Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Signal ; 26(12): 2658-66, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25152365

RESUMO

Tumor Necrosis Factor Receptor 2 (TNFR2) activates transcription factor κB (NF-κB) and c-Jun N-terminal kinase (JNK). Most of the biological activities triggered by TNFR2 depend on the recruitment of TNF Receptor-Associated Factor 2 (TRAF2) to the intracellular region of the receptor. The intracellular region of TNFR2 contains five highly conserved amino acid sequences, three of which appear implicated in receptor signaling. In this work we have studied the interaction of TNFR2 with TRAF proteins as well as the functional consequences of this interaction. We show that TRAF1, TRAF2 and TRAF3 bind to the receptor through two different binding sites located at conserved modules IV and V of its intracellular region, respectively. We also show that TRAF1 and TRAF3 have opposite effects to TRAF2 on NF-κB and JNK activation by TNFR2. Moreover, we show that TNFR2 is able to induce JNK activation in a TRAF2-independent fashion. This new receptor activity relies on a sequence located in the conserved module III. This region is also responsible for the ability of TNFR2 to induce TRAF2 degradation, thus emphasizing the role of conserved module III (amino acids 338-379) on receptor signaling and regulation. We show that only TNFR2 can induce TRAF2 degradation while TRAF1 or TRAF3 is not subjected to this regulatory mechanism and that TRAF1, but not TRAF3, is able to inhibit TRAF2 degradation.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/fisiologia , Linhagem Celular , Células HEK293 , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência , Transdução de Sinais/fisiologia
2.
Oncotarget ; 5(1): 224-36, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24318359

RESUMO

Tumor Necrosis Factor (TNF) interacts with two receptors known as TNFR1 and TNFR2. TNFR1 activation may result in either cell proliferation or cell death. TNFR2 activates Nuclear Factor-kappaB (NF-kB) and c-Jun N-terminal kinase (JNK) which lead to transcriptional activation of genes related to cell proliferation and survival. This depends on the binding of TNF Receptor Associated Factor 2 (TRAF2) to the receptor. TNFR2 also induces TRAF2 degradation. In this work we have investigated the structural features of TNFR2 responsible for inducing TRAF2 degradation and have studied the biological consequences of this activity. We show that when TNFR1 and TNFR2 are co-expressed, TRAF2 depletion leads to an enhanced TNFR1 cytotoxicity which correlates with the inhibition of NF-kB. NF-kB activation and TRAF2 degradation depend of different regions of the receptor since TNFR2 mutants at amino acids 343-349 fail to induce TRAF2 degradation and have lost their ability to enhance TNFR1-mediated cell death but are still able to activate NF-kB. Moreover, whereas NF-kB activation requires TRAF2 binding to the receptor, TRAF2 degradation appears independent of TRAF2 binding. Thus, TNFR2 mutants unable to bind TRAF2 are still able to induce its degradation and to enhance TNFR1-mediated cytotoxicity. To test further this receptor crosstalk we have developed a system stably expressing in cells carrying only endogenous TNFR1 the chimeric receptor RANK-TNFR2, formed by the extracellular region of RANK (Receptor activator of NF-kB) and the intracellular region of TNFR2.This has made possible to study independently the signals triggered by TNFR1 and TNFR2. In these cells TNFR1 is selectively activated by soluble TNF (sTNF) while RANK-TNFR2 is selectively activated by RANKL. Treatment of these cells with sTNF and RANKL leads to an enhanced cytotoxicity.


Assuntos
Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular , Fibroblastos , Células HEK293 , Humanos , Camundongos , Transdução de Sinais , Transfecção
3.
Cell Signal ; 24(6): 1297-305, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22374304

RESUMO

Tumor necrosis factor (TNF) is a key mediator in the inflammatory response which is implicated in the onset of a number of diseases. Research on TNF led to the characterization of the largest family of cytokines known until now, the TNF superfamily, which exert their biological effects through the interaction with transmembrane receptors of the TNFR superfamily. TNF itself exerts its biological effects interacting with two different receptors: TNFR1 and TNFR2. TNFR1 presents a death domain on its intracellular region. In contrast to TNFR1, TNFR2 does not have a death domain. Activation of TNFR1 implies the consecutive formation of two different TNF receptor signalling complexes. Complex I controls the expression of antiapoptotic proteins that prevent the triggering of cell death processes, whereas Complex II triggers cell death processes. TNFR2 only signals for antiapoptotic reactions. However, recent evidence indicates that TNFR2 also signals to induce TRAF2 degradation. TRAF2 is a key mediator in signal transduction of both TNFR1 and TNFR2. Thus, this novel signalling pathway has two important implications: on one hand, it represents an auto regulatory loop for TNFR2; on the other hand, when this signal is triggered TNFR1 activity is modified so that antiapoptotic pathways are inhibited and apoptotic reactions are enhanced.


Assuntos
Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais , Sequência de Aminoácidos , Animais , Apoptose , Humanos , Dados de Sequência Molecular , Receptores do Fator de Necrose Tumoral/metabolismo , Alinhamento de Sequência , Fator 1 Associado a Receptor de TNF/química , Fator 1 Associado a Receptor de TNF/imunologia , Fator 1 Associado a Receptor de TNF/metabolismo , Fator 2 Associado a Receptor de TNF/química , Fator 2 Associado a Receptor de TNF/imunologia , Fator 2 Associado a Receptor de TNF/metabolismo
4.
J Biol Chem ; 286(26): 22814-24, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21558270

RESUMO

Tumor necrosis factor receptor 2 (TNFR2) activates transcription factor κB (NF-κB) and c-Jun N-terminal kinase (JNK). The mechanisms mediating these activations are dependent on the recruitment of TNF receptor-associated factor 2 (TRAF2) to the intracellular region of the receptor. TNFR2 also induces TRAF2 degradation. We show that in addition to the well characterized TRAF2 binding motif 402-SKEE-405, the human receptor contains another sequence located at the C-terminal end (amino acids 425-439), which also recruits TRAF2 and activates NF-κB. In addition to that, human TNFR2 contains a conserved region (amino acids 338-379) which is responsible for TRAF2 degradation and therefore of terminating NF-κB signaling. TRAF2 degradation and the lack of NF-κB activation when both TNFR1 and TNFR2 are co-expressed results in an enhanced ability of TNFR1 to induce cell death, showing that the cross-talk between both receptors is of a great biological relevance. Induction of TRAF2 degradation appears to be independent of TRAF2 binding to the receptor. Amino acids 343-TGSSDSS-349 are essential for inducing TRAF2 degradation because deletion mutants of this region or point mutations at serine residues 345 and 346 or 348 and 349 obliterate the ability of TNFR2 to induce TRAF2 degradation.


Assuntos
NF-kappa B/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator 2 Associado a Receptor de TNF/metabolismo , Motivos de Aminoácidos , Animais , Células HEK293 , Humanos , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , NF-kappa B/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/genética , Fator 2 Associado a Receptor de TNF/genética
5.
J Proteome Res ; 9(9): 4649-60, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20597553

RESUMO

Microtubule interfering agents (MIAs) are antitumor drugs that inhibit microtubule dynamics, while kinesin spindle protein (KSP) inhibitors are substances that block the formation of the bipolar spindle during mitosis. All these compounds cause the accumulation of mitotic cells and subsequently cell death. We used two-dimensional gel electrophoresis (2DE) followed by MALDI-MS analysis to demonstrate that the MIAs vinblastine (Velban) and paclitaxel (Taxol), as well as the KSP inhibitor S-tritil-L-cysteine, induce the phosphorylation of annexin A2 in human lung carcinoma A549 cells. Further tandem mass spectrometry analysis using a combination of peptide fragmentation methods (CID and ETD) and multiple reaction monitoring (MRM) analysis determined that this modification occurs mainly at threonine 19. We show that MIAs and KSP inhibitors only induce this phosphorylation in cells capable of reaching the M phase. Furthermore, we demonstrate that CDK activity is required for the phosphorylation of annexin A2 induced by MIAs and KSP inhibitors. Finally, we have used double thymidine block synchronization to demonstrate that annexin A2 is not phosphorylated during a normal mitosis, indicating that this phosphorylation of annexin A2 is a specific response to these drugs.


Assuntos
Anexina A2/metabolismo , Cinesinas/antagonistas & inibidores , Mitose/efeitos dos fármacos , Proteômica/métodos , Moduladores de Tubulina/farmacologia , Sequência de Aminoácidos , Anexina A2/química , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Cisteína/análogos & derivados , Cisteína/farmacologia , Eletroforese em Gel Bidimensional , Humanos , Cinesinas/metabolismo , Dados de Sequência Molecular , Paclitaxel/farmacologia , Mapeamento de Peptídeos , Fosforilação/efeitos dos fármacos , Fosfotreonina/química , Fosfotreonina/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Vimblastina/farmacologia
6.
Cell Mol Life Sci ; 66(14): 2383-90, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19381437

RESUMO

Nuclear distribution gene C homolog (NudC) is a highly conserved gene. It has been identified in different species from fungi to mammals. The high degree of conservation, in special in the nudC domain, suggests that they are genes with essential functions. Most of the identified genes in the family have been implicated in cell division through the regulation of cytoplasmic dynein. As for mammalian genes, human NUDC has been implicated in the migration and proliferation of tumor cells and has therefore been considered a possible therapeutic target. There is evidence suggesting that mammalian NudC is also implicated in the regulation of the inflammatory response and in thrombopoiesis. The presence of these other functions not related to the interaction with molecular motors agrees with that these genes and their products are larger in size than their microbial orthologous, indicating that they have evolved to convey additional features.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Mamíferos/metabolismo , Proteínas Nucleares/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Proteínas de Ciclo Celular/genética , Citocinese , Dineínas/metabolismo , Humanos , Inflamação/enzimologia , Inflamação/metabolismo , Mamíferos/genética , Proteínas Nucleares/genética
7.
J Proteomics ; 71(6): 592-600, 2009 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-18832053

RESUMO

Microtubule interfering agents (MIAs) are anti-tumor drugs that inhibit microtubule dynamics, while kinesin spindle protein (KSP) inhibitors are substances that block the formation of the bipolar spindle during mitosis. All these compounds cause G2/M arrest and cell death. Using 2D-PAGE followed by Nano-LC-ESI-Q-ToF analysis, we found that MIAs such as vincristine (Oncovin) or paclitaxel (Taxol) and KSP inhibitors such as S-tritil-l-cysteine induce the phosphorylation of the nuclear protein p54(nrb) in HeLa cells. Furthermore, we demonstrate that cisplatin (Platinol), an anti-tumor drug that does not cause M arrest, does not induce this modification. We show that the G2/M arrest induced by MIAs is required for p54(nrb) phosphorylation. Finally, we demonstrate that CDK activity is required for MIA-induced phosphorylation of p54(nrb).


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Fase G2/fisiologia , Cinesinas/metabolismo , Mitose/fisiologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Fatores de Transcrição de Octâmero/metabolismo , Proteínas de Ligação a RNA/metabolismo , Moduladores de Tubulina/farmacologia , Linhagem Celular , Cisplatino/farmacologia , Cisteína/análogos & derivados , Cisteína/farmacologia , Proteínas de Ligação a DNA , Eletroforese em Gel Bidimensional , Fase G2/efeitos dos fármacos , Humanos , Cinesinas/antagonistas & inibidores , Microtúbulos , Moduladores de Mitose/farmacologia , Paclitaxel/farmacologia , Fosforilação , Espectrometria de Massas em Tandem , Vincristina/farmacologia
8.
Eur Cytokine Netw ; 18(4): 172-80, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17993451

RESUMO

Tumor necrosis factor (TNF) is known to be a mediator of a variety of cellular responses including apoptotic death or proliferation depending on the target cell and the environmental conditions. We show here that TNF triggers both growth and death signals in NIH 3T3 murine fibroblasts. In cells arrested in G(0) by serum deprivation, TNF drives approximately 50% of them to enter the cell cycle, but kills the cells that remain quiescent. The presence of serum prevents toxic effects of TNF, suggesting that TNF can cooperate to drive cells through the cell cycle, but is unable to do so by itself and alternatively it triggers death signals in cells unable to proliferate. Interestingly, TNF induces a similar toxic effect in cells forced to stay at the G(1)/S border, S or M phases. We have explored the TNF apoptotic pathway in arrested cells. This mechanism is not due to the loss of the anti-apoptotic capacity of NFkappaB and is mediated by mitochondria since Bcl-2 overexpression partially inhibits cell death. There are, however, interesting differences in the kinetics of mitochondrial events which indicate that this form of sensitization to TNF leads to an apoptotic mechanism different from that observed after sensitization by RNA synthesis inhibition.


Assuntos
Apoptose , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ciclo Celular , Proliferação de Células , Citocinas/metabolismo , DNA/metabolismo , Fibroblastos/metabolismo , Membranas Intracelulares/metabolismo , Camundongos , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Células NIH 3T3 , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Rodaminas/farmacologia
9.
Proteomics ; 7(18): 3299-304, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17708594

RESUMO

Paclitaxel (Ptx) is an antitumoural drug that inhibits microtubule dynamics, causes G2/M arrest and induces cell death. 2-D PAGE and MALDI-TOF-MS analysis of HeLa cells extracts revealed that Ptx up-regulates a form of the eukaryotic elongation factor 1Bgamma (eEF1Bgamma) and down-regulates another one. This event, linked to the lack of Ptx effect over eEF1Bgamma mRNA or protein levels suggested a PTM of this elongation factor. Further 2-D PAGE analysis followed by a phosphospecific staining with PRO-Q Diamond showed the staining of the Ptx up-regulated form only. Moreover, this Ptx up-regulated form of eEF1Bgamma disappears upon treatment with protein phosphatase. Thus, we demonstrate that human eEF1Bgamma phosphorylation is regulated by Ptx.


Assuntos
Paclitaxel/metabolismo , Fator 1 de Elongação de Peptídeos/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Eletroforese em Gel Bidimensional , Citometria de Fluxo , Células HeLa , Humanos , Dados de Sequência Molecular , Fator 1 de Elongação de Peptídeos/genética , Fosforilação , RNA Mensageiro/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
10.
FEBS Lett ; 581(16): 3057-62, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17555748

RESUMO

We report the characterization of a cDNA induced in mouse macrophages that encodes a 332-amino acid protein with extensive sequence identity with members of the mammalian nudC-like genes. The interaction between mNUDC and the regulatory beta subunit of platelet activating factor acetylhydrolase I (PAF-AH(I)) shown in this article indicates a new function of NUDC. Thus, we show that NUDC increases the catalytic activity of PAF-AH(I) and that this regulatory activity is located in the carboxyl terminal half of the protein which is highly conserved. This suggests a novel function for mammalian nudC-like genes as anti-inflammatory proteins.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Inflamação/genética , Macrófagos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ciclo Celular/isolamento & purificação , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Emericella/genética , Inflamação/enzimologia , Macrófagos/enzimologia , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/isolamento & purificação , Proteínas Nucleares/metabolismo , Ligação Proteica , Subunidades Proteicas/metabolismo , Homologia de Sequência de Aminoácidos
11.
Arch Biochem Biophys ; 461(1): 123-9, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17367746

RESUMO

Vincristine and paclitaxel are widely used antitumoral drugs that interfere with microtubule dynamics. We have previously demonstrated that vincristine induces phosphorylation of HSP27 at serine 82 in MCF-7 cells. In this report, we show that vincristine also causes phosphorylation of serines 78 and 15. Moreover, we demonstrate that phosphorylation of this chaperone is induced by the p38 signalling pathway while the JNK pathway is not implicated. Differences between vincristine and paclitaxel treatments are also appreciated. Thus, while vincristine induces a strong phosphorylation of the three serines, paclitaxel induces a weak phosphorylation of serine 78 and has no effect over serines 82 and 15 phosphorylation. Interestingly, pre-treatment of cells with a ten-fold excess of paclitaxel abolishes vincristine-induced phosphorylation of HSP27.


Assuntos
Proteínas de Choque Térmico/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Microtúbulos/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Moduladores de Tubulina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Linhagem Celular Tumoral , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/química , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Microtúbulos/enzimologia , Microtúbulos/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/química , Paclitaxel/farmacologia , Fosforilação/efeitos dos fármacos , Serina/metabolismo , Vincristina/farmacologia
12.
Cancer Lett ; 247(2): 273-82, 2007 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-16843591

RESUMO

Vincristine is an antitumor drug that inhibits microtubule polymerization, causes G2/M arrest and induces apoptosis. 2D-PAGE and MALDI-TOF-MS analysis of vincristine effects on MCF7 cells, revealed a vincristine upregulated form and a vincristine downregulated form of the antiapoptotic protein HSP27. These findings linked to the lack of vincristine effect over HSP27 mRNA, suggest a protein post-translational modification. Further assays indicated the presence of a phosphorylated peptide, containing serine 82, only in the vincristine upregulated form. Serine 82 phosphorylation was confirmed using specific antibodies. Thus, phosphorylation of HSP27 may play a role in the cellular response to vincristine.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas de Neoplasias/metabolismo , Vincristina/farmacologia , Sequência de Aminoácidos , Sequência de Bases , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Primers do DNA , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Humanos , Chaperonas Moleculares , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Fosforilação , Processamento de Proteína Pós-Traducional , Proteoma , Serina/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
13.
Breast Cancer Res Treat ; 98(1): 81-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16555127

RESUMO

Vinca alkaloids (VAs) such as Vincristine, Vinblastine and Vinorelbine are antineoplastic drugs that inhibit tubulin polymerisation into microtubules, induce mitotic G2/M arrest, activate c-Jun N-terminal kinase (JNK) and induce apoptosis. Although there are many studies evaluating the effect of VAs on breast cancer patients, until now little was known about how these compounds and estradiol signaling pathways might interfere. In this report, we show for the first time that VAs decreased ERalpha protein levels in the human breast cancer cell line MCF-7; VAs induced a parallel decrease in estrogen receptor mRNA. All the VAs tested inhibited estradiol (E2) mediated transactivation at ERE-driven promoters. E2 inhibited VAs-induced AP1 stimulation in MCF-7, but this inhibition was not observed when E2 is added 24 h in advance of VAs treatment. In contrast to the reported preventing effect over taxol-mediated apoptosis, E2 did not prevent VAs-induced cell death and interestingly, addition of E2 24 hours in advance of VAs treatment resulted in an increase of the number of cells undergoing apoptosis. Similar results were observed when E2 is replaced by other proliferation signals such as EGF. These results demonstrate that in the breast cancer cell-line MCF-7, E2-induced proliferation before VAs treatment enhances the apoptotical response to VAs which might have important implications in clinica.


Assuntos
Estradiol/metabolismo , Receptor alfa de Estrogênio/biossíntese , Regulação Neoplásica da Expressão Gênica , Alcaloides de Vinca/farmacologia , Apoptose , Linhagem Celular Tumoral , Células HeLa , Humanos , Potenciais da Membrana , Mitocôndrias/metabolismo , Plasmídeos/metabolismo , Transdução de Sinais , Transfecção , Vimblastina/farmacologia , Vincristina/farmacologia
14.
J Biol Chem ; 279(37): 38294-302, 2004 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15229223

RESUMO

Melatonin is an indole hormone produced mainly by the pineal gland. We have previously demonstrated that melatonin interferes with estrogen (E(2)) signaling in MCF7 cells by impairing estrogen receptor (ER) pathways. Here we present the characterization of its mechanism of action showing that melatonin is a specific inhibitor of E(2)-induced ERalpha-mediated transcription in both estrogen response element- and AP1-containing promoters, whereas ERbeta-mediated transactivation is not inhibited or even activated at certain promoters. We show that the sensitivity of MCF-7 cells to melatonin depends on the ERalpha/ERbeta ratio, and ectopic expression of ERbeta results in MCF-7 cells becoming insensitive to this hormone. Melatonin acts as a calmodulin antagonist inducing conformational changes in the ERalpha-calmodulin (CaM) complex, thus impairing the binding of E(2).ERalpha.CaM complex to DNA and, therefore, preventing ERalpha-dependent transcription. Moreover the mutant ERalpha (K302G,K303G), unable to bind calmodulin, becomes insensitive to melatonin. The effect of melatonin is specific since other related indoles neither interact with CaM nor inhibit ERalpha-mediated transactivation. Interestingly, melatonin does not affect the binding of coactivators to ERalpha, indicating that melatonin action is different from that of current therapeutic anti-estrogens used in breast cancer therapy. Thus, they target ERalpha at different levels, representing two independent ways to control ERalpha activity. It is, therefore, conceivably a synergistic pharmacological effect of melatonin and current anti-estrogen drugs.


Assuntos
Calmodulina/metabolismo , Melatonina/química , Receptores de Estrogênio/metabolismo , Calmodulina/química , Linhagem Celular Tumoral , DNA/química , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Estrogênios/metabolismo , Glutationa Transferase/metabolismo , Células HeLa , Humanos , Concentração Inibidora 50 , Luciferases/metabolismo , Melatonina/metabolismo , Modelos Químicos , Mutação , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Elementos de Resposta , Sensibilidade e Especificidade , Transdução de Sinais , Espectrometria de Fluorescência , Transcrição Gênica , Ativação Transcricional , Transfecção
15.
Endocrinology ; 144(7): 2967-76, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12810552

RESUMO

We have isolated and functionally characterized the exon 7-skipped variant (ERDeltaE7) of estrogen receptor (ER)alpha, which has emerged as the predominant variant expressed in multiple normal and tumoral tissues. However, to date no function has been established for this variant in mammalian cells. ERDeltaE7 exhibits a negligible ability to bind ligands, insensitivity to allosteric modulation by estrogen and antiestrogens, and loss of estrogen-dependent interaction with p160 coactivators such as SRC-1 and AIB1. ERDeltaE7 is able to form heterodimers with both ERalpha and ERbeta in a ligand-independent manner. Transient expression experiments in HeLa cells show that increasing amounts of ERDeltaE7 result in a progressive inhibition of the estrogen-dependent transcriptional activation by both wild-type ERalpha and ERbeta on estrogen response element-driven promoters. The inhibitory effect of ERDeltaE7 is due to the inhibition of binding of wild-type receptors to their responsive elements. Surprisingly, the activation function (AF)-1-dependent transactivation triggered by epithelial growth factor and phorbol-12-myristate-13-acetate is also abolished in ERDeltaE7 despite AF1 integrity, suggesting a cross-talk between AF1 and AF2 regions of the receptor. These results indicate that the naturally occurring variant ERDeltaE7 is a dominant negative receptor that, when expressed at high levels relative to wild-type ERs, might have profound effects on several estrogen-dependent functions.


Assuntos
Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Ativação Transcricional/fisiologia , Processamento Alternativo/genética , Sequência de Aminoácidos , Sequência de Bases , Dimerização , Moduladores de Receptor Estrogênico/metabolismo , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Estrogênios/metabolismo , Células HeLa , Humanos , Regiões Promotoras Genéticas/genética , Receptores de Estrogênio/química , Elementos de Resposta/genética
16.
Oral Oncol ; 38(4): 357-63, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12076699

RESUMO

The objective of this study is to describe the molecular alterations in carcinomas in one specific location of the head and neck, the hypopharynx. Thirty-seven hypopharyngeal squamous cell carcinomas were studied. The DNA from tumour and healthy tissue was evaluated for amplification of the 11q13 region and of the MYC and ERBB1 oncogenes, for integration of the Human Papillomavirus (HPV), and for loss of heterozygosity (LOH) at p53 and NAT2 loci. The most common alteration was the amplification of the 11q13 region (78% of the cases), followed by LOH at p53 locus (70%). MYC amplification was found in 19% of the cases, ERBB1 amplification in 29%, LOH at NAT2 locus in 25%, and integration of the HPV in 29%. 11q13 amplification was related with nodal metastases and higher tumour recurrence rates. These findings confirm that 11q13 amplification is one of the most frequent genetic alterations in hypopharyngeal squamous cell carcinomas, and that it may have prognostic significance in these tumours.


Assuntos
Carcinoma de Células Escamosas/genética , Genes erbB-1/genética , Genes myb/genética , Neoplasias Hipofaríngeas/genética , Adulto , Idoso , DNA Viral/análise , Amplificação de Genes , Heterozigoto , Humanos , Perda de Heterozigosidade/genética , Pessoa de Meia-Idade , Papillomaviridae/genética , Reação em Cadeia da Polimerase
17.
Mol Endocrinol ; 16(5): 947-60, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11981030

RESUMO

In the search for differences between ERalpha and ERbeta, we analyzed the interaction of both receptors with calmodulin (CaM) and demonstrated that ERalpha but not ERbeta directly interacts with CaM. Using transiently transfected HeLa cells, we examined the effect of the CaM antagonist N-(6-aminohexyl)-5-chloro-naphthalene sulfonilamide hydrochloride (W7) on the transactivation properties of ERalpha and ERbeta in promoters containing either estrogen response elements or activator protein 1 elements. Transactivation by ERalpha was dose-dependently inhibited by W7, whereas that of ERbeta was not inhibited or even activated at low W7 concentrations. In agreement with these results, transactivation of an estrogen response element containing promoter in MCF-7 cells (which express a high ERalpha/ERbeta ratio) was also inhibited by W7. In contrast, transactivation in T47D cells (which express a low ERalpha/ERbeta ratio) was not affected by this CaM antagonist. The sensitivity of MCF-7 cells to W7 was abolished when cells were transfected with increasing amounts of ERbeta, indicating that the sensitivity to CaM antagonists of estrogen-responsive tissues correlates with a high ERalpha/ERbeta ratio. Finally, substitution of lysine residues 302 and 303 of ERalpha for glycine rendered a mutant ERalpha unable to interact with CaM whose transactivation activity became insensitive to W7. Our results indicate that CaM antagonists are selective modulators of ER able to inhibit ERalpha-mediated activity, whereas ERbeta actions were not affected or even potentiated by W7.


Assuntos
Calmodulina/metabolismo , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Sítios de Ligação , Calmodulina/antagonistas & inibidores , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Estrogênios/farmacologia , Expressão Gênica , Células HeLa , Humanos , Lisina/genética , Mutagênese , Regiões Promotoras Genéticas , Receptores de Estrogênio/genética , Elementos de Resposta , Sulfonamidas/farmacologia , Fator de Transcrição AP-1/farmacologia , Ativação Transcricional/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA