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










Base de datos
Intervalo de año de publicación
1.
Cancer Sci ; 107(6): 734-45, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26998741

RESUMEN

The tumor suppressor p53 functions by inducing the transcription of a collection of target genes. We previously attempted to identify p53 target genes by microarray expression and ChIP-sequencing analyses. In this study, we describe a novel p53 target gene, FUCA1, which encodes a fucosidase. Although fucosidase, α-l-1 (FUCA1) has been reported to be a lysosomal protein, we detected it outside of lysosomes and observed that its activity is highest at physiological pH. As there is a reported association between fucosylation and tumorigenesis, we investigated the potential role of FUCA1 in cancer. We found that overexpression of FUCA1, but not a mutant defective in enzyme activity, suppressed the growth of cancer cells and induced cell death. Furthermore, we showed that FUCA1 reduced fucosylation and activation of epidermal growth factor receptor, and concomitantly suppressed epidermal growth factor signaling pathways. FUCA1 loss-of-function mutations are found in several cancers, its expression is reduced in cancers of the large intestine, and low FUCA1 expression is associated with poorer prognosis in several cancers. These results show that protein defucosylation mediated by FUCA1 is involved in tumor suppression.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo , alfa-L-Fucosidasa/genética , alfa-L-Fucosidasa/metabolismo , Muerte Celular , División Celular , Línea Celular , Supervivencia Celular , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fucosa/metabolismo , Humanos , Proteínas Mutantes/biosíntesis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neoplasias/enzimología , Neoplasias/genética , Transducción de Señal , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , alfa-L-Fucosidasa/biosíntesis
2.
Sci Rep ; 6: 19174, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26754925

RESUMEN

The transcription factors HSF1 and p53 both modulate the stress response, thereby protecting and facilitating the recovery of stressed cells, but both have the potential to promote tumor development. Here we show that a p53 target gene, IER5, encodes an activator of HSF1. IER5 forms a ternary complex with HSF1 and the phosphatase PP2A, and promotes the dephosphorylation of HSF1 at numbers of serine and threonine residues, generating a novel, hypo-phosphorylated active form of HSF1. IER5 is also transcriptionally upregulated in various cancers, although this upregulation is not always p53-dependent. The IER5 locus is associated with a so-called super enhancer, frequently associated with hyperactivated oncogenes in cancer cell lines. Enhanced expression of IER5 induces abnormal HSF1 activation in cancer cells and contributes to the proliferation of these cells under stressed conditions. These results reveal the existence of a novel IER5-mediated cancer regulation pathway that is responsible for the activation of HSF1 observed in various cancers.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas Nucleares/genética , Factores de Transcripción/metabolismo , Proliferación Celular , Daño del ADN , Elementos de Facilitación Genéticos , Expresión Génica , Regulación de la Expresión Génica , Factores de Transcripción del Choque Térmico , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Familia de Multigenes , Complejos Multiproteicos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Proteínas Nucleares/metabolismo , Fosforilación , Pronóstico , Regiones Promotoras Genéticas , Unión Proteica , Proteína Fosfatasa 2/metabolismo , Transducción de Señal , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Proc Natl Acad Sci U S A ; 111(52): 18691-6, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512506

RESUMEN

Communication between cancer cells and their microenvironment controls cancer progression. Although the tumor suppressor p53 functions in a cell-autonomous manner, it has also recently been shown to function in a non-cell-autonomous fashion. Although functional defects have been reported in p53 in stromal cells surrounding cancer, including mutations in the p53 gene and decreased p53 expression, the role of p53 in stromal cells during cancer progression remains unclear. We herein show that the expression of α-smooth muscle actin (α-SMA), a marker of cancer-associated fibroblasts (CAFs), was increased by the ablation of p53 in lung fibroblasts. CAFs enhanced the invasion and proliferation of lung cancer cells when cocultured with p53-depleted fibroblasts and required contact between cancer and stromal cells. A comprehensive analysis using a DNA chip revealed that tetraspanin 12 (TSPAN12), which belongs to the tetraspanin protein family, was derepressed by p53 knockdown. TSPAN12 knockdown in p53-depleted fibroblasts inhibited cancer cell proliferation and invasion elicited by coculturing with p53-depleted fibroblasts in vitro, and inhibited tumor growth in vivo. It also decreased CXC chemokine ligand 6 (CXCL6) secretion through the ß-catenin signaling pathway, suggesting that cancer cell contact with TSPAN12 in fibroblasts transduced ß-catenin signaling into fibroblasts, leading to the secretion of CXCL6 to efficiently promote invasion. These results suggest that stroma-derived p53 plays a pivotal role in epithelial cancer progression and that TSPAN12 and CXCL6 are potential targets for lung cancer therapy.


Asunto(s)
Fibroblastos/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Transducción de Señal , Tetraspaninas/metabolismo , Animales , Línea Celular Tumoral , Quimiocina CXCL6/genética , Quimiocina CXCL6/metabolismo , Fibroblastos/patología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Tetraspaninas/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(23): E2404-13, 2014 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-24912192

RESUMEN

The molecular mechanisms underlying the development of pancreatic neuroendocrine tumors (PanNETs) have not been well defined. We report here that the genomic region of the PHLDA3 gene undergoes loss of heterozygosity (LOH) at a remarkably high frequency in human PanNETs, and this genetic change is correlated with disease progression and poor prognosis. We also show that the PHLDA3 locus undergoes methylation in addition to LOH, suggesting that a two-hit inactivation of the PHLDA3 gene is required for PanNET development. We demonstrate that PHLDA3 represses Akt activity and Akt-regulated biological processes in pancreatic endocrine tissues, and that PHLDA3-deficient mice develop islet hyperplasia. In addition, we show that the tumor-suppressing pathway mediated by MEN1, a well-known tumor suppressor of PanNETs, is dependent on the pathway mediated by PHLDA3, and inactivation of PHLDA3 and MEN1 cooperatively contribute to PanNET development. Collectively, these results indicate the existence of a novel PHLDA3-mediated pathway of tumor suppression that is important in the development of PanNETs.


Asunto(s)
Genes Supresores de Tumor , Pérdida de Heterocigocidad , Tumores Neuroendocrinos/genética , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Animales , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Metilación de ADN , Humanos , Hiperplasia , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Masculino , Ratones , Ratones Noqueados , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pronóstico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
PLoS One ; 9(1): e86709, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466208

RESUMEN

The retinoblastoma susceptibility protein (pRB) is a phosphoprotein that regulates cell cycle progression at the G1/S transition. In quiescent and early G1 cells, pRB predominantly exists in the active hypophosphorylated form. The cyclin/cyclin-dependent protein kinase complexes phosphorylate pRB at the late G1 phase to inactivate pRB. This event leads to the dissociation and activation of E2F family transcriptional factors. At least 12 serine/threonine residues in pRB are phosphorylated in vivo. Although there have been many reports describing bulk phosphorylation of pRB, detail research describing the function of each phosphorylation site remains unknown. Besides its G1/S inhibitory function, pRB is involved in differentiation, prevention of cell death and control of tissue fate. To uncover the function of phosphorylation of pRB in various cellular conditions, we have been investigating phosphorylation of each serine/threonine residue in pRB with site-specific phospho-serine/threonine antibodies. Here we demonstrate that pRB is specifically phosphorylated at Ser612 in differentiated cells in a known kinase-independent manner. We also found that pRB phosphorylated at Ser612 still associates with E2F-1 and tightly binds to nuclear structures including chromatin. Moreover, expression of the Ser612Ala mutant pRB failed to induce differentiation. The findings suggest that phosphorylation of Ser612 provides a distinct function that differs from the function of phosphorylation of other serine/threonine residues in pRB.


Asunto(s)
Diferenciación Celular/fisiología , Fosforilación/fisiología , Proteína de Retinoblastoma/metabolismo , Serina/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor de Transcripción E2F1/metabolismo , Humanos , Células K562 , Proteínas Nucleares/metabolismo , Treonina/metabolismo , Células U937
7.
Genes Cells ; 19(2): 89-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24350565

RESUMEN

Retinoblastoma protein (pRB) controls cell cycle progression and cell cycle exit through interactions with cellular proteins. Many pRB-binding proteins, which function in gene transcription or modulation of chromatin structure, harbor LXCXE motifs in their binding domain for pRB. In this study, we found that nuclear mitotic apparatus protein (NuMA), a mitotic spindle organizer, interacts with pRB through LSCEE sequences located in its C-terminal region. siRNA-mediated down-regulation of pRB caused aberrant distribution of NuMA and alignment of spindle microtubules in mitotic cells. Abnormal organization of spindle microtubules was also accompanied by misalignment of an over-expressed NuMA mutant (mut-NuMA) with a defect in pRB binding caused by an LSGEK mutation. The mut-NuMA-over-expressing cells showed lower potency for survival than wild-type NuMA (wt-NuMA)-over-expressing cells during 2 weeks of culture. Interestingly, knockdown of pRB reduced the population of wt-NuMA-over-expressing cells to the same level as mut-NuMA cells after 2 weeks. Taken together, pRB may have a novel function in regulating the mitotic function of NuMA and spindle organization, which are required for proper cell cycle progression.


Asunto(s)
Antígenos Nucleares/metabolismo , Microtúbulos/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteína de Retinoblastoma/metabolismo , Huso Acromático/fisiología , Antígenos Nucleares/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo , Humanos , Microtúbulos/ultraestructura , Mitosis , Proteínas Asociadas a Matriz Nuclear/genética , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética , Proteína de Retinoblastoma/genética , Huso Acromático/ultraestructura
8.
Elife ; 2: e01228, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24302570

RESUMEN

Skeletal muscle degeneration is a complication arising from a variety of chronic diseases including advanced cancer. Pro-inflammatory cytokine TNF-α plays a pivotal role in mediating cancer-related skeletal muscle degeneration. Here, we show a novel function for retinoblastoma protein (Rb), where Rb causes sarcomeric disorganization. In human skeletal muscle myotubes (HSMMs), up-regulation of cyclin-dependent kinase 4 (CDK4) and concomitant phosphorylation of Rb was induced by TNF-α treatment, resulting in the translocation of phosphorylated Rb to the cytoplasm. Moreover, induced expression of the nuclear exporting signal (NES)-fused form of Rb caused disruption of sarcomeric organization. We identified mammalian diaphanous-related formin 1 (mDia1), a potent actin nucleation factor, as a binding partner of cytoplasmic Rb and found that mDia1 helps maintain the structural integrity of the sarcomere. These results reveal a novel non-nuclear function for Rb and suggest a potential mechanism of TNF-α-induced disruption of sarcomeric organization. DOI: http://dx.doi.org/10.7554/eLife.01228.001.


Asunto(s)
Citoplasma/metabolismo , Proteína de Retinoblastoma/metabolismo , Sarcómeros , Humanos , Músculo Esquelético/metabolismo , Fosforilación , Transporte de Proteínas , Factor de Necrosis Tumoral alfa/farmacología
9.
Mol Cell Biol ; 33(12): 2447-57, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23589328

RESUMEN

The p53 tumor suppressor protein is a transcription factor controlling various outcomes, such as growth arrest and apoptosis, through the regulation of different sets of target genes. The nuclear mitotic apparatus protein (NuMA) plays important roles in spindle pole organization during mitosis and in chromatin regulation in the nucleus during interphase. Although NuMA has been shown to colocalize with several nuclear proteins, including high-mobility-group proteins I and Y and GAS41, the role of NuMA during interphase remains unclear. Here we report that NuMA binds to p53 to modulate p53-mediated transcription. Acute and partial ablation of NuMA attenuates the induction of the proarrested p21 gene following DNA damage, subsequently causing impaired cell cycle arrest. Interestingly, NuMA knockdown had little effect on the induction of the p53-dependent proapoptotic PUMA gene. Furthermore, NuMA is required for the recruitment of cyclin-dependent kinase 8 (Cdk8), a component of the Mediator complex and a promoter of p53-mediated p21 gene function. These data demonstrate that NuMA is critical for the target selectivity of p53-mediated transcription.


Asunto(s)
Antígenos Nucleares/metabolismo , Quinasa 8 Dependiente de Ciclina/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antígenos Nucleares/genética , Neoplasias de la Mama/metabolismo , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Femenino , Fibrosarcoma/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Proteínas Asociadas a Matriz Nuclear/genética , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Transcripción Genética
10.
Proc Natl Acad Sci U S A ; 110(10): 3895-900, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23431171

RESUMEN

Monocytic leukemia zinc finger (MOZ)/KAT6A is a MOZ, Ybf2/Sas3, Sas2, Tip60 (MYST)-type histone acetyltransferase that functions as a coactivator for acute myeloid leukemia 1 protein (AML1)- and Ets family transcription factor PU.1-dependent transcription. We previously reported that MOZ directly interacts with p53 and is essential for p53-dependent selective regulation of p21 expression. We show here that MOZ is an acetyltransferase of p53 at K120 and K382 and colocalizes with p53 in promyelocytic leukemia (PML) nuclear bodies following cellular stress. The MOZ-PML-p53 interaction enhances MOZ-mediated acetylation of p53, and this ternary complex enhances p53-dependent p21 expression. Moreover, we identified an Akt/protein kinase B recognition sequence in the PML-binding domain of MOZ protein. Akt-mediated phosphorylation of MOZ at T369 has a negative effect on complex formation between PML and MOZ. As a result of PML-mediated suppression of Akt, the increased PML-MOZ interaction enhances p21 expression and induces p53-dependent premature senescence upon forced PML expression. Our research demonstrates that MOZ controls p53 acetylation and transcriptional activity via association with PML.


Asunto(s)
Histona Acetiltransferasas/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Secuencia de Bases , Células Cultivadas , Senescencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Técnicas de Inactivación de Genes , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/química , Histona Acetiltransferasas/deficiencia , Histona Acetiltransferasas/genética , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Leucemia Promielocítica Aguda/genética , Ratones , Modelos Biológicos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteína p53 Supresora de Tumor/química , Factores de Transcripción p300-CBP/metabolismo
11.
J Biol Chem ; 287(11): 7896-906, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22262832

RESUMEN

We previously showed that depletion of the retinoblastoma protein (RB) induces down-regulation of the adhesion molecule E-cadherin and thereby triggers the epithelial-mesenchymal transition. To further characterize the effect of RB inactivation on the phenotype of cancer cells, we have now examined RB expression in human breast cancer cell lines and clinical specimens. We found that RB-inactive cells exhibit a mesenchymal-like morphology and are highly invasive. We also found that ZEB proteins, transcriptional repressors of the E-cadherin gene, are markedly up-regulated in these cells in a manner sensitive to the miR-200 family of microRNAs. Moreover, depletion of ZEB in RB-inactive cells suppressed cell invasiveness and proliferation and induced epithelial marker expression. These results implicate ZEB in induction of the epithelial-mesenchymal transition, as well as in maintenance of the mesenchymal phenotype in RB-inactive cells. We also developed a screening program for inhibitors of ZEB1 expression and thereby identified several cyclin-dependent kinase inhibitors that blocked both ZEB1 expression and RB phosphorylation. Together, our findings suggest that RB inactivation contributes to tumor progression not only through loss of cell cycle control but also through up-regulation of ZEB expression and induction of an invasive phenotype.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/biosíntesis , Proteína de Retinoblastoma/biosíntesis , Factores de Transcripción/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cadherinas/biosíntesis , Cadherinas/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Proteínas de Homeodominio/genética , Humanos , MicroARNs/biosíntesis , MicroARNs/genética , Invasividad Neoplásica , Inhibidores de Proteínas Quinasas/farmacología , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Proteína de Retinoblastoma/genética , Factores de Transcripción/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
12.
J Biol Chem ; 286(50): 43013-25, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-22002055

RESUMEN

Genetic ablations of p73 have shown its implication in the development of the nervous system. However, the relative contribution of ΔNp73 and TAp73 isoforms in neuronal functions is still unclear. In this study, we have analyzed the expression of these isoforms during neuronal death induced by alteration of the amyloid-ß precursor protein function or cisplatin. We observed a concomitant up-regulation of a TAp73 isoform and a down-regulation of a ΔNp73 isoform. The shift in favor of the pro-apoptotic isoform correlated with an induction of the p53/p73 target genes such as Noxa. At a functional level, we showed that TAp73 induced neuronal death and that ΔNp73 has a neuroprotective role toward amyloid-ß precursor protein alteration or cisplatin. We investigated the mechanisms of p73 expression and found that the TAp73 expression was regulated at the promoter level. In contrast, regulation of ΔNp73 protein levels was regulated by phosphorylation at residue 86 and multiple proteases. Thus, this study indicates that tight transcriptional and post-translational mechanisms regulate the p73 isoform ratios that play an important role in neuronal survival.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Daño del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/genética , Apoptosis/fisiología , Células Cultivadas , Inmunoprecipitación de Cromatina , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Immunoblotting , Ratones , Neuronas/citología , Proteínas Nucleares/genética , Fosforilación , Isoformas de Proteínas/genética , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/genética
13.
J Biol Chem ; 286(20): 18251-60, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454683

RESUMEN

The common polymorphism of p53 at codon 72, either encoding proline or arginine, has drawn attention as a genetic factor associated with clinical outcome or cancer risk for the last 2 decades. We now show that these two polymorphic variants differ in protein structure, especially within the N-terminal region and, as a consequence, differ in post-translational modification at the N terminus. The arginine form (p53-72R) shows significantly enhanced phosphorylation at Ser-6 and Ser-20 compared with the proline form (p53-72P). We also show diminished Mdm2-mediated degradation of p53-72R compared with p53-72P, which is at least partly brought about by higher levels of phosphorylation at Ser-20 in p53-72R. Furthermore, enhanced p21 expression in p53-72R-expressing cells, which is dependent on phosphorylation at Ser-6, was demonstrated. Differential p21 expression between the variants was also observed upon activation of TGF-ß signaling. Collectively, we demonstrate a novel molecular difference and simultaneously suggest a difference in the tumor-suppressing function of the variants.


Asunto(s)
Codón , Predisposición Genética a la Enfermedad , Neoplasias , Polimorfismo Genético , Procesamiento Proteico-Postraduccional , Proteína p53 Supresora de Tumor , Línea Celular , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Mol Cell Biol ; 30(7): 1620-33, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20123963

RESUMEN

p53 phosphorylation at Ser46 following DNA damage is important for preferential transactivation of proapoptotic genes. Here, we report that ataxia-telangiectasia mutated (ATM) kinase is responsible for Ser46 phosphorylation of p53 during early-phase response to DNA damage. To elucidate the direct phosphorylation of p53 at Ser46 by ATM, an ATM mutant (ATM-AS) sensitive to ATP analogues was engineered. In vitro kinase assays revealed that p53 was phosphorylated at Ser46 by ATM-AS, even when ATP analogues were used as phosphate donors, although this phosphorylation site is not in an SQ motif, a consensus ATM site. Furthermore, Ser46 phosphorylation by ATM was dependent on the N- and C-terminal domains of p53, unlike Ser15 phosphorylation. Immunofluorescence analyses showed that Ser46-phosphorylated p53 was observed as foci in response to DNA damage and colocalized with gamma-H2AX or Ser1981-phosphorylated ATM. These results suggest that ATM phosphorylates a noncanonical serine residue on p53 by mechanisms different from those for the phosphorylation of Ser15.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Glutamina/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Serina , Treonina/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adenosina Trifosfato/análogos & derivados , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Secuencia de Bases , Proteínas de Ciclo Celular/genética , Línea Celular , ADN/genética , ADN/metabolismo , ADN/efectos de la radiación , Daño del ADN , Proteínas de Unión al ADN/genética , Glutamina/metabolismo , Humanos , Estructura Molecular , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Serina/genética , Serina/metabolismo , Treonina/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética
15.
J Mol Biol ; 394(3): 460-71, 2009 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19766654

RESUMEN

The p53 pathway is activated in response to various cellular stresses to protect cells from malignant transformation. We have previously shown that clathrin heavy chain (CHC), which is a cytosolic protein regulating endocytosis, is present in nuclei and binds to p53 to promote p53-mediated transcription. However, details of the binding interface between p53 and CHC remain unclear. Here, we report on the binding mode between p53 and CHC using mutation analyses and a structural model of the interaction generated by molecular dynamics. Structural modeling analyses predict that an Asn1288 residue in CHC is crucial for binding to p53. In fact, substitution of this Asn to Ala of CHC diminished its ability to interact with p53, leading to reduced activity to transactivate p53. Surprisingly, this mutation had little effect on receptor-mediated endocytosis. Thus, the function-specific mutation of CHC will clarify physiological roles of CHC in the regulation of the p53 pathway.


Asunto(s)
Cadenas Pesadas de Clatrina/química , Cadenas Pesadas de Clatrina/genética , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regiones no Traducidas 3' , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Asparagina/química , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Cadenas Pesadas de Clatrina/metabolismo , Secuencia Conservada , Endocitosis , Células HeLa , Humanos , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Dominios y Motivos de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Transferrina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Termodinámica , Proteína p53 Supresora de Tumor/química
16.
FEBS Lett ; 583(17): 2710-4, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19619542

RESUMEN

mdm2 and mdmx oncogenes play essential yet non-redundant roles in synergistic inactivation of the tumor suppressor, p53. While Mdm2 inhibits p53 activity mainly by augmenting its ubiquitination, the functional role of Mdmx on p53 ubiquitination remains obscure. In transfected H1299 cells, Mdmx augmented Mdm2-mediated ubiquitination of p53. In in vitro ubiquitination assays, the Mdmx/Mdm2 heteromeric complex, in comparison to the Mdm2 homomer, showed enhanced ubiquitinase activity toward p53 and the reduced auto-ubiquitination of Mdm2. Alteration of the substrate specificity via binding to Mdmx may contribute to efficient ubiquitination and inactivation of p53 by Mdm2.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Humanos , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Especificidad por Sustrato , Proteína p53 Supresora de Tumor/genética , Ubiquitinación
17.
Cancer Sci ; 100(7): 1291-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19432880

RESUMEN

The mdm2 and mdmx oncogenes play essential yet nonredundant roles in synergistic inactivatiosn of p53. However, the biochemical mechanism by which Mdmx synergizes with Mdm2 to inhibit p53 function remains obscure. Here we demonstrate that, using nonphosphorylatable mutants of Mdmx, the cooperative inhibition of p53 by Mdmx and Mdm2 was associated with cytoplasmic localization of p53, and with an increase of the interaction of Mdmx to p53 and Mdm2 in the cytoplasm. In addition, the Mdmx mutant cooperates with Mdm2 to induce ubiquitination of p53 at C-terminal lysine residues, and the integrity of the C-terminal lysines was partly required for the cooperative inhibition. The expression of subcellular localization mutants of Mdmx revealed that subcellular localization of Mdmx dictated p53 localization, and that cytoplasmic Mdmx tethered p53 in the cytoplasm and efficiently inhibited p53 activity. RNAi-mediated inhibition of Mdmx or introduction of the nuclear localization mutant of Mdmx reduced cytoplasmic retention of p53 in neuroblastoma cells, in which cytoplasmic sequestration of p53 is involved in its inactivation. Our data indicate that cytoplasmic tethering of p53 mediated by Mdmx contributes to p53 inactivation in some types of cancer cells.


Asunto(s)
Citoplasma/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Secuencia de Aminoácidos , Línea Celular Tumoral , Humanos , Datos de Secuencia Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
Cell ; 136(3): 535-50, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19203586

RESUMEN

p53 And Akt are critical players regulating tumorigenesis with opposite effects: whereas p53 transactivates target genes to exert its function as a tumor suppressor, Akt phosphorylates its substrates and transduces downstream survival signals. In addition, p53 and Akt negatively regulate each other to balance survival and death signals within a cell. We now identify PHLDA3 as a p53 target gene that encodes a PH domain-only protein. We find that PHLDA3 competes with the PH domain of Akt for binding of membrane lipids, thereby inhibiting Akt translocation to the cellular membrane and activation. Ablation of endogenous PHLDA3 results in enhanced Akt activity and decrease of p53-dependent apoptosis. We also demonstrate the suppression of anchorage-independent cell growth by PHLDA3. Loss of the PHLDA3 genomic locus was frequently observed in primary lung cancers, suggesting a role of PHLDA3 in tumor suppression. Our results reveal a new mode of coordination between the p53 and Akt pathways.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Proteínas Nucleares/metabolismo , Proteína Oncogénica v-akt/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Transducción de Señal
19.
J Biol Chem ; 284(1): 237-244, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19001415

RESUMEN

Upon DNA damage, p53 can induce either cell-cycle arrest or apoptosis. Here we show that monocytic leukemia zinc finger (MOZ) forms a complex with p53 to induce p21 expression and cell-cycle arrest. The levels of the p53-MOZ complex increased in response to DNA damage to levels that induce cell-cycle arrest. MOZ(-/-) mouse embryonic fibroblasts failed to arrest in G1 in response to DNA damage, and DNA damage-induced expression of p21 was impaired in MOZ(-/-) cells. These results suggest that MOZ is involved in regulating cell-cycle arrest in the G1 phase. Screening of tumor-associated p53 mutants demonstrated that the G279E mutation in p53 disrupts interactions between p53 and MOZ, but does not affect the DNA binding activity of p53. The leukemia-associated MOZ-CBP fusion protein inhibits p53-mediated transcription. These results suggest that inhibition of p53/MOZ-mediated transcription is involved in tumor pathogenesis and leukemogenesis.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Fase G1/fisiología , Regulación de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Histona Acetiltransferasas/genética , Humanos , Ratones , Ratones Noqueados , Unión Proteica/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína p53 Supresora de Tumor/genética
20.
Cancer Res ; 68(13): 5104-12, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18593909

RESUMEN

The retinoblastoma tumor suppressor protein (Rb) is mutated or expressed at very low levels in several tumor types, including retinoblastoma and osteosarcoma, as well as small cell lung, colon, prostate, bladder, and breast carcinomas. Loss or reduction of Rb expression is seen most commonly in high-grade breast adenocarcinomas, suggesting that a relationship may exist between loss of Rb function and a less-differentiated state, increased proliferation, and high metastatic potential. In this study, we found that knockdown of Rb by small interfering RNA in MCF7 breast cancer cells disrupts cell-cell adhesion and induces a mesenchymal-like phenotype. The epithelial-to-mesenchymal transition (EMT), a key event in embryonic morphogenesis, is implicated in the metastasis of primary tumors. Additionally, Rb is decreased during growth factor- and cytokine-induced EMT and overexpression of Rb inhibits the EMT in MCF10A human mammary epithelial cells. Ectopic expression and knockdown of Rb resulted in increased or reduced expression of E-cadherin, which is specifically involved in epithelial cell-cell adhesion. Other EMT-related transcriptional factors, including Slug and Zeb-1, are also induced by Rb depletion. Furthermore, we confirmed that Rb binds to an E-cadherin promoter sequence in association with the transcription factor activator protein-2alpha. Finally, in breast cancer specimens, we observed a concurrent down-regulation of Rb and E-cadherin expression in mesenchymal-like invasive cancers. These findings suggest that Rb inactivation contributes to tumor progression due to not only loss of cell proliferation control but also conversion to an invasive phenotype and that the inhibition of EMT is a novel tumor suppressor function of Rb.


Asunto(s)
Cadherinas/genética , Células Epiteliales/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Mesodermo/fisiología , ARN Interferente Pequeño/farmacología , Proteína de Retinoblastoma/antagonistas & inhibidores , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Progresión de la Enfermedad , Desarrollo Embrionario/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , Invasividad Neoplásica , Fenotipo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Transfección , Factor de Crecimiento Transformador beta/farmacología , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Proteínas de Unión al GTP rho/metabolismo , Proteínas de Unión al GTP rho/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...