Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biotechnol Biomed ; 6(1): 1-12, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36970578

RESUMO

The response of granulosa cells to Luteinizing Hormone (LH) and Follicle- Stimulating Hormone (FSH) is mediated mainly by cAMP/protein kinase A (PKA) signaling. Notably, the activity of the extracellular signal-regulated kinase (ERK) signaling cascade is elevated in response to these stimuli as well. We studied the involvement of the ERK cascade in LH- and FSH-induced steroidogenesis in two granulosa-derived cell lines, rLHR-4 and rFSHR-17, respectively. We found that stimulation of these cells with the appropriate gonadotropin induced ERK activation as well as progesterone production downstream of PKA. Inhibition of ERK activity enhanced gonadotropin-stimulated progesterone production, which was correlated with increased expression of the Steroidogenic Acute Regulatory Protein (StAR), a key regulator of progesterone synthesis. Therefore, it is likely that gonadotropin-stimulated progesterone formation is regulated by a pathway that includes PKA and StAR, and this process is down-regulated by ERK, due to attenuation of StAR expression. Our results suggest that activation of PKA signaling by gonadotropins not only induces steroidogenesis but also activates down-regulation machinery involving the ERK cascade. The activation of ERK by gonadotropins as well as by other agents may be a key mechanism for the modulation of gonadotropin-induced steroidogenesis.

2.
Cell Biol Int ; 46(7): 1021-1035, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35332606

RESUMO

The extracellular signal-regulated kinases (ERK) 1 and 2 (ERK1/2) are members of the mitogen-activated protein kinase family. Using various stimulated rodent cells and kinase activation techniques, we identified a 46-kDa ERK. The kinetics of activation of this ERK isoform was similar to that of ERK1 and ERK2 under most but not all circumstances. We purified this isoform from rat cells followed by its cloning. The sequence of this isoform revealed that it is an alternatively spliced version of the 44-kDa ERK1 and therefore we termed it ERK1b. Interestingly, this isoform had a 26-amino acid insertion between residues 340 and 341 of ERK1, which results from Intron 7 insertion to the sequence. Examining the expression pattern, we found that ERK1b is detected mainly in rat and particularly in Ras-transformed Rat1 cells. In this cell line, ERK1b was more sensitive to extracellular stimulation than ERK1 and ERK2. Moreover, unlike ERK1 and ERK2, ERK1b had a very low binding affinity to MEK1. This low interaction led to nuclear localization of this isoform when expressed together with MEK1 under conditions in which ERK1 and ERK2 are retained in the cytoplasm. In addition, ERK1b was not coimmunoprecipitated with MEK1. We identified a new, 46-kDa ERK alternatively spliced isoform. Our results indicate that this isoform is the major one to respond to exogenous stimulation in Ras-transformed cells, probably due to its differential regulation by MAPK/ERK kinase and by phosphatases.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno , Animais , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Isoformas de Proteínas/metabolismo , Ratos
3.
iScience ; 24(6): 102670, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34189435

RESUMO

ERK1c is an alternatively spliced isoform of ERK1 that specifically regulates mitotic Golgi fragmentation, which allows division of the Golgi during mitosis. We have previously shown that ERK1c translocates to the Golgi during mitosis where it is activated by a resident MEK1b to induce Golgi fragmentation. However, the mechanism of ERK1c functions in the Golgi remained obscure. Here, we searched for ERK1c substrates and identified HOOK3 as a mediator of ERK1c-induced mitotic Golgi fragmentation, which requires a second phosphorylation by AuroraA for its function. In cycling cells, HOOK3 interacts with microtubules (MTs) and links them to the Golgi. Early in mitosis, HOOK3 is phosphorylated by ERK1c and later by AuroraA, resulting in HOOK3 detachment from the MTs, and elevated interaction with GM130. This detachment modulates Golgi stability and allows fragmentation of the Golgi. This study demonstrates a novel mechanism of Golgi apparatus destabilization early in mitosis to allow mitotic progression.

4.
Sci Rep ; 8(1): 11830, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30087384

RESUMO

MAPK/ERK kinase (MEK) 1/2 are central signaling proteins that serve as specificity determinants of the MAPK/ERK cascade. More than twenty activating mutations have been reported for MEK1/2, and many of them are known to cause diseases such as cancers, arteriovenous malformation and RASopathies. Changes in their intrinsic activity do not seem to correlate with the severity of the diseases. Here we studied four MEK1/2 mutations using biochemical and molecular dynamic methods. Although the studied mutants elevated the activating phosphorylation of MEK they had no effect on the stimulated ERK1/2 phosphorylation. Studying the regulatory mechanism that may explain this lack of effect, we found that one type of mutation affects MEK stability and two types of mutations demonstrate a reduced sensitivity to PP2A. Together, our results indicate that some MEK mutations exert their function not only by their elevated intrinsic activity, but also by modulation of regulatory elements such as protein stability or dephosphorylation.


Assuntos
Regulação Enzimológica da Expressão Gênica , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 2/genética , Peptídeo Hidrolases/genética , Monoéster Fosfórico Hidrolases/genética , Animais , Células COS , Chlorocebus aethiops , Células HeLa , Humanos , MAP Quinase Quinase 1/química , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Mutação , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Peptídeo Hidrolases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Estabilidade Proteica
5.
Sci Signal ; 11(525)2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636389

RESUMO

The stimulated nuclear translocation of signaling proteins, such as MAPKs, is a necessity for the initiation and regulation of their physiological functions. Previously, we determined that nuclear translocation of the MAPKs p38 and JNK involves binding to heterodimers comprising importin 3 and either importin 7 or importin 9. Here, we identified the importin-binding region in p38 and JNK and developed a myristoylated peptide targeting this site that we called PERY. The PERY peptide specifically blocked the interaction of p38 and JNK with the importins, restricted their nuclear translocation, and inhibited phosphorylation of their nuclear (but not cytoplasmic) substrates. Through these effects, the PERY peptide reduced the proliferation of several (but not all) cancer cell lines in culture and inhibited the growth of a human breast cancer xenograft in mice. In addition, the PERY peptide substantially inhibited inflammation in mice, as manifested in models of colitis and colitis-associated colon cancer. The PERY peptide more effectively prevented colon cancer development than did a commercial p38 inhibitor. In vivo analysis further suggested that this effect was mediated by PERY peptide-induced prevention of the nuclear translocation of p38 in macrophages. Together, these results support the use of the nuclear translocation of p38 and JNK as a novel drug target to treat various cancers and inflammation-induced diseases.


Assuntos
Núcleo Celular/efeitos dos fármacos , Inflamação/prevenção & controle , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias/tratamento farmacológico , Peptídeos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Colite/induzido quimicamente , Colite/prevenção & controle , Sulfato de Dextrana , Feminino , Células HCT116 , Células HeLa , Humanos , Inflamação/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Células MCF-7 , Camundongos Nus , Neoplasias/metabolismo , Neoplasias/patologia , Homologia de Sequência de Aminoácidos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/genética
6.
Cell Physiol Biochem ; 43(4): 1533-1546, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29035881

RESUMO

BACKGROUND/AIMS: Results from recent studies suggest that extremely low frequency magnetic fields (ELF-MF) interfere with intracellular signaling pathways related to proliferative control. The mitogen-activated protein kinases (MAPKs), central signaling components that regulate essentially all stimulated cellular processes, include the extracellular signal-regulated kinases 1/2 (ERK1/2) that are extremely sensitive to extracellular cues. Anti-phospho-ERK antibodies serve as a readout for ERK1/2 activation and are able to detect minute changes in ERK stimulation. The objective of this study was to explore whether activation of ERK1/2 and other signaling cascades can be used as a readout for responses of a variety of cell types, both transformed and non-transformed, to ELF-MF. METHODS: We applied ELF-MF at various field strengths and time periods to eight different cell types with an exposure system housed in a tissue culture incubator and followed the phosphorylation of MAPKs and Akt by western blotting. RESULTS: We found that the phosphorylation of ERK1/2 is increased in response to ELF-MF. However, the phosphorylation of ERK1/2 is likely too low to induce ELF-MF-dependent proliferation or oncogenic transformation. The p38 MAPK was very slightly phosphorylated, but JNK or Akt were not. The effect on ERK1/2 was detected for exposures to ELF-MF strengths as low as 0.15 µT and was maximal at ∼10 µT. We also show that ERK1/2 phosphorylation is blocked by the flavoprotein inhibitor diphenyleneiodonium, indicating that the response to ELF-MF may be exerted via NADP oxidase similar to the phosphorylation of ERK1/2 in response to microwave radiation. CONCLUSIONS: Our results further indicate that cells are responsive to ELF-MF at field strengths much lower than previously suspected and that the effect may be mediated by NADP oxidase. However, the small increase in ERK1/2 phosphorylation is probably insufficient to affect proliferation and oncogenic transformation. Therefore, the results cannot be regarded as proof of the involvement of ELF-MF in cancer in general or childhood leukemia in particular.


Assuntos
Campos Eletromagnéticos , Ativação Enzimática , Sistema de Sinalização das MAP Quinases , Animais , Linhagem Celular , Linhagem Celular Tumoral , Campos Eletromagnéticos/efeitos adversos , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NADPH Oxidases/metabolismo , Neoplasias/etiologia , Neoplasias/metabolismo , Fosforilação
11.
12.
Oncotarget ; 7(31): 50490-50506, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27409167

RESUMO

Phosphoinositide (PIP) phosphatases such as myotubularins (MTMs) inhibit growth factor receptor signaling. However, the function of myotubularin-related protein 7 (MTMR7) in cancer is unknown. We show that MTMR7 protein was down-regulated with increasing tumor grade (G), size (T) and stage (UICC) in patients with colorectal cancer (CRC) (n=1786). The presence of MTMR7 in the stroma correlated with poor prognosis, whereas MTMR7 expression in the tumor was not predictive for patients' survival. Insulin reduced MTMR7 protein levels in human CRC cell lines, and CRC patients with type 2 diabetes mellitus (T2DM) or loss of imprinting (LOI) of insulin-like growth factor 2 (IGF2) had an increased risk for MTMR7 loss. Mechanistically, MTMR7 lowered PIPs and inhibited insulin-mediated AKT-ERK1/2 signaling and proliferation in human CRC cell lines. MTMR7 provides a novel link between growth factor signaling and cancer, and may thus constitute a potential marker or drug target for human CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Insulina/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Diabetes Mellitus Tipo 2/metabolismo , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Impressão Genômica , Humanos , Fator de Crescimento Insulin-Like II/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Metástase Neoplásica , Transdução de Sinais , Análise Serial de Tecidos , Distribuição Tecidual
13.
J Cell Sci ; 128(22): 4083-95, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26459638

RESUMO

Golgi fragmentation is a highly regulated process that allows division of the Golgi complex between the two daughter cells. The mitotic reorganization of the Golgi is accompanied by a temporary block in Golgi functioning, as protein transport in and out of the Golgi stops. Our group has previously demonstrated the involvement of the alternatively spliced variants ERK1c and MEK1b (ERK1 is also known as MAPK3, and MEK1 as MAP2K1) in mitotic Golgi fragmentation. We had also found that ERK1c translocates to the Golgi at the G2 to M phase transition, but the molecular mechanism underlying this recruitment remains unknown. In this study, we narrowed the translocation timing to prophase and prometaphase, and elucidated its molecular mechanism. We found that CDK1 phosphorylates Ser343 of ERK1c, thereby allowing the binding of phosphorylated ERK1c to a complex that consists of PI4KIIIß (also known as PI4KB) and the 14-3-3γ dimer (encoded by YWHAB). The stability of the complex is regulated by protein kinase D (PKD)-mediated phosphorylation of PI4KIIIß. The complex assembly induces the Golgi shuttling of ERK1c, where it is activated by MEK1b, and induces Golgi fragmentation. Our work shows that protein shuttling to the Golgi is not completely abolished at the G2 to M phase transition, thus integrating several independent Golgi-regulating processes into one coherent pathway.


Assuntos
Proteínas 14-3-3/metabolismo , Complexo de Golgi/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mitose/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Células HeLa , Humanos , Proteína Quinase 3 Ativada por Mitógeno/genética , Multimerização Proteica
14.
Mol Biol Cell ; 26(13): 2475-90, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25971798

RESUMO

ERK-regulated cell proliferation requires multiple phosphorylation events catalyzed first by MEK and then by casein kinase 2 (CK2), followed by interaction with importin7 and subsequent nuclear translocation of pERK. We report that genetic manipulation of a core component of the actin filaments of cancer cells, the tropomyosin Tm5NM1, regulates the proliferation of normal cells both in vitro and in vivo. Mouse embryo fibroblasts (MEFs) lacking Tm5NM1, which have reduced proliferative capacity, are insensitive to inhibition of ERK by peptide and small-molecule inhibitors, indicating that ERK is unable to regulate proliferation of these knockout (KO) cells. Treatment of wild-type MEFs with a CK2 inhibitor to block phosphorylation of the nuclear translocation signal in pERK resulted in greatly decreased cell proliferation and a significant reduction in the nuclear translocation of pERK. In contrast, Tm5NM1 KO MEFs, which show reduced nuclear translocation of pERK, were unaffected by inhibition of CK2. This suggested that it is nuclear translocation of CK2-phosphorylated pERK that regulates cell proliferation and this capacity is absent in Tm5NM1 KO cells. Proximity ligation assays confirmed a growth factor-stimulated interaction of pERK with Tm5NM1 and that the interaction of pERK with importin7 is greatly reduced in the Tm5NM1 KO cells.


Assuntos
Citoesqueleto de Actina/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Tropomiosina/fisiologia , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Caseína Quinase II/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Tropomiosina/genética , Tropomiosina/metabolismo
15.
Nat Commun ; 6: 6685, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25819065

RESUMO

A hallmark of the ERK1/2 functioning is their nuclear translocation, which is mainly required for the induction of proliferation. Activated ERK1/2 molecules that remain in the cytoplasm initiate other activities, including immediate feedback loops. Prevention of the nuclear translocation should therefore inhibit proliferation, without affecting cytoplasm-induced cellular processes. Here we present an NTS-derived myristoylated phosphomimetic peptide, which blocks the interaction of importin7 and ERK1/2, and consequently the nuclear translocation of the latter. In culture, the peptide induces apoptosis of melanoma cells inhibits the viability of other cancer cells, but has no effect on non-transformed, immortalized cells. It even inhibits the viability of PLX4032- and U0126-resistant melanoma cells. In xenograft models, the peptide inhibits several cancers, and acts much better than PLX4032 in preventing melanoma recurrence. This study provides a proof of concept for using the nuclear translocation of ERK1/2 as a drug target for the combat of various ERK1/2-related cancers.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Peptídeos/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Western Blotting , Células CHO , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cricetulus , Células HCT116 , Células HeLa , Humanos , Imuno-Histoquímica , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Camundongos Nus , Camundongos SCID , Microscopia de Fluorescência , Terapia de Alvo Molecular , Transplante de Neoplasias , Transporte Proteico/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Bioorg Med Chem ; 16(14): 6789-98, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18571926

RESUMO

We synthesized two carminic acid (7-alpha-d-glucopyranosyl-9,10-dihydro-3,5,6,8-tetrahydroxy-1-methyl-9,10-dioxo-2-anthracene carboxlic acid, CA)-GnRH conjugates to be used as a model for potential photoactive targeted compounds. CA was conjugated to the epsilon-amino group of [d-Lys(6)]GnRH through its carboxylic moiety or via a beta-alanine spacer (beta-ala). Redox potentials of CA and its conjugates were determined. We used electron spin resonance (ESR) and spin trapping techniques to study the light-stimulated redox properties of CA and its CA-GnRH conjugates. Upon irradiation, the compounds stimulated the formation of reactive oxygen species (ROS), that is, singlet oxygen ((1)O(2)) and oxygen radicals (O(2)(-*) and OH(*)). Both conjugates exhibited higher ROS production than the non-conjugated CA. The bioactivity properties of the CA conjugates and the parent peptide, [d-Lys(6)]GnRH, were tested on primary rat pituitary cells. We found that the conjugates preserved the bioactivity of GnRH as illustrated by their capability to induce ERK phosphorylation and LH release.


Assuntos
Carmim/análogos & derivados , Hormônio Liberador de Gonadotropina/química , Hipófise/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Carmim/química , Células Cultivadas , Espectroscopia de Ressonância de Spin Eletrônica , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Radicais Livres , Hormônio Luteinizante/metabolismo , Oxirredução , Fosforilação , Fotoquímica , Hipófise/citologia , Hipófise/metabolismo , Ratos , Oxigênio Singlete
17.
Mol Cell Biol ; 27(3): 803-17, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17101779

RESUMO

The mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) cascade plays a central role in intracellular signaling by many extracellular stimuli. One target of the ERK cascade is peroxisome proliferator-activated receptor gamma (PPARgamma), a nuclear receptor that promotes differentiation and apoptosis. It was previously demonstrated that PPARgamma activity is attenuated upon mitogenic stimulation due to phosphorylation of its Ser84 by ERKs. Here we show that stimulation by tetradecanoyl phorbol acetate (TPA) attenuates PPARgamma's activity in a MEK-dependent manner, even when Ser84 is mutated to Ala. To elucidate the mechanism of attenuation, we found that PPARgamma directly interacts with MEKs, which are the activators of ERKs, but not with ERKs themselves, both in vivo and in vitro. This interaction is facilitated by MEKs' phosphorylation and is mediated by the basic D domain of MEK1 and the AF2 domain of PPARgamma. Immunofluorescence microscopy and subcellular fractionation revealed that MEK1 exports PPARgamma from the nucleus, and this finding was supported by small interfering RNA knockdown of MEK1 and use of a cell-permeable interaction-blocking peptide, which prevented TPA-induced export of PPARgamma from the nucleus. Thus, we show here a novel mode of downregulation of PPARgamma by its MEK-dependent redistribution from the nucleus to the cytosol. This unanticipated role for the stimulation-induced nuclear shuttling of MEKs shows that MEKs can regulate additional signaling components besides the ERK cascade.


Assuntos
Núcleo Celular/metabolismo , Regulação para Baixo , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/metabolismo , PPAR gama/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Acil-CoA Oxidase/genética , Animais , Células CHO , Células COS , Núcleo Celular/efeitos dos fármacos , Chlorocebus aethiops , Cricetinae , Cricetulus , Citosol/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Células HeLa , Humanos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , PPAR gama/genética , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Ratos , Rosiglitazona , Serina/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Tiazolidinedionas/farmacologia , Ativação Transcricional/efeitos dos fármacos , Técnicas do Sistema de Duplo-Híbrido
18.
J Parasitol ; 91(4): 756-63, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17089740

RESUMO

The Schistosoma mansoni parasite life cycle involves complex developmental processes that enable it to cause severe hepatic damage. Protein phosphorylation has previously been implicated in the transformation of cercariae to schistosomula of S. mansoni. Here, we studied the possible involvement of surface (ecto) and shed (exo) protein kinases (PKs) in this developmental process. We found that ecto-PKs are indeed located on the surface of the schistosomula and can phosphorylate up to 5 distinct proteins at this location. Surface phosphorylation was sensitive to acetylcholine, which increased phosphorylation of 3 proteins and reduced phosphorylation of the other 2. The ecto-PKs can be shed from the surface into the incubation medium during parasite differentiation. The main exo-PK is CKII, as concluded from the substrate specificity of the PK, its inhibition by heparin, activation by spermin, and recognition by antibody directed to the anti--alpha-subunit of CKII in the incubation medium of the schistosomula. In spite of its similarity to the ecto-PKs, the activity of the exo-PK is not affected by addition of acetylcholine. These results indicate that ecto- and exo-PKs could be involved in the parasite's development or host-parasite interactions.


Assuntos
Acetilcolina/fisiologia , Caseína Quinase II/metabolismo , Proteínas Quinases/metabolismo , Schistosoma mansoni/enzimologia , Animais , Biomphalaria , Caseína Quinase II/química , Caseínas/metabolismo , Eletroforese em Gel de Poliacrilamida , Heparina/farmacologia , Camundongos , Fosforilação , Fosvitina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Schistosoma mansoni/metabolismo , Serina/metabolismo , Treonina/metabolismo , Tirosina/metabolismo
19.
Mol Cell Biol ; 24(22): 10000-15, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15509801

RESUMO

Extracellular signal-regulated kinases (ERKs) are signaling molecules that regulate many cellular processes. We have previously identified an alternatively spliced 46-kDa form of ERK1 that is expressed in rats and mice and named ERK1b. Here we report that the same splicing event in humans and monkeys causes, due to sequence differences in the inserted introns, the production of an ERK isoform that migrates together with the 42-kDa ERK2. Because of the differences of this isoform from ERK1b, we named it ERK1c. We found that its expression levels are about 10% of ERK1. ERK1c seems to be expressed in a wide variety of tissues and cells. Its activation by MEKs and inactivation by phosphatases are slower than those of ERK1, which is probably the reason for its differential regulation in response to extracellular stimuli. Unlike ERK1, ERK1c undergoes monoubiquitination, which is increased with elevated cell density concomitantly with accumulation of ERK1c in the Golgi apparatus. Elevated cell density also causes enhanced Golgi fragmentation, which is facilitated by overexpression of native ERK1c and is prevented by dominant-negative ERK1c, indicating that ERK1c mediates cell density-induced Golgi fragmentation. The differential regulation of ERK1c extends the signaling specificity of MEKs after stimulation by various extracellular stimuli.


Assuntos
Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Processamento Alternativo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Expressão Gênica , Células HeLa , Humanos , Cinética , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/química , Proteína Quinase 3 Ativada por Mitógeno/genética , Dados de Sequência Molecular , Peso Molecular , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Distribuição Tecidual , Fatores de Transcrição/metabolismo , Proteínas Elk-1 do Domínio ets
20.
Cancer Res ; 64(16): 5736-44, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15313914

RESUMO

A standard therapy used today for prostate cancer is androgen ablation by gonadotropin-releasing hormone analogs (GnRH-a). Although most patients respond to androgen ablation as an initial systemic therapy, nearly all cases will develop androgen resistance, the management of which is still a major challenge. Here, we report that GnRH-a can directly induce apoptosis of the androgen-independent prostate cancer-derived DU145 and PC3 cell lines. Using specific inhibitors, we found that the apoptotic effect of GnRH-a is mediated by c-Jun NH2-terminal kinase (JNK) and inhibited by the phosphatidylinositol 3'-kinase (PI3K)-protein kinase B (PKB) pathway. Indeed, in DU145 cells, GnRH-a activates the JNK cascade in a c-Src- and MLK3-dependent manner but does not involve protein kinase C and epidermal growth factor receptor. Concomitantly, GnRH-a reduces the activity of the PI3K-PKB pathway, which results in the dephosphorylation of PKB mainly in the nucleus. The reduction of PKB activity releases PKB-induced inhibition of MLK3 and thus further stimulates JNK activity and accelerates the apoptotic effect of GnRH-a. Interestingly, extracellular signal-regulated kinase is also activated by GnRH-a, and this occurs via a pathway that involves matrix metalloproteinases and epidermal growth factor receptor, but its activation does not affect JNK activation and the GnRH-a-induced apoptosis. Our results support a potential use of GnRH-a for the treatment of advanced prostate cancer and suggest that the outcome of this treatment can be amplified by using PI3K-PKB inhibitors.


Assuntos
Apoptose/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Apoptose/fisiologia , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Ativação Enzimática/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...