Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 15850, 2018 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-30374061

RESUMEN

A correction has been published and is appended to both the HTML and PDF versions of this paper. The error has not been fixed in the paper.

2.
Sci Rep ; 7: 46017, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28387346

RESUMEN

FoxM1b is a cell cycle-regulated transcription factor, whose over-expression is a marker for poor outcome in cancers. Its transcriptional activation function requires phosphorylation by Cdk1 or Cdk2 that primes FoxM1b for phosphorylation by Plk1, which triggers association with the co-activator CBP. FoxM1b also possesses transcriptional repression function. It represses the mammary differentiation gene GATA3 involving DNMT3b and Rb. We investigated what determines the two distinct functions of FoxM1b: activation and repression. We show that Rb binds to the C-terminal activation domain of FoxM1b. Analyses with phospho-defective and phospho-mimetic mutants of FoxM1b identified a critical role of the Plk1 phosphorylation sites in regulating the binding of FoxM1b to Rb and DNMT3b. That is opposite of what was seen for the interaction of FoxM1b with CBP. We show that, in addition to GATA3, FoxM1b also represses the mammary luminal differentiation marker FoxA1 by promoter-methylation, and that is regulated by the Plk1 phosphorylation sites in FoxM1b. Our results show that the Plk1 phosphorylation sites in FoxM1b serve as a regulator for its repressor function, and they provide insights into how FoxM1b inhibits differentiation genes and activates proliferation genes during cancer progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteína Forkhead Box M1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/metabolismo , Sitios de Unión , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Proteína Forkhead Box M1/química , Factor de Transcripción GATA3/genética , Humanos , Células MCF-7 , Mutación/genética , Fragmentos de Péptidos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos , Sialoglicoproteínas/metabolismo , ADN Metiltransferasa 3B , Quinasa Tipo Polo 1
3.
J Hepatol ; 63(2): 429-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25828473

RESUMEN

BACKGROUND & AIMS: Overexpression of FoxM1 correlates with poor prognosis in hepatocellular carcinoma (HCC). Moreover, the Ras-signaling pathway is found to be ubiquitously activated in HCC through epigenetic silencing of the Ras-regulators. We investigated the roles of FoxM1 in Ras-driven HCC, and on HCC cells with stem-like features. METHODS: We employed a transgenic mouse model that expresses the oncogenic Ras in the liver. That strain was crossed with a strain that harbor floxed alleles of FoxM1 and the MxCre gene that allows conditional deletion of FoxM1. FoxM1 alleles were deleted after development of HCC, and the effects on the tumors were analyzed. Also, FoxM1 siRNA was used in human HCC cell lines to determine its role in the survival of the HCC cells with stem cell features. RESULTS: Ras-driven tumors overexpress FoxM1. Deletion of FoxM1 inhibits HCC progression. There was increased accumulation of reactive oxygen species (ROS) in the FoxM1 deleted HCC cells. Moreover, FoxM1 deletion caused a disproportionate loss of the CD44+ and EpCAM+ HCC cells in the tumors. We show that FoxM1 directly activates expression of CD44 in human HCC cells. Moreover, the human HCC cells with stem cell features are addicted to FoxM1 for ROS-regulation and survival. CONCLUSION: Our results provide genetic evidence for an essential role of FoxM1 in the progression of Ras-driven HCC. In addition, FoxM1 is required for the expression of CD44 in HCC cells. Moreover, FoxM1 plays a critical role in the survival of the HCC cells with stem cell features by regulating ROS.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Células Madre/patología , Proteínas ras/genética , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/biosíntesis , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pronóstico , Transducción de Señal , Células Madre/metabolismo , Proteínas ras/biosíntesis
4.
Mol Cancer Ther ; 12(5): 759-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23427295

RESUMEN

The forkhead box transcription factor FOXM1 is considered to be a promising target for cancer therapy. However, the significance of FOXM1 in tumors harboring mutation in p53, which is very common, is unclear. In this study, we investigated the efficacy of FoxM1 targeting in spontaneous p53-null tumors using genetic ablation as well as using a peptide inhibitor of FOXM1. We show that conditional deletion of FoxM1 inhibits growth of the p53-null thymic lymphoma and sarcoma cells. In addition, deletion of FoxM1 induces apoptotic cell death of the p53-null tumors, accompanied by reduced expression of the FOXM1 target genes survivin and Bmi1. An ARF-derived peptide that inhibits the activity of FOXM1, by targeting it to the nucleolus, also induces apoptosis in the p53-null sarcoma and lymphoma, leading to a strong inhibition of their metastatic colonization. Together, our observations suggest that FOXM1 is critical for survival and growth of the p53-null lymphoma and sarcoma and provide proof-of-principle that FOXM1 is an effective therapeutic target for sarcoma and lymphoma carrying loss of function mutation in p53.


Asunto(s)
Factores de Transcripción Forkhead/antagonistas & inhibidores , Linfoma/genética , Linfoma/metabolismo , Sarcoma/genética , Sarcoma/metabolismo , Proteína p53 Supresora de Tumor/genética , Aloinjertos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/química , Modelos Animales de Enfermedad , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones , Ratones Noqueados , Péptidos/farmacología , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Survivin , Neoplasias del Timo/genética , Neoplasias del Timo/metabolismo
5.
Cell Rep ; 1(6): 715-29, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22813746

RESUMEN

Elevated expression of FoxM1 in breast cancer correlates with an undifferentiated tumor phenotype and a negative clinical outcome. However, a role for FoxM1 in regulating mammary differentiation was not known. Here, we identify another function of FoxM1, the ability to act as a transcriptional repressor, which plays an important role in regulating the differentiation of luminal epithelial progenitors. Regeneration of mammary glands with elevated levels of FoxM1 leads to aberrant ductal morphology and expansion of the luminal progenitor pool. Conversely, knockdown of FoxM1 results in a shift toward the differentiated state. FoxM1 mediates these effects by repressing the key regulator of luminal differentiation, GATA-3. Through association with DNMT3b, FoxM1 promotes methylation of the GATA-3 promoter in an Rb-dependent manner. This study identifies FoxM1 as a critical regulator of mammary differentiation with significant implications for the development of aggressive breast cancers.


Asunto(s)
Linaje de la Célula , Factores de Transcripción Forkhead/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , ADN (Citosina-5-)-Metiltransferasas , Metilación de ADN/genética , Femenino , Proteína Forkhead Box M1 , Factor de Transcripción GATA3/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Embarazo , Regiones Promotoras Genéticas/genética , Proteína de Retinoblastoma/metabolismo , Transcripción Genética , ADN Metiltransferasa 3B
6.
Cancer Res ; 71(12): 4292-302, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21507930

RESUMEN

Malignant neuroblastomas contain stem-like cells. These tumors also overexpress the Forkhead box transcription factor FoxM1. In this study, we investigated the roles of FoxM1 in the tumorigenicity of neuroblastoma. We showed that depletion of FoxM1 inhibits anchorage-independent growth and tumorigenicity in mouse xenografts. Moreover, knockdown of FoxM1 induces differentiation in neuroblastoma cells, suggesting that FoxM1 plays a role in the maintenance of the undifferentiated progenitor population. We showed that inhibition of FoxM1 in malignant neuroblastoma cells leads to the downregulation of the pluripotency genes sex determining region Y box 2 (Sox2) and Bmi1. We provided evidence that FoxM1 directly activates expression of Sox2 in neuroblastoma cells. By using a conditional deletion system and neurosphere cultures, we showed that FoxM1 is important for expression of Sox2 and Bmi1 in the mouse neural stem/progenitor cells and is critical for its self-renewal. Together, our observations suggested that FoxM1 plays an important role in the tumorigenicity of the aggressive neuroblastoma cells through maintenance of the undifferentiated state.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Células-Madre Neurales/fisiología , Neuroblastoma/etiología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proteína Forkhead Box M1 , Humanos , Ratones , Neuroblastoma/patología , Regiones Promotoras Genéticas , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología
7.
EMBO Mol Med ; 3(1): 21-34, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21204266

RESUMEN

The forkhead box M1b (FoxM1b) transcription factor is over-expressed in human cancers, and its expression often correlates with poor prognosis. Previously, using conditional knockout strains, we showed that FoxM1b is essential for hepatocellular carcinoma (HCC) development. However, over-expression of FoxM1b had only marginal effects on HCC progression. Here we investigated the effect of FoxM1b expression in the absence of its inhibitor Arf. We show that transgenic expression of FoxM1b in an Arf-null background drives hepatic fibrosis and metastasis of HCC. We identify novel mechanisms of FoxM1b that are involved in epithelial-mesenchymal transition, cell motility, invasion and a pre-metastatic niche formation. FoxM1b activates the Akt-Snail1 pathway and stimulates expression of Stathmin, lysyl oxidase, lysyl oxidase like-2 and several other genes involved in metastasis. Furthermore, we show that an Arf-derived peptide, which inhibits FoxM1b, impedes metastasis of the FoxM1b-expressing HCC cells. The observations indicate that FoxM1b is a potent activator of tumour metastasis and that the Arf-mediated inhibition of FoxM1b is a critical mechanism for suppression of tumour metastasis.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Metástasis de la Neoplasia , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/secundario , Transición Epitelial-Mesenquimal , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/genética , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Transgénicos , Proteína-Lisina 6-Oxidasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail , Estatmina/metabolismo , Factores de Transcripción/metabolismo
8.
Cancer Res ; 70(12): 5054-63, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20530690

RESUMEN

Inherent and acquired therapeutic resistance in breast cancer remains a major clinical challenge. In human breast cancer samples, overexpression of the oncogenic transcription factor FoxM1 has been suggested to be a marker of poor prognosis. In this study, we report that FoxM1 overexpression confers resistance to the human epidermal growth factor receptor 2 monoclonal antibody Herceptin and microtubule-stabilizing drug paclitaxel, both as single agents and in combination. FoxM1 altered microtubule dynamics to protect tumor cells from paclitaxel-induced apoptosis. Mechanistic investigations revealed that the tubulin-destabilizing protein Stathmin, whose expression also confers resistance to paclitaxel, is a direct transcriptional target of FoxM1. Significantly, attenuating FoxM1 expression by small interfering RNA or an alternate reading frame (ARF)-derived peptide inhibitor increased therapeutic sensitivity. Our findings indicate that targeting FoxM1 could relieve therapeutic resistance in breast cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Factores de Transcripción Forkhead/metabolismo , Paclitaxel/uso terapéutico , Anticuerpos Monoclonales Humanizados , Apoptosis , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Citometría de Flujo , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Fragmentos de Péptidos/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trastuzumab , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
9.
PLoS One ; 4(9): e6891, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19727397

RESUMEN

Oxygen is a key modulator of many cellular pathways, but current devices permitting in vitro oxygen modulation fail to meet the needs of biomedical research. A microfabricated insert for multiwell plates has been developed to more effectively control the temporal and spatial oxygen concentration to better model physiological phenomena found in vivo. The platform consists of a polydimethylsiloxane insert that nests into a standard multiwell plate and serves as a passive microfluidic gas network with a gas-permeable membrane aimed to modulate oxygen delivery to adherent cells. Equilibration time is on the order of minutes and a wide variety of oxygen profiles can be attained based on the device design, such as the cyclic profile achieved in this study, and even oxygen gradients to mimic those found in vivo. The proper biological consequences of the device's oxygen delivery were confirmed in cellular models via a proliferation assay and western analysis of the upregulation of hypoxia inducible transcription factor-1alpha. These experiments serve as a demonstration for the platform as a viable tool to increase experimental throughput and permit novel experimental possibilities in any biomedical research lab.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Oxígeno/metabolismo , Técnicas Biosensibles , Adhesión Celular , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Proliferación Celular , Dimetilpolisiloxanos/química , Diseño de Equipo , Gases , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Microfluídica , Modelos Biológicos , Factores de Tiempo
10.
EMBO J ; 28(19): 2908-18, 2009 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-19696738

RESUMEN

The transcription factor FoxM1 is over-expressed in most human malignancies. Although it is evident that FoxM1 has critical functions in tumour development and progression, the mechanisms by which FoxM1 participates in those processes are not understood. Here, we describe an essential role of FoxM1 in the regulation of oxidative stress that contributes to malignant transformation and tumour cell survival. We identify a negative feedback loop involving FoxM1 that regulates reactive oxygen species (ROS) in proliferating cells. We show that induction of FoxM1 by oncogenic Ras requires ROS. Elevated FoxM1, in turn, downregulates ROS levels by stimulating expression of ROS scavenger genes, such as MnSOD, catalase and PRDX3. FoxM1 depletion sensitizes cells to oxidative stress and increases oncogene-induced premature senescence. Moreover, tumour cells expressing activated AKT1 are 'addicted' to FoxM1, as they require continuous presence of FoxM1 for survival. Together, our results identify FoxM1 as a key regulator of ROS in dividing cells, and provide insights into the mechanism how tumour cells use FoxM1 to control oxidative stress to escape premature senescence and apoptosis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Factores de Transcripción Forkhead/metabolismo , Estrés Oxidativo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Genes ras , Humanos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Osteosarcoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Ann Neurol ; 57(1): 131-5, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15622542

RESUMEN

We discovered intronic mutations in two episodic ataxia type 2 (EA2) families: a four-nucleotide GAGT deletion at IVS41+(3-6) and a single nucleotide insertion (insT) at IVS24+3. We expressed minigenes harboring the mutations in cell lines to demonstrate exon skipping from the deletion mutation and the activation of a cryptic splice donor site from the insertion mutation. The identification of these disease-causing mutations expands the spectrum of EA2 mutations and emphasizes the importance of intronic sequences in regulating gene expression.


Asunto(s)
Ataxia/genética , Intrones , Mutación , Proteínas del Tejido Nervioso/genética , Adolescente , Adulto , Animales , Células COS , Canales de Calcio Tipo L , Niño , Chlorocebus aethiops , Análisis Mutacional de ADN/métodos , Exones , Salud de la Familia , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Empalme del ARN/fisiología , ARN Nuclear Pequeño/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transfección/métodos
12.
Science ; 304(5676): 1509-13, 2004 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-15105459

RESUMEN

The mechanisms controlling axon guidance are of fundamental importance in understanding brain development. Growing corticospinal and somatosensory axons cross the midline in the medulla to reach their targets and thus form the basis of contralateral motor control and sensory input. The motor and sensory projections appeared uncrossed in patients with horizontal gaze palsy with progressive scoliosis (HGPPS). In patients affected with HGPPS, we identified mutations in the ROBO3 gene, which shares homology with roundabout genes important in axon guidance in developing Drosophila, zebrafish, and mouse. Like its murine homolog Rig1/Robo3, but unlike other Robo proteins, ROBO3 is required for hindbrain axon midline crossing.


Asunto(s)
Axones/fisiología , Oftalmoplejía/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Rombencéfalo/crecimiento & desarrollo , Escoliosis/genética , Adulto , Empalme Alternativo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Potenciales Evocados Motores , Potenciales Evocados Somatosensoriales , Femenino , Lateralidad Funcional , Ligamiento Genético , Humanos , Hibridación in Situ , Imagen por Resonancia Magnética , Masculino , Bulbo Raquídeo/crecimiento & desarrollo , Bulbo Raquídeo/patología , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Morfogénesis , Mutación , Vías Nerviosas , Oftalmoplejía/patología , Oftalmoplejía/fisiopatología , Linaje , Estructura Terciaria de Proteína , Receptores de Superficie Celular , Receptores Inmunológicos/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rombencéfalo/patología , Escoliosis/patología , Escoliosis/fisiopatología , Análisis de Secuencia de ADN , Síndrome
13.
Pharmacol Biochem Behav ; 77(2): 365-70, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14751466

RESUMEN

We investigated the effects of methamphetamine (METH) on core body temperature (Tb) and motor activity (MA) with or without exposure to a peripheral immune challenge. Mice were exposed to an escalating METH treatment and then to a METH treatment known to cause neurotoxicity (binge METH treatment). This was followed by a challenge with lipopolysaccharide (LPS). Three days later, METH and saline-treated control groups were challenged with an acute test dose of METH (METH test). Animals exposed to the escalating METH treatment exhibited a significant increase in Tb only after the initial exposure to METH (Day 1) and following the METH test (Day 7). The hyperthermic effect produced by the METH test (Day 7) was reduced in mice previously exposed to combined exposure to binge METH and LPS treatments. The escalating METH treatment produced MA sensitization to the METH test. Animals treated with the binge METH, LPS injection or both treatments combined prevented MA sensitization to the METH test. These findings suggest that induction of peripheral endotoxemia in animals with a history of METH reduced the hyperthermic response to a subsequent challenge with METH.


Asunto(s)
Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Endotoxemia/fisiopatología , Endotoxemia/psicología , Metanfetamina/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA