Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Rep ; 38(2): 110223, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021072

RESUMO

MEK1 and MEK2, the only known activators of ERK, are attractive therapeutic candidates for both cancer and autoimmune diseases. However, how MEK signaling finely regulates immune cell activation is only partially understood. To address this question, we specifically delete Mek1 in hematopoietic cells in the Mek2 null background. Characterization of an allelic series of Mek mutants reveals the presence of distinct degrees of spontaneous B cell activation, which are inversely proportional to the levels of MEK proteins and ERK activation. While Mek1 and Mek2 null mutants have a normal lifespan, 1Mek1 and 1Mek2 mutants retaining only one functional Mek1 or Mek2 allele in hematopoietic cell lineages die from glomerulonephritis and lymphoproliferative disorders, respectively. This establishes that the fine-tuning of the ERK/MAPK pathway is critical to regulate B and T cell activation and function and that each MEK isoform plays distinct roles during lymphocyte activation and disease development.


Assuntos
Ativação Linfocitária/fisiologia , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , Alelos , Animais , Linfócitos B/metabolismo , Feminino , Humanos , Ativação Linfocitária/genética , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Transdução de Sinais/fisiologia , Linfócitos T/metabolismo
2.
Respir Res ; 16: 93, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26242183

RESUMO

BACKGROUND: Polo-like kinase 1 (Plk1) is a serine/threonine protein kinase that has been implicated in the regulation of mitosis. In addition, the activation of mitogen-activated protein kinase (MAPK) is a key event in the early stage of the growth factor response. The role of Plk1 in MAPK phosphorylation in cells has not been investigated. METHODS: Immunoblot analysis was used to evaluate Plk1 and MAPK phosphorylation in cells upon stimulation with platelet-derived growth factor (PDGF). We also generated stable Plk1 knockdown (KD) cells to assess the role of Plk1 in MAPK activation and cell proliferation. Furthermore, we used a non-phosphorylatable Plk1 mutant to determine the function of Plk1 phosphorylation in these processes. RESULTS: Treatment with PDGF increased Plk1 phosphorylation at Thr-210 (an indication of Plk1 activation) in human airway smooth muscle cells. Plk1 KD attenuated the PDGF-induced phosphorylation of MEK1/2 and ERK1/2 as well as cell proliferation. However, phosphorylation of Raf-1 and AKT upon stimulation with PDGF was not reduced in Plk1 KD cells. Furthermore, the expression of T210A Plk1 (alanine substitution at Thr-210) inhibited the PDGF-stimulated MEK1/2 phosphorylation, ERK1/2 phosphorylation and cell proliferation. CONCLUSIONS: Together, these findings suggest that Plk1 is activated upon growth factor stimulation, which may control the activation of MEK1/2 and ERK1/2, and smooth muscle cell proliferation.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Miócitos de Músculo Liso/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Brônquios/citologia , Brônquios/fisiologia , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Traqueia/citologia , Traqueia/fisiologia , Quinase 1 Polo-Like
3.
Arch Pharm Res ; 38(12): 2153-62, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26119076

RESUMO

It has been reported that alkaloids derived from Coptis chinensis exert anti-adipogenic activity on 3T3-L1 adipocytes by downregulating peroxisome proliferation-activity receptor-γ (PPAR-γ) and CCAAT/enhancer binding protein-α (C/EBP-α). However, the signaling-based mechanism of the inhibitory role of epiberberine in the early stages of 3T3-L1 adipocyte differentiation is uncharacterized. Here, we show that epiberberine had inhibitory effects on adipocyte differentiation and significantly decreased lipid accumulation by downregulating an adipocyte-specific transcription factor, sterol regulatory element-binding protein-1 (SREBP-1). Furthermore, we observed that epiberberine markedly suppressed the differentiation-mediated phosphorylation of components of both the Raf/mitogen-activated protein kinase 1 (MEK1)/extracellular signal-regulated protein kinase 1/2 (ERK1/2) and AMP-activated protein kinase-α1 (AMPKα)/Akt pathways. In addition, gene expression of fatty acid synthase (FAS) was significantly inhibited by treatment with epiberberine during adipogenesis. These results indicate that the anti-adipogenic mechanism of epiberberine is associated with inhibition of phosphorylation of Raf/MEK1/ERK1/2 and AMPKα/Akt, followed by downregulation of the major transcription factors of adipogenesis, such as PPAR-γ, C/EBP-α, and SREBP-1, and FAS. Taken together, this study suggests that the anti-adipogenic effect of epiberberine is mediated by downregulation of the Raf/MEK1/ERK1/2 and AMPKα/Akt pathways during 3T3-L1 adipocyte differentiation. Moreover, the anti-adipogenic effects of epiberberine were not accompanied by modulation of ß-catenin.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Adipogenia/fisiologia , Berberina/análogos & derivados , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteína Oncogênica v-akt/fisiologia , Quinases raf/fisiologia , Células 3T3-L1 , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Adipogenia/efeitos dos fármacos , Animais , Fármacos Antiobesidade/farmacologia , Berberina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/farmacologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteína Oncogênica v-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quinases raf/antagonistas & inibidores
4.
Gynecol Oncol ; 133(1): 128-37, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24434059

RESUMO

Mitogen-activated protein kinases (MAPKs) are a family of ubiquitous eukaryotic signal transduction enzymes which link extracellular stimuli to intracellular gene expression pathways. While several three-tiered MAPK cascades have been elucidated in mammals, the prototypical pathway involves a network of proteins and kinases including the Rat sarcoma protein (Ras), mitogen-activated protein kinase kinase kinase (Raf or MAP3K), mitogen-activated protein kinase kinase (MEK or MAP2K), and extracellular signal regulated protein kinase (ERK or MAPK). This MAPK cascade (the Ras/Raf/MEK/ERK pathway) is a receptor tyrosine kinase mediated signaling pathway that regulates cell proliferation, cell cycle progression, and cell migration. There are multiple molecular mechanisms of interaction and activation between the upstream nodes of the Ras/Raf/MEK/ERK cascade and other cell signaling pathways, all ultimately leading to the activation of the nuclear transcription factor ERK. Important downstream targets include MEK1/2, which comprise the final step leading to ERK transcription factor activation. While multiple conduits exist to activate ERK upstream of MEK, there is little redundancy downstream. Located at this pivotal intersection between a limited number of upstream activators and its exclusive downstream targets, MEK is an appealing molecular target of novel cancer therapies. MEK inhibitors are small molecules that inhibit MEK phosphorylation by binding to a pocket adjacent to the ATP binding site, decreasing both the amount of MEK activity, and the quantity of activated ERK in the cell. Unique allosteric noncompetitive binding sites of MEK inhibitors allow specific targeting of MEK enzymes and prevent cross-activation of other serine/threonine protein kinases through the conserved ATP binding site. This paper reviews the translational evidence in favor of MEK inhibitors in cancer, their role in gynecologic malignancies, and details regarding the status of the fourteen MEK inhibitors currently being clinically tested: trametinib, selumetinib, pimasertib, refametinib, PD-0325901, MEK162, TAK733, RO5126766, WX-554, RO4987655, cobimetinib, AZD8330, MSC2015103B, and ARRY-300.


Assuntos
Carcinoma/tratamento farmacológico , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Neoplasias Ovarianas/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Carcinoma/enzimologia , Neoplasias Colorretais/tratamento farmacológico , Feminino , Humanos , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Ovarianas/enzimologia
5.
Gastroenterology ; 146(3): 822-834.e7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24315826

RESUMO

BACKGROUND & AIMS: Kras signaling via mitogen-activated protein kinase (MAPK) is highly up-regulated in pancreatic cancer cells. We investigated whether MAPK signaling is required for the initiation and maintenance of pancreatic carcinogenesis in mice. METHODS: We studied the formation and maintenance of pancreatic intraepithelial neoplasia (PanINs) in p48Cre; TetO-KrasG12D; Rosa26(rtTa-IRES-EGFP) (iKras*) mice and LSL-KrasG12D mice bred with p48Cre mice (KC). Mice were given oral PD325901, a small-molecule inhibitor of MEK1 and MEK2 (factors in the MAPK signaling pathway), along with injections of cerulein to induce pancreatitis. Other mice were given PD325901 only after PanINs developed. Pancreatic tissues were collected and evaluated using histologic, immunohistochemical, immunofluorescence, and electron microscopy analyses. Acinar cells were isolated from the tissues and the effects of MEK1 and 2 inhibitors were assessed. RESULTS: PD325901 prevented PanIN formation, but not pancreatitis, in iKras* and KC mice. In iKras* or KC mice given PD325901 at 5 weeks after PanINs developed, PanINs regressed and acinar tissue regenerated. The regression occurred through differentiation of the PanIN cells to acini, accompanied by re-expression of the acinar transcription factor Mist1. CONCLUSIONS: In iKras* and KC mice, MAPK signaling is required for the initiation and maintenance of pancreatic cancer precursor lesions. MAPK signaling promotes formation of PanINs by enabling dedifferentiation of acinar cells into duct-like cells that are susceptible to transformation.


Assuntos
Células Acinares/patologia , Carcinoma in Situ/fisiopatologia , Desdiferenciação Celular/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Neoplasias Pancreáticas/fisiopatologia , Transdução de Sinais/fisiologia , Células Acinares/fisiologia , Animais , Carcinoma in Situ/patologia , Modelos Animais de Doenças , Feminino , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Mutantes , Neoplasias Pancreáticas/patologia , Pancreatite/fisiopatologia , Regulação para Cima/fisiologia
6.
Neuropharmacology ; 67: 78-87, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23154302

RESUMO

We investigated whether combinatorial post-injury treatment with progesterone (P4) and vitamin D hormone (VDH) would reduce ischemic injury more effectively than P4 alone in an oxygen glucose deprivation (OGD) model in primary cortical neurons and in a transient middle cerebral artery occlusion (tMCAO) model in rats. In the OGD model, P4 and VDH each showed neuroprotection individually, but combination of the "best" doses did not show substantial efficacy; instead, the lower dose of VDH in combination with P4 was the most effective. In the tMCAO model, P4 and VDH were given alone or in combination at different times post-occlusion for 7 days. In vivo data confirmed the in vitro findings and showed better infarct reduction at day 7 and functional outcomes (at 3, 5 and 7 days post-occlusion) after combinatorial treatment than when either agent was given alone. VDH, but not P4, upregulated heme oxygenase-1, suggesting a pathway for the neuroprotective effects of VDH differing from that of P4. The combination of P4 and VDH activated brain-derived neurotrophic factor and its specific receptor, tyrosine kinase receptor-B. Under specific conditions VDH potentiates P4's neuroprotective efficacy and should be considered as a potential partner of P4 in a low-cost, safe and effective combinatorial treatment for stroke.


Assuntos
Isquemia Encefálica/prevenção & controle , Fator Neurotrófico Derivado do Encéfalo/fisiologia , MAP Quinase Quinase 2/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Progesterona/administração & dosagem , Receptor trkB/fisiologia , Acidente Vascular Cerebral/prevenção & controle , Vitamina D/administração & dosagem , Animais , Células Cultivadas , Quimioterapia Combinada , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Resultado do Tratamento
7.
APMIS ; 121(4): 299-310, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23030595

RESUMO

Interaction between the immune system and cancer cells allows for the use of biological response modifiers, like OK-432, in cancer therapy. We have studied the involvement of monocytes (MOs) in the immune response to OK-432 by examining MCP-1, MIP-1α and MIP-1ß secretion, in vitro. OK-432-induced IL-6/TNF-α secretion has previously been shown to depend on mitogen-activated protein kinases (MAPKs) ERK1/2 and p38, and we therefore investigated the role of these MAPKs in OK-432-induced chemokine secretion. Here we demonstrate that pharmacological MEK1/2 kinase inhibition generally impaired chemokine secretion from MOs, whereas p38 MAPK inhibition in particular reduced MIP-1α production. Furthermore, simultaneous inhibition of MEK1/2 and Syk kinase was seen to have an additive impact on reduced MCP-1, MIP-1α and MIP-1ß secretion. Based on single cell flow cytometry analyses, OK-432, lipoteichoic acid (LTA) and lipopolysaccharide (LPS) were seen to induce p38 MAPK and NF-κB phosphorylation in MOs with different time kinetics. LTA and LPS have been shown to induce ERK1/2 phosphorylation, whereas the levels of phosphorylated ERK1/2 remained constant following OK-432 treatment at the time points tested. Toll-like receptors (TLRs) recognize pathogen-associated molecular patterns, and we demonstrate increased TLR2 cell surface levels on the MO population, most profoundly following stimulation with LTA and OK-432. Together these results indicate that modulation of MEK1/2 and p38 MAPK signalling could affect the response to OK-432 treatment, having the potential to improve its therapeutic potential within cancer and lymphangioma treatment.


Assuntos
Quimiocinas/metabolismo , Fatores Imunológicos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Picibanil/farmacologia , Butadienos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/fisiologia , Monócitos/imunologia , Monócitos/metabolismo , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinase Syk , Ácidos Teicoicos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Cell Cycle ; 11(17): 3237-49, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22895183

RESUMO

The p53R2 protein, a newly identified member of the ribonucleotide reductase family that provides nucleotides for DNA damage repair, is directly regulated by p53. We show that p53R2 is also regulated by a MEK2 (ERK kinase 2/MAP kinase kinase 2)-dependent pathway. Increased MEK1/2 phosphorylation by serum stimulation coincided with an increase in the RNR activity in U2OS and H1299 cells. The inhibition of MEK2 activity, either by treatment with a MEK inhibitor or by transfection with MEK2 siRNA, dramatically decreased the serum-stimulated RNR activity. Moreover, p53R2 siRNA, but not R2 siRNA, significantly inhibits serum-stimulated RNR activity, indicating that p53R2 is specifically regulated by a MEK2-dependent pathway. Co-immunoprecipitation analyses revealed that the MEK2 segment comprising amino acids 65-171 is critical for p53R2-MEK2 interaction, and the binding domain of MEK2 is required for MEK2-mediated increased RNR activity. Phosphorylation of MEK1/2 was greatly augmented by ionizing radiation, and RNR activity was concurrently increased. Ionizing radiation-induced RNR activity was markedly attenuated by transfection of MEK2 or p53R2 siRNA, but not R2 siRNA. These data show that MEK2 is an endogenous regulator of p53R2 and suggest that MEK2 may associate with p53R2 and upregulate its activity.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Reparo do DNA/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , MAP Quinase Quinase 2/metabolismo , Ribonucleotídeo Redutases/metabolismo , Anticorpos Monoclonais , Western Blotting , Linhagem Celular Tumoral , Raios gama , Vetores Genéticos/genética , Humanos , Imunoprecipitação , MAP Quinase Quinase 2/fisiologia , Fosforilação , Interferência de RNA , Contagem de Cintilação
9.
PLoS One ; 6(2): e17165, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21365009

RESUMO

Mitogen-activated protein kinase kinases (MKK or MEK) 1 and 2 are usually treated as redundant kinases. However, in assessing their relative contribution towards ERK-mediated biologic response investigators have relied on tests of necessity, not sufficiency. In response we developed a novel experimental model using lethal toxin (LeTx), an anthrax toxin-derived pan-MKK protease, and genetically engineered protease resistant MKK mutants (MKKcr) to test the sufficiency of MEK signaling in melanoma SK-MEL-28 cells. Surprisingly, ERK activity persisted in LeTx-treated cells expressing MEK2cr but not MEK1cr. Microarray analysis revealed non-overlapping downstream transcriptional targets of MEK1 and MEK2, and indicated a substantial rescue effect of MEK2cr on proliferation pathways. Furthermore, LeTx efficiently inhibited the cell proliferation and anchorage-independent growth of SK-MEL-28 cells expressing MKK1cr but not MEK2cr. These results indicate in SK-MEL-28 cells MEK1 and MEK2 signaling pathways are not redundant and interchangeable for cell proliferation. We conclude that in the absence of other MKK, MEK2 is sufficient for SK-MEL-28 cell proliferation. MEK1 conditionally compensates for loss of MEK2 only in the presence of other MKK.


Assuntos
Proliferação de Células , MAP Quinase Quinase 2/fisiologia , Melanoma/patologia , Neoplasias Cutâneas/patologia , Animais , Antígenos de Bactérias/metabolismo , Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Células CHO , Domínio Catalítico/efeitos dos fármacos , Domínio Catalítico/genética , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Proliferação de Células/efeitos dos fármacos , Análise por Conglomerados , Cricetinae , Cricetulus , Perfilação da Expressão Gênica , Humanos , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Melanoma/genética , Análise em Microsséries , Invasividade Neoplásica , Mutação Puntual/fisiologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Cutâneas/genética , Células Tumorais Cultivadas
10.
J Leukoc Biol ; 88(6): 1191-200, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20837746

RESUMO

IFN-ß and sIL-1Ra play crucial roles in the regulation of innate immunity and inflammation. IFN-ß, which is widely used to improve the course of relapsing, remitting multiple sclerosis, induces the production of sIL-1Ra in human monocytes through mechanisms that remain largely unknown. In this study, we identified PI3Kδ and MEK2 as key elements that control sIL-1Ra production in isolated human monocytes activated by IFN-ß. Blockade of MEK2, but not of MEK1, by inhibitors and siRNA prevented IFN-ß-induced PI3Kδ recruitment to the membrane, Akt phosphorylation, and sIL-1Ra production, suggesting that MEK2 acted upstream of PI3Kδ. Furthermore, ERK1/2, the only identified substrates of MEK1/2 to date, are dispensable for sIL-1Ra production in response to IFN-ß stimulation. Upon IFN-ß activation, MEK2 and PI3Kδ are translocated to monocyte membranes. These data suggest that MEK1 and MEK2 display different, nonredundant functions in IFN-ß signaling. That neither MEK1 nor ERK1/2 play a part in this mechanism is also an unexpected finding that gives rise to a better understanding of the MAPK signaling network. Together, these findings demonstrate that IFN-ß triggers an atypical MEK2/PI3Kδ signaling cascade to regulate sIL-1Ra expression in monocytes. The premise that MEK1 and MEK2 play a part in the induction of the proinflammatory cytokine, IL-1ß in human monocytes provides a rationale for an alternative, IFN-ß-mediated pathway to induce/enhance sIL-1Ra production and thus, to dampen inflammation.


Assuntos
Interferon beta/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/biossíntese , MAP Quinase Quinase 2/fisiologia , Monócitos/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/fisiologia , Classe I de Fosfatidilinositol 3-Quinases , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Humanos , MAP Quinase Quinase 1/fisiologia , Monócitos/efeitos dos fármacos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
11.
Oncol Rep ; 24(1): 251-5, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20514469

RESUMO

The mitogen-activated protein kinase kinase 1/2 (MEK1/2) signalling pathway plays a central role in tumour progression. Small molecules that inhibit MEK1/2 are therefore considered attractive candidates for anti-cancer drugs. However, the exact contributions of MEK1 and MEK2 to the development of pancreatic cancer remain to be established. To differentiate the functions of MEK1 and MEK2 in a cultured pancreatic cancer cell line, we utilised shRNA-mediated knockdown of their two mRNAs individually. We studied the effects of MEK1 and MEK2 knockdown on cell morphology, proliferation, mitotic arrest, and in vitro invasion capability in PC-1.0 cells. The results showed that inhibition of MEK1 expression was an effective and specific approach to inhibit cell proliferation and induce G0/G1 arrest. On the other hand, MEK2 knockdown specially altered cell morphology and inhibited the invasive ability of pancreatic cancer cells. Therefore, MEK1 and MEK2 mediate different biological responses in cultured pancreatic cancer cells. These proteins could become distinct targets for the inhibition of specific cellular functions in the treatment of pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Neoplasias Pancreáticas/enzimologia , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cricetinae , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Isoenzimas/fisiologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Mesocricetus , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Interferente Pequeno/farmacologia
12.
Aktuelle Urol ; 41(3): 178-83, 2010 May.
Artigo em Alemão | MEDLINE | ID: mdl-20446247

RESUMO

PURPOSE: Obesity is considered to be a risk factor for prostate cancer. Mitogenic actions of leptin, an adipocyte-derived hormone in a variety of cancer cell types have been identified. We have investigated the proliferative effects of leptin on human prostate cancer cells and assessed the role of tyrosine kinase signalling in mediating these actions. MATERIALS AND METHODS: Two human androgen-resistant prostate cancer cell lines and one androgen-sensitive human prostate adenocarcinoma cell line were treated with leptin (5-100 ng / mL) for up to 48 hours. Under serum-free conditions, cell proliferation was measured using an enzyme-linked colorimetric assay. Furthermore, phosphorylation of a downstream component of MAPK (ERK1 / 2) was detected by Western blotting and a specific inhibitor of MAPK (PD98059; 40 microM) was used to evaluate the role of this signalling pathway. RESULTS: Leptin dose-dependently increased the cell number in both androgen-resistant cell lines after 24 h and 48 h of incubation (percent of control: DU145 = 194.6 +/- 5.9 %, PC-3 = 177.9 +/- 6.8 %; 100 ng / mL leptin; 48 h; p < 0.001). Conversely, leptin's proliferative effect on the androgen-sensitive cell line was less pronounced (percent of control: LNCaP = 112.3 +/- 6.1 %; 100 ng / mL leptin; 48 h). Leptin also caused dose-dependent ERK1 / 2 phosphorylation in both androgen-resistant cell lines. In addition, pre-treatment with PD98059 inhibited these responses and attenuated leptin's mitogenic action. CONCLUSIONS: Data from this in vitro study suggest an association between obesity-associated hyperleptinemia and an increased risk for prostate cancer. Further investigations are necessary to clarify whether these data have clinical relevance regarding the use as a prognostic marker for predicting the timing of the occurrence of androgen resistency.


Assuntos
Adenocarcinoma/fisiopatologia , Adipocinas/fisiologia , Neoplasias Hormônio-Dependentes/fisiopatologia , Obesidade/fisiopatologia , Neoplasias da Próstata/fisiopatologia , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Adenocarcinoma/patologia , Western Blotting , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , Humanos , Técnicas In Vitro , Leptina/fisiologia , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Masculino , Neoplasias Hormônio-Dependentes/patologia , Obesidade/patologia , Fosforilação , Neoplasias da Próstata/patologia , Fatores de Risco
13.
Biochemistry ; 49(23): 4766-78, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20469933

RESUMO

Translocator protein TSPO is an 18 kDa protein implicated in numerous cell functions and is highly expressed in secretory and glandular tissues, especially in steroidogenic cells. TSPO expression is altered in pathological conditions such as certain cancers and neurological diseases. In search of the factors regulating Tspo expression, we recently showed that high levels of TSPO in steroidogenic cells may be due to high constitutive expression of protein kinase Cepsilon (PKCepsilon), while phorbol 12-myristate 13-acetate (PMA) activation of PKCepsilon drives inducible TSPO expression in nonsteroidogenic cells, likely through activator protein 1 (AP1). In this study, we aimed to identify the signal transduction pathway through which PKCepsilon regulates Tspo gene expression. The MEK1/2 specific inhibitor U0126, but not NFkappaB inhibitors, reduced basal Tspo promoter activity in TSPO-rich steroidogenic cells (MA-10 Leydig), as well as basal and PMA-induced Tspo promoter levels in TSPO-poor nonsteroidogenic cells (NIH-3T3 fibroblasts). AP1 and signal transducer and activation of transcription 3 (STAT3) have binding sites in the Tspo promoter and are downstream targets of PKCepsilon and MAPK (Raf-1-ERK1/2) pathways. PKCepsilon overexpression induced STAT3 phosphorylation in NIH-3T3 cells, while PKCepsilon knockdown reduced STAT3 and c-Jun phosphorylation in Leydig cells. MEK1/2, ERK2, c-Jun, and STAT3 knockdown reduced Tspo mRNA and protein levels in Leydig cells. Additionally, Raf-1 reduced Tspo mRNA levels in the same cells. MEK1/2, c-Jun, and STAT3 knockdown also reduced basal as well as PMA-induced Tspo mRNA levels in NIH-3T3 cells. Together, these results demonstrate that PKCepsilon regulates Tspo gene expression through a MAPK (Raf-1-MEK1/2-ERK1/2) signal transduction pathway, acting at least in part through c-Jun and STAT3 transcription factors.


Assuntos
Regulação Enzimológica da Expressão Gênica , Sistema de Sinalização das MAP Quinases/genética , Proteína Quinase C-épsilon/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptores de GABA/biossíntese , Receptores de GABA/genética , Fator de Transcrição STAT3/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Marcação de Genes/métodos , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Camundongos , Células NIH 3T3 , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-jun/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Receptores de GABA/deficiência , Fator de Transcrição STAT3/deficiência , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia
14.
Mol Cancer Ther ; 9(3): 606-16, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20179161

RESUMO

The HER-2 oncogene, a member of the erythroblastosis oncogene B (ERBB)-like oncogene family, has been shown to be amplified in many types of cancer, including breast cancer. However, the molecular mechanism of HER-2 overexpression is not completely understood. The phosphorylation of proteins on the serine or threonine residues that immediately precede proline (pSer/Thr-Pro) is specifically catalyzed by the prolyl isomerase Pin1 and is a key signaling mechanism in cell proliferation and transformation. Here, we found that Pin1 interacts with mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK) protein kinase 1, resulting in the induction of HER-2 expression. Pin1(-/-) mouse embryonic fibroblasts exhibited a decrease in epidermal growth factor (EGF)-induced MEK1/2 phosphorylation compared with Pin1(+/+) mouse embryonic fibroblast. In addition, a knockdown of Pin1 resulted in the inhibition of MEK1/2 phosphorylation induced by EGF in MCF-7 cells. Furthermore, PD98059, a specific inhibitor of MEK1/2, and Juglone, a potent Pin1 inhibitor, markedly suppressed the expression of activator protein-2alpha and the HER-2 promoter activity induced by EGF or 12-O-tetradecanoylphorbol-13-acetate in MCF-7 cells. Importantly, these inhibitors inhibited the neoplastic cell transformation induced by EGF in Pin1-overexpressing JB6 Cl41 cells, which showed enhanced cellular formation compared with the control cells. Therefore, Juglone and PD98059 inhibited the colony formation of MCF-7 breast cancer cells in soft agar. These results indicate that Pin1 amplifies EGF signaling in breast cancer cells through its interaction with MEK1 and then enhances HER-2 expression, suggesting that Pin1 plays an important role in the overexpression of HER-2 through Pin1-MEK1-activator protein-2alpha signaling in breast cancer.


Assuntos
Transformação Celular Neoplásica , MAP Quinase Quinase 1/metabolismo , Peptidilprolil Isomerase/fisiologia , Receptor ErbB-2/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Fator de Crescimento Epidérmico/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/metabolismo , MAP Quinase Quinase 2/fisiologia , Camundongos , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Peptidilprolil Isomerase de Interação com NIMA , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Fosforilação , Ligação Proteica/fisiologia , Receptor ErbB-2/metabolismo , Regulação para Cima/genética , Regulação para Cima/fisiologia
15.
Biochemistry ; 48(50): 11950-60, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19883121

RESUMO

The expression of the apolipoprotein A-I gene (apoA-I) in hepatocytes is repressed by pro-inflammatory cytokines such as IL-1beta and TNFalpha. In this work, we have demonstrated that treatment of HepG2 human hepatoma cells with chemical inhibitors for JNK, p38 protein kinases, and NFkappaB transcription factor abolishes the TNFalpha-mediated inhibition of human apoA-I gene expression in HepG2 cells. In addition, we have shown that TNFalpha decreases also the rate of secretion of apoA-I protein by HepG2 cells, and this effect depends on JNK and p38, but not on NFkappaB and MEK1/2 signaling pathways. The inhibitory effect of TNFalpha has been found to be mediated by the hepatic enhancer of the apoA-I gene. The decrease in the level of human apoA-I gene expression under the impact of TNFalpha appears to be partly mediated by the inhibition of HNF4alpha and PPARalpha gene expression. Treatment of HepG2 cells with PPARalpha antagonist (MK886) or LXR agonist (TO901317) abolishes the TNFalpha-mediated decrease in the level of apoA-I gene expression. PPARalpha agonist (WY-14643) abolishes the negative effect of TNFalpha on apoA-I gene expression in the case of simultaneous inhibition of MEK1/2, although neither inhibition of MEK1/2 nor addition of WY-14643 leads to the blocking of the TNFalpha-mediated decrease in the level of apoA-I gene expression individually. The ligand-dependent regulation of apoA-I gene expression by PPARalpha appears to be affected by the TNFalpha-mediated activation of MEK1/2 kinases, probably through PPARalpha phosphorylation. Treatment of HepG2 cells with PPARalpha and LXR synthetic agonists also blocks the inhibition of apoA-I protein secretion in HepG2 cells under the impact of TNFalpha. A chromatin immunoprecipitation assay demonstrates that TNFalpha leads to a 2-fold decrease in the level of PPARalpha binding with the apoA-I gene hepatic enhancer. At the same time, the level of LXRbeta binding with the apoA-I gene hepatic enhancer is increased 3-fold under the impact of TNFalpha. These results suggest that nuclear receptors HNF4alpha, PPARalpha, and LXRs are involved in the TNFalpha-mediated downregulation of human apoA-I gene expression and apoA-I protein secretion in HepG2 cells.


Assuntos
Apolipoproteína A-I/antagonistas & inibidores , Apolipoproteína A-I/genética , Regulação para Baixo/genética , Fator 4 Nuclear de Hepatócito/fisiologia , Receptores Nucleares Órfãos/fisiologia , PPAR alfa/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Apolipoproteína A-I/biossíntese , Linhagem Celular Tumoral , Fator 4 Nuclear de Hepatócito/metabolismo , Humanos , Ligantes , Receptores X do Fígado , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/fisiologia , NF-kappa B/fisiologia , Nafenopina/metabolismo , Nafenopina/farmacologia , Pirimidinas/metabolismo , Pirimidinas/farmacologia
16.
Biofactors ; 35(5): 407-16, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19565474

RESUMO

The extracellular signal-regulated kinase cascade is a central signaling pathway that is stimulated by various extracellular stimuli. The signals of these stimuli are then transferred by the cascade's components to a large number of targets at distinct subcellular compartments, which in turn induce and regulate a large number of cellular processes. To achieve these functions, the cascade exhibits versatile and dynamic subcellular distribution that allows proper temporal and spatial modulation of the appropriate processes. In this review, we discuss the intracellular localizations of different components of the ERK cascade, and the impact of these localizations on their activation and specificity.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Organelas/enzimologia , Transdução de Sinais/fisiologia , Animais , Cavéolas/enzimologia , Citoesqueleto/enzimologia , Endossomos/enzimologia , Adesões Focais/fisiologia , Complexo de Golgi/enzimologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mitocôndrias/enzimologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Quinases S6 Ribossômicas/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Quinases raf/fisiologia
17.
Toxicol Appl Pharmacol ; 240(1): 55-65, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19595700

RESUMO

Bromocriptine (Bc) produces pituitary tumoral mass regression which induces the cellular death that was classically described as apoptosis. However, recent works have related that other mechanisms of cell death could also be involved in the maintenance of physiological and pathological pituitary homeostasis. The aim of this study was to evaluate and characterize the different types of cell death in the involution induced by Bc in experimental rat pituitary tumors. The current study demonstrated that Bc induced an effective regression of estrogen induced pituitary tumors by a mechanism identified as parapoptosis. This alternative cell death was ultrastructurally recognized by extensive cytoplasmic vacuolization and an increased cell electron density, represented around 25% of the total pituitary cells counted. Furthermore, the results obtained from biochemical assays did not correspond to the criteria of apoptosis or necrosis. We also investigated the participation of p38, ERK1/2 and PKC delta in the parapoptotic pathway. An important observation was the significant increase in phosphorylated forms of these MAPKs, the holoenzyme and catalytic fragments of PKC delta in nuclear fractions after Bc administration compared to control and estrogen treated rats. Furthermore, the immunolocalization at ultrastructural level of these kinases showed a similar distribution pattern, with a prevalent localization at nuclear level in lactotrophs from Bc treated rats. In summary, we determined that parapoptosis is the predominant cell death type involved in the regression of pituitary tumors in response to Bc treatment, and may cause the activation of PKC delta, ERK1/2 and p38.


Assuntos
Apoptose/efeitos dos fármacos , Bromocriptina/uso terapêutico , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/patologia , Prolactinoma/tratamento farmacológico , Prolactinoma/patologia , Animais , Apoptose/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , MAP Quinase Quinase 2/fisiologia , Masculino , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Neoplasias Hipofisárias/enzimologia , Prolactinoma/enzimologia , Proteína Quinase C-delta/fisiologia , Ratos , Ratos Wistar , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
18.
J Hepatol ; 51(1): 67-76, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19447520

RESUMO

BACKGROUND/AIMS: Based on the observation of biphasic induction of SGK1 expression in the regenerating liver, we investigated the role of SGK1 in the regulation of MEK/ERK signaling pathway which plays a crucial role in regulating growth and survival signaling. METHODS: To determine the role of SGK1 in the activation of MEK/ERK signaling cascade, we infected primary hepatocytes with recombinant adenoviral vector encoding SGK1, and assessed its effect on the MEK/ERK signaling pathway. RESULTS: Partial hepatectomy resulted in the biphasic transcriptional induction of SGK1 in regenerating liver tissues. Infection of primary hepatocytes with an adenoviral vector encoding SGK1 enhanced the ERK phosphorylation under serum-starved conditions and this was blocked by the expression of kinase-dead SGK1. SGK1 was found to physically interact with ERK1/2 as well as MEK1/2. Furthermore, SGK1 mediated the phosphorylation of ERK2 on Ser(29) in a serum-dependent manner. Replacement of Ser(29) to aspartic acid, which mimics the phosphorylation of Ser(29), enhanced the ERK2 activity as well as the MEK/ERK complexes formation. CONCLUSIONS: SGK1 expression during liver regeneration is a part of a signaling pathway that is necessary for enhancing ERK signaling activation through modulating the MEK/ERK complex formation.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Proteínas Imediatamente Precoces/fisiologia , Regeneração Hepática/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Células Cultivadas , Humanos , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley
19.
Oncogene ; 28(12): 1485-95, 2009 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-19198628

RESUMO

Ras genes are commonly mutated in human cancers of the skin and other tissues. Oncogenic Ras signals through multiple effector pathways, including the Erk1/2 mitogen-activated protein kinase (MAPK), phosphatidylinositol-3 kinase (PI3K) and the Ral guanine nucleotide exchange factor (RalGEF) cascades. In epidermis, the activation of oncogenic Ras induces hyperplasia and inhibits differentiation, features characteristic of squamous cell carcinoma. The downstream effector pathways required for oncogenic Ras effects in epidermis, however, are undefined. In this study, we investigated the direct contribution of Mek1 and Mek2 MAPKKs to oncogenic Ras signaling. The response of murine epidermis to conditionally active oncogenic Ras was unimpaired by deletion of either Mek1 or Mek2 MAPKKs individually. In contrast, Ras effects were entirely abolished by combined deletion of all Mek1/2 alleles, whereas epidermis retaining only one allele of either Mek1 or Mek2 showed intermediate responsiveness. Thus, the effects of oncogenic Ras on proliferation and differentiation in skin show a gene dosage-dependent requirement for the Erk1/2 MAPK cascade at the level of Mek1/2 MAPKKs.


Assuntos
Dosagem de Genes , Genes ras/fisiologia , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 2/genética , Neoplasias Cutâneas/etiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Hiperplasia , Integrases/fisiologia , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais , Pele/patologia
20.
PLoS One ; 4(2): e4600, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19240800

RESUMO

Mechanical ventilation, a fundamental therapy for acute lung injury, worsens pulmonary vascular permeability by exacting mechanical stress on various components of the respiratory system causing ventilator associated lung injury. We postulated that MK2 activation via p38 MAP kinase induced HSP25 phosphorylation, in response to mechanical stress, leading to actin stress fiber formation and endothelial barrier dysfunction. We sought to determine the role of p38 MAP kinase and its downstream effector MK2 on HSP25 phosphorylation and actin stress fiber formation in ventilator associated lung injury. Wild type and MK2(-/-) mice received mechanical ventilation with high (20 ml/kg) or low (7 ml/kg) tidal volumes up to 4 hrs, after which lungs were harvested for immunohistochemistry, immunoblotting and lung permeability assays. High tidal volume mechanical ventilation resulted in significant phosphorylation of p38 MAP kinase, MK2, HSP25, actin polymerization, and an increase in pulmonary vascular permeability in wild type mice as compared to spontaneous breathing or low tidal volume mechanical ventilation. However, pretreatment of wild type mice with specific p38 MAP kinase or MK2 inhibitors abrogated HSP25 phosphorylation and actin polymerization, and protected against increased lung permeability. Finally, MK2(-/-) mice were unable to phosphorylate HSP25 or increase actin polymerization from baseline, and were resistant to increases in lung permeability in response to HV(T) MV. Our results suggest that p38 MAP kinase and its downstream effector MK2 mediate lung permeability in ventilator associated lung injury by regulating HSP25 phosphorylation and actin cytoskeletal remodeling.


Assuntos
Actinas/metabolismo , Permeabilidade Capilar , Proteínas de Choque Térmico/fisiologia , Lesão Pulmonar/etiologia , MAP Quinase Quinase 2/fisiologia , Proteínas de Neoplasias/fisiologia , Respiração Artificial/efeitos adversos , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Pulmão , Lesão Pulmonar/metabolismo , Camundongos , Camundongos Knockout , Chaperonas Moleculares , Fosforilação , Estresse Mecânico , Ventiladores Mecânicos/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA