Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Commun Biol ; 5(1): 101, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35091687

RESUMEN

The MITF transcription factor and the RAS/RAF/MEK/ERK pathway are two interconnected main players in melanoma. Understanding how MITF activity is regulated represents a key question since its dynamic modulation is involved in the phenotypic plasticity of melanoma cells and their resistance to therapy. By investigating the role of ARAF in NRAS-driven mouse melanoma through mass spectrometry experiments followed by a functional siRNA-based screen, we unexpectedly identified MITF as a direct ARAF partner. Interestingly, this interaction is conserved among the RAF protein kinase family since BRAF/MITF and CRAF/MITF complexes were also observed in the cytosol of NRAS-mutated mouse melanoma cells. The interaction occurs through the kinase domain of RAF proteins. Importantly, endogenous BRAF/MITF complexes were also detected in BRAF-mutated human melanoma cells. RAF/MITF complexes modulate MITF nuclear localization by inducing an accumulation of MITF in the cytoplasm, thus negatively controlling its transcriptional activity. Taken together, our study highlights a new level of regulation between two major mediators of melanoma progression, MITF and the MAPK/ERK pathway, which appears more complex than previously anticipated.


Asunto(s)
Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo , Quinasas raf/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Factor de Transcripción Asociado a Microftalmía/genética , Quinasas raf/genética
2.
Nat Commun ; 13(1): 3284, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672398

RESUMEN

While apneas are associated with multiple pathological and fatal conditions, the underlying molecular mechanisms remain elusive. We report that a mutated form of the transcription factor Mafa (Mafa4A) that prevents phosphorylation of the Mafa protein leads to an abnormally high incidence of breath holding apneas and death in newborn Mafa4A/4A mutant mice. This apneic breathing is phenocopied by restricting the mutation to central GABAergic inhibitory neurons and by activation of inhibitory Mafa neurons while reversed by inhibiting GABAergic transmission centrally. We find that Mafa activates the Gad2 promoter in vitro and that this activation is enhanced by the mutation that likely results in increased inhibitory drives onto target neurons. We also find that Mafa inhibitory neurons are absent from respiratory, sensory (primary and secondary) and pontine structures but are present in the vicinity of the hypoglossal motor nucleus including premotor neurons that innervate the geniohyoid muscle, to control upper airway patency. Altogether, our data reveal a role for Mafa phosphorylation in regulation of GABAergic drives and suggest a mechanism whereby reduced premotor drives to upper airway muscles may cause apneic breathing at birth.


Asunto(s)
Apnea , Neuronas Motoras , Animales , Factores de Transcripción Maf de Gran Tamaño , Ratones , Neuronas Motoras/fisiología , Fosforilación , Regiones Promotoras Genéticas
3.
Mol Cell Biol ; 27(1): 31-43, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17074813

RESUMEN

The B-Raf proto-oncogene encodes several isoforms resulting from alternative splicing in the hinge region upstream of the kinase domain. The presence of exon 8b in the B2-Raf(8b) isoform and exon 9b in the B3-Raf(9b) isoform differentially regulates B-Raf by decreasing and increasing MEK activating and oncogenic activities, respectively. Using different cell systems, we investigated here the molecular basis of this regulation. We show that exons 8b and 9b interfere with the ability of the B-Raf N-terminal region to interact with and inhibit the C-terminal kinase domain, thus modulating the autoinhibition mechanism in an opposite manner. Exons 8b and 9b are flanked by two residues reported to down-regulate B-Raf activity upon phosphorylation. The S365A mutation increased the activity of all B-Raf isoforms, but the effect on B2-Raf(8b) was more pronounced. This was correlated to the high level of S365 phosphorylation in this isoform, whereas the B3-Raf(9b) isoform was poorly phosphorylated on this residue. In contrast, S429 was equally phosphorylated in all B-Raf isoforms, but the S429A mutation activated B2-Raf(8b), whereas it inhibited B3-Raf(9b). These results indicate that phosphorylation on both S365 and S429 participate in the differential regulation of B-Raf isoforms through distinct mechanisms. Finally, we show that autoinhibition and phosphorylation represent independent but convergent mechanisms accounting for B-Raf regulation by alternative splicing.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas B-raf/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Embrión de Pollo , Humanos , MAP Quinasa Quinasa 1/metabolismo , Datos de Secuencia Molecular , Células PC12 , Fosforilación , Unión Proteica , Isoformas de Proteínas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas B-raf/química , Ratas , Homología de Secuencia de Aminoácido
4.
Front Endocrinol (Lausanne) ; 11: 563267, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33101198

RESUMEN

Epidemiologic analyses have shed light on an association between type 2 diabetes (T2D) and pancreatic ductal adenocarcinoma (PDAC). Recent data also suggest a potential relationship between T2D and insulinoma. Under rare circumstances, type 1 diabetes (T1D) can also be implicated in tumorigenesis. The biological mechanisms underlying such relationships are extremely complex. Some genetic factors contributing to the development of T2D are shared with pancreatic exocrine and endocrine tumors. Obesity and overweight can also contribute to the initiation and severity of T2D, while aging may influence both endocrine and exocrine tumors. Finally, pharmacological treatments of T2D may have an impact on PDAC. On the other hand, some treatments for insulinoma can trigger diabetes. In the present minireview, we discuss the cellular and molecular mechanisms that could explain these interactions. This analysis may help to define new potential therapeutic strategies.


Asunto(s)
Envejecimiento/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulinoma/metabolismo , Obesidad/metabolismo , Neoplasias Pancreáticas/metabolismo , Envejecimiento/genética , Envejecimiento/patología , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Humanos , Insulinoma/genética , Insulinoma/patología , Obesidad/genética , Obesidad/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Riesgo
5.
EMBO Mol Med ; 11(8): e9830, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31328883

RESUMEN

Medulloblastoma (MB) is a pediatric tumor of the cerebellum divided into four groups. Group 3 is of bad prognosis and remains poorly characterized. While the current treatment involving surgery, radiotherapy, and chemotherapy often fails, no alternative therapy is yet available. Few recurrent genomic alterations that can be therapeutically targeted have been identified. Amplifications of receptors of the TGFß/Activin pathway occur at very low frequency in Group 3 MB. However, neither their functional relevance nor activation of the downstream signaling pathway has been studied. We showed that this pathway is activated in Group 3 MB with some samples showing a very strong activation. Beside genetic alterations, we demonstrated that an ActivinB autocrine stimulation is responsible for pathway activation in a subset of Group 3 MB characterized by high PMEPA1 levels. Importantly, Galunisertib, a kinase inhibitor of the cognate receptors currently tested in clinical trials for Glioblastoma patients, showed efficacy on orthotopically grafted MB-PDX. Our data demonstrate that the TGFß/Activin pathway is active in a subset of Group 3 MB and can be therapeutically targeted.


Asunto(s)
Comunicación Autocrina , Neoplasias Cerebelosas/metabolismo , Subunidades beta de Inhibinas/metabolismo , Meduloblastoma/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidades beta de Inhibinas/genética , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/genética , Meduloblastoma/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Fosforilación , Pirazoles/farmacología , Quinolinas/farmacología , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta3/genética , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Nat Med ; 24(12): 1877-1886, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30374200

RESUMEN

Preventing the immune escape of tumor cells by blocking inhibitory checkpoints, such as the interaction between programmed death ligand-1 (PD-L1) and programmed death-1 (PD-1) receptor, is a powerful anticancer approach. However, many patients do not respond to checkpoint blockade. Tumor PD-L1 expression is a potential efficacy biomarker, but the complex mechanisms underlying its regulation are not completely understood. Here, we show that the eukaryotic translation initiation complex, eIF4F, which binds the 5' cap of mRNAs, regulates the surface expression of interferon-γ-induced PD-L1 on cancer cells by regulating translation of the mRNA encoding the signal transducer and activator of transcription 1 (STAT1) transcription factor. eIF4F complex formation correlates with response to immunotherapy in human melanoma. Pharmacological inhibition of eIF4A, the RNA helicase component of eIF4F, elicits powerful antitumor immune-mediated effects via PD-L1 downregulation. Thus, eIF4A inhibitors, in development as anticancer drugs, may also act as cancer immunotherapies.


Asunto(s)
Antígeno B7-H1/genética , Factor 4F Eucariótico de Iniciación/genética , Melanoma/terapia , Factor de Transcripción STAT1/genética , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno B7-H1/uso terapéutico , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia , Interferón gamma/genética , Interferón gamma/inmunología , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/uso terapéutico , Biosíntesis de Proteínas , Transducción de Señal/efectos de los fármacos , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología
7.
Cancer Cell ; 33(3): 435-449.e6, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29533784

RESUMEN

Cancer cells often express differentiation programs unrelated to their tissue of origin, although the contribution of these aberrant phenotypes to malignancy is poorly understood. An aggressive subgroup of medulloblastoma, a malignant pediatric brain tumor of the cerebellum, expresses a photoreceptor differentiation program normally expressed in the retina. We establish that two photoreceptor-specific transcription factors, NRL and CRX, are master regulators of this program and are required for tumor maintenance in this subgroup. Beyond photoreceptor lineage genes, we identify BCL-XL as a key transcriptional target of NRL and provide evidence substantiating anti-BCL therapy as a rational treatment opportunity for select MB patients. Our results highlight the utility of studying aberrant differentiation programs in cancer and their potential as selective therapeutic vulnerabilities.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proteínas del Ojo/genética , Proteínas de Homeodominio/genética , Meduloblastoma/genética , Transactivadores/genética , Animales , Diferenciación Celular/genética , Neoplasias Cerebelosas/genética , Humanos , Ratones Desnudos , Retina/patología , Transcripción Genética/genética
8.
Mol Cell Biol ; 23(6): 1983-93, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12612072

RESUMEN

It is widely thought that the biological outcomes of Raf-1 activation are solely attributable to the activation of the MEK/extracellular signal-regulated kinase (ERK) pathway. However, an increasing number of reports suggest that some Raf-1 functions are independent of this pathway. In this report we show that mutation of the amino-terminal 14-3-3 binding site of Raf-1 uncouples its ability to activate the MEK/ERK pathway from the induction of cell transformation and differentiation. In NIH 3T3 fibroblasts and COS-1 cells, mutation of serine 259 resulted in Raf-1 proteins which activated the MEK/ERK pathway as efficiently as v-Raf. However, in contrast to v-Raf, RafS259 mutants failed to transform. They induced morphological alterations and slightly accelerated proliferation in NIH 3T3 fibroblasts but were not tumorigenic in mice and behaved like wild-type Raf-1 in transformation assays measuring loss of contact inhibition or anchorage-independent growth. Curiously, the RafS259 mutants inhibited focus induction by an activated MEK allele, suggesting that they can hyperactivate negative-feedback pathways. In primary cultures of postmitotic chicken neuroretina cells, RafS259A was able to sustain proliferation to a level comparable to that sustained by the membrane-targeted transforming Raf-1 protein, RafCAAX. In contrast, RafS259A was only a poor inducer of neurite formation in PC12 cells in comparison to RafCAAX. Thus, RafS259 mutants genetically separate MEK/ERK activation from the ability of Raf-1 to induce transformation and differentiation. The results further suggest that RafS259 mutants inhibit signaling pathways required to promote these biological processes.


Asunto(s)
Transformación Celular Neoplásica/genética , Quinasa 1 de Quinasa de Quinasa MAP , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas 14-3-3 , Células 3T3/metabolismo , Células 3T3/trasplante , Células 3T3/ultraestructura , Transporte Activo de Núcleo Celular , Alelos , Animales , Sitios de Unión , Células COS/metabolismo , Células COS/ultraestructura , Diferenciación Celular/genética , División Celular/efectos de los fármacos , Chlorocebus aethiops , Inhibición de Contacto , Activación Enzimática , Retroalimentación Fisiológica , Genes Reporteros , Ratones , Ratones SCID , Proteína Quinasa 3 Activada por Mitógenos , Mutagénesis Sitio-Dirigida , Células PC12/citología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-raf/fisiología , Ratas , Transfección , Tirosina 3-Monooxigenasa/metabolismo
9.
Mol Cell Oncol ; 4(6): e1344758, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29209643

RESUMEN

Using mouse genetics, we recently showed that BRAF has a critical role in initiation of NRAS-driven melanoma that cannot be compensated by CRAF. In contrast, RAF proteins display compensatory functions in fully established tumors and ARAF can sustain proliferation in the absence of BRAF and CRAF, highlighting an addiction to RAF signaling in NRAS-driven melanoma.

10.
Nat Commun ; 8: 15262, 2017 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-28497782

RESUMEN

NRAS and its effector BRAF are frequently mutated in melanoma. Paradoxically, CRAF but not BRAF was shown to be critical for various RAS-driven cancers, raising the question of the role of RAF proteins in NRAS-induced melanoma. Here, using conditional ablation of Raf genes in NRAS-induced mouse melanoma models, we investigate their contribution in tumour progression, from the onset of benign tumours to malignant tumour maintenance. We show that BRAF expression is required for ERK activation and nevi development, demonstrating a critical role in the early stages of NRAS-driven melanoma. After melanoma formation, single Braf or Craf ablation is not sufficient to block tumour growth, showing redundant functions for RAF kinases. Finally, proliferation of resistant cells emerging in the absence of BRAF and CRAF remains dependent on ARAF-mediated ERK activation. These results reveal specific and compensatory functions for BRAF and CRAF and highlight an addiction to RAF signalling in NRAS-driven melanoma.


Asunto(s)
Melanoma/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Sistema de Señalización de MAP Quinasas/genética , Melanoma/genética , Melanoma/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas ras/genética
11.
Cancer Res ; 63(12): 3061-5, 2003 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12810628

RESUMEN

A high frequency of activating BRAF somatic mutations have been identified recently in malignant melanoma and nevi indicating that BRAF activation could be an early and critical step in the initiation of melanocytic neoplasia. To determine whether BRAF mutations could be an earlier event occurring at the germline level, we screened the entire BRAF coding region for germline mutations in 80 independent melanoma-prone families or patients with multiple primary melanoma without a familial history. We identified 13 BRAF variants, 4 of which were silent mutations in coding regions and 9 nucleotide substitutions in introns. None of these BRAF variants segregated with melanoma in the 11 melanoma families studied. Moreover, there was no significant difference in the frequency of heterozygotes for BRAF variants between melanoma cases and controls when they were compared. Our data suggest that BRAF is unlikely to be a melanoma susceptibility gene.


Asunto(s)
Melanoma/genética , Proteínas Proto-Oncogénicas c-raf/fisiología , Neoplasias Cutáneas/genética , Segregación Cromosómica , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Familia , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Mutación de Línea Germinal , Humanos , Intrones/genética , Proteínas de Neoplasias/genética , Neoplasias Primarias Múltiples/genética , Neoplasias de Tejido Nervioso/genética , Síndromes Neoplásicos Hereditarios/genética , Nevo Pigmentado/genética , Mutación Puntual , Proteínas Proto-Oncogénicas B-raf , Proteínas Proto-Oncogénicas c-raf/genética
12.
FEBS Lett ; 579(17): 3547-54, 2005 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-15963504

RESUMEN

Basic-leucine zipper transcription factors of the Maf family are key regulators of various developmental and differentiation processes. We previously reported that the phosphorylation status of MafA is a critical determinant of its biological functions. Using Western blot and mass spectrometry analysis, we now show that MafA is phosphorylated by p38 MAP kinase and identify three phosphoacceptor sites: threonine 113 and threonine 57, evolutionarily conserved residues located in the transcription activating domain, and serine 272. Mutation of these residues severely impaired MafA biological activity. Furthermore, we show that p38 also phosphorylates MafB and c-Maf. Together, these findings suggest that the p38 MAP kinase pathway is a novel regulator of large Maf transcription factors.


Asunto(s)
Cristalino/enzimología , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Secuencia de Aminoácidos , Animales , Diferenciación Celular/genética , Pollos , Proteínas de Unión al ADN/fisiología , Humanos , Cristalino/citología , Ratones , Datos de Secuencia Molecular , Mutación , Fosforilación , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-maf , Codorniz , Serina/genética , Treonina/genética , Factores de Transcripción/genética
13.
Nat Commun ; 6: 5953, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25601028

RESUMEN

How metabolism regulators play roles during early development remains elusive. Here we show that PFKFB4 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4), a glycolysis regulator, is critical for controlling dorsal ectoderm global patterning in gastrulating frog embryos via a non-glycolytic function. PFKFB4 is required for dorsal ectoderm progenitors to proceed towards more specified fates including neural and non-neural ectoderm, neural crest or placodes. This function is mediated by Akt signalling, a major pathway that integrates cell homeostasis and survival parameters. Restoring Akt signalling rescues the loss of PFKFB4 in vivo. In contrast, glycolysis is not essential for frog development at this stage. Our study reveals the existence of a PFKFB4-Akt checkpoint that links cell homeostasis to the ability of progenitor cells to undergo differentiation, and uncovers glycolysis-independent functions of PFKFB4.


Asunto(s)
Embrión no Mamífero/embriología , Embrión no Mamífero/enzimología , Proteína Oncogénica v-akt/metabolismo , Fosfofructoquinasa-2/metabolismo , Animales , Glucólisis/genética , Glucólisis/fisiología , Proteína Oncogénica v-akt/genética , Fosfofructoquinasa-2/genética
14.
Gene Expr Patterns ; 4(1): 35-46, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14678826

RESUMEN

Maf proteins are basic-leucine zipper transcription factors belonging to the AP1 superfamily. Several developmental processes require Maf proteins yet, the redundancy or complementarity of their respective roles in common processes has been only partially investigated. We present for the first time a complete comparative analysis of maf gene expression patterns in vertebrates. Expression of c-maf, mafB/kreisler, mafA/L-maf, mafF, mafG and mafK was analyzed by whole-mount in situ hybridization within chick embryos and their extraembryonic tissues ranging from embryonic day (E) 1 to 7. We carefully examined the extent of overlap between distinct maf genes and report that the developing lens, kidney, pancreas and apoptotic zones of limb buds show sustained co-expression of large maf genes. Small maf genes also exhibit overlap, for example in the dermomyotome. We also describe so far unidentified sites of maf gene expression. mafA is found in the developing neural tube and dorsal root ganglia. c-maf hybridization is detected in the neuroretina, the notochord and the endothelium of extraembryonic blood vessels.


Asunto(s)
Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Proteínas Proto-Oncogénicas/genética , Animales , Embrión de Pollo , Gástrula/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Esbozos de los Miembros/embriología , Esbozos de los Miembros/metabolismo , Factor de Transcripción MafF , Factor de Transcripción MafK , Mesodermo/metabolismo , Proteínas Nucleares/genética , Páncreas/embriología , Páncreas/metabolismo , Sistema Nervioso Periférico/embriología , Sistema Nervioso Periférico/metabolismo , Proteínas Proto-Oncogénicas c-maf , Proteínas Represoras/genética , Retina/embriología , Retina/metabolismo , Médula Espinal/embriología , Médula Espinal/metabolismo
15.
Ann N Y Acad Sci ; 973: 145-8, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12485852

RESUMEN

We have recently reported that two Rho family GTPases, Rac1 and Cdc42, are intimately involved in the control of cell survival of murine fibroblasts linked to adherence to the extracellular matrix. Inhibition of either Rac1 or Cdc42 signaling in adherent cells mimics the loss of anchorage and efficiently induces apoptosis in both immortalized and primary cells. In both cases cell death is dependent on the wild-type p53 tumor suppressor and is accompanied by activation of endogenous p53. Here, we describe that the inhibition of Rac1 or Cdc42 signaling leads to MAPK ERK activation via a pathway involving PI(3)K, Akt, Raf, and MEK, but not Ras. The moderate level of ERK activation that accompanies anoikis is an essential component of proapoptotic signaling; whereas sustained, high-intensity ERK signaling promotes survival in the same experimental system.


Asunto(s)
Anoicis/fisiología , Apoptosis/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Activación Enzimática , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt , Proteína de Unión al GTP cdc42/antagonistas & inhibidores , Proteínas de Unión al GTP rho/fisiología
16.
Neuropeptides ; 37(2): 98-104, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12747941

RESUMEN

The vasoactive intestinal peptide (VIP) has been shown to regulate cell proliferation and differentiation in many cell types. We previously reported that this neuropeptide inhibited proliferation in HT29 adenocarcinoma cells cultured in serum-containing medium. In addition, it has been demonstrated that VIP induced a potent stimulation of intracellular cAMP production in these cells cultured either in the absence or in the presence of serum. We also demonstrated that VIP induced phosphorylation of the small GTPase Rap1 in these cancerogenous cells. In the present study, the effects of VIP on the proliferation of HT29 cells cultured in the absence of growth factors and various concomitant signalling events were investigated. Under serum-free conditions VIP stimulates HT29 cell proliferation and induced a time- and concentration-dependent ERK activation. Furthermore, VIP induced the activation of the small GTPase Rap1 and of a 95 kDa isoform of the serine/threonine kinase B-Raf. Ras GTPase is also activated in VIP-stimulated cells. We hypothesize that VIP-induced proliferation in HT29 adenocarcinoma cells may involve a cAMP-Rap1/Ras-B-Raf-ERK signalling pathway.


Asunto(s)
Transducción de Señal/efectos de los fármacos , Péptido Intestinal Vasoactivo/farmacología , Proteínas de Unión al GTP rap1/genética , Proteínas ras/genética , División Celular/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Células HT29 , Humanos , Indicadores y Reactivos , Plásmidos/genética
17.
J Nucleic Acids ; 2012: 639062, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22007291

RESUMEN

Among the 518 protein kinases encoded by the human kinome, several of them act as oncoproteins in human cancers. Like other eukaryotic genes, oncogenes encoding protein kinases are frequently subjected to alternative splicing in coding as well as noncoding sequences. In the present paper, we will illustrate how alternative splicing can significantly impact on the physiological functions of oncogenic protein kinases, as demonstrated by mouse genetic model studies. This includes examples of membrane-bound tyrosine kinases receptors (FGFR2, Ret, TrkB, ErbB4, and VEGFR) as well as cytosolic protein kinases (B-Raf). We will further discuss how regular alternative splicing events of these kinases are in some instances implicated in oncogenic processes during tumor progression (FGFR, TrkB, ErbB2, Abl, and AuroraA). Finally, we will present typical examples of aberrant splicing responsible for the deregulation of oncogenic kinases activity in cancers (AuroraB, Jak2, Kit, Met, and Ron).

18.
Cell Rep ; 2(4): 774-80, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23022482

RESUMEN

B-Raf and C-Raf kinases have emerged as critical players in melanoma. However, little is known about their role during development and homeostasis of the melanocyte lineage. Here, we report that knockout of B-raf and C-raf genes in this lineage results in normal pigmentation at birth with no defect in migration, proliferation, or differentiation of melanoblasts in mouse hair follicles. In contrast, the double raf knockout mice displayed hair graying resulting from a defect in cell-cycle entry of melanocyte stem cells (MSCs) and their subsequent depletion in the hair follicle bulge. Therefore, Raf signaling is dispensable for early melanocyte lineage development, but necessary for MSC maintenance.


Asunto(s)
Melanocitos/citología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Células Madre/citología , Animales , Diferenciación Celular , Linaje de la Célula , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Folículo Piloso/fisiología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas B-raf/deficiencia , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-raf/deficiencia , Proteínas Proto-Oncogénicas c-raf/genética , Transducción de Señal , Factor de Células Madre/metabolismo , Xenopus/crecimiento & desarrollo
19.
Clin Cancer Res ; 18(1): 263-72, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22096025

RESUMEN

PURPOSE: The emergence of skin tumors in patients treated with sorafenib or with more recent BRAF inhibitors is an intriguing and potentially serious event. We carried out a clinical, pathologic, and molecular study of skin lesions occurring in patients receiving sorafenib. EXPERIMENTAL DESIGN: Thirty-one skin lesions from patients receiving sorafenib were characterized clinically and pathologically. DNA extracted from the lesions was screened for mutation hot spots of HRAS, NRAS, KiRAS, TP53, EGFR, BRAF, AKT1, PI3KCA, TGFBR1, and PTEN. Biological effect of sorafenib was studied in vivo in normal skin specimen and in vitro on cultured keratinocytes. RESULTS: We observed a continuous spectrum of lesions: from benign to more inflammatory and proliferative lesions, all seemingly initiated in the hair follicles. Eight oncogenic HRAS, TGFBR1, and TP53 mutations were found in 2 benign lesions, 3 keratoacanthomas (KA) and 3 KA-like squamous cell carcinoma (SCC). Six of them correspond to the typical UV signature. Treatment with sorafenib led to an increased keratinocyte proliferation and a tendency toward increased mitogen-activated protein kinase (MAPK) pathway activation in normal skin. Sorafenib induced BRAF-CRAF dimerization in cultured keratinocytes and activated CRAF with a dose-dependent effect on MAP-kinase pathway activation and on keratinocyte proliferation. CONCLUSION: Sorafenib induces keratinocyte proliferation in vivo and a time- and dose-dependent activation of the MAP kinase pathway in vitro. It is associated with a spectrum of lesions ranging from benign follicular cystic lesions to KA-like SCC. Additional and potentially preexisting somatic genetic events, like UV-induced mutations, might influence the evolution of benign lesions to more proliferative and malignant tumors.


Asunto(s)
Bencenosulfonatos/efectos adversos , Mutación/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Piridinas/efectos adversos , Receptores de Factores de Crecimiento Transformadores beta/genética , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/genética , Proteína p53 Supresora de Tumor/genética , Adulto , Anciano , Antineoplásicos/efectos adversos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Células Cultivadas , Femenino , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Masculino , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Receptor Tipo I de Factor de Crecimiento Transformador beta , Transducción de Señal , Piel/efectos de los fármacos , Piel/efectos de la radiación , Neoplasias Cutáneas/diagnóstico , Sorafenib , Rayos Ultravioleta/efectos adversos , Quinasas raf/genética , Proteínas ras/genética
20.
PLoS One ; 5(12): e15272, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21203559

RESUMEN

The B-raf proto-oncogene exerts essential functions during development and adulthood. It is required for various processes, such as placental development, postnatal nervous system myelination and adult learning and memory. The mouse B-raf gene encodes several isoforms resulting from alternative splicing of exons 8b and 9b located in the hinge region upstream of the kinase domain. These alternative sequences modulate the biochemical and biological properties of B-Raf proteins. To gain insight into the physiological importance of B-raf alternative splicing, we generated two conditional knockout mice of exons 8b and 9b. Homozygous animals with a constitutive deletion of either exon are healthy and fertile, and survive up to 18 months without any visible abnormalities, demonstrating that alternative splicing is not essential for embryonic development and brain myelination. However, behavioural analyses revealed that expression of exon 9b-containing isoforms is required for B-Raf function in hippocampal-dependent learning and memory. In contrast, mice mutated on exon 8b are not impaired in this function. Interestingly, our results suggest that exon 8b is present only in eutherians and its splicing is differentially regulated among species.


Asunto(s)
Empalme Alternativo , Regulación de la Expresión Génica , Hipocampo/metabolismo , Aprendizaje , Memoria , Proteínas Proto-Oncogénicas B-raf/metabolismo , Animales , Exones , Miedo , Hipocampo/patología , Homocigoto , Ratones , Ratones Noqueados , Vaina de Mielina/química , Células 3T3 NIH , Filogenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA