Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
1.
Acta Neuropathol Commun ; 8(1): 41, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32245394

RESUMEN

Meningioma represents the most common primary brain tumor in adults. Recently several non-NF2 mutations in meningioma have been identified and correlated with certain pathological subtypes, locations and clinical observations. Alterations of cellular pathways due to these mutations, however, have largely remained elusive. Here we report that the Krueppel like factor 4 (KLF4)-K409Q mutation in skull base meningiomas triggers a distinct tumor phenotype. Transcriptomic analysis of 17 meningioma samples revealed that KLF4K409Q mutated tumors harbor an upregulation of hypoxia dependent pathways. Detailed in vitro investigation further showed that the KLF4K409Q mutation induces HIF-1α through the reduction of prolyl hydroxylase activity and causes an upregulation of downstream HIF-1α targets. Finally, we demonstrate that KLF4K409Q mutated tumors are susceptible to mTOR inhibition by Temsirolimus. Taken together, our data link the KLF4K409Q mediated upregulation of HIF pathways to the clinical and biological characteristics of these skull base meningiomas possibly opening new therapeutic avenues for this distinct meningioma subtype.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Hipoxia Tumoral/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Ratones , Ratones Desnudos , Mutación , Trasplante de Neoplasias , Prolil Hidroxilasas , Inhibidores de Proteínas Quinasas/farmacología , RNA-Seq , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sirolimus/análogos & derivados , Sirolimus/farmacología , Neoplasias de la Base del Cráneo , Regulación hacia Arriba
2.
Cancer Res ; 78(7): 1805-1819, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29339541

RESUMEN

Lung cancer is the leading cause of cancer-related death worldwide, in large part due to its high propensity to metastasize and to develop therapy resistance. Adaptive responses to hypoxia and epithelial-mesenchymal transition (EMT) are linked to tumor metastasis and drug resistance, but little is known about how oxygen sensing and EMT intersect to control these hallmarks of cancer. Here, we show that the oxygen sensor PHD3 links hypoxic signaling and EMT regulation in the lung tumor microenvironment. PHD3 was repressed by signals that induce EMT and acted as a negative regulator of EMT, metastasis, and therapeutic resistance. PHD3 depletion in tumors, which can be caused by the EMT inducer TGFß or by promoter methylation, enhanced EMT and spontaneous metastasis via HIF-dependent upregulation of the EGFR ligand TGFα. In turn, TGFα stimulated EGFR, which potentiated SMAD signaling, reinforcing EMT and metastasis. In clinical specimens of lung cancer, reduced PHD3 expression was linked to poor prognosis and to therapeutic resistance against EGFR inhibitors such as erlotinib. Reexpression of PHD3 in lung cancer cells suppressed EMT and metastasis and restored sensitivity to erlotinib. Taken together, our results establish a key function for PHD3 in metastasis and drug resistance and suggest opportunities to improve patient treatment by interfering with the feedforward signaling mechanisms activated by PHD3 silencing.Significance: This study links the oxygen sensor PHD3 to metastasis and drug resistance in cancer, with implications for therapeutic improvement by targeting this system. Cancer Res; 78(7); 1805-19. ©2018 AACR.


Asunto(s)
Antineoplásicos/uso terapéutico , Transición Epitelial-Mesenquimal/genética , Clorhidrato de Erlotinib/uso terapéutico , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Crecimiento Transformador alfa/metabolismo , Células A549 , Animales , Proteínas Reguladoras de la Apoptosis , Hipoxia de la Célula/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Femenino , Células HCT116 , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/genética , Ratones , Ratones Desnudos , Proteínas Mitocondriales/metabolismo , Metástasis de la Neoplasia/genética , Microambiente Tumoral/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Cell ; 25(1): 118-31, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24434214

RESUMEN

Cbx4 is a polycomb group protein that is also a SUMO E3 ligase, but its potential roles in tumorigenesis remain to be explored. Here, we report that Cbx4, but not other members of the Cbx family, enhances hypoxia-induced vascular endothelial growth factor (VEGF) expression and angiogenesis in hepatocellular carcinoma (HCC) cells through enhancing HIF-1α sumoylations at K391 and K477 in its two SUMO-interacting motifs-dependent mechanisms and increasing transcriptional activity of HIF-1. The Cbx4 expression is significantly correlated with VEGF expression, angiogenesis, and the overall survival of HCC patients and also in subcutaneously and orthotopically transplanted mice HCC models. Collectively, our findings demonstrate that Cbx4 plays a critical role in tumor angiogenesis by governing HIF-1α protein.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/metabolismo , Neovascularización Patológica/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Western Blotting , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/patología , Inmunoprecipitación de Cromatina , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica/fisiología , Xenoinjertos , Humanos , Inmunohistoquímica , Ligasas , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/patología , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína SUMO-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA