Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 528(7583): 570-4, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26675719

RESUMEN

Variant rs351855-G/A is a commonly occurring single-nucleotide polymorphism of coding regions in exon 9 of the fibroblast growth factor receptor FGFR4 (CD334) gene (c.1162G>A). It results in an amino-acid change at codon 388 from glycine to arginine (p.Gly388Arg) in the transmembrane domain of the receptor. Despite compelling genetic evidence for the association of this common variant with cancers of the bone, breast, colon, prostate, skin, lung, head and neck, as well as soft-tissue sarcomas and non-Hodgkin lymphoma, the underlying biological mechanism has remained elusive. Here we show that substitution of the conserved glycine 388 residue to a charged arginine residue alters the transmembrane spanning segment and exposes a membrane-proximal cytoplasmic signal transducer and activator of transcription 3 (STAT3) binding site Y(390)-(P)XXQ(393). We demonstrate that such membrane-proximal STAT3 binding motifs in the germline of type I membrane receptors enhance STAT3 tyrosine phosphorylation by recruiting STAT3 proteins to the inner cell membrane. Remarkably, such germline variants frequently co-localize with somatic mutations in the Catalogue of Somatic Mutations in Cancer (COSMIC) database. Using Fgfr4 single nucleotide polymorphism knock-in mice and transgenic mouse models for breast and lung cancers, we validate the enhanced STAT3 signalling induced by the FGFR4 Arg388-variant in vivo. Thus, our findings elucidate the molecular mechanism behind the genetic association of rs351855 with accelerated cancer progression and suggest that germline variants of cell-surface molecules that recruit STAT3 to the inner cell membrane are a significant risk for cancer prognosis and disease progression.


Asunto(s)
Membrana Celular/metabolismo , Mutación de Línea Germinal , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Exones/genética , Femenino , Técnicas de Sustitución del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Fosforilación , Fosfotirosina/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/química , Transducción de Señal
2.
Nature ; 507(7493): 508-12, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24553136

RESUMEN

Tumour metastasis is the primary cause of mortality in cancer patients and remains the key challenge for cancer therapy. New therapeutic approaches to block inhibitory pathways of the immune system have renewed hopes for the utility of such therapies. Here we show that genetic deletion of the E3 ubiquitin ligase Cbl-b (casitas B-lineage lymphoma-b) or targeted inactivation of its E3 ligase activity licenses natural killer (NK) cells to spontaneously reject metastatic tumours. The TAM tyrosine kinase receptors Tyro3, Axl and Mer (also known as Mertk) were identified as ubiquitylation substrates for Cbl-b. Treatment of wild-type NK cells with a newly developed small molecule TAM kinase inhibitor conferred therapeutic potential, efficiently enhancing anti-metastatic NK cell activity in vivo. Oral or intraperitoneal administration using this TAM inhibitor markedly reduced murine mammary cancer and melanoma metastases dependent on NK cells. We further report that the anticoagulant warfarin exerts anti-metastatic activity in mice via Cbl-b/TAM receptors in NK cells, providing a molecular explanation for a 50-year-old puzzle in cancer biology. This novel TAM/Cbl-b inhibitory pathway shows that it might be possible to develop a 'pill' that awakens the innate immune system to kill cancer metastases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Asesinas Naturales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/patología , Metástasis de la Neoplasia/inmunología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico , Femenino , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Masculino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/inmunología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Warfarina/farmacología , Warfarina/uso terapéutico , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
3.
Mol Cell ; 35(3): 291-304, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19683494

RESUMEN

We analyzed the G-actin-regulated transcriptome by gene expression analysis using previously characterized actin-binding drugs. We found many known MAL/MRTF-dependent target genes of serum response factor (SRF), as well as additional directly regulated genes. Surprisingly, several putative antiproliferative target genes were identified, including mig6/errfi-1, a negative regulator of the EGFR family. Mig6 induction occurred through actin-MAL-SRF signaling, and MAL was inducibly recruited to and activated a mig6 promoter element. Upregulation of Mig6 by lipid agonists such as LPA and S1P or actin drugs involved MAL and correlated with decreased activation of EGFR, MAPK/Erk, and c-fos. Mig6 depletion restored EGFR signaling and provided a proliferative advantage. Overexpression of MAL exhibited strong antiproliferative effects requiring the domains for SRF binding and transactivation, which supports antagonistic functions of MAL on growth-promoting signals. Our results show the existence of negatively acting transcriptional networks between pro- and antiproliferative signaling pathways toward SRF.


Asunto(s)
Actinas/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Receptores ErbB/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas de Transporte de Membrana/fisiología , Proteínas de la Mielina/fisiología , Proteolípidos/fisiología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular , Proliferación Celular , Cicloheximida/farmacología , Citocalasina D/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , ARN Mensajero/metabolismo , Tiazolidinas/farmacología , Proteínas Supresoras de Tumor
4.
Bioorg Med Chem Lett ; 26(2): 424-428, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26704265

RESUMEN

Activation of various interacting stress kinases, particularly the c-Jun N-terminal kinases (JNK), and a concomitant phosphorylation of insulin receptor substrate 1 (IRS-1) at serine 307 play a central role both in insulin resistance and in ß-cell dysfunction. IRS-1 phosphorylation is stimulated by elevated free fatty acid levels through different pathways in obesity. A series of novel pyrido[2,3-d]pyrimidin-7-one derivatives were synthesized as potential antidiabetic agents, preventing IRS-1 phosphorylation at serine 307 in a cellular model of lipotoxicity and type 2 diabetes.


Asunto(s)
Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Fosforilación/efectos de los fármacos , Pirimidinas/química , Pirimidinas/farmacología , Serina/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo
5.
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
6.
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
7.
Nat Rev Cancer ; 6(4): 321-30, 2006 04.
Artículo en Inglés | MEDLINE | ID: mdl-16557283

RESUMEN

Human polo-like kinase 1 (PLK1) is essential during mitosis and in the maintenance of genomic stability. PLK1 is overexpressed in human tumours and has prognostic potential in cancer, indicating its involvement in carcinogenesis and its potential as a therapeutic target. The use of different PLK1 inhibitors has increased our knowledge of mitotic regulation and allowed us to assess their ability to suppress tumour growth in vivo. We address the structural features of the kinase domain and the unique polo-box domain of PLK1 that are most suited for drug development and discuss our current understanding of the therapeutic potential of PLK1.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Silenciador del Gen , Neoplasias/terapia , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Predicción , Humanos , Mitosis , Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Quinasa Tipo Polo 1
8.
Mol Cancer ; 13: 13, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24461128

RESUMEN

The advent of effective targeted therapeutics has led to increasing emphasis on precise biomarkers for accurate patient stratification. Here, we describe the role of ACK1, a non-receptor tyrosine kinase in abrogating migration and invasion in KRAS mutant lung adenocarcinoma. Bosutinib, which inhibits ACK1 at 2.7 nM IC50, was found to inhibit cell migration and invasion but not viability in a panel of non-small cell lung cancer (NSCLC) cell lines. Knockdown of ACK1 abrogated bosutinib-induced inhibition of cell migration and invasion specifically in KRAS mutant cells. This finding was further confirmed in an in vivo zebrafish metastatic model. Tissue microarray data on 210 Singaporean lung adenocarcinomas indicate that cytoplasmic ACK1 was significantly over-expressed relative to paired adjacent non-tumor tissue. Interestingly, ACK1 expression in "normal" tissue adjacent to tumour, but not tumour, was independently associated with poor overall and relapse-free survival. In conclusion, inhibition of ACK1 with bosutinib attenuates migration and invasion in the context of KRAS mutant NSCLC and may fulfil a therapeutic niche through combinatorial treatment approaches.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Nitrilos/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Quinolinas/farmacología , Proteínas ras/genética , Animales , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Invasividad Neoplásica/patología , Proteínas Proto-Oncogénicas p21(ras) , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
9.
PLoS Genet ; 7(12): e1002400, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22174695

RESUMEN

Pituitary tumors are common intracranial neoplasms, yet few germline abnormalities have been implicated in their pathogenesis. Here we show that a single nucleotide germline polymorphism (SNP) substituting an arginine (R) for glycine (G) in the FGFR4 transmembrane domain can alter pituitary cell growth and hormone production. Compared with FGFR4-G388 mammosomatotroph cells that support prolactin (PRL) production, FGFR4-R388 cells express predominantly growth hormone (GH). Growth promoting effects of FGFR4-R388 as evidenced by enhanced colony formation was ascribed to Src activation and mitochondrial serine phosphorylation of STAT3 (pS-STAT3). In contrast, diminished pY-STAT3 mediated by FGFR4-R388 relieved GH inhibition leading to hormone excess. Using a knock-in mouse model, we demonstrate the ability of FGFR4-R385 to promote GH pituitary tumorigenesis. In patients with acromegaly, pituitary tumor size correlated with hormone excess in the presence of the FGFR4-R388 but not the FGFR4-G388 allele. Our findings establish a new role for the FGFR4-G388R polymorphism in pituitary oncogenesis, providing a rationale for targeting Src and STAT3 in the personalized treatment of associated disorders.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes src , Hormona del Crecimiento/genética , Neoplasias Hipofisarias/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Factor de Transcripción STAT3/genética , Serina/genética , Animales , Dasatinib , Regulación Neoplásica de la Expresión Génica , Técnicas de Sustitución del Gen , Hormona del Crecimiento/metabolismo , Humanos , Ratones , Mitocondrias/genética , Fosforilación , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Polimorfismo de Nucleótido Simple , Prolactina/genética , Prolactina/metabolismo , Pirimidinas/farmacología , Ratas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Serina/metabolismo , Tiazoles/farmacología
10.
Biochem Biophys Res Commun ; 442(3-4): 139-46, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23665325

RESUMEN

NEK9 is known to play a role in spindle assembly and in the control of centrosome separation, but the consequences of NEK9 targeting in cancer cells remain to be elucidated. In this study, we used siRNA to investigate the consequences of targeting NEK9 in glioblastoma and kidney cancer cells as a first step in assessing its potential as an anti-cancer therapeutic target. Live cell imaging revealed that NEK9 depletion of U1242 glioblastoma and Caki2 kidney carcinoma cells resulted in failure of cytokinesis. Interestingly, NEK9-depleted Caki2 cells overrode mitosis under incorrect chromosome alignment and were converted to a micronucleated phenotype, leading to cell death. Whereas, the RPE1 normal epithelium cell line was refractory to abnormal mitosis upon NEK9 knockdown. Nocodazole-induced mitotic arrest was compromised after NEK9 depletion, indicating that NEK9 has an important role in mitotic checkpoint system. Taken together, we propose that NEK9 inhibition represents a novel anti-cancer strategy by induction of mitotic catastrophe via impairment of spindle dynamics, cytokinesis and mitotic checkpoint control.


Asunto(s)
Neoplasias del Sistema Nervioso Central/terapia , Glioblastoma/terapia , Neoplasias Renales/terapia , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Terapia Molecular Dirigida , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Huso Acromático/fisiología , Apoptosis/genética , Apoptosis/fisiología , Línea Celular Tumoral , Citocinesis/genética , Citocinesis/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Puntos de Control de la Fase M del Ciclo Celular/genética , Micronúcleos con Defecto Cromosómico , Quinasas Relacionadas con NIMA , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Interferencia de ARN , Huso Acromático/genética
11.
BMC Cancer ; 13: 197, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23597096

RESUMEN

BACKGROUND: Tumor-associated macrophages (TAM) promote malignant progression, yet the repertoire of oncogenic factors secreted by TAM has not been clearly defined. We sought to analyze which EGFR- and STAT3-activating factors are secreted by monocytes/macrophages exposed to tumor cell-secreted factors. METHODS: Following exposure of primary human monocytes and macrophages to supernatants of a variety of tumor cell lines, we have analyzed transcript and secreted protein levels of EGFR family ligands and of STAT3 activators. To validate our findings, we have analyzed TAM infiltration levels, systemic and local protein levels as well as clinical data of primary breast cancer patients. RESULTS: Primary human monocytes and macrophages respond to tumor cell-derived factors by secreting EGFR- and STAT3-activating ligands, thus inducing two important oncogenic pathways in carcinoma cells. Tumor cell-secreted factors trigger two stereotype secretory profiles in peripheral blood monocytes and differentiated macrophages: monocytes secrete epiregulin (EREG) and oncostatin-M (OSM), while macrophages secrete heparin-binding EGF-like growth factor (HB-EGF) and OSM. HB-EGF and OSM cooperatively induce tumor cell chemotaxis. HB-EGF and OSM are co-expressed by TAM in breast carcinoma patients, and plasma levels of both ligands correlate strongly. Elevated HB-EGF levels accompany TAM infiltration, tumor growth and dissemination in patients with invasive disease. CONCLUSIONS: Our work identifies systemic markers for TAM involvement in cancer progression, with the potential to be developed into molecular targets in cancer therapy.


Asunto(s)
Neoplasias de la Mama/sangre , Carcinoma/sangre , Quimiotaxis , Factor de Crecimiento Epidérmico/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Oncostatina M/metabolismo , Neoplasias de la Mama/patología , Carcinoma/secundario , Línea Celular Tumoral , Células Cultivadas , Epirregulina , Femenino , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Metástasis Linfática , Macrófagos/metabolismo , Monocitos/metabolismo , Comunicación Paracrina , Factor de Transcripción STAT3/metabolismo
12.
Nat Med ; 12(5): 568-73, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648858

RESUMEN

The growing number of recently identified negative feedback regulators of receptor tyrosine kinases (RTKs) highlights the importance of signal attenuation and modulation for correct signaling outcome. Mitogen-inducible gene 6 (Mig6 also known as RALT or Gene 33) is a multiadaptor protein thought to be involved in the regulation of RTK and stress signaling. Here, we show that deletion of the mouse gene encoding Mig6 (designated Errfi1, which stands for ERBB receptor feedback inhibitor 1) causes hyperactivation of endogenous epidermal growth factor receptor (EGFR) and sustained signaling through the mitogen-activated protein kinase (MAPK) pathway, resulting in overproliferation and impaired differentiation of epidermal keratinocytes. Furthermore, Errfi1-/- mice develop spontaneous tumors in various organs and are highly susceptible to chemically induced formation of skin tumors. A tumor-suppressive role for Mig6 is supported by our finding that MIG6 is downregulated in various human cancers. Inhibition of endogenous Egfr signaling with the Egfr inhibitor gefitinib (Iressa) or replacement of wild-type Egfr with the kinase-deficient protein encoded by the hypomorphic Egfr(wa2) allele completely rescued skin defects in Erffi1-/- mice. Carcinogen-induced tumors displayed by Errfi1-/- mice were highly sensitive to gefitinib. These results indicate that Mig6 is a specific negative regulator of Egfr signaling in skin morphogenesis and is a novel tumor suppressor of Egfr-dependent carcinogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Receptores ErbB/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Morfogénesis , Neoplasias/metabolismo , Piel/crecimiento & desarrollo , Piel/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proliferación Celular , Gefitinib , Humanos , Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Quinazolinas/metabolismo , Piel/citología , Piel/metabolismo
13.
Int J Cancer ; 131(2): E45-55, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22020623

RESUMEN

Resistance to chemotherapeutic agents constitutes a major problem in the treatment of cancer. Over the past years, multi-targeted protein kinase inhibitors such as Gleevec, Sunitinib and Sorafenib are gaining wider acceptance for cancer treatment. These drugs show anti-tumor activity in vitro and in patients. Extended usage of these drugs in therapy commonly results in disease progression due to formation of resistance caused by rearrangements and accumulation of mutations in the unstable cancer cell genome. However, the underlying drug-specific mechanisms for the development of resistance remain elusive. Hence, a detailed understanding of the molecular genetic events involved in this processes is pivotal to counteract are not directly targeted by Sunitinib (unpublished data). Therefore, development of specific or multi-targeted inhibitors for these kinases for combinatorial therapy with e.g., an IL-8 neutralizing antibody might circumvent or substantially delay Sunitinib resistance formation and enhance survival prognosis. PRKX, TTBK2 and RSK4 expression. The specific reduction of these genes employing siRNA was sufficient to sensitize the kidney- and melanoma-cell lines against Sunitinib. In line with the elevated expression of PRKX, TTBK2 or RSK4, this sensitization effect was strikingly higher in the Sunitinib resistant cell lines, suggesting an expression-based mechanism of these genes to trigger Sunitinib resistance. Hence, we propose that PRKX, TTBK2 and RSK4 are potential resistance markers in Sunitinib therapy and might therefore represent targets for the development of novel strategies to overcome resistance.


Asunto(s)
Resistencia a Antineoplásicos/genética , Indoles/farmacología , Neoplasias Renales/genética , Melanoma/genética , Proteínas Serina-Treonina Quinasas/genética , Pirroles/farmacología , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Sunitinib
14.
Int J Cancer ; 131(12): 2808-19, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22511343

RESUMEN

A major obstacle in the successful treatment of cancer is the occurrence of chemoresistance. Cancer cells surviving chemotherapy and giving rise to a recurrence of the tumor are termed cancer stem cells and can be identified by elevated levels of certain stem cell markers. Eradication of this cell population is a priority objective in cancer therapy. Here, we report elevated levels of stem cell markers in MCF-7 mammospheres. Likewise, an upregulation of HER2 and its differential expression within individual cells of mammospheres was observed. Sorting for HER2(high) and HER2(low) cells revealed an upregulation of stem cell markers NANOG, OCT4 and SOX2 in the HER2(low) cell fraction. Accordingly, HER2(low) cells also showed reduced proliferation, ductal-like outgrowths and an increased number of colonies in matrigel. Xenografts from subcutaneously injected HER2(low) sorted cells exihibited earlier onset but slower growth of tumors and an increase in stem cell markers compared to tumors developed from the HER2(high) fraction. Treatment of mammospheres with salinomycin reduced the expression of SOX2 indicating a selective targeting of cancer stem cells. Trastuzumab however, did not reduce the expression of SOX2 in mammospheres. Furthermore, a combinatorial treatment of mammospheres with trastuzumab and salinomycin was superior to single treatment with each drug. Thus, targeting HER2 expressing tumors with anti-HER2 therapies will not necessarily eliminate cancer stem cells and may lead to a more aggressive cancer cell phenotype. Our study demonstrates efficient killing of both HER2 positive cells and cancer stem cells, hence opening a possibility for a new combinatorial treatment strategy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Receptor ErbB-2/metabolismo , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cartilla de ADN , Femenino , Humanos , Piranos/administración & dosificación , Piranos/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Trastuzumab
15.
Cancer Cell ; 1(2): 117-23, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12086869

RESUMEN

Cancer therapy directed at specific, frequently occurring molecular alterations in signaling pathways of cancer cells has been validated through the clinical development and regulatory approval of agents such as Herceptin for the treatment of advanced breast cancer and Gleevec for chronic myelogenous leukemia and gastrointestinal stromal tumors. While most novel, target-directed cancer drugs have pregenomic origins, one can anticipate a postgenomic wave of sophisticated "smart drugs" to fundamentally change the treatment of all cancers. With these prospects, interest in this new class of therapeutics extends from basic research scientists to practicing oncologists and their patients. An extension of the initial successes in molecular oncology will occur more quickly and successfully through an appreciation of lessons learned with the first group of agents in their progress through clinical development.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos , Inhibidores Enzimáticos/administración & dosificación , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Proteínas de Fusión bcr-abl , Humanos , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular
16.
Hepatology ; 51(4): 1383-90, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20044804

RESUMEN

UNLABELLED: The mitogen-inducible gene-6 (mig-6) is a multi-adaptor protein implicated in the regulation of the HER family of receptor tyrosine kinases. We have reported recently that mig-6 is a negative regulator of epidermal growth factor receptor (EGFR)-dependent skin morphogenesis and tumor formation in vivo. In the liver, ablation of mig-6 leads to an increase in EGFR protein levels, suggesting that mig-6 is a negative regulator of EGFR function. In line with this observation, primary hepatocytes isolated from mig-6 knockout and wild-type control mice display sustained mitogenic signaling in response to EGF. In order to explore the role of mig-6 in the liver in vivo, we analyzed liver regeneration in mig-6 knockout and wild-type control mice. Interestingly, mig-6 knockout mice display enhanced hepatocyte proliferation in the initial phases after partial hepatectomy. This phenotype correlates with activation of endogenous EGFR signaling, predominantly through the protein kinase B pathway. In addition, mig-6 is an endogenous inhibitor of EGFR signaling and EGF-induced tumor cell migration in human liver cancer cell lines. Moreover, mig-6 is down-regulated in human hepatocellular carcinoma and this correlates with increased EGFR expression. CONCLUSION: Our data implicate mig-6 as a regulator of EGFR activity in hepatocytes and as a suppressor of EGFR signaling in human liver cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Carcinoma Hepatocelular/patología , Receptores ErbB/fisiología , Hepatocitos/fisiología , Neoplasias Hepáticas/patología , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Proteínas Supresoras de Tumor
17.
Eur J Clin Invest ; 41(9): 987-94, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21382022

RESUMEN

BACKGROUND: Prostate disease incidence is expected to rise among developing nations secondary to increased prevalence of obesity and the elderly. Although many case-control studies have associated obesity to prostate cancer aggressiveness, few have correlated markers of prostate pathology to biomarkers of visceral obesity and insulin resistance, using an apparently healthy cohort. This study aims to fill this gap. MATERIALS AND METHODS: The 219 consenting adult Arab men, aged 30-70 years, were included in this cross-sectional study. Demographics were noted and anthropometrics measured. Fasting blood samples were extracted, and glycaemic and lipid profile were determined using routine laboratory methods. Serum adipocytokines and inflammatory markers were measured using multiplex assays. Total prostate-specific antigen (tPSA), free PSA (fPSA), parathyroid-related protein (PTHrP) and endoglin were measured using enzyme-linked immunosorbent assays. RESULTS: Serum triglycerides and waist-hip ratio (WHR) were significantly and positively associated with circulating (tPSA) levels in all subjects (P < 0·01). Systolic blood pressure (SBP), adiponectin, active plasminogen activator inhibitor-1 (aPAI-1) and insulin-like growth factor-1 (IGF-1) had significant inverse associations to tPSA. Stepwise linear regression revealed that adiponectin, IGF-1, WHR and PTHrP explained 30% of variance in tPSA levels (P < 0·0001), while SBP, resistin and BMI explained 18·7% of variance in endoglin (P = 0·001). CONCLUSIONS: The associations of adiponectin and WHR strengthen the link between insulin resistance and visceral adiposity to prostate volume markers among apparently healthy Arab men. Follow-up studies are needed to extend these preliminary findings so that early interventions can be provided to those at increased risk.


Asunto(s)
Biomarcadores/sangre , Obesidad Abdominal/sangre , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/sangre , Relación Cintura-Cadera , Adiponectina/sangre , Adulto , Anciano , Antígenos CD/sangre , Árabes , Índice de Masa Corporal , Estudios Transversales , Endoglina , Humanos , Resistencia a la Insulina/fisiología , Modelos Lineales , Masculino , Persona de Mediana Edad , Proteína Relacionada con la Hormona Paratiroidea/sangre , Receptores de Superficie Celular/sangre , Factores de Riesgo , Triglicéridos/sangre
18.
J Cell Biol ; 173(2): 301-10, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16636149

RESUMEN

The transactivation of enhanced growth factor receptor (EGFR) by G protein-coupled receptor (GPCR) ligands is recognized as an important signaling mechanism in the regulation of complex biological processes, such as cancer development. Estrogen (E2), which is a steroid hormone that is intimately implicated in breast cancer, has also been suggested to function via EGFR transactivation. In this study, we demonstrate that E2-induced EGFR transactivation in human breast cancer cells is driven via a novel signaling system controlled by the lipid kinase sphingosine kinase-1 (SphK1). We show that E2 stimulates SphK1 activation and the release of sphingosine 1-phosphate (S1P), by which E2 is capable of activating the S1P receptor Edg-3, resulting in the EGFR transactivation in a matrix metalloprotease-dependent manner. Thus, these findings reveal a key role for SphK1 in the coupling of the signals between three membrane-spanning events induced by E2, S1P, and EGF. They also suggest a new signal transduction model across three individual ligand-receptor systems, i.e., "criss-cross" transactivation.


Asunto(s)
Receptores ErbB/metabolismo , Estrógenos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Línea Celular Tumoral , Receptores ErbB/efectos de los fármacos , Femenino , Humanos , Modelos Biológicos , Activación Transcripcional
20.
Clin Cancer Res ; 15(6): 2058-66, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19240166

RESUMEN

PURPOSE: Establishment of antiapoptotic signaling pathways in tumor cells is a major cause for the failure of chemotherapy against cancer. To investigate the underlying mechanisms, we developed an experimental approach that is based on the genetic plasticity of cancer cells and the selection for cell survival on treatment with chemotherapeutic agents. EXPERIMENTAL DESIGN: Gene expression changes of surviving cell clones were analyzed by macroarrays. Involvement of fibroblast growth factor receptor 4 (FGFR4) in antiapoptotic pathways was elucidated by apoptosis assays, small interfering RNA experiments, and an antagonistic antibody. RESULTS: We show that FGFR4 gene expression is up-regulated in doxorubicin-treated, apoptosis-resistant cancer cell clones. Ectopic expression of FGFR4 in cancer cells led to reduced apoptosis sensitivity on treatment with doxorubicin or cyclophosphamide, whereas knockdown of endogenous FGFR4 expression in breast cancer cell lines had the opposite effect. FGFR4 overexpression resulted in Bcl-xl up-regulation at both mRNA and protein levels. Knockdown of FGFR4 expression by small interfering RNA caused a decrease in phospho-extracellular signal-regulated kinase 1/2 levels and reduced Bcl-xl expression. Moreover, an antagonistic FGFR4 antibody suppressed the resistance of cancer cells with endogenous FGFR4 expression against apoptosis-inducing chemotherapeutic agents. CONCLUSION: Based on these findings, we propose an antiapoptotic signaling pathway that is initiated by FGFR4 and regulating the expression of Bcl-xl through the mitogen-activated protein kinase cascade. Our findings are exemplary for a novel strategy toward the elucidation of diverse signaling pathways that define antiapoptotic potential in cancer cells. These observations open new avenues toward the diagnosis of chemoresistant tumors and therapies targeting FGFR4-overexpressing cancers.


Asunto(s)
Resistencia a Antineoplásicos , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/genética , Línea Celular Tumoral , Doxorrubicina/farmacología , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Humanos , Sistema de Señalización de MAP Quinasas , ARN Mensajero/análisis , Regulación hacia Arriba , Proteína bcl-X/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA