Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell Biol ; 35(18): 3145-62, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26149384

RESUMEN

Ezrin is a key regulator of cancer metastasis that links the extracellular matrix to the actin cytoskeleton and regulates cell morphology and motility. We discovered a small-molecule inhibitor, NSC305787, that directly binds to ezrin and inhibits its function. In this study, we used a nano-liquid chromatography-tandem mass spectrometry (nano-LC-MS-MS)-based proteomic approach to identify ezrin-interacting proteins that are competed away by NSC305787. A large number of the proteins that interact with ezrin were implicated in protein translation and stress granule dynamics. We validated direct interaction between ezrin and the RNA helicase DDX3, and NSC305787 blocked this interaction. Downregulation or long-term pharmacological inhibition of ezrin led to reduced DDX3 protein levels without changes in DDX3 mRNA. Ectopic overexpression of ezrin in low-ezrin-expressing osteosarcoma cells caused a notable increase in DDX3 protein levels. Ezrin inhibited the RNA helicase activity of DDX3 but increased its ATPase activity. Our data suggest that ezrin controls the translation of mRNAs preferentially with a structured 5' untranslated region, at least in part, by sustaining the protein level of DDX3 and/or regulating its function. Therefore, our findings suggest a novel function for ezrin in regulation of gene translation that is distinct from its canonical role as a cytoskeletal scaffold at the cell membrane.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , ARN Helicasas DEAD-box/metabolismo , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Adamantano/análogos & derivados , Adamantano/farmacología , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Cromatografía Liquida , Proteínas del Citoesqueleto/antagonistas & inhibidores , ARN Helicasas DEAD-box/genética , Humanos , Ratones , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Unión Proteica/efectos de los fármacos , Proteómica , Quinolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño , Resonancia por Plasmón de Superficie , Espectrometría de Masas en Tándem
3.
PLoS One ; 9(12): e114260, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25479232

RESUMEN

BACKGROUND: The erythroblastosis virus E26 transforming sequences (ETS) family of transcription factors consists of a highly conserved group of genes that play important roles in cellular proliferation, differentiation, migration and invasion. Chromosomal translocations fusing ETS factors to promoters of androgen responsive genes have been found in prostate cancers, including the most clinically aggressive forms. ERG and ETV1 are the most commonly translocated ETS proteins. Over-expression of these proteins in prostate cancer cells results in a more invasive phenotype. Inhibition of ETS activity by small molecule inhibitors may provide a novel method for the treatment of prostate cancer. METHODS AND FINDINGS: We recently demonstrated that the small molecule YK-4-279 inhibits biological activity of ETV1 in fusion-positive prostate cancer cells leading to decreased motility and invasion in-vitro. Here, we present data from an in-vivo mouse xenograft model. SCID-beige mice were subcutaneously implanted with fusion-positive LNCaP-luc-M6 and fusion-negative PC-3M-luc-C6 tumors. Animals were treated with YK-4-279, and its effects on primary tumor growth and lung metastasis were evaluated. YK-4-279 treatment resulted in decreased growth of the primary tumor only in LNCaP-luc-M6 cohort. When primary tumors were grown to comparable sizes, YK-4-279 inhibited tumor metastasis to the lungs. Expression of ETV1 target genes MMP7, FKBP10 and GLYATL2 were reduced in YK-4-279 treated animals. ETS fusion-negative PC-3M-luc-C6 xenografts were unresponsive to the compound. Furthermore, YK-4-279 is a chiral molecule that exists as a racemic mixture of R and S enantiomers. We established that (S)-YK-4-279 is the active enantiomer in prostate cancer cells. CONCLUSION: Our results demonstrate that YK-4-279 is a potent inhibitor of ETV1 and inhibits both the primary tumor growth and metastasis of fusion positive prostate cancer xenografts. Therefore, YK-4-279 or similar compounds may be evaluated as a potential therapeutic tool for treatment of human prostate cancer at different stages.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Indoles/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Factores de Transcripción/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Metástasis de la Neoplasia , Proteínas de Fusión Oncogénica/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transactivadores/genética , Factores de Transcripción/genética , Regulador Transcripcional ERG , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Med Chem ; 57(24): 10290-303, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25432018

RESUMEN

EWS-FLI1 is an oncogenic fusion protein implicated in the development of Ewing's sarcoma family tumors (ESFT). Using our previously reported lead compound 2 (YK-4-279), we designed and synthesized a focused library of analogues. The functional inhibition of the analogues was measured by an EWS-FLI1/NR0B1 reporter luciferase assay and a paired cell screening approach measuring effects on growth inhibition for human cells containing EWS-FLI1 (TC32 and TC71) and control PANC1 cell lines devoid of the oncoprotein. Our data revealed that substitution of electron donating groups at the para-position on the phenyl ring was the most favorable for inhibition of EWS-FLI1 by analogs of 2. Compound 9u (with a dimethylamino substitution) was the most active inhibitor with GI50 = 0.26 ± 0.1 µM. Further, a correlation of growth inhibition (EWS-FLI1 expressing TC32 cells) and the luciferase reporter activity was established (R(2) = 0.84). Finally, we designed and synthesized a biotinylated analogue and determined the binding affinity for recombinant EWS-FLI1 (Kd = 4.8 ± 2.6 µM).


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Sarcoma de Ewing/tratamiento farmacológico , Compuestos de Anilina/síntesis química , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Humanos , Indoles/síntesis química , Luciferasas/metabolismo , Modelos Moleculares , Estructura Molecular , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
5.
Bioorg Med Chem ; 22(1): 478-87, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24326277

RESUMEN

Respiratory failure due to pulmonary metastasis is the major cause of death for patients with osteosarcoma. However, the molecular basis for metastasis of osteosarcoma is poorly understood. Recently, ezrin, a member of the ERM family of proteins, has been associated with osteosarcoma metastasis to the lungs. The small molecule NSC 668394 was identified to bind to ezrin, inhibit in vitro and in vivo cell migration, invasion, and metastatic colony survival. Reported herein are the design and synthesis of analogues of NSC 668394, and subsequent functional ezrin inhibition studies. The binding affinity was characterized by surface plasmon resonance technique. Cell migration and invasion activity was determined by electrical cell impedance methodology. Optimization of a series of heterocyclic-dione analogues led to the discovery of compounds 21k and 21m as potential novel antimetastatic agents.


Asunto(s)
Antineoplásicos/síntesis química , Proteínas del Citoesqueleto/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular , Proteínas del Citoesqueleto/antagonistas & inhibidores , Diseño de Fármacos , Humanos , Osteosarcoma/metabolismo , Osteosarcoma/patología
6.
Am J Transl Res ; 5(3): 254-68, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23634237

RESUMEN

The discovery of chromosomal translocations in prostate cancer has greatly enhanced our understanding of prostate cancer biology. Genomic rearrangements involving the ETS family of transcription factors are estimated to be present in 50-70% of prostate cancer cases. These rearrangements fuse the ETS factors with promoters of genes that are androgen regulated. Thus, the expression of ETS factors, such as ERG, ETV1, ETV4 and ETV5, is mediated by androgen. In-vitro and in-vivo studies suggest that overexpression of ETS proteins increase cell proliferation and confer an invasive phenotype to prostate cancer cells. Epidemiological studies demonstrate that ETS-fusion positive patients exhibit tumors corresponding to a more advanced disease. The ability of ETS factors to serve as markers for screening and diagnosing prostate cancer patients is being investigated, and the results have been largely positive to date. Additionally, ETS factors present an excellent opportunity as therapeutic targets and several strategies have been devised to directly target ETS proteins or their binding partners and downstream effectors.

7.
Neoplasia ; 14(4): 297-310, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22577345

RESUMEN

We previously associated the cytoskeleton linker protein, Ezrin, with the metastatic phenotype of pediatric sarcomas, including osteosarcoma and rhabdomyosarcoma. These studies have suggested that Ezrin contributes to the survival of cancer cells after their arrival at secondary metastatic locations. To better understand this role in metastasis, we undertook two noncandidate analyses of Ezrin function including a microarray subtraction of high-and low-Ezrin-expressing cells and a proteomic approach to identify proteins that bound the N-terminus of Ezrin in tumor lysates. Functional analyses of these data led to a novel and unifying hypothesis that Ezrin contributes to the efficiency of metastasis through regulation of protein translation. In support of this hypothesis, we found Ezrin to be part of the ribonucleoprotein complex to facilitate the expression of complex messenger RNA in cells and to bind with poly A binding protein 1 (PABP1; PABPC1). The relevance of these findings was supported by our identification of Ezrin and components of the translational machinery in pseudopodia of highly metastatic cells during the process of cell invasion. Finally, two small molecule inhibitors recently shown to inhibit the Ezrin metastatic phenotype disrupted the Ezrin/PABP1 association. Taken together, these results provide a novel mechanistic basis by which Ezrin may contribute to metastasis.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Invasividad Neoplásica , Biosíntesis de Proteínas/fisiología , Western Blotting , Línea Celular , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoprecipitación , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , ARN Interferente Pequeño , Espectrometría de Masas en Tándem , Transfección
8.
PLoS One ; 6(4): e19343, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559405

RESUMEN

BACKGROUND: Genomic rearrangements involving the ETS family of transcription factors occur in 40-70% of prostate cancer cases. ERG and ETV1 are the most common ETS members observed in these genetic alterations. The high prevalence of these rearrangements and their biological significance represents a novel therapeutic target for the treatment of prostate cancer. METHODS AND FINDINGS: We recently reported the development of YK-4-279, a small molecule inhibitor of EWS-FLI1 oncoprotein in Ewing's Sarcoma. Since ERG and ETV1 belong to the same class of ETS factors as FLI1, we tested the ability of YK-4-279 to inhibit biological functions of ERG and ETV1 proteins in prostate cancer. YK-4-279 inhibited ERG and ETV1 mediated transcriptional activity in a luciferase assay. YK-4-279 also decreased ERG and ETV1 downstream target mRNA and protein expression in ETV1-fusion positive LNCaP and ERG fusion positive VCaP cells. YK-4-279 reduced the motility of LNCaP cells in a scratch assay and the invasive phenotype of both LNCaP and VCaP cells in a HUVEC invasion assay. Fusion-negative PC3 cells were unresponsive to YK-4-279. SiRNA mediated ERG knockdown in VCaP cells resulted in a loss of drug responsiveness. Concurrently, transient ERG expression in PC-3 cells resulted in increased invasive potential, which was reduced by YK-4-279. CONCLUSION: These data demonstrate that YK-4-279 inhibits ERG and ETV1 biological activity in fusion-positive prostate cancer cells leading to decreased motility and invasion. Therefore, YK-4-279 may have an impact on metastasis in prostate cancer and it may be further evaluated for its clinical applications in prostate cancer in addition to Ewing's sarcoma.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al ADN/metabolismo , Indoles/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Inmunoprecipitación de Cromatina , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Cinética , Masculino , Invasividad Neoplásica , Regulador Transcripcional ERG
9.
J Vis Exp ; (50)2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21490581

RESUMEN

Metastatic dissemination of malignant cells requires degradation of basement membrane, attachment of tumor cells to vascular endothelium, retraction of endothelial junctions and finally invasion and migration of tumor cells through the endothelial layer to enter the bloodstream as a means of transport to distant sites in the host(1-3). Once in the circulatory system, cancer cells adhere to capillary walls and extravasate to the surrounding tissue to form metastatic tumors(4,5). The various components of tumor cell-endothelial cell interaction can be replicated in vitro by challenging a monolayer of human umbilical vein endothelial cells (HUVEC) with cancer cells. Studies performed with electron and phase-contrast microscopy suggest that the in vitro sequence of events fairly represent the in vivo metastatic process(6). Here, we describe an electrical-impedance based technique that monitors and quantifies in real-time the invasion of endothelial cells by malignant tumor cells. Giaever and Keese first described a technique for measuring fluctuations in impedance when a population of cells grow on the surface of electrodes(7,8). The xCELLigence instrument, manufactured by Roche, utilizes a similar technique to measure changes in electrical impedance as cells attach and spread in a culture dish covered with a gold microelectrode array that covers approximately 80% of the area on the bottom of a well. As cells attach and spread on the electrode surface, it leads to an increase in electrical impedance(9-12). The impedance is displayed as a dimensionless parameter termed cell-index, which is directly proportional to the total area of tissue-culture well that is covered by cells. Hence, the cell-index can be used to monitor cell adhesion, spreading, morphology and cell density. The invasion assay described in this article is based on changes in electrical impedance at the electrode/cell interphase, as a population of malignant cells invade through a HUVEC monolayer (Figure 1). The disruption of endothelial junctions, retraction of endothelial monolayer and replacement by tumor cells lead to large changes in impedance. These changes directly correlate with the invasive capacity of tumor cells, i.e., invasion by highly aggressive cells lead to large changes in cell impedance and vice versa. This technique provides a two-fold advantage over existing methods of measuring invasion, such as boyden chamber and matrigel assays: 1) the endothelial cell-tumor cell interaction more closely mimics the in vivo process, and 2) the data is obtained in real-time and is more easily quantifiable, as opposed to end-point analysis for other methods.


Asunto(s)
Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/patología , Electrofisiología/métodos , Células Endoteliales/patología , Osteosarcoma/irrigación sanguínea , Osteosarcoma/patología , Comunicación Celular/fisiología , Impedancia Eléctrica , Electrofisiología/instrumentación , Humanos , Invasividad Neoplásica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...