Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
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.
Cell Mol Life Sci ; 72(20): 3983-98, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25975225

RESUMEN

The family of RAF kinases transduces extracellular information to the nucleus, and their activation is crucial for cellular regulation on many levels, ranging from embryonic development to carcinogenesis. B-RAF and C-RAF modulate neurogenesis and neuritogenesis during chicken inner ear development. C-RAF deficiency in humans is associated with deafness in the rare genetic insulin-like growth factor 1 (IGF-1), Noonan and Leopard syndromes. In this study, we show that RAF kinases are expressed in the developing inner ear and in adult mouse cochlea. A homozygous C-Raf deletion in mice caused profound deafness with no evident cellular aberrations except for a remarkable reduction of the K(+) channel Kir4.1 expression, a trait that suffices as a cause of deafness. To explore the role of C-Raf in cellular protection and repair, heterozygous C-Raf (+/-) mice were exposed to noise. A reduced C-RAF level negatively affected hearing preservation in response to noise through mechanisms involving the activation of JNK and an exacerbated apoptotic response. Taken together, these results strongly support a role for C-RAF in hearing protection.


Asunto(s)
Oído Interno/metabolismo , Pérdida Auditiva/genética , Ruido , Proteínas Proto-Oncogénicas c-raf/fisiología , Animales , Apoptosis/genética , Cóclea/metabolismo , Oído Interno/embriología , Femenino , Pérdida Auditiva/metabolismo , Masculino , Ratones , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Transducción de Señal
3.
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
4.
Cancer Cell ; 12(2): 145-59, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17692806

RESUMEN

Progression of non-small-cell lung cancer (NSCLC) to metastasis is poorly understood. Two genetic approaches were used to evaluate the role of adherens junctions in a C-RAF driven mouse model for NSCLC: conditional ablation of the cdh1 gene and expression of dominant-negative (dn) E-cadherin. Disruption of E-cadherin caused massive formation of intratumoral vessels that was reversible in the early phase of induction. Vascularized tumors grew more rapidly, developed invasive fronts, and gave rise to micrometastasis. beta-catenin was identified as a critical effector of E-cadherin disruption leading to upregulation of VEGF-A and VEGF-C. In vivo, lung tumor cells with disrupted E-cadherin expressed beta-catenin target genes normally found in other endodermal lineages suggesting that reprogramming may be involved in metastatic progression.


Asunto(s)
Adenocarcinoma/secundario , Cadherinas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/secundario , Adhesión Celular , Neoplasias Pulmonares/irrigación sanguínea , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas c-raf/fisiología , Adenocarcinoma/etiología , Adenocarcinoma/metabolismo , Adenoma/etiología , Adenoma/patología , Uniones Adherentes , Animales , Antígenos CD , Apoptosis , Biomarcadores/metabolismo , Cadherinas/genética , Carcinoma de Pulmón de Células no Pequeñas/etiología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Células Cultivadas , Progresión de la Enfermedad , Endodermo/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Técnica del Anticuerpo Fluorescente , Genes Dominantes , Immunoblotting , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Luciferasas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-raf/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
5.
J Biol Chem ; 288(49): 35237-52, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24158441

RESUMEN

In metazoans, the highly conserved MAPK signaling pathway regulates cell fate decision. Aberrant activation of this pathway has been implicated in multiple human cancers and some developmental disorders. KSR1 functions as an essential scaffold that binds the individual components of the cascade and coordinates their assembly into multiprotein signaling platforms. The mechanism of KSR1 regulation is highly complex and not completely understood. In this study, we identified Tyr(728) as a novel regulatory phosphorylation site in KSR1. We show that Tyr(728) is phosphorylated by LCK, uncovering an additional and unexpected link between Src kinases and MAPK signaling. To understand how phosphorylation of Tyr(728) may regulate the role of KSR1 in signal transduction, we integrated structural modeling and biochemical studies. We demonstrate that Tyr(728) is involved in maintaining the conformation of the KSR1 kinase domain required for binding to MEK. It also affects phosphorylation and activation of MEK by RAF kinases and consequently influences cell proliferation. Moreover, our studies suggest that phosphorylation of Tyr(728) may affect the intrinsic kinase activity of KSR1. Together, we propose that phosphorylation of Tyr(728) may regulate the transition between the scaffolding and the catalytic function of KSR1 serving as a control point used to fine-tune cellular responses.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proliferación Celular , Activación Enzimática , Humanos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Conformación Proteica , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Tirosina/química
6.
Cancer Cell ; 9(1): 9-12, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16413467

RESUMEN

RAF research is booming since the discovery of mutant B-RAF in approximately 8% of human cancer. One reason for the excitement is the availability of RAF-targeted therapies. RAF inhibitors have been developed because RAF functions at a convergence point of signal transduction. Two recent papers by the groups of Rosen and Marais dramatically advance our understanding of RAF oncogenes in human tumors. The results confirm that the mitogenic cascade (RAF-MEK-ERK) is essential for RAF transformation, that RAF kinases work in concert, and that RAF-transformed cells are hooked on MEK, making them sensitive to growth inhibition by kinase inhibitors.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/fisiología , Transducción de Señal/fisiología , Quinasas raf/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática , Genes ras , Humanos , Mutación , Quinasas raf/antagonistas & inhibidores , Quinasas raf/genética
7.
J Biol Chem ; 287(34): 28445-55, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22711539

RESUMEN

Inhibitor of apoptosis (IAPs) proteins are characterized by the presence of evolutionarily conserved baculoviral inhibitor of apoptosis repeat (BIR) domains, predominantly known for their role in inhibiting caspases and, thereby, apoptosis. We have shown previously that multi-BIR domain-containing IAPs, cellular IAPs, and X-linked IAP can control tumor cell migration by directly regulating the protein stability of C-RAF kinase. Here, we extend our observations to a single BIR domain containing IAP family member melanoma-IAP (ML-IAP). We show that ML-IAP can directly bind to C-RAF and that ML-IAP depletion leads to an increase in C-RAF protein levels, MAPK activation, and cell migration in melanoma cells. Thus, our results unveil a thus far unknown role for ML-IAP in controlling C-RAF stability and cell migration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Proteínas Inhibidoras de la Apoptosis/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Estabilidad de Enzimas/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Melanoma/genética , Melanoma/patología , Proteínas de Neoplasias/genética , Unión Proteica/genética , Proteínas Proto-Oncogénicas c-raf/genética
8.
J Biol Chem ; 287(27): 23128-40, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22605333

RESUMEN

The maternally imprinted Ras-related tumor suppressor gene DiRas3 is lost or down-regulated in more than 60% of ovarian and breast cancers. The anti-tumorigenic effect of DiRas3 is achieved through several mechanisms, including inhibition of cell proliferation, motility, and invasion, as well as induction of apoptosis and autophagy. Re-expression of DiRas3 in cancer cells interferes with the signaling through Ras/MAPK and PI3K. Despite intensive research, the mode of interference of DiRas3 with the Ras/RAF/MEK/ERK signal transduction is still a matter of speculation. In this study, we show that DiRas3 associates with the H-Ras oncogene and that activation of H-Ras enforces this interaction. Furthermore, while associated with DiRas3, H-Ras is able to bind to its effector protein C-RAF. The resulting multimeric complex consisting of DiRas3, C-RAF, and active H-Ras is more stable than the two protein complexes H-Ras·C-RAF or H-Ras·DiRas3, respectively. The consequence of this complex formation is a DiRas3-mediated recruitment and anchorage of C-RAF to components of the membrane skeleton, suppression of C-RAF/B-RAF heterodimerization, and inhibition of C-RAF kinase activity.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Citoesqueleto/metabolismo , Dimerización , Genes Supresores de Tumor/fisiología , Humanos , Complejos Multiproteicos/metabolismo , Prenilación/fisiología , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al GTP rho/genética
9.
J Biol Chem ; 286(20): 17934-44, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21317286

RESUMEN

BAD (Bcl-2 antagonist of cell death) belongs to the proapoptotic BH3-only subfamily of Bcl-2 proteins. Physiological activity of BAD is highly controlled by phosphorylation. To further analyze the regulation of BAD function, we investigated the role of recently identified phosphorylation sites on BAD-mediated apoptosis. We found that in contrast to the N-terminal phosphorylation sites, the serines 124 and 134 act in an antiapoptotic manner because the replacement by alanine led to enhanced cell death. Our results further indicate that RAF kinases represent, besides PAK1, BAD serine 134 phosphorylating kinases. Importantly, in the presence of wild type BAD, co-expression of survival kinases, such as RAF and PAK1, leads to a strongly increased proliferation, whereas substitution of serine 134 by alanine abolishes this process. Furthermore, we identified BAD serine 134 to be strongly involved in survival signaling of B-RAF-V600E-containing tumor cells and found that phosphorylation of BAD at this residue is critical for efficient proliferation in these cells. Collectively, our findings provide new insights into the regulation of BAD function by phosphorylation and its role in cancer signaling.


Asunto(s)
Apoptosis , Proliferación Celular , Sistema de Señalización de MAP Quinasas , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Sustitución de Aminoácidos , Supervivencia Celular/genética , Células HEK293 , Células HeLa , Humanos , Mutación Missense , Neoplasias/genética , Fosforilación , Proteínas Proto-Oncogénicas B-raf/genética , Proteína Letal Asociada a bcl/genética , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
10.
J Biol Chem ; 286(18): 16491-503, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21454547

RESUMEN

The serine/threonine kinase RAF is a central component of the MAPK cascade. Regulation of RAF activity is highly complex and involves recruitment to membranes and association with Ras and scaffold proteins as well as multiple phosphorylation and dephosphorylation events. Previously, we identified by molecular modeling an interaction between the N-region and the RKTR motif of the kinase domain in RAF and assigned a new function to this tetrapeptide segment. Here we found that a single substitution of each basic residue within the RKTR motif inhibited catalytic activity of all three RAF isoforms. However, the inhibition and phosphorylation pattern of C-RAF and A-RAF differed from B-RAF. Furthermore, substitution of the first arginine led to hyperphosphorylation and accumulation of A-RAF and C-RAF in plasma membrane fraction, indicating that this residue interferes with the recycling process of A-RAF and C-RAF but not B-RAF. In contrast, all RAF isoforms behave similarly with respect to the RKTR motif-dependent dimerization. The exchange of the second arginine led to exceedingly increased dimerization as long as one of the protomers was not mutated, suggesting that substitution of this residue with alanine may result in similar a structural rearrangement of the RAF kinase domain, as has been found for the C-RAF kinase domain co-crystallized with a dimerization-stabilizing RAF inhibitor. In summary, we provide evidence that each of the basic residues within the RKTR motif is indispensable for correct RAF function.


Asunto(s)
Membrana Celular/enzimología , Mutación Missense , Multimerización de Proteína/fisiología , Quinasas raf/metabolismo , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Células COS , Membrana Celular/genética , Chlorocebus aethiops , Humanos , Estructura Terciaria de Proteína , Quinasas raf/genética
11.
Biochim Biophys Acta ; 1810(2): 162-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21081150

RESUMEN

BACKGROUND: BAD protein (Bcl-2 antagonist of cell death) belongs to the BH3-only subfamily of proapoptotic proteins and is proposed to function as the sentinel of the cellular health status. Physiological activity of BAD is regulated by phosphorylation, association with 14-3-3 proteins, binding to membrane lipids and pore formation. Since the functional role of the BAD C-terminal part has not been considered so far, we have investigated here the interplay of the structure and function of this region. METHODS: The structure of the regulatory C-terminal part of human BAD was analyzed by CD spectroscopy. The channel-forming activity of full-length BAD and BAD peptides was carried out by lipid bilayer measurements. Interactions between proteins and peptides were monitored by the surface plasmon resonance technique. In aqueous solution, C-terminal part of BAD exhibits a well-ordered structure and stable conformation. In a lipid environment, the helical propensity considerably increases. The interaction of the C-terminal segment of BAD with the isolated BH3 domain results in the formation of permanently open pores whereby the phosphorylation of serine 118 within the BH3 domain is necessary for effective pore formation. In contrast, phosphorylation of serine 99 in combination with 14-3-3 association suppresses formation of channels. C-terminal part of BAD controls BAD function by structural transitions, lipid binding and phosphorylation. Conformational changes of this region upon membrane interaction in conjunction with phosphorylation of the BH3 domain suggest a novel mechanism for regulation of BAD. GENERAL SIGNIFICANCE: Multiple signaling pathways mediate inhibition and activation of cell death via BAD.


Asunto(s)
Membrana Dobles de Lípidos/química , Conformación Proteica , Estructura Terciaria de Proteína , Proteína Letal Asociada a bcl/química , Proteínas 14-3-3/química , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Secuencia de Aminoácidos , Dicroismo Circular , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Membrana Dobles de Lípidos/metabolismo , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Resonancia por Plasmón de Superficie , Agua/química , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/metabolismo
12.
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
13.
Nat Cell Biol ; 7(8): 837-43, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16041367

RESUMEN

Ras proteins control the signalling pathways that are responsible for normal growth and malignant transformation. Raf protein kinases are direct Ras effector proteins that initiate the mitogen-activated protein kinase (MAPK) cascade, which mediates diverse biological functions such as cell growth, survival and differentiation. Here we show that prohibitin, a ubiquitously expressed and evolutionarily conserved protein is indispensable for the activation of the Raf-MEK-ERK pathway by Ras. The membrane targeting and activation of C-Raf by Ras needs prohibitin in vivo. In addition, direct interaction with prohibitin is required for C-Raf activation. C-Raf kinase fails to interact with the active Ras induced by epidermal growth factor in the absence of prohibitin. Moreover, in prohibitin-deficient cells the adhesion complex proteins cadherin and beta-catenin relocalize to the plasma membrane and thereby stabilize adherens junctions. Our data show an unexpected role of prohibitin in the activation of the Ras-Raf signalling pathway and in modulating epithelial cell adhesion and migration.


Asunto(s)
Movimiento Celular/fisiología , Células Epiteliales/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Represoras/fisiología , Quinasas raf/metabolismo , Proteínas ras/fisiología , Proteínas 14-3-3/metabolismo , Cadherinas/metabolismo , Caveolas/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Forma de la Célula/efectos de los fármacos , Forma de la Célula/genética , Proteínas del Citoesqueleto/metabolismo , Citosol/metabolismo , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/ultraestructura , Flavonoides/farmacología , Células HeLa , Humanos , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Prohibitinas , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transactivadores/metabolismo , Transfección , beta Catenina , Quinasas raf/genética , Proteínas ras/metabolismo
14.
Am J Respir Cell Mol Biol ; 45(4): 692-703, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21278326

RESUMEN

Fibrocytes comprise a recently described cell type of blood-derived, fibroblast-like cells that are recruited from the circulation to sites of wound repair, vascular remodeling, or fibrotic tissue remodeling. We recently showed that the stable prostacyclin analogue treprostinil, a clinically approved drug for pulmonary arterial hypertension (PAH), significantly reduced the recruitment of fibrocytes to sites of vascular remodeling in experimental hypoxic pulmonary hypertension. Here we report on the molecular mechanism underlying the inhibitory action of treprostinil on the adhesion and differentiation of human fibrocytes. Human fibrocytes expressed the prostanoid receptors, prostaglandin I (IP) receptors and prostaglandin E subtype receptors (EP2 and EP4). The generation of intracellular cyclic adenosine monophosphate (cAMP) by treprostinil reduced the expression of the integrins CD49 and CD29 when freshly isolated human peripheral blood mononuclear cells were treated with treprostinil. Cell-matrix adhesion was significantly impaired by treatment with treprostinil. We present evidence for a treprostinil/cAMP-induced downstream suppression of extracellular regulated kinase (ERK) that is transmitted via a protein kinase A-independent pathway through Rap proteins, which sequester Ras. The resulting dephosphorylated state of c-Raf limits the activity of ERK. The cell-matrix adhesion assay with the ERK inhibitor further confirmed that the adhesion of fibrocytes was impaired. Thus our data suggest that treprostinil inhibits the adhesion and differentiation of fibrocytes by limiting the activity of ERK via the cAMP-Rap axis.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Epoprostenol/análogos & derivados , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Leucocitos Mononucleares/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas ras/metabolismo , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Epoprostenol/farmacología , Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipertensión Pulmonar Primaria Familiar , Humanos , Hipertensión Pulmonar/enzimología , Integrina alfa5/metabolismo , Integrina beta1/metabolismo , Leucocitos Mononucleares/enzimología , Mutación , Fosforilación , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptores de Prostaglandina/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Proteínas de Unión al GTP rap1/genética
15.
Immunol Cell Biol ; 89(7): 812-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21221124

RESUMEN

Macrophages exert a wide variety of functions, which necessitate a high level of plasticity on the chromatin level. In the work presented here, we analyzed the role of the polycomb group protein Bmi1 during the acute response of bone marrow derived macrophages (BMDM) to lipopolysaccharide (LPS). Unexpectedly, we observed that Bmi1 was rapidly induced at the protein level and transiently phosphorylated upon LPS treatment. The induction of Bmi1 was dependent on MAP-kinase signaling. LPS treatment of BMDM in the absence of Bmi1 resulted in a pronounced increase in expression of the anti-inflammatory cytokine interleukin-10 (IL-10). Our results identify Bmi1 as a repressor of IL-10 expression during macrophage activation.


Asunto(s)
Interleucina-10/biosíntesis , Activación de Macrófagos , Macrófagos/inmunología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Interleucina-10/metabolismo , Lipopolisacáridos/inmunología , Sistema de Señalización de MAP Quinasas , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Complejo Represivo Polycomb 1 , Interferencia de ARN , ARN Interferente Pequeño , Receptor Toll-Like 4/metabolismo
16.
BMC Microbiol ; 11: 163, 2011 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-21745384

RESUMEN

BACKGROUND: Specific cell targeting is an important, yet unsolved problem in bacteria-based therapeutic applications, like tumor or gene therapy. Here, we describe the construction of a novel, internalin A and B (InlAB)-deficient Listeria monocytogenes strain (Lm-spa+), which expresses protein A of Staphylococcus aureus (SPA) and anchors SPA in the correct orientation on the bacterial cell surface. RESULTS: This listerial strain efficiently binds antibodies allowing specific interaction of the bacterium with the target recognized by the antibody. Binding of Trastuzumab (Herceptin®) or Cetuximab (Erbitux®) to Lm-spa+, two clinically approved monoclonal antibodies directed against HER2/neu and EGFR/HER1, respectively, triggers InlAB-independent internalization into non-phagocytic cancer cell lines overexpressing the respective receptors. Internalization, subsequent escape into the host cell cytosol and intracellular replication of these bacteria are as efficient as of the corresponding InlAB-positive, SPA-negative parental strain. This specific antibody/receptor-mediated internalization of Lm-spa+ is shown in the murine 4T1 tumor cell line, the isogenic 4T1-HER2 cell line as well as the human cancer cell lines SK-BR-3 and SK-OV-3. Importantly, this targeting approach is applicable in a xenograft mouse tumor model after crosslinking the antibody to SPA on the listerial cell surface. CONCLUSIONS: Binding of receptor-specific antibodies to SPA-expressing L. monocytogenes may represent a promising approach to target L. monocytogenes to host cells expressing specific receptors triggering internalization.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Endocitosis , Listeria monocytogenes/patogenicidad , Proteínas de la Membrana/deficiencia , Proteína Estafilocócica A/metabolismo , Animales , Línea Celular Tumoral , Receptores ErbB/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Unión Proteica , Receptor ErbB-2/inmunología , Proteína Estafilocócica A/genética
17.
Cell Microbiol ; 12(7): 891-905, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20088950

RESUMEN

We have investigated how Bacillus anthracis lethal toxin (LT) triggers caspase-3 activation and the formation of thick actin cables in human endothelial cells. By DNA array analysis we show that LT has a major impact on the cell transcriptome and we identify key host genes involved in LT cytotoxic effects. Indeed, upregulation of TRAIL and downregulation of XIAP both participate in LT-induced caspase-3 activation. LT induces a downregulation of the immediate early gene and master regulator of transcription egr1. Importantly, its re-expression in LT-intoxicated cells blocks caspase-3 activation. In parallel, we found that the formation of actin cables induced by LT occurs in the absence of direct activation of RhoA/ROCK signalling. We show that knock-down of cortactin and rhophilin-2 under conditions of calponin-1 expression defines the minimal set of genes regulated by LT for actin cable formation. Together our data establish that the modulation of the cell transcriptome by LT plays a key role in triggering human endothelial cell toxicity.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Apoptosis/efectos de los fármacos , Caspasa 3/genética , Línea Celular , Células Endoteliales/citología , Regulación de la Expresión Génica/genética , Humanos , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Proteína Inhibidora de la Apoptosis Ligada a X/genética
18.
Oncoimmunology ; 10(1): 1885778, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33643696

RESUMEN

Non-small cell lung cancers (NSCLCs) establish a highly immunosuppressive tumor microenvironment supporting cancer growth. To interfere with cancer-mediated immunosuppression, selective immune-checkpoint inhibitors (ICIs) have been approved as a standard-of-care treatment for NSCLCs. However, the majority of patients poorly respond to ICI-based immunotherapies. Oncolytic viruses are amongst the many promising immunomodulatory treatments tested as standalone therapy or in combination with ICIs to improve therapeutic outcome. Previously, we demonstrated the oncolytic and immunomodulatory efficacy of low-pathogenic influenza Aviruses (IAVs) against NSCLCs in immunocompetent transgenic mice with alung-specific overexpression of active Raf kinase (Raf-BxB). IAV infection not only resulted in significant primary virus-induced oncolysis, but also caused afunctional reversion of tumor-associated macrophages (TAMs) comprising additional anti-cancer activity. Here we show that NSCLCs as well as TAMs and cytotoxic immune cells overexpress IC molecules of the PD-L2/PD-1 and B7-H3 signaling axes. Thus, we aimed to combine oncolytic IAV-infection with ICIs to exploit the benefits of both anti-cancer approaches. Strikingly, IAV infection combined with the novel B7-H3 ICI led to increased levels of M1-polarized alveolar macrophages and increased lung infiltration by cytotoxic Tlymphocytes, which finally resulted in significantly improved oncolysis of about 80% of existing tumors. In contrast, application of clinically approved α-PD-1 IC antibodies alone or in combination with oncolytic IAV did not provide additional oncolytic or immunomodulatory efficacy. Thus, individualized therapy with synergistically acting oncolytic IAV and B7-H3 ICI might be an innovative future approach to target NSCLCs that are resistant to approved ICIs in patients.


Asunto(s)
Gripe Humana , Neoplasias Pulmonares , Virus Oncolíticos , Orthomyxoviridae , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico , Pulmón , Neoplasias Pulmonares/terapia , Ratones , Virus Oncolíticos/genética , Microambiente Tumoral
19.
J Biol Chem ; 284(41): 28004-28020, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19667065

RESUMEN

BAD is a proapoptotic member of the Bcl-2 protein family that is regulated by phosphorylation in response to survival factors. Although much attention has been devoted to the identification of phosphorylation sites in murine BAD, little data are available with respect to phosphorylation of human BAD protein. Using mass spectrometry, we identified here besides the established phosphorylation sites at serines 75, 99, and 118 several novel in vivo phosphorylation sites within human BAD (serines 25, 32/34, 97, and 124). Furthermore, we investigated the quantitative contribution of BAD targeting kinases in phosphorylating serine residues 75, 99, and 118. Our results indicate that RAF kinases represent, besides protein kinase A, PAK, and Akt/protein kinase B, in vivo BAD-phosphorylating kinases. RAF-induced phosphorylation of BAD was reduced to control levels using the RAF inhibitor BAY 43-9006. This phosphorylation was not prevented by MEK inhibitors. Consistently, expression of constitutively active RAF suppressed apoptosis induced by BAD and the inhibition of colony formation caused by BAD could be prevented by RAF. In addition, using the surface plasmon resonance technique, we analyzed the direct consequences of BAD phosphorylation by RAF with respect to association with 14-3-3 and Bcl-2/Bcl-X(L) proteins. Phosphorylation of BAD by active RAF promotes 14-3-3 protein association, in which the phosphoserine 99 represented the major binding site. Finally, we show here that BAD forms channels in planar bilayer membranes in vitro. This pore-forming capacity was dependent on phosphorylation status and interaction with 14-3-3 proteins. Collectively, our findings provide new insights into the regulation of BAD function by phosphorylation.


Asunto(s)
Canales Iónicos/química , Canales Iónicos/metabolismo , Proteína Letal Asociada a bcl/química , Proteína Letal Asociada a bcl/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Canales Iónicos/genética , Membrana Dobles de Lípidos/metabolismo , Espectrometría de Masas , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Alineación de Secuencia , Proteína Letal Asociada a bcl/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Quinasas p21 Activadas/metabolismo , Quinasas raf/genética , Quinasas raf/metabolismo
20.
Cancer Res ; 80(19): 4199-4211, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32816854

RESUMEN

Although NF-κB is known to play a pivotal role in lung cancer, contributing to tumor growth, microenvironmental changes, and metastasis, the epigenetic regulation of NF-κB in tumor context is largely unknown. Here we report that the IKK2/NF-κB signaling pathway modulates metastasis-associated protein 2 (MTA2), a component of the nucleosome remodeling and deacetylase complex (NuRD). In triple transgenic mice, downregulation of IKK2 (Sftpc-cRaf-IKK2DN) in cRaf-induced tumors in alveolar epithelial type II cells restricted tumor formation, whereas activation of IKK2 (Sftpc-cRaf-IKK2CA) supported tumor growth; both effects were accompanied by altered expression of MTA2. Further studies employing genetic inhibition of MTA2 suggested that in primary tumor growth, independent of IKK2, MTA2/NuRD corepressor complex negatively regulates NF-κB signaling and tumor growth, whereas later dissociation of MTA2/NuRD complex from the promoter of NF-κB target genes and IKK2-dependent positive regulation of MTA2 leads to activation of NF-κB signaling, epithelial-mesenchymal transition, and lung tumor metastasis. These findings reveal a previously unrecognized biphasic role of MTA2 in IKK2/NF-κB-driven primary-to-metastatic lung tumor progression. Addressing the interaction between MTA2 and NF-κB would provide potential targets for intervention of tumor growth and metastasis. SIGNIFICANCE: These findings strongly suggest a prominent role of MTA2 in primary tumor growth, lung metastasis, and NF-κB signaling modulatory functions.


Asunto(s)
Histona Desacetilasas/metabolismo , Neoplasias Pulmonares/patología , FN-kappa B/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Histona Desacetilasas/genética , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inflamación/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/genética , Proteínas Represoras/genética , Transducción de Señal , Transactivadores/genética , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA