Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Cancer ; 14: 54, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25884419

RESUMEN

BACKGROUND: Squamous cell carcinoma (SCC) is the most common type of tongue and larynx cancer and a common type of lung cancer. In this study, we attempted to specifically evaluate the signaling pathway underlying HGF/Met induced EGFR ligand release in SSCs. The Met proto-oncogene encodes for a tyrosine kinase receptor which is often hyperactivated in human cancers. Met activation correlates with poor patient outcome. Several studies revealed a role of Met in receptor-crosstalk inducing either activation of other receptors, or inducing their resistance to targeted cancer treatments. In an epithelial tumor cell line screen we recently showed that the Met ligand HGF blocks the EGFR tyrosine kinase and at the same time activates transcriptional upregulation and accumulation in the supernatant of the EGFR ligand amphiregulin (Oncogene 32:3846-56, 2013). In the present work we describe the pathway responsible for the amphiregulin induction. FINDINGS: Amphiregulin is transcriptionally upregulated and is released into the supernatant. We show that Erk2 but not Erk1 mediates amphiregulin upregulation upon treatment with monocyte derived HGF. A siRNA knockdown of Erk2 completely abolishes amphiregulin release in squamous cell carcinomas. CONCLUSIONS: These results identify Erk2 as the key downstream signal transducer between Met activation and EGFR ligand upregulation in squamous cell carcinoma cell lines derived from tongue, larynx and lung.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Anfirregulina/metabolismo , Comunicación Celular , Línea Celular Tumoral , Células Endoteliales/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Modelos Biológicos , Comunicación Paracrina , Unión Proteica , Proto-Oncogenes Mas , Receptor ErbB-2/metabolismo
2.
J Biol Chem ; 289(39): 26804-26816, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25096573

RESUMEN

Tumors are often greatly dependent on signaling cascades promoting cell growth or survival and may become hypersensitive to inactivation of key components within these signaling pathways. Ras and RAF mutations found in human cancer confer constitutive activity to these signaling molecules thereby converting them into an oncogenic state. RAF dimerization is required for normal Ras-dependent RAF activation and is required for the oncogenic potential of mutant RAFs. Here we describe a new mouse model for lung tumor development to investigate the role of B-RAF in oncogenic C-RAF-mediated adenoma initiation and growth. Conditional elimination of B-RAF in C-RAF BxB-expressing embryonic alveolar epithelial type II cells did not block adenoma formation. However, loss of B-RAF led to significantly reduced tumor growth. The diminished tumor growth upon B-RAF inactivation was due to reduced cell proliferation in absence of senescence and increased apoptosis. Furthermore, B-RAF elimination inhibited C-RAF BxB-mediated activation of the mitogenic cascade. In line with these data, mutation of Ser-621 in C-RAF BxB abrogated in vitro the dimerization with B-RAF and blocked the ability to activate the MAPK cascade. Taken together these data indicate that B-RAF is an important factor in oncogenic C-RAF-mediated tumorigenesis.


Asunto(s)
Adenoma/enzimología , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/enzimología , Neoplasias Pulmonares/enzimología , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Alveolos Pulmonares/enzimología , Mucosa Respiratoria/enzimología , Adenoma/genética , Adenoma/patología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/genética , Alveolos Pulmonares/patología , Mucosa Respiratoria/patología
3.
Neoplasia ; 14(12): 1164-77, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23308049

RESUMEN

Although progenitor cells of the conducting airway have been spatially localized and some insights have been gained regarding their molecular phenotype, relatively little is known about the mechanisms regulating their maintenance, activation, and differentiation. This study investigates the potential roles of E-cadherin in mouse Clara cells, as these cells were shown to represent the progenitor/stem cells of the conducting airways and have been implicated as the cell of origin of human non-small cell lung cancer. Postnatal inactivation of E-cadherin affected Clara cell differentiation and compromised airway regeneration under injury conditions. In steady-state adult lung, overexpression of the dominant negative E-cadherin led to an expansion of the bronchiolar stem cells and decreased differentiation concomitant with canonical Wnt signaling activation. Expansion of the bronchiolar stem cell pool was associated with an incessant proliferation of neuroepithelial body.associated Clara cells that ultimately gave rise to bronchiolar hyperplasia. Despite progressive hyperplasia, only a minority of the mice developed pulmonary solid tumors, suggesting that the loss of E-cadherin function leads to tumor formation when additional mutations are sustained. The present study reveals that E-cadherin plays a critical role in the regulation of proliferation and homeostasis of the epithelial cells lining the conducting airways.


Asunto(s)
Bronquiolos/metabolismo , Bronquiolos/patología , Cadherinas/metabolismo , Neoplasias Pulmonares/metabolismo , Lesiones Precancerosas/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Células Madre/metabolismo , Animales , Cadherinas/genética , Diferenciación Celular , Proliferación Celular , Homeostasis , Hiperplasia , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Lesiones Precancerosas/patología , Regeneración , Células Madre/patología , Uteroglobina/metabolismo , Vía de Señalización Wnt
4.
PLoS One ; 6(12): e29093, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22194995

RESUMEN

Growth factor induced signaling cascades are key regulatory elements in tissue development, maintenance and regeneration. Perturbations of these cascades have severe consequences, leading to developmental disorders and neoplastic diseases. As a major function in signal transduction, activating mutations in RAF family kinases are the cause of human tumorigenesis, where B-RAF V600E has been identified as the prevalent mutant. In order to address the oncogenic function of B-RAF V600E, we have generated transgenic mice expressing the activated oncogene specifically in lung alveolar epithelial type II cells. Constitutive expression of B-RAF V600E caused abnormalities in alveolar epithelium formation that led to airspace enlargements. These lung lesions showed signs of tissue remodeling and were often associated with chronic inflammation and low incidence of lung tumors. The inflammatory cell infiltration did not precede the formation of the lung lesions but was rather accompanied with late tumor development. These data support a model where the continuous regenerative process initiated by oncogenic B-RAF-driven alveolar disruption provides a tumor-promoting environment associated with chronic inflammation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Transformación Celular Neoplásica/patología , Neoplasias Pulmonares/patología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Muerte Celular , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Colágeno/metabolismo , Transición Epitelial-Mesenquimal , Células Caliciformes/metabolismo , Células Caliciformes/patología , Humanos , Hiperplasia , Inmunohistoquímica , Inflamación/patología , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
5.
Neoplasia ; 13(11): 1005-18, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22131876

RESUMEN

Here we describe a novel conditional mouse lung tumor model for investigation of the pathogenesis of human lung cancer. On the basis of the frequent involvement of the Ras-RAF-MEK-ERK signaling pathway in human non-small cell lung carcinoma (NSCLC), we have explored the target cell availability, reversibility, and cell type specificity of transformation by oncogenic C-RAF. Targeting expression to alveolar type II cells or to Clara cells, the two likely precursors of human NSCLC, revealed differential tumorigenicity between these cells. Whereas expression of oncogenic C-RAF in alveolar type II cells readily induced multifocal macroscopic lung tumors independent of the developmental state, few tumors with type II pneumocytes features and incomplete penetrance were found when targeted to Clara cells. Induced tumors did not progress and were strictly dependent on the initiating oncogene. Deinduction of mice resulted in tumor regression due to autophagy rather than apoptosis. Induction of autophagic cell death in regressing lung tumors suggests the use of autophagy enhancers as a treatment choice for patients with NSCLC.


Asunto(s)
Autofagia/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Transformación Celular Neoplásica/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-raf/genética , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/fisiología , Expresión Génica/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones Transgénicos , Transfección , Trasplante Heterólogo , Carga Tumoral/genética , Células Tumorales Cultivadas
6.
PLoS One ; 4(6): e6029, 2009 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-19551151

RESUMEN

BACKGROUND: Metastasis is a process by which cancer cells learn to form satellite tumors in distant organs and represents the principle cause of death of patients with solid tumors. NSCLC is the most lethal human cancer due to its high rate of metastasis. METHODOLOGY/PRINCIPAL FINDINGS: Lack of a suitable animal model has so far hampered analysis of metastatic progression. We have examined c-MYC for its ability to induce metastasis in a C-RAF-driven mouse model for non-small-cell lung cancer. c-MYC alone induced frank tumor growth only after long latency at which time secondary mutations in K-Ras or LKB1 were detected reminiscent of human NSCLC. Combination with C-RAF led to immediate acceleration of tumor growth, conversion to papillary epithelial cells and angiogenic switch induction. Moreover, addition of c-MYC was sufficient to induce macrometastasis in liver and lymph nodes with short latency associated with lineage switch events. Thus we have generated the first conditional model for metastasis of NSCLC and identified a gene, c-MYC that is able to orchestrate all steps of this process. CONCLUSIONS/SIGNIFICANCE: Potential markers for detection of metastasis were identified and validated for diagnosis of human biopsies. These markers may represent targets for future therapeutic intervention as they include genes such as Gata4 that are exclusively expressed during lung development.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Hígado/metabolismo , Neoplasias Hepáticas/patología , Pulmón/citología , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Mutación , Proteínas Proto-Oncogénicas c-raf/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA