Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 30(2): 488-499, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36477079

RESUMO

Cisplatin and other platinum-based anticancer agents are among the most widely used chemotherapy drugs in the treatment of different types of cancer. However, it is common to find patients who respond well to treatment at first but later relapse due to the appearance of resistance to cisplatin. Among the mechanisms responsible for this phenomenon is the increase in DNA damage repair. Here, we elucidate the effect of cisplatin on the MRN (MRE11-RAD50-NBS1) DNA damage sensor complex. We found that the tumor suppressor FBXW7 is a key factor in controlling the turnover of the MRN complex by inducing its degradation through lysosomes. Inhibition of lysosomal enzymes allowed the detection of the association of FBXW7-dependent ubiquitylated MRN with LC3 and the autophagy adaptor p62/SQSTM1 and the localization of MRN in lysosomes. Furthermore, cisplatin-induced cell death increased MRN degradation, suggesting that this complex is one of the targets that favor cell death. These findings open the possibility of using the induction of the degradation of the MRN complex after genotoxic damage as a potential therapeutic strategy to eliminate tumor cells.


Assuntos
Cisplatino , Enzimas Reparadoras do DNA , Humanos , Cisplatino/farmacologia , Proteína 7 com Repetições F-Box-WD/metabolismo , Proteína Homóloga a MRE11 , Enzimas Reparadoras do DNA/genética , Proteínas de Ciclo Celular/genética , Proteínas Nucleares/metabolismo , Proteínas de Ligação a DNA/metabolismo , Hidrolases Anidrido Ácido/metabolismo
2.
Cancers (Basel) ; 12(4)2020 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-32316282

RESUMO

Too much of a good thing can become a bad thing. An example is FBXW7, a well-known tumor suppressor that may also contribute to tumorigenesis. Here, we reflect on the results of three laboratories describing the role of FBXW7 in the degradation of p53 and the possible implications of this finding in tumor cell development. We also speculate about the function of FBXW7 as a key player in the cell fate after DNA damage and how this could be exploited in the treatment of cancer disease.

3.
FASEB J ; 33(10): 11420-11430, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31337255

RESUMO

Eukaryotic cells have developed sophisticated mechanisms to ensure the integrity of the genome and prevent the transmission of altered genetic information to daughter cells. If this control system fails, accumulation of mutations would increase risk of diseases such as cancer. Ubiquitylation, an essential process for protein degradation and signal transduction, is critical for ensuring genome integrity as well as almost all cellular functions. Here, we investigated the role of the SKP1-Cullin-1-F-box protein (SCF)-[F-box and tryptophan-aspartic acid (WD) repeat domain containing 7 (FBXW7)] ubiquitin ligase in cell proliferation by searching for targets implicated in this process. We identified a hitherto-unknown FBXW7-interacting protein, p53, which is phosphorylated by glycogen synthase kinase 3 at serine 33 and then ubiquitylated by SCF(FBXW7) and degraded. This ubiquitylation is carried out in normally growing cells but primarily after DNA damage. Specifically, we found that SCF(FBXW7)-specific targeting of p53 is crucial for the recovery of cell proliferation after UV-induced DNA damage. Furthermore, we observed that amplification of FBXW7 in wild-type p53 tumors reduced the survival of patients with breast cancer. These results provide a rationale for using SCF(FBXW7) inhibitors in the treatment of this subset of tumors.-Galindo-Moreno, M., Giráldez, S., Limón-Mortés, M. C., Belmonte-Fernández, A., Reed, S. I., Sáez, C., Japón, M. Á., Tortolero, M., Romero, F. SCF(FBXW7)-mediated degradation of p53 promotes cell recovery after UV-induced DNA damage.


Assuntos
Dano ao DNA/genética , Proteína 7 com Repetições F-Box-WD/genética , Proteína Supressora de Tumor p53/genética , Animais , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Chlorocebus aethiops , Proteínas F-Box/genética , Células HCT116 , Células HEK293 , Humanos , Mutação/genética , Fosforilação/genética , Domínios Proteicos/genética , Proteólise , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética
4.
FASEB J ; 31(7): 2925-2936, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28360195

RESUMO

Polo-like kinase 1 (PLK1) is a serine/threonine kinase involved in several stages of the cell cycle, including the entry and exit from mitosis, and cytokinesis. Furthermore, it has an essential role in the regulation of DNA replication. Together with cyclin A, PLK1 also promotes CDH1 phosphorylation to trigger its ubiquitination and degradation, allowing cell cycle progression. The PLK1 levels in different type of tumors are very high compared to normal tissues, which is consistent with its role in promoting proliferation. Therefore, several PLK1 inhibitors have been developed and tested for the treatment of cancer. Here, we further analyzed PLK1 degradation and found that cytoplasmic PLK1 is ubiquitinated and subsequently degraded by the SCFßTrCP/proteasome. This procedure is triggered when heat shock protein (HSP) 90 is inhibited with geldanamycin, which results in misfolding of PLK1. We also identified CDK1 as the major kinase involved in this degradation. Our work shows for the first time that HSP90 inhibition arrests cell cycle progression at the G1/S transition. This novel mechanism inhibits CDH1 degradation through CDK1-dependent PLK1 destruction by the SCFßTrCP/proteasome. In these conditions, CDH1 substrates do not accumulate and cell cycle arrests, providing a novel pathway for regulation of the cell cycle at the G1-to-S boundary.-Giráldez, S., Galindo-Moreno, M., Limón-Mortés, M. C., Rivas, A. C., Herrero-Ruiz, J., Mora-Santos, M., Sáez, C., Japón, M. Á., Tortolero, M., Romero, F. G1/S phase progression is regulated by PLK1 degradation through the CDK1/ßTrCP axis.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/fisiologia , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Animais , Proteína Quinase CDC2/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular , Clonagem Molecular , Regulação Enzimológica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Plasmídeos , Mutação Puntual , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas Contendo Repetições de beta-Transducina/genética , Quinase 1 Polo-Like
5.
Eur J Cancer ; 49(2): 500-10, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22819078

RESUMO

Pituitary tumour transforming gene (pttg1) encodes Securin, a protein involved in the inhibition of sister chromatid separation binding to Separase until the onset of anaphase. Separase is a cysteine-protease that degrades cohesin to segregate the sister chromatids to opposite poles of the cell. The amount of Securin is strongly regulated because it should allow Separase activation when it is degraded by the anaphase promoting complex/cyclosome, should arrest the cell cycle after DNA damage, when it is degraded through SKP1-CUL1-ßTrCP ubiquitin ligase, and its overexpression induces tumour formation and correlates with metastasis in multiple tumours. Securin is a phosphoprotein that contains 32 potentially phosphorylatable residues. We mutated and analysed most of them, and found a single mutant, hSecT60A, that showed enhanced oncogenic properties. Our fluorescence activated cell sorting analysis, fluorescence in situ hybridisation assays, tumour cell migration and invasion experiments and gene expression by microarrays analysis clearly involved hSecT60A in chromosomal instability and cell invasion. These results show, for the first time, that a single mutation in pttg1 is sufficient to trigger the oncogenic properties of Securin. The finding of this point mutation in patients might be used as an effective strategy for early detection of cancer.


Assuntos
Transformação Celular Neoplásica/genética , Instabilidade Cromossômica , Proteínas de Neoplasias/genética , Neoplasias/genética , Mutação Puntual , Animais , Células COS , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Chlorocebus aethiops , Expressão Gênica , Células HCT116 , Células HeLa , Humanos , Análise em Microsséries , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Securina , Treonina/genética , Transfecção
6.
J Biol Chem ; 286(34): 30047-56, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21757741

RESUMO

PTTG1, also known as securin, is an inactivating partner of separase, the major effector for chromosome segregation during mitosis. At the metaphase-to-anaphase transition, securin is targeted for proteasomal destruction by the anaphase-promoting complex or cyclosome, allowing activation of separase. In addition, securin is overexpressed in metastatic or genomically instable tumors, suggesting a relevant role for securin in tumor progression. Stability of securin is regulated by phosphorylation; some phosphorylated forms are degraded out of mitosis, by the action of the SKP1-CUL1-F-box protein (SCF) complex. The kinases targeting securin for proteolysis have not been identified, and mechanistic insight into the cause of securin accumulation in human cancers is lacking. Here, we demonstrate that glycogen synthase kinase-3ß (GSK3ß) phosphorylates securin to promote its proteolysis via SCF(ßTrCP) E3 ubiquitin ligase. Importantly, a strong correlation between securin accumulation and GSK3ß inactivation was observed in breast cancer tissues, indicating that GSK3ß inactivation may account for securin accumulation in breast cancers.


Assuntos
Neoplasias da Mama/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Mitose , Proteínas de Neoplasias/metabolismo , Animais , Neoplasias da Mama/genética , Células COS , Chlorocebus aethiops , Feminino , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Fosforilação , Estabilidade Proteica , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Securina
7.
J Cell Sci ; 121(11): 1825-31, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18460583

RESUMO

Securin is a chaperone protein with bifunctional properties. It binds to separase to inhibit premature sister chromatid separation until the onset of anaphase, and it also takes part in cell-cycle arrest after UV irradiation. At metaphase-to-anaphase transition, securin is targeted for proteasomal destruction by the anaphase-promoting complex or cyclosome (APC/C), allowing activation of separase. However, although securin is reported to undergo proteasome-dependent degradation after UV irradiation, the ubiquitin ligase responsible for securin ubiquitylation has not been well characterized. In this study, we show that UV radiation induced a marked reduction of securin in both the nucleus and cytoplasm. Moreover, we show that GSK-3beta inhibitors prevent securin degradation, and that CUL1 and betaTrCP are involved in this depletion. We also confirmed that SKP1-CUL1-betaTrCP (SCF(betaTrCP)) ubiquitylates securin in vivo, and identified a conserved and unconventional betaTrCP recognition motif (DDAYPE) in the securin primary amino acid sequence of humans, nonhuman primates and rodents. Furthermore, downregulation of betaTrCP caused an accumulation of securin in non-irradiated cells. We conclude that SCF(betaTrCP) is the E3 ubiquitin ligase responsible for securin degradation after UV irradiation, and that it is involved in securin turnover in nonstressed cells.


Assuntos
Proteínas de Neoplasias/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos da radiação , Raios Ultravioleta , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Células COS , Ciclo Celular/fisiologia , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Chlorocebus aethiops , Proteínas Culina/metabolismo , Citoplasma/metabolismo , Citoplasma/efeitos da radiação , Regulação para Baixo/fisiologia , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células HeLa , Humanos , Proteínas de Neoplasias/efeitos da radiação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Securina , Ubiquitinação/fisiologia
8.
Mol Cell Biol ; 26(11): 4017-27, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16705156

RESUMO

Sister chromatid segregation is triggered at the metaphase-to-anaphase transition by the activation of the protease separase. For most of the cell cycle, separase activity is kept in check by its association with the inhibitory chaperone securin. Activation of separase occurs at anaphase onset, when securin is targeted for destruction by the anaphase-promoting complex or cyclosome E3 ubiquitin protein ligase. This results in the release of the cohesins from chromosomes, which in turn allows the segregation of sister chromatids to opposite spindle poles. Here we show that human securin (hSecurin) forms a complex with enzymatically active protein phosphatase 2A (PP2A) and that it is a substrate of the phosphatase, both in vitro and in vivo. Treatment of cells with okadaic acid, a potent inhibitor of PP2A, results in various hyperphosphorylated forms of hSecurin which are extremely unstable, due to the action of the Skp1/Cul1/F-box protein complex ubiquitin ligase. We propose that PP2A regulates hSecurin levels by counteracting its phosphorylation, which promotes its degradation. Misregulation of this process may lead to the formation of tumors, in which overproduction of hSecurin is often observed.


Assuntos
Proteínas de Neoplasias/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas Ligases SKP Culina F-Box/metabolismo , Animais , Células COS , Ciclo Celular , Chlorocebus aethiops , Ativação Enzimática , Células HCT116 , Células HeLa , Humanos , Fosforilação , Ligação Proteica , Proteína Fosfatase 2 , Securina , Especificidade por Substrato , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA