Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Cell Death Differ ; 23(3): 509-20, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26358154

RESUMO

The tumor-suppressor p53 is a transcription factor that prevents cancer development and is involved in regulation of various physiological processes. This is mediated both by induction of cell cycle arrest and apoptosis and by controlling the expression of a plethora of target genes, including secreted proteins. It has been demonstrated that p53 may exert its effect in non-cell-autonomous manner by modulating the expression of genes that encode for secreted factors. In this study, we utilized our microarray data to identify and characterize novel p53 target genes expressed in human liver cells and associated with steroid hormones processing and transfer. We identified the steroid hormones binding factors, sex hormone-binding globulin (SHBG), corticosteroid-binding globulin (CBG) and cytochrome P450 family 21 subfamily A polypeptide 2, as novel p53 target genes. Their expression and secretion was increased following p53 activation in various hepatic cells. We observed that p53 wild-type mice exhibited higher levels of CBG compared with their p53 null counterparts. We demonstrated that the induction of the steroid hormones binding factors can be mediated by binding to specific p53 responsive elements within their promoters. In addition, utilizing conditioned medium experiments we have shown that p53-dependent induction of SHBG secretion from liver cells enhances apoptosis of breast cancer cells. Moreover, depletion of SHBG abolished the induction of breast cancer cells death. The newly identified p53 target genes suggest a novel non-cell-autonomous tumor-suppressive regulation mediated by p53 that is central for maintaining organism homeostasis.


Assuntos
Fígado/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose , Células Hep G2 , Humanos , Células MCF-7 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Globulina de Ligação a Hormônio Sexual/genética , Globulina de Ligação a Hormônio Sexual/metabolismo , Esteroide 21-Hidroxilase/genética , Esteroide 21-Hidroxilase/metabolismo , Transcortina/genética , Transcortina/metabolismo , Transcrição Gênica
2.
Waste Manag ; 45: 298-305, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26054962

RESUMO

In batteries associated with waste electrical and electronic equipment (WEEE), battery systems can be found with a higher content of valuable and critical raw materials like cobalt and rare earth elements (REE) relative to the general mix of portable batteries. Based on a material flow model, this study estimates the flows of REE and cobalt associated to WEEE and the fate of these metals in the end-of-life systems. In 2011, approximately 40 Mg REE and 325 Mg cobalt were disposed of with WEEE-batteries. The end-of-life recycling rate for cobalt was 14%, for REE 0%. The volume of waste batteries can be expected to grow, but variation in the battery composition makes it difficult to forecast the future secondary raw material potential. Nevertheless, product specific treatment strategies ought to be implemented throughout the stages of the value chain.


Assuntos
Cobalto/análise , Fontes de Energia Elétrica , Resíduo Eletrônico/análise , Metais Terras Raras/análise , Reciclagem/métodos , Gerenciamento de Resíduos/métodos , Alemanha , Modelos Teóricos
4.
Cell Death Differ ; 21(9): 1419-31, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24832469

RESUMO

p53 loss of heterozygosity (p53LOH) is frequently observed in Li-Fraumeni syndrome (LFS) patients who carry a mutant (Mut) p53 germ-line mutation. Here, we focused on elucidating the link between p53LOH and tumor development in stem cells (SCs). Although adult mesenchymal stem cells (MSCs) robustly underwent p53LOH, p53LOH in induced embryonic pluripotent stem cells (iPSCs) was significantly attenuated. Only SCs that underwent p53LOH induced malignant tumors in mice. These results may explain why LFS patients develop normally, yet acquire tumors in adulthood. Surprisingly, an analysis of single-cell sub-clones of iPSCs, MSCs and ex vivo bone marrow (BM) progenitors revealed that p53LOH is a bi-directional process, which may result in either the loss of wild-type (WT) or Mut p53 allele. Interestingly, most BM progenitors underwent Mutp53LOH. Our results suggest that the bi-directional p53LOH process may function as a cell-fate checkpoint. The loss of Mutp53 may be regarded as a DNA repair event leading to genome stability. Indeed, gene expression analysis of the p53LOH process revealed upregulation of a specific chromatin remodeler and a burst of DNA repair genes. However, in the case of loss of WTp53, cells are endowed with uncontrolled growth that promotes cancer.


Assuntos
Alelos , Perda de Heterozigosidade , Células-Tronco/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Proteína Supressora de Tumor p53/metabolismo
5.
Cell Death Differ ; 20(5): 774-83, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23412343

RESUMO

Proper regulation of white and brown adipogenic differentiation is important for maintaining an organism's metabolic profile in a homeostatic state. The recent observations showing that the p53 tumor suppressor plays a role in metabolism raise the question of whether it is involved in the regulation of white and brown adipocyte differentiation. By using several in vitro models, representing various stages of white adipocyte differentiation, we found that p53 exerts a suppressive effect on white adipocyte differentiation in both mouse and human cells. Moreover, our in vivo analysis indicated that p53 is implicated in protection against diet-induced obesity. In striking contrast, our data shows that p53 exerts a positive regulatory effect on brown adipocyte differentiation. Abrogation of p53 function in skeletal muscle committed cells reduced their capacity to differentiate into brown adipocytes and histological analysis of brown adipose tissue revealed an impaired morphology in both embryonic and adult p53-null mice. Thus, depending on the specific adipogenic differentiation program, p53 may exert a positive or a negative effect. This cell type dependent regulation reflects an additional modality of p53 in maintaining a homeostatic state, not only in the cell, but also in the organism at large.


Assuntos
Adipócitos Marrons/metabolismo , Adipogenia , Tecido Adiposo Marrom/metabolismo , Obesidade/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Células 3T3 , Adipócitos Marrons/citologia , Tecido Adiposo Marrom/citologia , Tecido Adiposo Branco/citologia , Tecido Adiposo Branco/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Dieta , Metabolismo Energético , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
6.
Cell Death Differ ; 20(2): 312-20, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22996684

RESUMO

The process of somatic cell reprogramming is gaining increasing interest as reprogrammed cells are considered to hold a great therapeutic potential. However, with current technologies this process is relatively inefficient. Recent studies reported that inhibition of the p53 tumor suppressor profoundly facilitates reprogramming and attributed this effect to the ability of p53 to restrict proliferation and induce apoptosis. Given that mesenchymal-to-epithelial transition (MET) was recently shown to be necessary for reprogramming of fibroblasts, we investigated whether p53 counteracts reprogramming by affecting MET. We found that p53 restricts MET during the early phases of reprogramming and that this effect is primarily mediated by the ability of p53 to inhibit Klf4-dependent activation of epithelial genes. Moreover, transcriptome analysis revealed a large transcriptional signature enriched with epithelial genes, which is markedly induced by Klf4 exclusively in p53(-/-) cells. We also found that the expression of the epithelial marker E-Cadherin negatively correlates with p53 activity in a variety of mesenchymal cells even before the expression of reprogramming factors. Finally, we demonstrate that the inhibitory effect of p53 on MET is mediated by p21. We conclude that inhibition of the p53-p21 axis predisposes mesenchymal cells to the acquisition of epithelial characteristics and renders them more prone to reprogramming. Our study uncovers a novel mechanism by which p53 restrains reprogramming and highlights the role of p53 in regulating cell plasticity.


Assuntos
Transição Epitelial-Mesenquimal , Proteína Supressora de Tumor p53/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Células Cultivadas , Reprogramação Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
9.
Cancer Gene Ther ; 18(1): 2-11, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20966976

RESUMO

Three decades of p53 research have led to many advances in understanding the basic biology of normal and cancer cells. Nonetheless, the detailed functions of p53 in normal cells, and even more so in cancer cells, remain obscure. A major breakthrough is the realization that mutant p53 has a life of its own: it contributes to cancer not only through loss of activity, but also through gain of specific 'mutant functions'. This new focus on mutant p53 is the rationale behind the meeting series dedicated to advances on mutant p53 biology. This review provides an overview of results presented at the Fourth International Workshop on Mutant p53, held in Akko, Israel in March 2009. New roles and functions of p53 relevant for tumor suppressions were presented, including the regulation of microRNAs networks, the modulation of cell-stroma interactions and the induction of senescence. A main focus of the meeting was the rapidly growing body of knowledge on autonomous properties of mutant p53 and on their oncogenic 'gain of function' impact. Importantly, the meeting highlighted that, 30 years after p53 discovery, research on mutant p53 is entering the clinical and translational era. Two major steps forward in this respect are a better understanding of the active mechanism of small drugs targeting mutant p53 in tumor cells and an improved definition of the prognostic and predictive value of mutant p53 in human cancer.


Assuntos
Neoplasias/genética , Neoplasias/terapia , Proteína Supressora de Tumor p53/genética , Congressos como Assunto , Humanos , Israel
10.
Cell Death Differ ; 18(2): 271-81, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20689556

RESUMO

A mutation within one allele of the p53 tumor suppressor gene can inactivate the remaining wild-type allele in a dominant-negative manner and in some cases can exert an additional oncogenic activity, known as mutant p53 'gain of function' (GOF). To study the role of p53 mutations in prostate cancer and to discriminate between the dominant-negative effect and the GOF activity of mutant p53, we measured, using microarrays, the expression profiles of three immortalized prostate epithelial cultures expressing wild-type, inactivated p53 or mutated p53. Analysis of these gene expression profiles showed that both inactivated p53 and p53(R175H) mutant expression resulted in the upregulation of cell cycle progression genes. A second group, which was upregulated exclusively by mutant p53(R175H), was predominantly enriched in developmental genes. This group of genes included the Twist1, a regulator of metastasis and epithelial-mesenchymal transition (EMT). Twist1 levels were also elevated in metastatic prostate cancer-derived cell line DU145, in immortalized lung fibroblasts and in a subset of lung cancer samples, all in a mutant p53-dependent manner. p53(R175H) mutant bearing immortalized epithelial cells showed typical features of EMT, such as higher expression of mesenchymal markers, lower expression of epithelial markers and enhanced invasive properties in vitro. The mechanism by which p53(R175H) mutant induces Twist1 expression involves alleviation of the epigenetic repression. Our data suggest that Twist1 expression might be upregulated following p53 mutation in cancer cells.


Assuntos
Transição Epitelial-Mesenquimal , Proteínas Nucleares/metabolismo , Neoplasias da Próstata/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Substituição de Aminoácidos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Epigênese Genética , Histonas/metabolismo , Humanos , Masculino , Mutação , Proteínas Nucleares/genética , Complexo Repressor Polycomb 1 , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia , Proteína 1 Relacionada a Twist/genética , Regulação para Cima
11.
Oncogene ; 28(50): 4469-79, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-19855428

RESUMO

The Lats2 tumor suppressor protein has been implicated earlier in promoting p53 activation in response to mitotic apparatus stress, by preventing Mdm2-driven p53 degradation. We now report that Lats2 also has a role in an ATR-Chk1-mediated stress check point in response to oncogenic H-Ras. Activated mutant H-Ras triggers the translocation of Lats2 from centrosomes into the nucleus, coupled with an increase in Lats2 protein levels. This leads to the induction of p53 activity, upregulation of proapoptotic genes, downregulation of antiapoptotic genes and eventually apoptotic cell death. Many of the cells that survive apoptosis undergo senescence. However, a fraction of the cells escape this checkpoint mechanism, despite maintaining a high mutant H-Ras expression. These escapers display increased genome instability, as evidenced by a substantial fraction of cells with micronuclei and cells with polyploid genomes. Interestingly, such cells show markedly reduced levels of Lats2, in conjunction with enhanced hypermethylation of the Lats2 gene promoter. Our findings suggest that Lats2 might have an important role in quenching H-Ras-induced transformation, whereas silencing of Lats2 expression might serve as a mechanism to enable tumor progression.


Assuntos
Transformação Celular Neoplásica , Genes ras , Proteínas Serina-Treonina Quinases/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/fisiologia , Quinase 1 do Ponto de Checagem , Inativação Gênica , Humanos , Mutação , Proteínas Quinases/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/análise , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
12.
Oncogene ; 28(29): 2678-89, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19483728

RESUMO

These investigations demonstrate that expression of the inhibitor of apoptosis family member, survivin, is dramatically increased during immortalization of nontransformed human fibroblasts that were transduced with telomerase reverse transcriptase (hTERT). Expression of survivin in immortalized fibroblasts peaked during G(2)/M phase of the cell cycle. However, the upregulation of survivin was dissociated from the rate of proliferation and proportion of G(2)/M cells. Depletion of survivin from immortal fibroblasts increased sensitivity to stress-induced apoptosis and resulted in an accumulation of cells with 4N DNA content. Conversely, overexpression of survivin in mortal fibroblasts conferred resistance to apoptosis. In contrast, very low levels of survivin in proliferating parental fibroblasts had no bearing on sensitivity to apoptosis. The upregulation of survivin did not appear to be a direct consequence of hTERT transduction. However, repression of hTERT resulted in the rapid downregulation of survivin in telomerase-immortalized fibroblasts and tumor cell lines, but not in cells immortalized via an Alternative Lengthening of Telomeres mechanism. These results have important therapeutic implications, as telomerase and survivin are both broadly expressed in human cancers. Selection during the immortalization process for cells expressing high levels of survivin may account for the abundance of survivin in diverse tumor types.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Telomerase/metabolismo , Apoptose , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Fibroblastos/metabolismo , Humanos , Proteínas Inibidoras de Apoptose , Survivina , Telomerase/genética , Transdução Genética , Regulação para Cima
13.
Eur J Cancer ; 45(3): 443-53, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19008096

RESUMO

The p73 locus gene has a complex structure encoding a plethora of isoforms. The different DeltaN truncated isoforms of p73 may exert different activities depending on the cellular context. The beta isoform of DeltaNp73 seems to have a particular pattern of action even if its role in cell cycle and mitosis is still under investigation. To gain further knowledge of DeltaNp73beta's function, we investigated the effects of its over-expression in tumour cellular models, using the tetracycline-inducible expression system. In the human lung carcinoma cell line H1299, DeltaNp73beta over-expression resulted in suppression of cell growth and in cell death. Surprisingly stable over-expression of DeltaNp73beta impaired the genomic stability of tumour cells, leading to the formation of tetraploid cells. The cells become enlarged and multinucleate, with incorrect mitotic figures, and died by apoptotic-independent pathways. Our data suggest that DeltaNp73beta-induced aberrant mitosis evades the control of the mitotic spindle assay checkpoint, leading to tetraploidy and cell death through mitotic catastrophe rather than apoptosis. The various C-terminal regions of DeltaNp73 may influence the final cellular phenotype and we assume that the beta one in particular could be important in both cell growth control and regulation of mitosis.


Assuntos
Apoptose/genética , Proteínas de Ligação a DNA/genética , Mitose/genética , Proteínas Nucleares/genética , Poliploidia , Carcinoma de Pequenas Células do Pulmão/genética , Proteínas Supressoras de Tumor/genética , Apoptose/fisiologia , Ciclo Celular/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/genética , Humanos , Mitose/fisiologia , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Carcinoma de Pequenas Células do Pulmão/metabolismo , Proteínas Supressoras de Tumor/metabolismo
14.
Oncogene ; 28(6): 933-6, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19060923

RESUMO

The p53 tumor suppressor serves as a crucial barrier against cancer development. In tumor cells and their progenitors, p53 suppresses cancer in a cell-autonomous manner. However, p53 also possesses non-cell-autonomous activities. For example, p53 of stromal fibroblasts can modulate the spectrum of proteins secreted by these cells, rendering their microenvironment less supportive of the survival and spread of adjacent tumor cells. We now report that epithelial tumor cells can suppress p53 induction in neighboring fibroblasts, an effect reproducible by tumor cell-conditioned medium. The ability to suppress fibroblast p53 activation is acquired by epithelial cells in the course of neoplastic transformation. Specifically, stable transduction of immortalized epithelial cells by mutant H-Ras and p53-specific short inhibitory RNA endows them with the ability to quench fibroblast p53 induction. Importantly, human cancer-associated fibroblasts are more susceptible to this suppression than normal fibroblasts. These findings underscore a mechanism whereby epithelial cancer cells may overcome the non-cell-autonomous tumor suppressor function of p53 in stromal fibroblasts.


Assuntos
Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Transformação Celular Neoplásica , Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/metabolismo , Genes Supressores de Tumor , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , RNA Interferente Pequeno/metabolismo
15.
Oncogene ; 27(54): 6866-74, 2008 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18820708

RESUMO

Human protease-activated receptor 1 (hPar1) is a bona fide receptor of the hemostatic protease thrombin, and has a central function in tumor progression. Inactivation of the tumor suppressor gene p53 is one of the most common genomic alterations occurring in cancer. Here, we address the interrelations between p53 and hPar1 in cancer. We demonstrate an inverse correlation between hPar1 gene expression and wild-type (wt) p53 levels, and a direct correlation with levels of the mutant (mt) p53. Bioinformatic search revealed the presence of at least two p53 motifs in the hPar1 promoter. Indeed, temperature-sensitive (ts) p53 forms reduced hPar1 promoter activity on wt p53 expression. Ectopic introduction of the p53R175H mutant into cells lacking p53 caused a moderate two-fold induction of hPar1 promoter activity. Chromatin immunoprecipitation (ChIP) analyses confirmed a physical association between the p53 protein and hPar1 chromatin fragments. In parallel, PAR1 function is attenuated by p53, as shown by inhibition of pFAK levels and a Matrigel invasion assay. Ectopic reinforcement of hPar1 rescued the inhibition conferred by p53, confirming that p53 directly affects hPar1 expression and function. Altogether, we provide evidence for a direct binding between p53 and hPar1 chromatin, and assign hPar1 as a target of p53.


Assuntos
Genes p53 , Neoplasias/genética , Receptor PAR-1/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Cromatina/genética , Biologia Computacional , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Regiões Promotoras Genéticas , Neoplasias da Próstata/genética , Interferência de RNA , RNA Mensageiro/genética , Receptor PAR-1/genética , Supressão Genética
16.
Oncogene ; 26(52): 7302-12, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17533371

RESUMO

The tumor suppressor p53 is a key modulator of the cellular stress response, inducing cell-cycle arrest, apoptosis, senescence and cell differentiation. To evaluate further the molecular mechanism underlying p53 function, the transcriptional profiles of proliferating and senescent WI-38 cells, both wild-type p53 expressers and counterparts with an inactivated p53, were compared by DNA microarray analysis. In particular, the amyloid-beta precursor-like protein 1 (APLP1) is induced in senescent cells in a p53-dependent manner. APLP1 was confirmed to be a novel transcriptional target of p53 by in vivo and in vitro characterization of a p53 responsive element found in the first intron of the APLP1 gene locus. APLP1 knockdown experiments demonstrate that APLP1 is required for the proliferation of fibroblastic and epithelial cells. Moreover, depletion of APLP1 expression diminishes stress-induced apoptosis of neural cells, whereas ectopic APLP1 expression augments apoptosis. Based on these data, a mechanism is proposed whereby p53-dependent induction of APLP1 is involved in neural cell death, and which may exacerbate neuronal cell loss in some acute or chronic neurodegenerative disorders.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Perfilação da Expressão Gênica , Neuroblastoma/metabolismo , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Morte Celular , Proliferação de Células , Células Cultivadas , Senescência Celular/fisiologia , Imunoprecipitação da Cromatina , Fibroblastos/metabolismo , Humanos , Immunoblotting , Luciferases/metabolismo , Pulmão/embriologia , Pulmão/metabolismo , Neuroblastoma/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/genética
17.
Oncogene ; 26(15): 2202-11, 2007 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-17401429

RESUMO

In addition to the loss of wild-type p53 activity, a high percentage of tumor cells accumulate mutant p53 protein isoforms. Whereas the hallmark of the wild-type p53 is its tumor suppressor activities, tumor-associated mutant p53 proteins acquire novel functions enabling them to promote a large spectrum of cancer phenotypes. During the last years, it became clear that tumor-associated mutant p53 proteins are not only distinct from the wild-type p53, but they also represent a heterogeneous population of proteins with a variety of structure-function features. One of the major mechanisms underlying mutant p53 gain of function is the ability to regulate gene expression. Although a large number of specific target genes were identified, the molecular basis for this regulation is not fully elucidated. This review describes the present knowledge about the transcriptional activities of mutant p53 and the mechanisms that might underlie its target gene specificity.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Oncogenes , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Humanos , Mutação , Transcrição Gênica
18.
Cell Death Differ ; 13(12): 2140-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16729031

RESUMO

The tumor suppressor p53 plays a pivotal role in suppressing tumorigenesis by inducing genomic stability, cell cycle arrest or apoptosis. AIF is a mitochondrial protein, which, upon translocation to the nucleus, can participate in apoptosis, primarily in a caspase-independent contexts. We now report that AIF gene expression is subject to positive transcriptional regulation by p53. Interestingly, unlike most known p53 target genes, the AIF gene is regulated by basal levels of p53, and activation of p53 by genotoxic stress does not result in a substantial further increase in AIF expression. The AIF gene harbors a p53 responsive element, which is bound by p53 within cells. p53 drives efficient induction of large-scale DNA fragmentation, a hallmark of AIF activity. Importantly, caspase-independent death is compromised in cells lacking functional p53, in line with the known role of AIF in this process. Thus, in addition to its documented effects on caspase-dependent apoptosis, p53 may also sensitize cells to caspase-independent death through positive regulation of AIF expression. Moreover, in the absence of overt apoptotic signals, the constitutive induction of AIF by p53 may underpin a cytoprotective maintenance role, based on the role of AIF in ensuring proper mitochondrial function.


Assuntos
Fator de Indução de Apoptose/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Fator de Indução de Apoptose/genética , Caspases/genética , Caspases/metabolismo , Linhagem Celular Tumoral , Fragmentação do DNA , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Íntrons/genética , Íntrons/fisiologia , Mitocôndrias/fisiologia , Proteína Supressora de Tumor p53/genética , Regulação para Cima/genética , Regulação para Cima/fisiologia
19.
Oncogene ; 25(3): 359-69, 2006 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-16170349

RESUMO

Tumor-associated mutant forms of p53 can exert an antiapoptotic gain of function activity, which confers a selective advantage upon tumor cells harboring such mutations. We report that mutant p53 suppresses the expression of the MSP (MST-1/HGFL) gene, encoding the ligand of the receptor tyrosine kinase RON, implicated in a variety of cellular responses. Mutant p53 associates with the MSP gene promoter and represses its transcriptional activity, leading to a decrease in mRNA levels and a subsequent decrease in the levels of secreted MSP protein. Forced downregulation of MSP expression in H1299 cells, derived from a large-cell lung carcinoma, confers increased resistance against etoposide-induced cell death. These antiapoptotic consequences of MSP downregulation seemingly conflict with the well-documented ability of the RON receptor to promote cell survival and tumor progression when aberrantly hyperactive. Yet, they are consistent with the fact that reduced MSP expression was observed in many types of human cancer, including large-cell lung carcinoma. Thus, repression of MSP gene expression by mutant p53 may contribute to oncogenesis in a cell type-specific manner.


Assuntos
Apoptose/fisiologia , MAP Quinase Quinase Quinases/genética , Mutação , Serina Endopeptidases/genética , Proteína Supressora de Tumor p53/fisiologia , Sequência de Bases , Western Blotting , Linhagem Celular , Imunoprecipitação da Cromatina , Primers do DNA , Regulação para Baixo , Proteínas de Membrana , Proteína Supressora de Tumor p53/genética
20.
FEBS Lett ; 559(1-3): 152-8, 2004 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-14960324

RESUMO

While the stress-response-associated importance of the p53 tumor suppressor is well established, recent studies have also linked p53 with several basic parameters in the normal behavior of cells. Here, we present evidence that basal p53 expression in WI38 human embryonic lung fibroblasts restricts growth rate and mediates density-dependent inhibition of growth and the associated G1 phase arrest of the cell cycle by affecting the density-dependent regulation of p16/INK4a. Additionally, we show that prolonged culturing of hTert-immortalized WI38 cells leads to a loss of density-dependent growth inhibition that correlates with p27/KIP deregulation as well as the previously shown INK4a locus silencing, and to an onset of contact-induced, p53-dependent cell death.


Assuntos
Inibição de Contato , Fibroblastos/citologia , Proteína Supressora de Tumor p53/fisiologia , Técnicas de Cultura de Células , Proteínas de Ciclo Celular/fisiologia , Morte Celular , Divisão Celular , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p27 , Fase G1 , Inativação Gênica , Humanos , Proteínas Supressoras de Tumor/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA