cdc42 de Ligação ao GTP/farmacologia , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA