Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Cancer Res ; 19(5): 784-798, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33500360

RESUMEN

Several master transcription factors (TF) can activate the epithelial-to-mesenchymal transition (EMT). However, their individual and combinatorial contributions to EMT in breast cancer are not defined. We show that overexpression of EMT-TFs individually in epithelial cells upregulated endogenous SNAI2, ZEB1/2, TCF4, and TWIST1/2 as a result of positive feedback mediated in part by suppression of their negative regulator miRNAs miR200s/203/205. We identified TCF4 as a potential new target of miR200s. Expression of ZEB1/2 strongly correlated with the mesenchymal phenotype in breast cancer cells, with the CD24-/CD44+ stemness profile, and with lower expression of core epithelial genes in human breast tumors. Knockdown of EMT-TFs identified the key role of ZEB1 and its functional cooperation with other EMT-TFs in the maintenance of the mesenchymal state. Inducible ZEB1+2 knockdown in xenograft models inhibited pulmonary metastasis, emphasizing their critical role in dissemination from primary site and in extravasation. However, ZEB1+2 depletion one-week after intravenous injection did not inhibit lung colonization, suggesting that ZEB1/2 and EMT are not essential for macrometastatic outgrowth. These results provide strong evidence that EMT is orchestrated by coordinated expression of several EMT-TFs and establish ZEB1 as a key master regulator of EMT and metastasis in breast cancer. IMPLICATIONS: The EMT program is orchestrated by coordinated expression of multiple EMT transcription factors, whereas ZEB1 integrates the EMT master regulatory network and plays the major role in promoting EMT and metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factores de Transcripción/metabolismo , Animales , Neoplasias de la Mama/genética , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular/fisiología , Transición Epitelial-Mesenquimal , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , MicroARNs/genética , MicroARNs/metabolismo , Metástasis de la Neoplasia , Factores de Transcripción/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
2.
Echocardiography ; 26(7): 865-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20003023

RESUMEN

Real time two-dimensional transthoracic and transesophageal echocardiography demonstrated a mobile echolucent mass attached to the pulmonary valve in a 25-year-old adult, 20 years following balloon pulmonary valvuloplasty. The mass was surgically excised and pathology showed it to be a cyst.


Asunto(s)
Cateterismo/efectos adversos , Quistes/etiología , Quistes/cirugía , Ecocardiografía Doppler/métodos , Estenosis de la Válvula Pulmonar/cirugía , Válvula Pulmonar/diagnóstico por imagen , Válvula Pulmonar/cirugía , Adulto , Quistes/diagnóstico por imagen , Humanos , Masculino , Estenosis de la Válvula Pulmonar/complicaciones , Resultado del Tratamiento
3.
Mol Cancer Ther ; 15(8): 1809-22, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27235164

RESUMEN

Recent findings suggest that the inhibition of Aurora A (AURKA) kinase may offer a novel treatment strategy against metastatic cancers. In the current study, we determined the effects of AURKA inhibition by the small molecule inhibitor MLN8237 both as a monotherapy and in combination with the microtubule-targeting drug eribulin on different stages of metastasis in triple-negative breast cancer (TNBC) and defined the potential mechanism of its action. MLN8237 as a single agent and in combination with eribulin affected multiple steps in the metastatic process, including migration, attachment, and proliferation in distant organs, resulting in suppression of metastatic colonization and recurrence of cancer. Eribulin application induces accumulation of active AURKA in TNBC cells, providing foundation for the combination therapy. Mechanistically, AURKA inhibition induces cytotoxic autophagy via activation of the LC3B/p62 axis and inhibition of pAKT, leading to eradication of metastases, but has no effect on growth of mammary tumor. Combination of MLN8237 with eribulin leads to a synergistic increase in apoptosis in mammary tumors, as well as cytotoxic autophagy in metastases. These preclinical data provide a new understanding of the mechanisms by which MLN8237 mediates its antimetastatic effects and advocates for its combination with eribulin in future clinical trials for metastatic breast cancer and early-stage solid tumors. Mol Cancer Ther; 15(8); 1809-22. ©2016 AACR.


Asunto(s)
Aurora Quinasa A/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Azepinas/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Furanos/farmacología , Cetonas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res ; 75(2): 344-55, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25421577

RESUMEN

KAP1 (TRIM28) is a transcriptional regulator in embryonic development that controls stem cell self-renewal, chromatin organization, and the DNA damage response, acting as an essential corepressor for KRAB family zinc finger proteins (KRAB-ZNF). To gain insight into the function of this large gene family, we developed an antibody that recognizes the conserved zinc fingers linker region (ZnFL) in multiple KRAB-ZNF. Here, we report that the expression of many KRAB-ZNF along with active SUMOlyated KAP1 is elevated widely in human breast cancers. KAP1 silencing in breast cancer cells reduced proliferation and inhibited the growth and metastasis of tumor xenografts. Conversely, KAP1 overexpression stimulated cell proliferation and tumor growth. In cells where KAP1 was silenced, we identified multiple downregulated genes linked to tumor progression and metastasis, including EREG/epiregulin, PTGS2/COX2, MMP1, MMP2, and CD44, along with downregulation of multiple KRAB-ZNF proteins. KAP1-dependent stabilization of KRAB-ZNF required direct interactions with KAP1. Together, our results show that KAP1-mediated stimulation of multiple KRAB-ZNF contributes to the growth and metastasis of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Represoras/biosíntesis , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Neoplasias de la Mama/genética , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Pollos , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Proteínas Represoras/metabolismo , Sumoilación , Proteína 28 que Contiene Motivos Tripartito , Regulación hacia Arriba , Dedos de Zinc
5.
Mol Cancer Res ; 12(5): 681-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24574519

RESUMEN

UNLABELLED: The prometastatic protein NEDD9 (neural precursor cell expressed, developmentally downregulated 9) is highly expressed in many cancers and is required for mesenchymal individual cell migration and progression to the invasive stage. Nevertheless, the molecular mechanisms of NEDD9-driven migration and the downstream targets effecting metastasis are not well defined. In the current study, knockdown of NEDD9 in highly metastatic tumor cells drastically reduces their migratory capacity due to disruption of actin dynamics at the leading edge. Specifically, NEDD9 deficiency leads to a decrease in the persistence and stability of lamellipodial protrusions similar to knockdown of cortactin (CTTN). Mechanistically, it was shown that NEDD9 binds to and regulates acetylation of CTTN in an Aurora A kinase (AURKA)/HDAC6-dependent manner. The knockdown of NEDD9 or AURKA results in an increase in the amount of acetylated CTTN and a decrease in the binding of CTTN to F-actin. Overexpression of the deacetylation mimicking (9KR) mutant of CTTN is sufficient to restore actin dynamics at the leading edge and migration proficiency of the tumor cells. Inhibition of AURKA and HDAC6 activity by alisertib and Tubastatin A in xenograft models of breast cancer leads to a decrease in the number of pulmonary metastases. Collectively, these findings identify CTTN as the key downstream component of NEDD9-driven migration and metastatic phenotypes. IMPLICATIONS: This study provides a mechanistic platform for therapeutic interventions based on AURKA and HDAC6 inhibition for patients with metastatic breast cancer to prevent and/or eradicate metastases.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aurora Quinasa A/metabolismo , Cortactina/metabolismo , Histona Desacetilasas/metabolismo , Fosfoproteínas/metabolismo , Acetilación , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Femenino , Células HEK293 , Xenoinjertos , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Seudópodos/metabolismo , Seudópodos/patología , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cancer Res ; 12(1): 69-81, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24202705

RESUMEN

UNLABELLED: The scaffolding protein NEDD9 is an established prometastatic marker in several cancers. Nevertheless, the molecular mechanisms of NEDD9-driven metastasis in cancers remain ill-defined. Here, using a comprehensive breast cancer tissue microarray, it was shown that increased levels of NEDD9 protein significantly correlated with the transition from carcinoma in situ to invasive carcinoma. Similarly, it was shown that NEDD9 overexpression is a hallmark of highly invasive breast cancer cells. Moreover, NEDD9 expression is crucial for the protease-dependent mesenchymal invasion of cancer cells at the primary site but not at the metastatic site. Depletion of NEDD9 is sufficient to suppress invasion of tumor cells in vitro and in vivo, leading to decreased circulating tumor cells and lung metastases in xenograft models. Mechanistically, NEDD9 localized to invasive pseudopods and was required for local matrix degradation. Depletion of NEDD9 impaired invasion of cancer cells through inactivation of membrane-bound matrix metalloproteinase MMP14 by excess TIMP2 on the cell surface. Inactivation of MMP14 is accompanied by reduced collagenolytic activity of soluble metalloproteinases MMP2 and MMP9. Reexpression of NEDD9 is sufficient to restore the activity of MMP14 and the invasive properties of breast cancer cells in vitro and in vivo. Collectively, these findings uncover critical steps in NEDD9-dependent invasion of breast cancer cells. IMPLICATIONS: This study provides a mechanistic basis for potential therapeutic interventions to prevent metastasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/patología , Neoplasias Pulmonares/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Fosfoproteínas/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Neoplasias de la Mama/genética , Carcinoma in Situ/genética , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Células MCF-7 , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos NOD , Invasividad Neoplásica/genética , Trasplante de Neoplasias , Células Neoplásicas Circulantes , Fosfoproteínas/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Análisis de Matrices Tisulares , Inhibidor Tisular de Metaloproteinasa-2/genética , Trasplante Heterólogo
7.
Cancer Res ; 73(10): 3168-80, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23539442

RESUMEN

Aurora A kinase (AURKA) is overexpressed in 96% of human cancers and is considered an independent marker of poor prognosis. While the majority of tumors have elevated levels of AURKA protein, few have AURKA gene amplification, implying that posttranscriptional mechanisms regulating AURKA protein levels are significant. Here, we show that NEDD9, a known activator of AURKA, is directly involved in AURKA stability. Analysis of a comprehensive breast cancer tissue microarray revealed a tight correlation between the expression of both proteins, significantly corresponding with increased prognostic value. A decrease in AURKA, concomitant with increased ubiquitination and proteasome-dependent degradation, occurs due to depletion or knockout of NEDD9. Reexpression of wild-type NEDD9 was sufficient to rescue the observed phenomenon. Binding of NEDD9 to AURKA is critical for AURKA stabilization, as mutation of S296E was sufficient to disrupt binding and led to reduced AURKA protein levels. NEDD9 confers AURKA stability by limiting the binding of the cdh1-substrate recognition subunit of APC/C ubiquitin ligase to AURKA. Depletion of NEDD9 in tumor cells increases sensitivity to AURKA inhibitors. Combination therapy with NEDD9 short hairpin RNAs and AURKA inhibitors impairs tumor growth and distant metastasis in mice harboring xenografts of breast tumors. Collectively, our findings provide rationale for the use of AURKA inhibitors in treatment of metastatic tumors and predict the sensitivity of the patients to AURKA inhibitors based on NEDD9 expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Fosfoproteínas/fisiología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Aurora Quinasa A , Aurora Quinasas , Línea Celular Tumoral , Estabilidad de Enzimas , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Fosfoproteínas/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Serina-Treonina Quinasas/química , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA