Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 518(7538): 240-4, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25409150

RESUMEN

Broad and deep tumour genome sequencing has shed new light on tumour heterogeneity and provided important insights into the evolution of metastases arising from different clones. There is an additional layer of complexity, in that tumour evolution may be influenced by selective pressure provided by therapy, in a similar fashion to that occurring in infectious diseases. Here we studied tumour genomic evolution in a patient (index patient) with metastatic breast cancer bearing an activating PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha, PI(3)Kα) mutation. The patient was treated with the PI(3)Kα inhibitor BYL719, which achieved a lasting clinical response, but the patient eventually became resistant to this drug (emergence of lung metastases) and died shortly thereafter. A rapid autopsy was performed and material from a total of 14 metastatic sites was collected and sequenced. All metastatic lesions, when compared to the pre-treatment tumour, had a copy loss of PTEN (phosphatase and tensin homolog) and those lesions that became refractory to BYL719 had additional and different PTEN genetic alterations, resulting in the loss of PTEN expression. To put these results in context, we examined six other patients also treated with BYL719. Acquired bi-allelic loss of PTEN was found in one of these patients, whereas in two others PIK3CA mutations present in the primary tumour were no longer detected at the time of progression. To characterize our findings functionally, we examined the effects of PTEN knockdown in several preclinical models (both in cell lines intrinsically sensitive to BYL719 and in PTEN-null xenografts derived from our index patient), which we found resulted in resistance to BYL719, whereas simultaneous PI(3)K p110ß blockade reverted this resistance phenotype. We conclude that parallel genetic evolution of separate metastatic sites with different PTEN genomic alterations leads to a convergent PTEN-null phenotype resistant to PI(3)Kα inhibition.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Tiazoles/farmacología , Alelos , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fosfatidilinositol 3-Quinasa Clase I , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Pérdida de Heterocigocidad/efectos de los fármacos , Pérdida de Heterocigocidad/genética , Ratones , Ratones Desnudos , Fosfohidrolasa PTEN/metabolismo , Tiazoles/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Proc Natl Acad Sci U S A ; 113(11): 3030-5, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929372

RESUMEN

Loss of the tumor suppressor gene PTEN is implicated in breast cancer progression and resistance to targeted therapies, and is thought to promote tumorigenesis by activating PI3K signaling. In a transgenic model of breast cancer, Pten suppression using a tetracycline-regulatable short hairpin (sh)RNA cooperates with human epidermal growth factor receptor 2 (HER2/neu), leading to aggressive and metastatic disease with elevated signaling through PI3K and, surprisingly, the mitogen-activated protein kinase (MAPK) pathway. Restoring Pten function is sufficient to down-regulate both PI3K and MAPK signaling and triggers dramatic tumor regression. Pharmacologic inhibition of MAPK signaling produces similar effects to Pten restoration, suggesting that the MAPK pathway contributes to the maintenance of advanced breast cancers harboring Pten loss.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias Mamarias Experimentales/fisiopatología , Proteínas de Neoplasias/fisiología , Fosfohidrolasa PTEN/deficiencia , Receptor ErbB-2/fisiología , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Genes erbB-2 , Humanos , MAP Quinasa Quinasa 1/fisiología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Desnudos , Ratones Transgénicos , Metástasis de la Neoplasia , Proteínas de Neoplasias/deficiencia , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal/fisiología
3.
Br J Cancer ; 110(4): 958-66, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24300972

RESUMEN

BACKGROUND: Circulating tumour cells (CTCs) have an important role in metastatic processes, but details of their basic characteristics remain elusive. We hypothesised that CD44-expressing CTCs show a mesenchymal phenotype and high potential for survival in hepatocellular carcinoma (HCC). METHODS: Circulating CD44(+)CD90(+) cells, previously shown to be tumour-initiating cells, were sorted from human blood and their genetic characteristics were compared with those of tumour cells from primary tissues. The mechanism underlying the high survival potential of CD44-expressing cells in the circulatory system was investigated in vitro. RESULTS: CD44(+)CD90(+) cells in the blood acquired epithelial-mesenchymal transition, and CD44 expression remarkably increased from the tissue to the blood. In Li7 and HLE cells, the CD44(high) population showed higher anoikis resistance and sphere-forming ability than did the CD44(low) population. This difference was found to be attributed to the upregulation of Twist1 and Akt signal in the CD44(high) population. Twist1 knockdown showed remarkable reduction in anoikis resistance, sphere formation, and Akt signal in HLE cells. In addition, mesenchymal markers and CD44s expression were downregulated in the Twist1 knockdown. CONCLUSIONS: CD44s symbolises the acquisition of a mesenchymal phenotype regulating anchorage-independent capacity. CD44s-expressing tumour cells in peripheral blood are clinically important therapeutic targets in HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Receptores de Hialuranos/metabolismo , Neoplasias Hepáticas/patología , Células Neoplásicas Circulantes/patología , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Anoicis/genética , Apoptosis , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , Neoplasias Hepáticas/metabolismo , Mesodermo/citología , Proteínas Nucleares/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Antígenos Thy-1/metabolismo , Proteína 1 Relacionada con Twist/biosíntesis
4.
Br J Cancer ; 109(2): 379-86, 2013 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-23778530

RESUMEN

BACKGROUND: Multiple early gastric cancers (EGCs) may develop in 6-14% of patients even after achieving curative endoscopic submucosal dissection (ESD); however, a useful biomarker for predicting recurrence is not available. The present study investigated whether the expression of CD44 variant 9 (CD44v9), a functional cancer stem cell marker, in the primary gastric cancer tissue represents an indicator of recurrence. METHODS: Eighty-eight patients who underwent ESD for EGC from 2008 to 2010 were enrolled and monitored for recurrence for 3 years. The expression levels of CD44v9 in the tissue of initial EGCs were evaluated by immunohistochemistry, and the recurrence rate was compared between CD44v9-positive and CD44v9-negative groups. The mucin phenotype and expression of microRNA-21 (miR-21) and programmed cell death protein 4 (PDCD4) were also analysed. RESULTS: The recurrence rate of EGC was significantly higher in the CD44v9-positive group than in the CD44v9-negative group (hazard ratio (HR), 21.8; 95% confidence interval (CI), 5.71-83.1). However, mucin phenotypes and the expression of miR-21 and PDCD4 did not predict recurrence after ESD. Meanwhile, grade of gastric atrophy was also identified as a significant marker of multiple recurrence (HR, 4.95; 95% CI, 1.30-18.8). CONCLUSION: CD44 variant 9 expression represents a potential predictive marker for recurrence in EGC.


Asunto(s)
Adenocarcinoma/diagnóstico , Biomarcadores de Tumor , Receptores de Hialuranos/fisiología , Recurrencia Local de Neoplasia/diagnóstico , Neoplasias Gástricas/diagnóstico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Progresión de la Enfermedad , Endoscopía , Femenino , Gastrectomía/métodos , Humanos , Receptores de Hialuranos/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Resultado del Tratamiento
5.
Nat Med ; 4(8): 915-22, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9701243

RESUMEN

Neurofibromatosis type 2 (NF2) protein, also known as merlin or schwannomin, is a tumor suppressor, and NF2 is mutated in most schwannomas and meningiomas. Although these tumors are dependent on NF2, some lack detectable NF2 mutations, which indicates that alternative mechanisms exist for inactivating merlin. Here, we demonstrate cleavage of merlin by the ubiquitous protease calpain and considerable activation of the calpain system resulting in the loss of merlin expression in these tumors. Increased proteolysis of merlin by calpain in some schwannomas and meningiomas exemplifies tumorigenesis linked to the calpain-mediated proteolytic pathway.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Calpaína/metabolismo , Genes de la Neurofibromatosis 2 , Glioma/metabolismo , Proteínas de la Membrana/metabolismo , Meningioma/metabolismo , Neurilemoma/metabolismo , Secuencia de Bases , Línea Celular , Cartilla de ADN , Activación Enzimática , Glutatión Transferasa/biosíntesis , Humanos , Proteínas de la Membrana/biosíntesis , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Neurofibromina 2 , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Moldes Genéticos , Transcripción Genética , Transfección , Células Tumorales Cultivadas
6.
J Cell Biol ; 141(2): 409-18, 1998 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-9548719

RESUMEN

The small GTPase Rho is believed to regulate the actin cytoskeleton and cell adhesion through its specific targets. We previously identified the Rho targets: protein kinase N, Rho-associated kinase (Rho-kinase), and the myosin-binding subunit (MBS) of myosin phosphatase. We found that in MDCK epithelial cells, MBS accumulated at the tetradecanoylphorbol-13-acetate (TPA)-induced membrane ruffling area, where moesin, a member of the ERM (ezrin, radixin, and moesin) family, was localized. Neither membrane ruffling nor an accumulation of moesin and MBS at the free-end plasma membrane was induced when MDCK cells were stimulated with TPA after the microinjection of C3, which ADP-ribosylates and inactivates Rho. MBS was colocalized with moesin at the cell-cell contact sites in MDCK cells. We also found that moesin was coimmunoprecipitated with MBS from MDCK cells. Recombinant MBS interacted with the amino-terminal domains of moesin and ezrin. Myosin phosphatase composed of the catalytic subunit and MBS showed phosphatase activity toward moesin, which was phosphorylated by Rho-kinase. The phosphatase activity was inhibited when MBS was phosphorylated by Rho-kinase. These results suggest that MBS is recruited with moesin to the plasma membrane and that myosin phosphatase and Rho-kinase regulate the phosphorylation state of moesin downstream of Rho.


Asunto(s)
Toxinas Botulínicas , Proteínas de Microfilamentos , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/fisiología , Animales , Encéfalo/metabolismo , Bovinos , Línea Celular , Membrana Celular/química , Membrana Celular/enzimología , Membrana Celular/ultraestructura , Sistema Libre de Células , Proteínas del Citoesqueleto , Perros , Células Epiteliales , Péptidos y Proteínas de Señalización Intracelular , Fosfatasa de Miosina de Cadena Ligera , Miosinas/metabolismo , Fosfoproteínas Fosfatasas/análisis , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas/análisis , Proteínas/genética , Proteínas Recombinantes de Fusión , Transducción de Señal/fisiología , Acetato de Tetradecanoilforbol/farmacología , Quinasas Asociadas a rho
7.
J Cell Biol ; 155(5): 755-62, 2001 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-11714729

RESUMEN

CD44 is a widely distributed cell surface adhesion molecule and is implicated in diverse biological processes. However, the nature of intracellular signaling triggered by CD44 remains to be elucidated. Here, we show that CD44 undergoes sequential proteolytic cleavage in the ectodomain and intracellular domain, resulting in the release of a CD44 intracellular domain (ICD) fragment. Consequently, CD44ICD acts as a signal transduction molecule, where it translocates to the nucleus and activates transcription mediated through the 12-O-tetradecanoylphorbol 13-acetate-responsive element, which is found in numerous genes involved in diverse cellular processes. Expression of an uncleavable CD44 mutant as well as metalloprotease inhibitor treatment blocks CD44-mediated transcriptional activation. In search of the underlying mechanism, we have found that CD44ICD potentiates transactivation mediated by the transcriptional coactivator CBP/p300. Furthermore, we show that cells expressing CD44ICD produce high levels of CD44 messenger RNA, suggesting that the CD44 gene is one of the potential targets for transcriptional activation by CD44ICD. These observations establish a novel CD44 signaling pathway and shed new light on the functional link between proteolytic processing of an adhesion molecule at the cell surface and transcriptional activation in the nucleus.


Asunto(s)
Receptores de Hialuranos/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular/fisiología , Animales , Fraccionamiento Celular , Línea Celular , Genes Reporteros , Humanos , Ionomicina/farmacología , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transactivadores/metabolismo , Transcripción Genética/fisiología , Activación Transcripcional
8.
J Cell Biol ; 149(5): 1073-86, 2000 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-10831611

RESUMEN

The mitotic apparatus plays a pivotal role in dividing cells to ensure each daughter cell receives a full set of chromosomes and complement of cytoplasm during mitosis. A human homologue of the Drosophila warts tumor suppressor, h-warts/LATS1, is an evolutionarily conserved serine/threonine kinase and a dynamic component of the mitotic apparatus. We have identified an interaction of h-warts/LATS1 with zyxin, a regulator of actin filament assembly. Zyxin is a component of focal adhesion, however, during mitosis a fraction of cytoplasmic-dispersed zyxin becomes associated with h-warts/LATS1 on the mitotic apparatus. We found that zyxin is phosphorylated specifically during mitosis, most likely by Cdc2 kinase, and that the phosphorylation regulates association with h-warts/LATS1. Furthermore, microinjection of truncated h-warts/LATS1 protein, including the zyxin-binding portion, interfered with localization of zyxin to mitotic apparatus, and the duration of mitosis of these injected cells was significantly longer than that of control cells. These findings suggest that h-warts/LATS1 and zyxin play a crucial role in controlling mitosis progression by forming a regulatory complex on mitotic apparatus.


Asunto(s)
Actinas/metabolismo , Proteínas de Drosophila , Metaloproteínas/metabolismo , Proteínas Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/metabolismo , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Proteína Quinasa CDC2/metabolismo , Células COS/citología , Células COS/metabolismo , Proteínas del Citoesqueleto , Expresión Génica/fisiología , Genes Supresores de Tumor/fisiología , Glicoproteínas , Células HeLa , Humanos , Metaloproteínas/genética , Metaloproteínas/inmunología , Mitosis/fisiología , Datos de Secuencia Molecular , Fragmentos de Péptidos/inmunología , Fosforilación , Plásmidos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Dedos de Zinc/genética , Zixina
9.
J Cell Biol ; 143(5): 1249-58, 1998 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-9832553

RESUMEN

Rho-associated kinase (Rho-kinase), which is activated by the small GTPase Rho, regulates formation of stress fibers and focal adhesions, myosin fiber organization, and neurite retraction through the phosphorylation of cytoskeletal proteins, including myosin light chain, the ERM family proteins (ezrin, radixin, and moesin) and adducin. Rho-kinase was found to phosphorylate a type III intermediate filament (IF) protein, glial fibrillary acidic protein (GFAP), exclusively at the cleavage furrow during cytokinesis. In the present study, we examined the roles of Rho-kinase in cytokinesis, in particular organization of glial filaments during cytokinesis. Expression of the dominant-negative form of Rho-kinase inhibited the cytokinesis of Xenopus embryo and mammalian cells, the result being production of multinuclei. We then constructed a series of mutant GFAPs, where Rho-kinase phosphorylation sites were variously mutated, and expressed them in type III IF-negative cells. The mutations induced impaired segregation of glial filament (GFAP filament) into postmitotic daughter cells. As a result, an unusually long bridge-like cytoplasmic structure formed between the unseparated daughter cells. Alteration of other sites, including the cdc2 kinase phosphorylation site, led to no remarkable defect in glial filament separation. These results suggest that Rho-kinase is essential not only for actomyosin regulation but also for segregation of glial filaments into daughter cells which in turn ensures correct cytokinetic processes.


Asunto(s)
División Celular/genética , División Celular/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Secuencia de Aminoácidos , Animales , Dominio Catalítico/genética , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Filamentos Intermedios/metabolismo , Filamentos Intermedios/ultraestructura , Péptidos y Proteínas de Señalización Intracelular , Células L , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Transfección , Xenopus , Quinasas Asociadas a rho
10.
Oncogene ; 26(17): 2395-406, 2007 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-17130845

RESUMEN

The serine protease Omi/HtrA2 was initially regarded as a proapoptotic molecule that proteolyses several proteins to induce cell death. Recent studies, however, indicate that loss of Omi protease activity increases susceptibility to stress-induced cell death. These complicated findings suggest that the protease activity of Omi is involved not only in apoptosis but also in cellular homeostasis. However, the targets which Omi uses to mediate this novel process are unknown. Previously, we showed that WARTS (WTS)/large tumor-suppressor 1 mitotic kinase interacts with the protein/discs-large protein/zonula (PDZ) domain of Omi and promotes its protease activity. We now report that WTS is a substrate for Omi protease activity, thus it is not only a regulator but also a downstream target of this protease. Interaction with Omi PDZ domain is required for WTS to be proteolysed. When caspase-9-deficient mouse embryonic fibroblasts (MEFs) were treated with staurosporine, WTS was proteolysed by activated endogenous Omi without induction of cell death. Therefore, protease activity of Omi and proteolysis of WTS are not necessarily required for cell death. We found that depletion of Omi from HeLa cells results in accelerated cell proliferation despite no significant change in the duration of mitosis. The depletion of WTS showed the same effect on S phase progression. Therefore, WTS proteolytic fragment(s) generated by Omi may act as an inhibitor of G1/S progression. Our data reveal a role for Omi-mediated processing of WTS in negative regulation of cell cycle progression at interphase, suggesting a novel function of Omi other than apoptosis.


Asunto(s)
Proliferación Celular , Proteínas Mitocondriales/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Serina Endopeptidasas/fisiología , Animales , Apoptosis/fisiología , Células COS , Chlorocebus aethiops , Células HeLa , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Interfase/fisiología , Especificidad por Sustrato
11.
J Dent Res ; 97(7): 779-786, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29494307

RESUMEN

This study investigated a case of spindle cell carcinoma (SpCC) in tongue pathological lesions. The patient experienced a local recurrence and distant metastasis after surgical intervention. Although standard chemotherapy was administered, a granulomatous mass continued to develop. This aggressive growth led to survival of the tumor. Secondary debulking surgery was performed to improve the patient's quality of life at the request of the patient. Using a tissue sample derived from the secondary debulking surgery, we performed an analysis of the tumor's cell surface antigens, differentiation potential, metastatic ability, and inhibition potential by anticancer reagents. In vitro analysis revealed that the cell population grown under adherent culture conditions expressed the mesenchymal stem cell (MSC) markers CD73, CD90, and CD105. The cell line established from this SpCC contained colony-forming unit fibroblasts (CFU-Fs) and exhibited multipotent differentiation into several mesenchymal lineages, including bone, cartilage, and fat. The SpCC cells also displayed vigorous mobilization. These characteristics suggested that they had the differentiation potential of mesenchymal cells, especially MSCs, rather than that of epithelial cells. The surgical specimen analyzed in this study resisted the molecular target reagent cetuximab, which is an epidermal growth factor receptor inhibitor. This clinical insight revealed that chemotherapy-resistant SpCC cells have different characteristics compared to most other cancer cells, which are sensitive to cetuximab. Our cell death assay revealed that SpCC cell death was induced by the anticancer drug imatinib, which is known to inhibit protein tyrosine kinase activity of ABL, platelet-derived growth factor receptor α (PDGFRα), and KIT. Here, we report recurrent SpCC with characteristics of MSCs and potential for treatment with imatinib.


Asunto(s)
Carcinoma/patología , Células Madre Mesenquimatosas/patología , Recurrencia Local de Neoplasia/patología , Neoplasias de la Lengua/patología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/análisis , Carcinoma/terapia , Técnicas de Cultivo de Célula , Muerte Celular , Diferenciación Celular , Movimiento Celular , Terapia Combinada , Resistencia a Antineoplásicos , Citometría de Flujo , Humanos , Recurrencia Local de Neoplasia/terapia , Procedimientos Quirúrgicos Orales , Calidad de Vida , Células Madre , Neoplasias de la Lengua/terapia , Células Tumorales Cultivadas
12.
Oncogene ; 25(1): 79-90, 2006 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-16170361

RESUMEN

The adenovirus type 5 gene E1A is known to suppress tumorigenicity by transcriptionally downregulating HER-2/neu (HER2) or by inducing apoptosis. We show here that E1A also suppressed the tumorigenicity of the low-HER2-expressing ovarian cancer cell line OVCAR-3 by decreasing cell proliferation. We further found that the mechanism responsible for this reduced proliferation is the presence of PEA15 (phosphoprotein enriched in astrocytes), which is upregulated by E1A in ovarian cancer; PEA15 promotes translocation of ERK from the nucleus to the cytoplasm, leading to inhibition of ERK-dependent transcription and proliferation. Indeed, siRNA-mediated knockdown of PEA15 expression in OVCAR-3 stable E1A transfectants resulted in a nuclear accumulation of the active form of ERK, followed by an increase in Elk-1 activity, DNA synthesis, and anchorage-independent growth. Finally, PEA15 by itself suppressed colony formation in breast and ovarian cancer cell lines, in which E1A is known to have antitumor activity. We conclude that part of the antitumor effect of E1A in ovarian cancer results from cytoplasmic sequestration of the activated form of ERK by PEA15.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Apoptosis , Citoplasma/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/tratamiento farmacológico , Fosfoproteínas/metabolismo , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis , Western Blotting , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , ADN/metabolismo , Regulación hacia Abajo , Activación Enzimática , Femenino , Vectores Genéticos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Microscopía Fluorescente , Neoplasias Ováricas/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor ErbB-2/metabolismo , Factores de Tiempo , Transcripción Genética , Transfección , Regulación hacia Arriba
13.
Mol Cell Biol ; 20(14): 5107-18, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10866667

RESUMEN

MBD1 is a mammalian protein that binds symmetrically methylated CpG sequences and regulates gene expression in association with DNA methylation. This protein possesses a conserved sequence, named methyl-CpG binding domain (MBD), among a family of methyl-CpG binding proteins that mediate the biological consequences of the methylation. In addition, MBD1 has at least five isoforms due to alternative splicing events, resulting in the presence of CXXC1, CXXC2, and CXXC3 in MBD1 isoforms v1 (MBD1v1) and MBD1v2, and CXXC1 and CXXC2 in MBD1v3 and -v4. In the present study, we have investigated the significance of MBD, CXXC, and the C-terminal transcriptional repression domain (TRD) in MBD1. A bacterially expressed MBD binds efficiently to densely methylated rather than to sparsely methylated DNAs. In both methylation-deficient Drosophila melanogaster SL2 cells and mammalian CHO-K1 cells, MBD1v1 represses transcription preferentially from both unmethylated and sparsely methylated promoters, while MBD1v3 inhibits densely methylated but not unmethylated promoter activities. The CXXC3 sequence in MBD1v1 is responsible for the ability to bind unmethylated promoter. Furthermore, we have constructed mutant-type MBD1s in which the functionally important residues Arg22, Arg30, Asp32, Tyr34, Arg44, Ser45, and Tyr52 are changed to alanine to investigate the correlation between the structure and function of the MBD in MBD1. Excepting those for Ser45 and Tyr52, none of the recombinant MBD mutants bound to the densely methylated or unmethylated DNAs, and green fluorescent protein-fused MBD1 mutants did not localize properly in the nucleus. All the MBD1v1 and -v3 mutants lost the activity of methylation-dependent gene repression. Based on these findings we have concluded that MBD1 acts as a transcriptional regulator depending on the density of methyl-CpG pairs through the cooperation of MBD, CXXC, and TRD sequences.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética , Secuencias de Aminoácidos , Animales , Sitios de Unión , Células CHO , Núcleo Celular/genética , Núcleo Celular/metabolismo , Islas de CpG , Cricetinae , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , ADN/metabolismo , Metilación de ADN , Regulación de la Expresión Génica , Humanos , Mutagénesis , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factores de Transcripción
14.
Mol Cell Biol ; 19(9): 6415-26, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10454587

RESUMEN

DNA methylation of promoter-associated CpG islands is involved in the transcriptional repression of vertebrate genes. To investigate the mechanisms underlying gene inactivation by DNA methylation, we characterized a human MBD1 protein, one of the components of MeCP1, which possesses a methyl-CpG binding domain (MBD) and cysteine-rich (CXXC) domains. Four novel MBD1 isoforms (MBD1v1, MBD1v2, MBD1v3, and MBD1v4) were identified by the reverse transcription-PCR method. We found that these transcripts were alternatively spliced in the region of CXXC domains and the C terminus. Green fluorescent protein-fused MBD1 was localized to multiple foci on the human genome, mostly in the euchromatin regions, and particularly concentrated in the pericentromeric region of chromosome 1. Both the MBD sequence and genome methylation were required for proper localization of the MBD1 protein. We further investigated whether MBD1 isoforms are responsible for transcriptional repression of human genes. A bacterially expressed MBD1 protein bound preferentially to methylated DNA fragments containing CpG islands from the tumor suppressor genes p16, VHL, and E-cadherin and from an imprinted SNRPN gene. All MBD1 isoforms inhibited promoter activities of these genes via methylation. Interestingly, MBD1 isoforms v1 and v2 containing three CXXC domains also suppressed unmethylated promoter activities in mammalian cells. These effects were further manifested in Drosophila melanogaster cells, which lack genome methylation. Sp1-activated transcription of methylated p16 and SNRPN promoters was inhibited by all of the MBD1 isoforms, whereas the isoforms v1 and v2 reduced Sp1-activated transcription from unmethylated promoters as well. These findings suggested that the MBD1 isoforms have different roles in methylation-mediated transcriptional silencing in euchromatin.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión/genética , Células COS , Línea Celular , Núcleo Celular/metabolismo , Islas de CpG , Cartilla de ADN/genética , Proteínas de Unión al ADN/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Eucromatina , Células HeLa , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transcripción Genética
15.
Oncogene ; 36(9): 1276-1286, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-27546618

RESUMEN

The transcription factor nuclear factor-κB (NF-κB) has important roles for tumorigenesis, but how it regulates cancer stem cells (CSCs) remains largely unclear. We identified insulin-like growth factor 2 (IGF2) is a key target of NF-κB activated by HER2/HER3 signaling to form tumor spheres in breast cancer cells. The IGF2 receptor, IGF1 R, was expressed at high levels in CSC-enriched populations in primary breast cancer cells. Moreover, IGF2-PI3K (IGF2-phosphatidyl inositol 3 kinase) signaling induced expression of a stemness transcription factor, inhibitor of DNA-binding 1 (ID1), and IGF2 itself. ID1 knockdown greatly reduced IGF2 expression, and tumor sphere formation. Finally, treatment with anti-IGF1/2 antibodies blocked tumorigenesis derived from the IGF1Rhigh CSC-enriched population in a patient-derived xenograft model. Thus, NF-κB may trigger IGF2-ID1-IGF2-positive feedback circuits that allow cancer stem-like cells to appear. Then, they may become addicted to the circuits. As the circuits are the Achilles' heels of CSCs, it will be critical to break them for eradication of CSCs.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinogénesis , Femenino , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor II del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Clasificación del Tumor , Invasividad Neoplásica , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Pronóstico , Transducción de Señal , Esferoides Celulares , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Cell Biol ; 216(3): 641-656, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28193700

RESUMEN

Phosphatase and tensin homologue (PTEN) protein levels are critical for tumor suppression. However, the search for a recurrent cancer-associated gene alteration that causes PTEN degradation has remained futile. In this study, we show that Importin-11 (Ipo11) is a transport receptor for PTEN that is required to physically separate PTEN from elements of the PTEN degradation machinery. Mechanistically, we find that the E2 ubiquitin-conjugating enzyme and IPO11 cargo, UBE2E1, is a limiting factor for PTEN degradation. Using in vitro and in vivo gene-targeting methods, we show that Ipo11 loss results in degradation of Pten, lung adenocarcinoma, and neoplasia in mouse prostate with aberrantly high levels of Ube2e1 in the cytoplasm. These findings explain the correlation between loss of IPO11 and PTEN protein in human lung tumors. Furthermore, we find that IPO11 status predicts disease recurrence and progression to metastasis in patients choosing radical prostatectomy. Thus, our data introduce the IPO11 gene as a tumor-suppressor locus, which is of special importance in cancers that still retain at least one intact PTEN allele.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , beta Carioferinas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Enzimas Ubiquitina-Conjugadoras/metabolismo
17.
Nucleic Acids Res ; 29(4): E23, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11160943

RESUMEN

A tetracycline-controlled gene expression system provides a powerful tool to dissect the functions of gene products. However, it often appears difficult to establish cell lines or transgenic animals stably expressing tetracycline-dependent transactivators, possibly as a result of toxicity of the transactivator domains used. In order to overcome this problem, we developed a novel tetracycline-dependent transactivator that works efficiently in mammalian cells. This transactivator is a fusion of the tet reverse repressor mutant and the transcriptional activating domain of human E2F4, which is ubiquitously expressed in vivo. We demonstrate here that this tetracycline-regulated gene expression system provides a two log transcriptional activation in mammalian cells as assessed by northern blot and luciferase analyses. Combining this system with green fluorescent protein reporter systems or microarray gene expression profiling will facilitate the study of gene function.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Tetraciclina/farmacología , Transactivadores/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Proteínas de Unión al ADN/genética , Doxiciclina/farmacología , Factor de Transcripción E2F4 , Escherichia coli , Perfilación de la Expresión Génica/métodos , Genes Reporteros/genética , Humanos , Cinética , Mutación/genética , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/química , Transactivadores/genética , Factores de Transcripción/genética , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
J Natl Cancer Inst ; 90(4): 307-15, 1998 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-9486817

RESUMEN

BACKGROUND: CD44 is a cell surface glycoprotein implicated in such diverse biologic processes as lymphocyte activation and homing, extracellular matrix adhesion, and cellular migration. Primary transcripts of the CD44 gene can be alternatively spliced to produce a variety of messenger RNA (mRNA) species. The standard form of CD44 mRNA contains sequences from at least 20 genomic exons; variant mRNAs contain sequences from one or more additional exons (v1-10). Predominant expression of a specific CD44 variant, i.e., CD44v8-10, in several human carcinomas has been described previously. In this study, we developed a novel molecular approach for detecting cancer cells that overexpress CD44v8-10 mRNA. METHODS: After finding that CD44v8-10 was predominantly expressed in non-small-cell lung and bladder carcinomas and that CD44v10 was predominantly expressed in leukocytes, we developed a competitive reverse transcription-polymerase chain reaction assay (CC-RT-PCR) that allows quantification of the relative expression of these two mRNA species in clinical specimens (i.e., determination of a v8-10/v10 ratio). CC-RT-PCR analysis was applied to pleural effusion specimens from patients with benign or malignant lung diseases as well as to spontaneously voided urine samples from patients with benign or malignant urologic diseases. RESULTS: Fifty two of 54 samples from patients with benign diseases expressed CD44v10 predominantly (v8-10/v10 ratio < or = 0.65), whereas 46 of 61 samples from patients with malignant diseases expressed CD44v8-10 predominantly (v8-10/v10 ratio > 1.00) (two-sided P < .001). CC-RT-PCR detected predominant expression of CD44v8-10 in cytologically negative samples from 11 patients who were later diagnosed with malignant disease. CONCLUSIONS: CC-RT-PCR analysis of CD44v8-10 expression could be an important adjunct to cytologic examination in cancer diagnosis, especially in detecting exfoliated cancer cells in pleural effusions and urine.


Asunto(s)
Receptores de Hialuranos/genética , Neoplasias/diagnóstico , ARN Mensajero/análisis , ARN Neoplásico/análisis , Neoplasias de la Mama/diagnóstico , Diagnóstico Diferencial , Humanos , Inmunohistoquímica , Neoplasias Laríngeas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Neoplasias/inmunología , Neoplasias/orina , Derrame Pleural/patología , Reacción en Cadena de la Polimerasa/métodos , Reproducibilidad de los Resultados , Transcripción Genética , Neoplasias de la Vejiga Urinaria/diagnóstico
19.
J Natl Cancer Inst ; 86(5): 344-9, 1994 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-8308926

RESUMEN

BACKGROUND: Heritable germline mutations of the p53 gene have been described in patients with Li-Fraumeni syndrome, occasionally in nonfamilial malignancies such as multifocal osteosarcoma, in a small subgroup of young patients with two or more primary malignancies, and in patients with sporadic breast carcinoma. We recently reported that multifocal gliomas are frequently associated with other primary malignancies, and we hypothesized that genetic alterations may account for this phenomenon. PURPOSE: We examined the frequency of germline p53 gene mutations in patients with glioma and either multifocality of lesions, history of an additional primary (different) malignancy, or a family history of cancer. METHODS: Lymphocytes from 51 glioma patients were analyzed for germline p53 gene mutations using RNA-polymerase chain reaction analysis, single-strand conformation polymorphism, and gene sequencing techniques. RESULTS: Germline p53 gene mutations were detected in six of 19 patients with multifocal glioma, including two with family history of cancer, one with another primary malignancy, and two with all three risk factors; one of four patients with unifocal glioma, another primary malignancy, and a family history of cancer; and two of 15 patients with unifocal glioma and a family history of cancer but no second malignancies. No mutations were detected in the patient with unifocal glioma and another malignancy or in the 12 control patients with unifocal glioma and no second malignancies or family history of cancer. Patients having mutations were younger than other patients in the same group. CONCLUSIONS: Germline p53 mutations are frequent in patients with multifocal glioma, glioma and another primary malignancy, and glioma associated with a family history of cancer, particularly if these factors are combined. IMPLICATIONS: Relatives at high risk can be identified for genetic counseling, early cancer detection, and possible enrollment in chemoprevention trials.


Asunto(s)
Neoplasias Encefálicas/genética , Genes p53/genética , Mutación de Línea Germinal/genética , Glioma/genética , Adulto , Anciano , Secuencia de Bases , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Mutación Puntual
20.
Cancer Res ; 48(16): 4633-8, 1988 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-3135116

RESUMEN

The expression and phosphotyrosine activity of pp60v-src were measured in the B31 avian sarcoma virus-transformed rat cell line by flow cytometry using monoclonal antibodies against pp60v-src (EB7) and phosphotyrosine (1G2). Although the immunocytochemical staining was markedly heterogeneous, binding of both antibodies was significantly greater to B31 cells than to untransformed Rat 1 cells. Binding of 1G2 to phosphotyrosine residues was specific; it was entirely inhibited by adding excess phenylphosphate but was not affected by phosphoserine or phosphothreonine. The relationship between the amount of phosphorylated tyrosine measured by our FCM technique and total cellular phosphotyrosine measured by phosphoamino acid analysis was linear in vanadate-treated BALB/c 3T3 cells. Treatment of B31 cells for 48 h with herbimycin A, a benzenoid ansamycin antibiotic, to decrease the expression and tyrosine kinase activity of pp60v-src caused reductions of 42% in anti-pp60v-src and 58% in anti-phosphotyrosine antibody immunofluorescence. DNA staining with the fluorescent dye propidium iodide showed no cell cycle specificity in the binding of either antibody. Herbimycin A also caused the transformed cell line to revert to the morphology, actin configuration, and growth behavior of untransformed cells; these changes were reversed within 12 h after removal of the drug. Flow cytometric evaluation of tyrosine kinase expression and activity was fast and easy, and the results correlated well with other measures of cell phenotype. This technique can be used to quantitate the effects of drugs on oncogenic proteins such as pp60v-src and their associated tyrosine kinase activity.


Asunto(s)
Citometría de Flujo , Proteínas Tirosina Quinasas/análisis , Proteínas de los Retroviridae/análisis , Actinas/análisis , Aminoácidos/análisis , Animales , Benzoquinonas , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Lactamas Macrocíclicas , Proteína Oncogénica pp60(v-src) , Quinonas/farmacología , Ratas , Rifabutina/análogos & derivados , Vanadatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA