Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
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.
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
4.
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
5.
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
6.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...