Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 6: 20471, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26839216

RESUMEN

Using a series of immunoprecipitation (IP)-tandem mass spectrometry (LC-MS/MS) experiments and reciprocal BLAST, we conducted a fly-human cross-species comparison of the phosphoinositide-3-kinase (PI3K) interactome in a drosophila S2R+ cell line and several NSCLC and human multiple myeloma cell lines to identify conserved interacting proteins to PI3K, a critical signaling regulator of the AKT pathway. Using H929 human cancer cells and drosophila S2R+ cells, our data revealed an unexpected direct binding of Corkscrew, the drosophila ortholog of the non-receptor protein tyrosine phosphatase type II (SHP2) to the Pi3k21B (p60) regulatory subunit of PI3K (p50/p85 human ortholog) but no association with Pi3k92e, the human ortholog of the p110 catalytic subunit. The p85-SHP2 association was validated in human cell lines, and formed a ternary regulatory complex with GRB2-associated-binding protein 2 (GAB2). Validation experiments with knockdown of GAB2 and Far-Western blots proved the direct interaction of SHP2 with p85, independent of adaptor proteins and transfected FLAG-p85 provided evidence that SHP2 binding on p85 occurred on the SH2 domains. A disruption of the SHP2-p85 complex took place after insulin/IGF1 stimulation or imatinib treatment, suggesting that the direct SHP2-p85 interaction was both independent of AKT activation and positively regulates the ERK signaling pathway.


Asunto(s)
Dípteros/metabolismo , Drosophila/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteómica/métodos , Animales , Línea Celular , Proteínas de Drosophila/metabolismo , Humanos , Proteínas de Insectos/metabolismo , Unión Proteica , Espectrometría de Masas en Tándem
2.
Elife ; 42015 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-25686219

RESUMEN

Treatment of EGFR-mutant lung cancer with erlotinib results in dramatic tumor regression but it is invariably followed by drug resistance. In characterizing early transcriptional changes following drug treatment of mutant EGFR-addicted cells, we identified the stem cell transcriptional regulator SOX2 as being rapidly and specifically induced, both in vitro and in vivo. Suppression of SOX2 sensitizes cells to erlotinib-mediated apoptosis, ultimately decreasing the emergence of acquired resistance, whereas its ectopic expression reduces drug-induced cell death. We show that erlotinib relieves EGFR-dependent suppression of FOXO6, leading to its induction of SOX2, which in turn represses the pro-apoptotic BH3-only genes BIM and BMF. Together, these observations point to a physiological feedback mechanism that attenuates oncogene addiction-mediated cell death associated with the withdrawal of growth factor signaling and may therefore contribute to the development of resistance.


Asunto(s)
Supervivencia Celular/fisiología , Receptores ErbB/antagonistas & inhibidores , Factores de Transcripción Forkhead/fisiología , Neoplasias Pulmonares/metabolismo , Factores de Transcripción SOXB1/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/fisiología , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Receptores ErbB/genética , Clorhidrato de Erlotinib/farmacología , Humanos , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/fisiología
3.
Cancer Res ; 72(13): 3228-37, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22552284

RESUMEN

The phosphoinositide 3-kinase (PI3K)/AKT and RAF/MEK/ERK signaling pathways are activated in a wide range of human cancers. In many cases, concomitant inhibition of both pathways is necessary to block proliferation and induce cell death and tumor shrinkage. Several feedback systems have been described in which inhibition of one intracellular pathway leads to activation of a parallel signaling pathway, thereby decreasing the effectiveness of single-agent targeted therapies. In this study, we describe a feedback mechanism in which MEK inhibition leads to activation of PI3K/AKT signaling in EGFR and HER2-driven cancers. We found that MEK inhibitor-induced activation of PI3K/AKT resulted from hyperactivation of ERBB3 as a result of the loss of an inhibitory threonine phosphorylation in the conserved juxtamembrane domains of EGFR and HER2. Mutation of this amino acid led to increased ERBB receptor activation and upregulation of the ERBB3/PI3K/AKT signaling pathway, which was no longer responsive to MEK inhibition. Taken together, these results elucidate an important, dominant feedback network regulating central oncogenic pathways in human cancer.


Asunto(s)
Retroalimentación , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Línea Celular , Activación Enzimática , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Proteínas Tirosina Quinasas Receptoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem
4.
Cancer Discov ; 2(3): 227-35, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22448344

RESUMEN

UNLABELLED: BRAF mutations occur in 10-15% of colorectal cancers (CRCs) and confer adverse outcome. While RAF inhibitors such as vemurafenib (PLX4032) have proven effective in BRAF mutant melanoma, they are surprisingly ineffective in BRAF mutant CRCs, and the reason for this disparity remains unclear. Compared to BRAF mutant melanoma cells, BRAF mutant CRC cells were less sensitive to vemurafenib, and P-ERK suppression was not sustained in response to treatment. Although transient inhibition of phospho-ERK by vemurafenib was observed in CRC, rapid ERK re-activation occurred through EGFR-mediated activation of RAS and CRAF. BRAF mutant CRCs expressed higher levels of phospho-EGFR than BRAF mutant melanomas, suggesting that CRCs are specifically poised for EGFR-mediated resistance. Combined RAF and EGFR inhibition blocked reactivation of MAPK signaling in BRAF mutant CRC cells and markedly improved efficacy in vitro and in vivo. These findings support evaluation of combined RAF and EGFR inhibition in BRAF mutant CRC patients. SIGNIFICANCE: BRAF valine 600 (V600) mutations occur in 10% to 15% of colorectal cancers, yet these tumors show a surprisingly low clinical response rate (~5%) to selective RAF inhibitors such as vemurafenib, which have produced dramatic response rates (60%­80%) in melanomas harboring the identical BRAF V600 mutation. We found that EGFR-mediated MAPK pathway reactivation leads to resistance to vemurafenib in BRAF-mutant colorectal cancers and that combined RAF and EGFR inhibition can lead to sustained MAPK pathway suppression and improved efficacy in vitro and in tumor xenografts.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Receptores ErbB/metabolismo , Indoles/farmacología , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/farmacología , Quinasas raf/antagonistas & inhibidores , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Células HT29 , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 71(18): 5965-75, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21775521

RESUMEN

Phosphatiditylinositide-3-kinase (PI3K) is activated in some cancers by direct mutation, but it is activated more commonly in cancer by mutation of upstream acting receptor tyrosine kinases (TK). At present, there is no systematic method to determine which TK signaling cascades activate PI3K in certain cancers, despite the likely utility of such information to help guide selection of tyrosine kinase inhibitor (TKI) drug strategies for personalized therapy. Here, we present a quantitative liquid chromatography tandem mass spectrometry approach that identifies upstream activators of PI3K both in vitro and in vivo. Using non-small cell lung carcinoma to illustrate this approach, we show a correct identification of the mechanism of PI3K activation in several models, thereby identifying the most appropriate TKI to downregulate PI3K signaling. This approach also determined the molecular mechanisms and adaptors required for PI3K activation following inhibition of the mTOR kinase TORC1. We further validated the approach in breast cancer cells with mutational activation of PIK3CA, where tandem mass spectrometry detected and quantitatively measured the abundance of a helical domain mutant (E545K) of PIK3CA connected to PI3K activation. Overall, our findings establish a mass spectrometric approach to identify functional interactions that govern PI3K regulation in cancer cells. Using this technique to define the pathways that activate PI3K signaling in a given tumor could help inform clinical decision making by helping guide personalized therapeutic strategies for different patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Neoplasias Pulmonares/enzimología , Espectrometría de Masas en Tándem/métodos , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Cromatografía Liquida/métodos , Fosfatidilinositol 3-Quinasa Clase I , Activación Enzimática , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones Desnudos , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Sirolimus/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Transfección
6.
Sci Transl Med ; 3(75): 75ra26, 2011 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-21430269

RESUMEN

Lung cancers harboring mutations in the epidermal growth factor receptor (EGFR) respond to EGFR tyrosine kinase inhibitors, but drug resistance invariably emerges. To elucidate mechanisms of acquired drug resistance, we performed systematic genetic and histological analyses of tumor biopsies from 37 patients with drug-resistant non-small cell lung cancers (NSCLCs) carrying EGFR mutations. All drug-resistant tumors retained their original activating EGFR mutations, and some acquired known mechanisms of resistance including the EGFR T790M mutation or MET gene amplification. Some resistant cancers showed unexpected genetic changes including EGFR amplification and mutations in the PIK3CA gene, whereas others underwent a pronounced epithelial-to-mesenchymal transition. Surprisingly, five resistant tumors (14%) transformed from NSCLC into small cell lung cancer (SCLC) and were sensitive to standard SCLC treatments. In three patients, serial biopsies revealed that genetic mechanisms of resistance were lost in the absence of the continued selective pressure of EGFR inhibitor treatment, and such cancers were sensitive to a second round of treatment with EGFR inhibitors. Collectively, these results deepen our understanding of resistance to EGFR inhibitors and underscore the importance of repeatedly assessing cancers throughout the course of the disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Genotipo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Receptores ErbB/genética , Clorhidrato de Erlotinib , Femenino , Gefitinib , Humanos , Masculino , Persona de Mediana Edad , Mutación , Fenotipo , Quinazolinas/uso terapéutico
7.
Clin Cancer Res ; 16(14): 3523-5, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20538763

RESUMEN

HER2 amplification and PIK3CA mutation were validated as biomarkers for sensitivity to the single-agent phosphoinositide 3-kinase (PI3K) inhibitor, GDC-0941, in breast cancer models. A novel expression profile was developed to identify other breast cancers sensitive to PI3K inhibitors. These expression studies highlighted feedback networks connecting TORC1, PI3K, and mitogen-activated protein kinase (MAPK) pathways, and underscored the potential for combination therapies.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Indazoles/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Sulfonamidas/farmacología , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Receptor ErbB-2/metabolismo , Factores de Transcripción/metabolismo
8.
Cancer Cell ; 17(1): 77-88, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20129249

RESUMEN

MET amplification activates ERBB3/PI3K/AKT signaling in EGFR mutant lung cancers and causes resistance to EGFR kinase inhibitors. We demonstrate that MET activation by its ligand, HGF, also induces drug resistance, but through GAB1 signaling. Using high-throughput FISH analyses in both cell lines and in patients with lung cancer, we identify subpopulations of cells with MET amplification prior to drug exposure. Surprisingly, HGF accelerates the development of MET amplification both in vitro and in vivo. EGFR kinase inhibitor resistance, due to either MET amplification or autocrine HGF production, was cured in vivo by combined EGFR and MET inhibition. These findings highlight the potential to prospectively identify treatment naive, patients with EGFR-mutant lung cancer who will benefit from initial combination therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Resistencia a Antineoplásicos/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas/genética , Receptores de Factores de Crecimiento/genética , Transducción de Señal/fisiología , Animales , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Amplificación de Genes , Expresión Génica , Factor de Crecimiento de Hepatocito , Humanos , Hibridación Fluorescente in Situ , Neoplasias Pulmonares/metabolismo , Ratones , Mutación , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-met , Quinazolinonas/farmacología , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA