Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 109(42): E2875-84, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-23027969

RESUMO

Although the protooncogene c-Jun plays a critical role in cell proliferation, cell death, and malignant transformation, DNA microarray screens have identified only a few human cancer types with aberrant expression of c-Jun. Here, we show that c-Jun accumulation is robustly elevated in human glioblastoma and that this increase contributes to the malignant properties of the cells. Most importantly, the increase in c-Jun protein accumulation occurs with no corresponding increase in c-Jun mRNA or the half-life of the c-Jun protein but, rather, in the translatability of the transcript. The c-Jun 5'UTR harbors a potent internal ribosomal entry site (IRES) with a virus-like IRES domain that directs cap-independent translation in glioblastoma cells. Accumulation of c-Jun is not dependent on MAPK activity but can be stimulated by a cytoskeleton-dependent pathway. Our findings provide evidence that human c-Jun is an IRES-containing cellular transcript that contributes to cancer development through translational activation. This previously undescribed mechanism of c-Jun regulation might also be relevant to other types of human cancer and offers unique potential targets for therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Glioblastoma/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ribossomos/metabolismo , Animais , Western Blotting , Células Cultivadas , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Luciferases , Biossíntese de Proteínas/genética , Ratos , Ratos Sprague-Dawley
2.
Int J Cancer ; 130(12): 2801-11, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21732343

RESUMO

Recently, we discovered that the loss of E-cadherin induces c-Jun protein expression, which is a member of the AP-1 transcription factor family and a key player in the processes of cell proliferation and tumor development and also found in elevated levels in melanomas. Notably, the mRNA level of c-Jun was not affected, suggesting that c-Jun is regulated at post-transcriptional level. Here, we present data that suggest that the dynamic cytoskeletal network, linked to E-cadherin, is involved in the regulation of the c-Jun protein and transcriptional activity. In a signaling cascade, the loss of E-cadherin activates the transcriptional regulator ETS-1 and consequently leads to the induction of RhoC expression that stabilizes c-Jun in melanoma. The link between RhoC and c-Jun seems to be indirect via the cytoskeleton. We conclude that the loss of E-cadherin mediated cell-adhesion induces c-Jun protein expression in a multistep process, offering several possibilities for therapeutic intervention.


Assuntos
Melanoma/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Humanos , Nocodazol/farmacologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Transcrição Gênica , Ativação Transcricional , Proteínas rho de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/biossíntese , Proteína rhoB de Ligação ao GTP/biossíntese , Proteína de Ligação a GTP rhoC
3.
J Cell Sci ; 123(Pt 3): 351-9, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20053634

RESUMO

Evolution of the uricotelic system for ammonia detoxification required a mechanism for tissue-specific subcellular localization of glutamine synthetase (GS). In uricotelic vertebrates, GS is mitochondrial in liver cells and cytoplasmic in brain. Because these species contain a single copy of the GS gene, it is not clear how tissue-specific subcellular localization is achieved. Here we show that in chicken, which utilizes the uricotelic system, the GS transcripts of liver and brain cells are identical and, consistently, there is no difference in the amino acid sequence of the protein. The N-terminus of GS, which constitutes a 'weak' mitochondrial targeting signal (MTS), is sufficient to direct a chimeric protein to the mitochondria in hepatocytes and to the cytoplasm in astrocytes. Considering that a weak MTS is dependent on a highly negative mitochondrial membrane potential (DeltaPsi) for import, we examined the magnitude of DeltaPsi in hepatocytes and astrocytes. Our results unexpectedly revealed that DeltaPsi in hepatocytes is considerably more negative than that of astrocytes and that converting the targeting signal into 'strong' MTS abolished the capability to confer tissue-specific subcellular localization. We suggest that evolutional selection of weak MTS provided a tool for differential targeting of an identical protein by taking advantage of tissue-specific differences in DeltaPsi.


Assuntos
Encéfalo/metabolismo , Glutamato-Amônia Ligase/química , Glutamato-Amônia Ligase/metabolismo , Fígado/metabolismo , Mitocôndrias/metabolismo , Processamento Alternativo , Sequência de Aminoácidos , Animais , Encéfalo/ultraestrutura , Células Cultivadas , Galinhas , Glutamato-Amônia Ligase/genética , Imunoprecipitação , Fígado/ultraestrutura , Espectrometria de Massas , Potencial da Membrana Mitocondrial/fisiologia , Microscopia Eletrônica de Transmissão , Mitocôndrias/ultraestrutura , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
4.
Cancers (Basel) ; 11(11)2019 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-31744174

RESUMO

Microtubules are highly dynamic structures, which consist of α- and ß-tubulin heterodimers. They are essential for a number of cellular processes, including intracellular trafficking and mitosis. Tubulin-binding chemotherapeutics are used to treat different types of tumors, including malignant melanoma. The transcription factor c-Jun is a central driver of melanoma development and progression. Here, we identify the microtubule network as a main regulator of c-Jun activity. Monomeric α-tubulin fosters c-Jun protein stability by protein-protein interaction. In addition, this complex formation is necessary for c-Jun's nuclear localization sequence binding to importin 13, and consequent nuclear import and activity of c-Jun. A reduction in monomeric α-tubulin levels by treatment with the chemotherapeutic paclitaxel resulted in a decline in the nuclear accumulation of c-Jun in melanoma cells in an experimental murine model and in patients' tissues. These findings add important knowledge to the mechanism of the action of microtubule-targeting drugs and indicate the newly discovered regulation of c-Jun by the microtubule cytoskeleton as a novel therapeutic target for melanoma and potentially also other types of cancer.

5.
Mol Cell Endocrinol ; 252(1-2): 142-7, 2006 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-16687209

RESUMO

The cytoskeleton is a dynamic network that undergoes restructuring during a variety of cellular events including cell contact formation, cell invasion and the mitotic phase of the cell cycle. Here, we review the contribution of the cytoskeletal network to the inductive activity of glucocorticoids by focusing on the hormonal control of glutamine synthetase in the chick neural retina. Depolymerization of the cytoskeleton in cells of the intact retinal tissue inhibits the hormonal induction of glutamine synthetase, but does not alter the cellular amount of the glucocorticoid-receptor protein or the ability of the receptor molecules to translocate into the nucleus. Inhibition of glutamine synthetase induction occurs via a mechanism that involves elevation of c-Jun protein accumulation and repression of glucocorticoid-receptor transcriptional activity. Unlike growth factors and other c-Jun inducing stimuli that control the transcription of the c-Jun gene, depolymerization of the cytoskeleton elevates c-Jun accumulation by upregulating the translation of the c-Jun transcript. We postulate that the cytoskeletal-dependent increase in c-Jun accumulation is involved in cell contact control of both cell proliferation and transcriptional activity of the glucocorticoid-receptor protein.


Assuntos
Comunicação Celular/fisiologia , Citoesqueleto/fisiologia , Glucocorticoides/fisiologia , Animais , Divisão Celular , Genes jun , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiologia
6.
Cancer Biol Ther ; 17(5): 486-97, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27050748

RESUMO

A fundamental event in the development and progression of malignant melanoma is the de-regulation of cancer-relevant transcription factors. We recently showed that c-Jun is a main regulator of melanoma progression and, thus, is the most important member of the AP-1 transcription factor family in this disease. Surprisingly, no cancer-related specific c-Jun target genes in melanoma were described in the literature, so far. Therefore, we focused on pre-existing ChIP-Seq data (Encyclopedia of DNA Elements) of 3 different non-melanoma cell lines to screen direct c-Jun target genes. Here, a specific c-Jun antibody to immunoprecipitate the associated promoter DNA was used. Consequently, we identified 44 direct c-Jun targets and a detailed analysis of 6 selected genes confirmed their deregulation in malignant melanoma. The identified genes were differentially regulated comparing 4 melanoma cell lines and normal human melanocytes and we confirmed their c-Jun dependency. Direct interaction between c-Jun and the promoter/enhancer regions of the identified genes was confirmed by us via ChIP experiments. Interestingly, we revealed that the direct regulation of target gene expression via c-Jun can be independent of the existence of the classical AP-1 (5´-TGA(C/G)TCA-3´) consensus sequence allowing for the subsequent down- or up-regulation of the expression of these cancer-relevant genes. In summary, the results of this study indicate that c-Jun plays a crucial role in the development and progression of malignant melanoma via direct regulation of cancer-relevant target genes and that inhibition of direct c-Jun targets through inhibition of c-Jun is a potential novel therapeutic option for treatment of malignant melanoma.


Assuntos
Melanoma/genética , Proteínas Proto-Oncogênicas c-jun/genética , Linhagem Celular Tumoral , Humanos , Melanoma/metabolismo
7.
Oncotarget ; 7(37): 59932-59944, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27494896

RESUMO

MicroRNAs have been shown to act as oncogenes or tumor suppressers via various cellular pathways. Specifically, in breast cancer, upregulation of miR-10b is positively associated with aggressiveness of tumors. However, the mechanism by which miR-10b contributes to cell malignancy is largely unknown. Here we show that at the receiving end of the miR-10b pathway is the proto-oncogene c-Jun, a transcription factor that plays a critical role in stimulation of cell proliferation and tumor progression. c-Jun is known to be translationally activated by loss of cell contacts or restructuring of the cytoskeleton. A comprehensive analysis of miRNA expression exhibited a significant increase in miR-10b expression. This was supported by analysis of breast cancer cells, which showed that loss of E-cadherin in metastatic cells is accompanied by elevation of miR-10b and interestingly, by a marked increase in accumulation of c-Jun. Silencing miR-10b in metastatic breast cancer cells leads to a decline in c-Jun expression, whereas overexpression of miR-10b in HaCaT cells is sufficient to elevate the accumulation of c-Jun. The increase in c-Jun protein accumulation in metastatic cells is not accompanied by an increase in c-Jun mRNA and is not dependent on MAPK activity. Knockdown and overexpression experiments revealed that the increase is mediated by NF1 and RhoC, downstream targets of miR-10b that affect cytoskeletal dynamics through the ROCK pathway. Overall, we show the ability of miR-10b to activate the expression of c-Jun through RhoC and NF1, which represents a novel pathway for promoting migration and invasion of human cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Citoesqueleto/metabolismo , MicroRNAs/genética , Neurofibromina 1/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteína de Ligação a GTP rhoC/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Carcinogênese , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Metástase Neoplásica , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-jun/genética , RNA Interferente Pequeno/genética , Regulação para Cima
8.
FEBS Lett ; 579(25): 5527-34, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16213501

RESUMO

Glutamine synthetase (GS) plays a key role in two major biochemical pathways: In liver GS catalyzes ammonia detoxification, whereas in neural tissues it also functions in recycling of the neurotransmitter glutamate. In most species the GS gene gives rise to a cytoplasmic protein in both liver and neural tissues. However, in species that utilize the ureosmotic or uricotelic system for ammonia detoxification, the enzyme is cytoplasmic in neural tissues, but mitochondrial in liver cells. Since most vertebrates have a single copy of the GS gene, it is not clear how tissue-specific subcellular localization is achieved. Here we show that in the ureosmotic elasmobranch, Squalus acanthias (spiny dogfish), two different GS transcripts are generated by tissue-specific alternative splicing. The liver transcript contains an alternative exon that is not present in the neural one. This exon leads to acquisition of an upstream in-frame start codon and formation of a mitochondrial targeting signal (MTS). Therefore, the liver product is targeted to the mitochondria while the neural one is retained in the cytoplasm. These findings present a mechanism in which alternative splicing of an MTS-encoding exon is used to generate tissue-specific subcellular localization.


Assuntos
Processamento Alternativo , Glutamato-Amônia Ligase/análise , Glutamato-Amônia Ligase/genética , Squalus acanthias/genética , Sequência de Aminoácidos , Animais , Citoplasma/enzimologia , Glutamato-Amônia Ligase/metabolismo , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Isoenzimas/análise , Isoenzimas/genética , Isoenzimas/metabolismo , Fígado/citologia , Fígado/metabolismo , Mitocôndrias/enzimologia , Dados de Sequência Molecular , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo , Squalus acanthias/metabolismo , Transcrição Gênica , Ureia/metabolismo
9.
Mol Biol Cell ; 20(7): 2121-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19193763

RESUMO

Loss of E-cadherin-mediated cell-cell contacts can elicit a signaling pathway that leads to acquisition of an invasive phenotype. Here, we show that at the receiving end of this pathway is the proto-oncogene c-Jun, a member of the activator protein-1 family of transcription factors that play a key role in stimulation of cell proliferation and tumor promotion. Cell separation or abrogation of E-cadherin-mediated cell-cell contacts both cause a dramatic increase in accumulation of the c-Jun protein. Unlike growth factors that enhance the expression of c-Jun by activating the transcription of the c-jun gene, the cell contact-dependent increase in c-Jun accumulation is not accompanied by a corresponding increase in c-Jun mRNA or c-Jun protein stability but rather in the translatability of the c-Jun transcript. Consistently, the increase in c-Jun accumulation is not dependent on activation of the mitogen-activated protein kinase or beta-catenin pathways but is mediated by signals triggered by the restructured cytoskeleton. Depolymerization of the cytoskeleton can mimic the effect of cell separation and cause a dramatic increase in c-Jun accumulation, whereas Taxol inhibits the cell contact-dependent increase. This novel mechanism of c-Jun regulation seems to underlie the robust overexpression of c-Jun in tumor cells of patients with colon carcinoma.


Assuntos
Caderinas/metabolismo , Comunicação Celular , Proteínas Proto-Oncogênicas c-jun/genética , Regulação para Cima/genética , Animais , Linhagem Celular , Galinhas , Citoesqueleto/metabolismo , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Biossíntese de Proteínas , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
10.
J Biol Chem ; 283(1): 110-119, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17984088

RESUMO

Restriction of glutamine synthetase to the nervous system is mainly achieved through the mutual function of the glucocorticoid receptor and the neural restrictive silencing factor, NRSF/REST. Glucocorticoids induce glutamine synthetase expression in neural tissues while NRSF/REST represses the hormonal response in non-neural cells. NRSF/REST is a modular protein that contains two independent repression domains, at the N and C termini of the molecule, and is dominantly expressed in nonneural cells. Neural tissues express however splice variants, REST4/5, which contain the repression domain at the N, but not at the C terminus of the molecule. Here we show that full-length NRSF/REST or its C-terminal domain can inhibit almost completely the induction of gene transcription by glucocorticoids. By contrast, the N-terminal domain not only fails to repress the hormonal response but rather stimulates it markedly. The inductive activity of the N-terminal domain is mediated by hBrm, which is recruited to the promoter only in the concomitant presence of GR. Importantly, a similar inductive activity is also exerted by the splice variant REST4. These findings raise the possibility that NRSF/REST exhibits a dual role in regulation of glutamine synthetase. It represses gene induction in nonneural cells and enhances the hormonal response, via its splice variant, in the nervous system.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sítios de Ligação/genética , Western Blotting , Células COS , Linhagem Celular Tumoral , Células Cultivadas , Galinhas , Chlorocebus aethiops , Vetores Genéticos/genética , Células HeLa , Humanos , Imunoprecipitação , Regiões Promotoras Genéticas/genética , Receptores de Glucocorticoides/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Retina/citologia , Retina/efeitos dos fármacos , Retina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Leveduras/genética , Leveduras/metabolismo
11.
J Neurochem ; 83(3): 574-80, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12390519

RESUMO

Clearance of synaptic glutamate by glial cells is required for the normal function of excitatory synapses and for prevention of neurotoxicity. Although the regulatory role of glial glutamate transporters in glutamate clearance is well established, little is known about the influence of glial glutamate metabolism on this process. This study examines whether glutamine synthetase (GS), a glial-specific enzyme that amidates glutamate to glutamine, affects the uptake of glutamate. Retinal explants were incubated in the presence of [(14)C]glutamate and glutamate uptake was assessed by measurement of the amount of radioactively labeled molecules within the cells and the amount of [(14)C]glutamine released to the medium. An increase in GS expression in Müller glial cells, caused by induction of the endogenous gene, did not affect the amount of glutamate accumulated within the cells, but led to a dramatic increase in the amount of glutamine released. This increase, which was directly correlated with the level of GS expression, was dependent on the presence of external sodium ions, and could be completely abolished by methionine sulfoximine, a specific inhibitor of GS activity. Our results demonstrate that GS activity significantly influences the uptake of glutamate by the neural retina and suggest that this enzyme may represent an important target for neuroprotective strategies.


Assuntos
Espaço Extracelular/metabolismo , Glutamato-Amônia Ligase/metabolismo , Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Retina/metabolismo , Animais , Embrião de Galinha , Indução Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Glutamato-Amônia Ligase/antagonistas & inibidores , Ácido Glutâmico/farmacocinética , Técnicas In Vitro , Metionina Sulfoximina/farmacologia , Neuroglia/classificação , Neuroglia/enzimologia , Retina/citologia , Retina/embriologia , Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa