Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
2.
Oncogene ; 35(2): 228-40, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25961923

RESUMO

The p53 inhibitor, MDM4 (MDMX) is a cytoplasmic protein with p53-activating function under DNA damage conditions. Particularly, MDM4 promotes phosphorylation of p53 at Ser46, a modification that precedes different p53 activities. We investigated the mechanism by which MDM4 promotes this p53 modification and its consequences in untransformed mammary epithelial cells and tissues. In response to severe DNA damage, MDM4 stimulates p53Ser46(P) by binding and stabilizing serine-threonine kinase HIPK2. Under these conditions, the p53-inhibitory complex, MDM4/MDM2, dissociates and this allows MDM4 to promote p53/HIPK2 functional interaction. Comparative proteomic analysis of DNA damage-treated cells versus -untreated cells evidenced a diffuse downregulation of proteins with anti-apoptotic activity, some of which were targets of p53Ser46(P)/HIPK2 repressive activity. Importantly, MDM4 depletion abolishes the downregulation of these proteins indicating the requirement of MDM4 to promote p53-mediated transcriptional repression. Consistently, MDM4-mediated HIPK2/p53 activation precedes HIPK2/p53 nuclear translocation and activity. Noteworthy, repression of these proteins was evident also in mammary glands of mice subjected to γ-irradiation and was significantly enhanced in transgenic mice overexpressing MDM4. This study evidences the flexibility of MDM2/MDM4 heterodimer, which allows the development of a positive activity of cytoplasmic MDM4 towards p53-mediated transcriptional function. Noteworthy, this activity uncovers coordinated repression of molecules with shared anti-apoptotic function which precedes active cell apoptosis and that are frequently overexpressed and/or markers of tumour phenotype in human cancer.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/metabolismo , Dano ao DNA/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Sequência de Bases , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Citoplasma/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Células HCT116 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Serina/metabolismo , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
3.
Oncogene ; 35(7): 887-96, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-25961924

RESUMO

Human Mena (hMENA), an actin regulatory protein of the ENA/VASP family, cooperates with ErbB receptor family signaling in breast cancer. It is overexpressed in high-risk preneoplastic lesions and in primary breast tumors where it correlates with HER2 overexpression and an activated status of AKT and MAPK. The concomitant overexpression of hMENA and HER2 in breast cancer patients is indicative of a worse prognosis. hMENA is expressed along with alternatively expressed isoforms, hMENA(11a) and hMENAΔv6 with opposite functions. A novel role for the epithelial-associated hMENA(11a) isoform in sustaining HER3 activation and pro-survival pathways in HER2-overexpressing breast cancer cells has been identified by reverse phase protein array and validated in vivo in a series of breast cancer tissues. As HER3 activation is crucial in mechanisms of cell resistance to PI3K inhibitors, we explored whether hMENA(11a) is involved in these resistance mechanisms. The specific hMENA(11a) depletion switched off the HER3-related pathway activated by PI3K inhibitors and impaired the nuclear accumulation of HER3 transcription factor FOXO3a induced by PI3K inhibitors, whereas PI3K inhibitors activated hMENA(11a) phosphorylation and affected its localization. At the functional level, we found that hMENA(11a) sustains cell proliferation and survival in response to PI3K inhibitor treatment, whereas hMENA(11a) silencing increases molecules involved in cancer cell apoptosis. As shown in three-dimensional cultures, hMENA(11a) contributes to resistance to PI3K inhibition because its depletion drastically reduced cell viability upon treatment with PI3K inhibitor BEZ235. Altogether, these results indicate that hMENA(11a) in HER2-overexpressing breast cancer cells sustains HER3/AKT axis activation and contributes to HER3-mediated resistance mechanisms to PI3K inhibitors. Thus, hMENA(11a) expression can be proposed as a marker of HER3 activation and resistance to PI3K inhibition therapies, to select patients who may benefit from these combined targeted treatments. hMENA(11a) activity could represent a new target for antiproliferative therapies in breast cancer.


Assuntos
Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas dos Microfilamentos/metabolismo , Receptor ErbB-3/genética , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Eletroforese em Gel Bidimensional , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , Proteínas dos Microfilamentos/genética , Inibidores de Fosfoinositídeo-3 Quinase , Isoformas de Proteínas , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Receptor ErbB-2/genética , Transfecção
4.
Oncogene ; 34(19): 2493-504, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24998848

RESUMO

The TP53 tumor-suppressor gene is frequently mutated in human cancer. Missense mutations can add novel functions (gain-of-function, GOF) that promote tumor malignancy. Here we report that mutant (mut) p53 promotes tumor malignancy by suppressing the expression of a natural occurring anti-inflammatory cytokine, the secreted interleukin-1 receptor antagonist (sIL-1Ra, IL1RN). We show that mutp53 but not wild-type (wt) p53 suppresses the sIL-1Ra production in conditioned media of cancer cells. Moreover, mutp53, but not wtp53, binds physically the sIL-1Ra promoter and the protein-protein interaction with the transcriptional co-repressor MAFF (v-MAF musculoaponeurotic fibrosarcoma oncogene family, protein F) is required for mutp53-induced sIL-1Ra suppression. Remarkably, when exposed to IL-1 beta (IL-1ß) inflammatory stimuli, mutp53 sustains a ready-to-be-activated in vitro and in vivo cancer cells' response through the sIL-1Ra repression. Taken together, these results identify sIL-1Ra as a novel mutp53 target gene, whose suppression might be required to generate a chronic pro-inflammatory tumor microenvironment through which mutp53 promotes tumor malignancy.


Assuntos
Proteínas de Ligação a DNA/genética , Inflamação/genética , Proteína Antagonista do Receptor de Interleucina 1/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral , Células HT29 , Células Hep G2 , Humanos , Inflamação/imunologia , Proteína Antagonista do Receptor de Interleucina 1/biossíntese , Proteína Antagonista do Receptor de Interleucina 1/genética , Interleucina-1beta/farmacologia , Células MCF-7 , Fator de Transcrição MafF/metabolismo , Mutação , Neoplasias/genética , Neoplasias/mortalidade , Proteínas Nucleares/metabolismo , Prognóstico , Regiões Promotoras Genéticas/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno , Microambiente Tumoral/imunologia
5.
Cell Death Dis ; 5: e1414, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25210797

RESUMO

Che-1/AATF is an RNA polymerase II-binding protein that is involved in the regulation of gene transcription, which undergoes stabilization and accumulation in response to DNA damage. We have previously demonstrated that following apoptotic induction, Che-1 protein levels are downregulated through its interaction with the E3 ligase HDM2, which leads to Che-1 degradation by ubiquitylation. This interaction is mediated by Pin1, which determines a phosphorylation-dependent conformational change. Here we demonstrate that HIPK2, a proapoptotic kinase, is involved in Che-1 degradation. HIPK2 interacts with Che-1 and, upon genotoxic stress, phosphorylates it at specific residues. This event strongly increases HDM2/Che-1 interaction and degradation of Che-1 protein via ubiquitin-dependent proteasomal system. In agreement with these findings, we found that HIPK2 depletion strongly decreases Che-1 ubiquitylation and degradation. Notably, Che-1 overexpression strongly counteracts HIPK2-induced apoptosis. Our results establish Che-1 as a new HIPK2 target and confirm its important role in the cellular response to DNA damage.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Proteínas de Transporte/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas de Transporte/genética , Dano ao DNA , Humanos , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteólise , Proteínas Repressoras/genética , Ubiquitina/metabolismo , Ubiquitinação
6.
Cell Death Dis ; 4: e729, 2013 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-23868062

RESUMO

The transcription factor Pax8, a member of the Paired-box gene family, is a critical regulator required for proper development and differentiation of thyroid follicular cells. Despite being Pax8 well characterized with respect to its role in regulating genes responsible for thyroid differentiation, its involvement in cell survival and proliferation has been hypothesized but remains unclear. Here, we show that Pax8 overexpression significantly increases proliferation and colony-forming efficiency of Fischer rat thyroid line 5 epithelial cells, although it is not sufficient to overcome their hormone dependence. More interestingly, we show that Pax8-specific silencing induces apoptosis through a p53-dependent pathway that involves caspase-3 activation and cleavage of poly(ADP)ribose polymerase. Our data indicate that tumor protein 53 induced nuclear protein 1 (tp53inp1), a positive regulator of p53-dependent cell cycle arrest and apoptosis, is a transcriptional target of Pax8 and is upregulated by Pax8 knockdown. Remarkably, tp53inp1 silencing significantly abolishes Pax8-induced apoptosis thus suggesting that tp53inp1 may be the mediator of the observed effects. In conclusion, our data highlight that Pax8 is required for the survival of differentiated epithelial cells and its expression levels are able to modulate the proliferation rate of such cells.


Assuntos
Proliferação de Células , Sobrevivência Celular , Células Epiteliais/fisiologia , Fatores de Transcrição Box Pareados/fisiologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Ciclo Celular , Linhagem Celular , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX8 , Interferência de RNA , Ratos
7.
Eur J Histochem ; 57(1): e6, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23549465

RESUMO

The protein insulin-like growth factor II mRNA binding protein 3 (IMP3) is an important factor for cell migration and adhesion in malignancies. Recent studies have shown a remarkable overexpression of IMP3 in different human malignant neoplasms and also revealed it as an important prognostic marker in some tumor entities. The purpose of this study is to compare IMP3 immunostaining in squamous cellular skin tumor and determine whether IMP3 can aid in the differential diagnosis of these lesions. To our knowledge, IMP3 expression has not been investigated in skin squamous cell proliferations thus far. Immunohistochemical staining for IMP3 was performed on slides organized by samples from 67 patients, 34 with keratoacanthoma and 33 with primary squamous cell carcinoma (16 invasive and 17 in situ). The majority of our KAs (25/34) were negative for IMP-3 staining. The majority of SCCs (19/33) are positive for IMP3 staining. The percentage of IMP3 positive cells increases significantly in group SCC (p=0.0111), and in particular in the SCC in situ group (p=0.0021) with respect to the KA group.  IMP3 intensity staining increases significantly in SCCs (p=0.0213), and particularly in SCCs (p=0.008) with respect to KA. Our data show that IMP3 expression is different in keratoacanthomas with respect to squamous cell carcinoma. IMP3 assessment and staining pattern, together with a careful histological study, can be useful in the differential diagnosis between KA e SCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Regulação Neoplásica da Expressão Gênica , Ceratoacantoma/metabolismo , Proteínas de Neoplasias/biossíntese , Proteínas de Ligação a RNA/biossíntese , Neoplasias Cutâneas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/patologia , Feminino , Humanos , Imuno-Histoquímica , Ceratoacantoma/patologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estudos Retrospectivos , Neoplasias Cutâneas/patologia
8.
Oncogene ; 30(48): 4802-13, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21602882

RESUMO

Homeodomain-interacting protein kinase 2 (HIPK2) is an emerging player in cell response to genotoxic agents that senses damage intensity and contributes to the cell's choice between cell cycle arrest and apoptosis. Phosphorylation of p53 at S46, an apoptosis-specific p53 posttranslational modification, is the most characterized HIPK2 function in response to lethal doses of ultraviolet (UV), ionizing radiation or different anticancer drugs, such as cisplatin, roscovitine and doxorubicin (DOX). Indeed, like p53, HIPK2 has been shown to contribute to the effectiveness of these treatments. Interestingly, p53-independent mechanisms of HIPK2-induced apoptosis were described for UV and tumor growth factor-ß treatments; however, it is unknown whether these mechanisms are relevant for the responses to anticancer drugs. Because of the importance of the so-called 'p53-independent apoptosis and drug response' in human cancer chemotherapy, we asked whether p53-independent factor(s) might be involved in HIPK2-mediated chemosensitivity. Here, we show that HIPK2 depletion by RNA interference induces resistance to different anticancer drugs even in p53-null cells, suggesting the involvement of HIPK2 targets other than p53 in response to chemotherapy. In particular, we found that HIPK2 phosphorylates and promotes proteasomal degradation of ΔNp63α, a prosurvival ΔN isoform of the p53 family member, p63. Indeed, effective cell response to different genotoxic agents was shown to require phosphorylation-induced proteasomal degradation of ΔNp63α. In DOX-treated cells, we show that HIPK2 depletion interferes with ΔNp63α degradation, and expression of a HIPK2-resistant ΔNp63α-Δ390 mutant induces chemoresistance. We identify T397 as the ΔNp63α residue phosphorylated by HIPK2, and show that the non-phosphorylatable ΔNp63α-T397A mutant is not degraded in the face of either HIPK2 overexpression or DOX treatment. These results indicate ΔNp63α as a novel target of HIPK2 in response to genotoxic drugs.


Assuntos
Proteínas de Transporte/metabolismo , Dano ao DNA , Proteínas Serina-Treonina Quinases/metabolismo , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Humanos , Fosforilação , Proteólise , Proteína Supressora de Tumor p53/metabolismo
9.
Cell Prolif ; 42(3): 373-84, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19438900

RESUMO

INTRODUCTION/OBJECTIVES: The serine/threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) is a co-regulator of an increasing number of transcription factors and cofactors involved in DNA damage response and development. We and others have cloned HIPK2 as an interactor of the p53 oncosuppressor, and have studied the role of this interaction in cell response to stress. Nevertheless, our original cloning of HIPK2 as a p53-binding protein, was aimed at discovering partners of p53 involved in cell differentiation and development, still controversial p53 functions. To this aim, we used p53 as bait in yeast two-hybrid screening of a cDNA library from mouse embryo (day 11 postcoitus) when p53 is highly expressed. METHODS AND RESULTS: In this study, we directly explored whether HIPK2 and p53 cooperate in cell differentiation. By measuring HIPK2 expression and activity in skeletal muscle and haemopoietic differentiation, we observed inverse behaviour of HIPK2 and p53--excluding cooperation activity of these two factors in this event. However, by HIPK2 depletion experiments, we showed that drastic HIPK2 suppression promotes cell-cycle arrest by induction of the cyclin-dependent kinase inhibitor p21(Waf-1/Cip-1). HIPK2 activity is independent of DNA damage and takes place in cell-cycle-arresting conditions, such as terminal differentiation, growth factor deprivation, and G(0) resting. CONCLUSIONS: HIPK2 was found to be involved in cell-cycle regulation dependent on p21(Waf-1/Cip-1) and independent of DNA damage.


Assuntos
Proteínas de Transporte/fisiologia , Proliferação de Células , Dano ao DNA , Proteínas Serina-Treonina Quinases/fisiologia , Apoptose/fisiologia , Sequência de Bases , Western Blotting , Células da Medula Óssea/citologia , Proteínas de Transporte/genética , Diferenciação Celular , Células Cultivadas , Primers do DNA , Humanos , Músculo Esquelético/citologia , Proteínas Serina-Treonina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Int J Biol Markers ; 22(4): 302-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18161662

RESUMO

Mutations in the TP53 gene are the most common genetic alterations in cancer. Accumulation of mutated protein may induce circulating anti-p53 antibodies (anti-p53Ab) in sera of cancer patients. The aim of our work was to evaluate the presence and prognostic value of anti-p53Ab in gastric cancer patients and to investigate whether their presence is related to p53 overexpression in tumor tissue. Anti-p53Ab were analyzed in sera from 111 patients with gastric carcinoma and from 64 healthy donors by ELISA. p53 expression was also quantified by ELISA in biopsies of 54 gastric cancers and 22 healthy gastric mucosas. Significant anti-p53Ab levels were found in 15.3% of patients, whereas none of the 64 donor sera were positive. High levels of p53 expression were detected only in tumor tissue, in 72.2% of cases. A significant correlation was observed between anti-p53Ab and high levels of mutated p53 in tissue (p<0.05). The survival time of serum-positive patients was significantly longer than that of patients with low/negative serum levels, with a survival rate of 41.2% and 14.9%, respectively, over 48 months (p<0.05). Thus, detection of serum anti-p53Ab in gastric cancer patients can be useful to identify a subset of patients with better prognosis.


Assuntos
Adenocarcinoma/sangue , Adenocarcinoma/diagnóstico , Regulação Neoplásica da Expressão Gênica , Genes p53 , Neoplasias Gástricas/sangue , Neoplasias Gástricas/diagnóstico , Proteína Supressora de Tumor p53/química , Adenocarcinoma/genética , Adulto , Idoso , Biópsia , Estudos de Casos e Controles , Progressão da Doença , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Gástricas/genética , Proteína Supressora de Tumor p53/imunologia
12.
Cell Death Differ ; 13(9): 1554-63, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16341121

RESUMO

HMGA gene overexpression and rearrangements are frequent in several tumours, but their oncogenic function is still unclear. Here we report of a physical and functional interaction between High Mobility Group A1 (HMGA1) protein and p53 oncosuppressor. We found that HMGA1 binds p53 in vitro and in vivo, and both proteins are present in the same complexes bound to the Bax gene promoter. HMGA1 interferes with the p53-mediated transcription of p53 effectors Bax and p21(waf1) while cooperates with p53 in the transcriptional activation of the p53 inhibitor mdm2. This transcriptional modulation is associated with a reduced p53-dependent apoptosis in cells expressing exogenous HMGA1 and p53, or in cells expressing endogenously the proteins and in which p53 was activated by UV-irradiation. Furthermore, antisense inhibition of HMGA1b expression dramatically increases the UV-induced p53-mediated apoptosis. These data define a new physical and functional interaction between HMGA1 and p53 that modulates transcription of p53 target genes and inhibits apoptosis.


Assuntos
Apoptose , Proteína HMGA1b/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transcrição Gênica , Ativação Transcricional , Raios Ultravioleta , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
13.
Mol Biol Cell ; 15(8): 3751-7, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15181149

RESUMO

We recently demonstrated that the p53 oncosuppressor associates to centrosomes in mitosis and this association is disrupted by treatments with microtubule-depolymerizing agents. Here, we show that ATM, an upstream activator of p53 after DNA damage, is essential for p53 centrosomal localization and is required for the activation of the postmitotic checkpoint after spindle disruption. In mitosis, p53 failed to associate with centrosomes in two ATM-deficient, ataxiatelangiectasia-derived cell lines. Wild-type ATM gene transfer reestablished the centrosomal localization of p53 in these cells. Furthermore, wild-type p53 protein, but not the p53-S15A mutant, not phosphorylatable by ATM, localized at centrosomes when expressed in p53-null K562 cells. Finally, Ser15 phosphorylation of endogenous p53 was detected at centrosomes upon treatment with phosphatase inhibitors, suggesting that a p53 dephosphorylation step at centrosome contributes to sustain the cell cycle program in cells with normal mitotic spindles. When dissociated from centrosomes by treatments with spindle inhibitors, p53 remained phosphorylated at Ser15. AT cells, which are unable to phosphorylate p53, did not undergo postmitotic proliferation arrest after nocodazole block and release. These data demonstrate that ATM is required for p53 localization at centrosome and support the existence of a surveillance mechanism for inhibiting DNA reduplication downstream of the spindle assembly checkpoint


Assuntos
Centrossomo/química , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Serina/metabolismo , Proteína Supressora de Tumor p53/análise , Proteína Supressora de Tumor p53/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular , Linhagem Celular , Centrossomo/metabolismo , Proteínas de Ligação a DNA , Humanos , Mutação/genética , Nocodazol/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Serina/genética , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Tubulina (Proteína)/análise , Proteína Supressora de Tumor p53/química , Proteínas Supressoras de Tumor
14.
Cell Death Differ ; 11(6): 596-607, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15150542

RESUMO

Stimulation of the Ras/MAPK cascade can either activate p53 and promote replicative senescence and apoptosis, or degrade p53 and promote cell survival. Here we show that p53 can directly counteract the Ras/MAPK signaling by inactivating ERK2/MAPK. This inactivation is due to a caspase cleavage of the ERK2 protein and contributes to p53-mediated growth arrest. We found that in Ras-transformed cells, growth arrest induced by p53, but not p21(Waf1), is associated with a strong reduction in ERK2 activity, phosphorylation, and protein half-life, and with the appearance of caspase activity. Likewise, DNA damage-induced cell cycle arrest correlates with p53-dependent ERK2 downregulation and caspase activation. Furthermore, caspase inhibitors or expression of a caspase-resistant ERK2 mutant interfere with ERK2 cleavage and restore proliferation in the presence of p53 activation, indicating that caspase-mediated ERK2 degradation contributes to p53-induced growth arrest. These findings strongly point to ERK2 as a novel p53 target in growth suppression.


Assuntos
Caspases/metabolismo , Divisão Celular/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Caspase 3 , Divisão Celular/efeitos dos fármacos , Linhagem Celular Transformada , Dano ao DNA/fisiologia , Regulação para Baixo , Doxorrubicina/farmacologia , Camundongos
15.
J Biol Chem ; 276(22): 19205-13, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11376010

RESUMO

Growing evidence indicates a central role for p53 in mediating cell cycle arrest in response to mitotic spindle defects so as to prevent rereplication in cells in which the mitotic division has failed. Here we report that a transient inhibition of spindle assembly induced by nocodazole, a tubulin-depolymerizing drug, triggers a stable activation of p53, which can transduce a cell cycle inhibitory signal even when the spindle-damaging agent is removed and the spindle is allowed to reassemble. Cells transiently exposed to nocodazole continue to express high levels of p53 and p21 in the cell cycle that follows the transient exposure to nocodazole and become arrested in G(1), regardless of whether they carry a diploid or polyploid genome after mitotic exit. We also show that p53 normally associates with centrosomes in mitotic cells, whereas nocodazole disrupts this association. Together these results suggest that the induction of spindle damage, albeit transient, interferes with the subcellular localization of p53 at specific mitotic locations, which in turn dictates cell cycle arrest in the offspring of such defective mitoses.


Assuntos
Centrossomo/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Anáfase , Antineoplásicos/farmacologia , Western Blotting , Ciclo Celular , Linhagem Celular , Separação Celular , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Fase G1 , Humanos , Células K562 , Metáfase , Microscopia de Fluorescência , Mitose , Nocodazol/farmacologia , Ploidias , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fatores de Tempo , Transfecção , Tubulina (Proteína)/metabolismo , Regulação para Cima
16.
FEBS Lett ; 490(3): 163-70, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11223031

RESUMO

Most genes are members of a family. It is generally believed that a gene family derives from an ancestral gene by duplication and divergence. The tumor suppressor p53 was a striking exception to this established rule. However, two new p53 homologs, p63 and p73, have recently been described [1-6]. At the sequence level, p63 and p73 are more similar to each other than each is to p53, suggesting the possibility that the ancestral gene is a gene resembling p63/p73, while p53 is phylogenetically younger [1,2].The complexity of the family has also been enriched by the alternatively spliced forms of p63 and p73, which give rise to a complex network of proteins involved in the control of cell proliferation, apoptosis and development [1,2,4,7-9]. In this review we will mainly focus on similarities and differences as well as relationships among p63, p73 and p53.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Proteínas de Membrana , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transativadores , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Dano ao DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Fatores de Transcrição E2F , Deleção de Genes , Genes Supressores de Tumor , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Oncogênicas/metabolismo , Fosfoproteínas/química , Fosfoproteínas/genética , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/metabolismo , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor
17.
J Biol Chem ; 276(8): 5570-6, 2001 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-11096075

RESUMO

During normal cell cycles, the function of mitotic cyclin-cdk1 complexes, as well as of cdc25C phosphatase, is required for G2 phase progression. Accordingly, the G2 arrest induced by DNA damage is associated with a down-regulation of mitotic cyclins, cdk1, and cdc25C phosphatase expression. We found that the promoter activity of these genes is repressed in the G2 arrest induced by DNA damage. We asked whether the CCAAT-binding NF-Y modulates mitotic cyclins, cdk1, and cdc25C gene transcription during this type of G2 arrest. In our experimental conditions, the integrity of the CCAAT boxes of cyclin B1, cyclin B2, and cdc25C promoters, as well as the presence of a functional NF-Y complex, is strictly required for the transcriptional inhibition of these promoters. Furthermore, a dominant-negative p53 protein, impairing doxorubicin-induced G2 arrest, prevents transcriptional down-regulation of the mitotic cyclins, cdk1, and cdc25C genes. We conclude that, as already demonstrated for cdk1, NF-Y mediates the transcriptional inhibition of the mitotic cyclins and the cdc25C genes during p53-dependent G2 arrest induced by DNA damage. These data suggest a transcriptional regulatory role of NF-Y in the G2 checkpoint after DNA damage.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Proteínas de Ciclo Celular/genética , Ciclina B/genética , Fase G2/fisiologia , Regiões Promotoras Genéticas/genética , Fosfatases cdc25/genética , Fator de Ligação a CCAAT/genética , Células Cultivadas , Ciclina B1 , Dano ao DNA , Regulação para Baixo , Doxorrubicina/farmacologia , Mitose , Músculo Esquelético/citologia , Transcrição Gênica , Proteína Supressora de Tumor p53/genética
18.
Anticancer Res ; 20(5B): 3497-502, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11131653

RESUMO

It has recently been shown that tumor cells can retain the ability to undergo senescence, while the capacity of bypassing senescence has been associated with tumor progression. In this report, we showed that v-Ha-ras-mediated transformation of already immortal C2C12 myoblasts can be associated with senesence, in a low amount during in vitro passages and, to a higher extent, affer cellular stress (cell culture alkalinkation), or DNA damage (doxorubicin treatment). The capacity to undergo replicative senescence is associated with a strong increase of wt-p53 transcriptional activity and p21WAF1 up-regulation. These biochemical activities are down-modulated in the cells that evade the massive replicative senescence after stressing stimuli. Altogether, these findings show that active ras can cause senescence during the transformation of already immortal cells in associaton with p53/p21WAF1 pathway activation and support the hypothesis that p53/p21WAF1 functional activity is important in maintaining the integrity of the senescence pathway during cellular transformation.


Assuntos
Transformação Celular Neoplásica/genética , Senescência Celular/genética , Ciclinas/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes ras , Proteína Supressora de Tumor p53/fisiologia , Animais , Divisão Celular/genética , Linhagem Celular Transformada , Transformação Celular Neoplásica/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Ciclinas/genética , Genes p53 , Camundongos , Músculos/citologia , Músculos/metabolismo , Músculos/fisiologia , Transdução de Sinais/genética , Estresse Fisiológico/genética , Estresse Fisiológico/patologia , Ativação Transcricional , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Regulação para Cima
19.
J Cell Biol ; 151(6): 1295-304, 2000 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-11121443

RESUMO

The p53 oncosuppressor protein regulates cell cycle checkpoints and apoptosis, but increasing evidence also indicates its involvement in differentiation and development. We had previously demonstrated that in the presence of differentiation-promoting stimuli, p53-defective myoblasts exit from the cell cycle but do not differentiate into myocytes and myotubes. To identify the pathways through which p53 contributes to skeletal muscle differentiation, we have analyzed the expression of a series of genes regulated during myogenesis in parental and dominant-negative p53 (dnp53)-expressing C2C12 myoblasts. We found that in dnp53-expressing C2C12 cells, as well as in p53(-/-) primary myoblasts, pRb is hypophosphorylated and proliferation stops. However, these cells do not upregulate pRb and have reduced MyoD activity. The transduction of exogenous TP53 or Rb genes in p53-defective myoblasts rescues MyoD activity and differentiation potential. Additionally, in vivo studies on the Rb promoter demonstrate that p53 regulates the Rb gene expression at transcriptional level through a p53-binding site. Therefore, here we show that p53 regulates myoblast differentiation by means of pRb without affecting its cell cycle-related functions.


Assuntos
Músculo Esquelético/citologia , Fatores de Regulação Miogênica/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Camundongos , Camundongos Mutantes , Modelos Biológicos , Músculo Esquelético/metabolismo , Regiões Promotoras Genéticas , Processamento Pós-Transcricional do RNA , Proteína do Retinoblastoma/genética , Transdução de Sinais , Células-Tronco , Transcrição Gênica , Proteína Supressora de Tumor p53/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA