Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 96(5): 816-25, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25865493

RESUMEN

Transcription factors operate in developmental processes to mediate inductive events and cell competence, and perturbation of their function or regulation can dramatically affect morphogenesis, organogenesis, and growth. We report that a narrow spectrum of amino-acid substitutions within the transactivation domain of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a leucine zipper-containing transcription factor of the AP1 superfamily, profoundly affect development. Seven different de novo missense mutations involving conserved residues of the four GSK3 phosphorylation motifs were identified in eight unrelated individuals. The distinctive clinical phenotype, for which we propose the eponym Aymé-Gripp syndrome, is not limited to lens and eye defects as previously reported for MAF/Maf loss of function but includes sensorineural deafness, intellectual disability, seizures, brachycephaly, distinctive flat facial appearance, skeletal anomalies, mammary gland hypoplasia, and reduced growth. Disease-causing mutations were demonstrated to impair proper MAF phosphorylation, ubiquitination and proteasomal degradation, perturbed gene expression in primary skin fibroblasts, and induced neurodevelopmental defects in an in vivo model. Our findings nosologically and clinically delineate a previously poorly understood recognizable multisystem disorder, provide evidence for MAF governing a wider range of developmental programs than previously appreciated, and describe a novel instance of protein dosage effect severely perturbing development.


Asunto(s)
Catarata/genética , Sordera/genética , Glucógeno Sintasa Quinasa 3/genética , Discapacidad Intelectual/genética , Proteínas Proto-Oncogénicas c-maf/genética , Catarata/patología , Síndrome de Down/genética , Síndrome de Down/patología , Humanos , Discapacidad Intelectual/patología , Mutación , Fenotipo , Fosforilación , Convulsiones/genética , Convulsiones/patología
2.
Hum Mol Genet ; 23(19): 5243-50, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24858910

RESUMEN

Retinoblastoma is a non-hereditary as well as an inherited pediatric tumor of the developing retina resulting from the inactivation of both copies of the RB1 tumor suppressor gene. Familial retinoblastoma is a highly penetrant genetic disease that usually develops by carrying germline mutations that inactivate one allele of the RB1 gene, leading to multiple retinoblastomas. However, large and complete germline RB1 deletions are associated with low or no tumor risk for reasons that remain unknown. In this study, we define a minimal genomic region associated with this low penetrance. This region encompasses few genes including MED4 a subunit of the mediator complex. We further show that retinoblastoma RB1 -/- cells cannot survive in the absence of MED4, both in vitro and in orthotopic xenograft models in vivo, therefore identifying MED4 as a survival gene in retinoblastoma. We propose that the contiguous loss of the adjacent retinoblastoma gene, MED4, explains the low penetrance in patients with large deletions that include both RB1 and MED4. Our findings also point to another synthetic lethal target in tumors with inactivated RB1 and highlight the importance of collateral damage in carcinogenesis.


Asunto(s)
Eliminación de Gen , Complejo Mediador/genética , Penetrancia , Proteína de Retinoblastoma/genética , Retinoblastoma/genética , Animales , Apoptosis/genética , Muerte Celular/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Hibridación Genómica Comparativa , Femenino , Expresión Génica , Técnicas de Inactivación de Genes , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Linaje , Interferencia de ARN , Retinoblastoma/mortalidad , Retinoblastoma/patología , Ensayo de Tumor de Célula Madre
5.
Neuro Oncol ; 25(5): 899-912, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36273330

RESUMEN

BACKGROUND: Intensive chemotherapeutic regimens with craniospinal irradiation have greatly improved survival in medulloblastoma patients. However, survival markedly differs among molecular subgroups and their biomarkers are unknown. Through unbiased screening, we found Schlafen family member 11 (SLFN11), which is known to improve response to DNA damaging agents in various cancers, to be one of the top prognostic markers in medulloblastomas. Hence, we explored the expression and functions of SLFN11 in medulloblastoma. METHODS: SLFN11 expression for each subgroup was assessed by immunohistochemistry in 98 medulloblastoma patient samples and by analyzing transcriptomic databases. We genetically or epigenetically modulated SLFN11 expression in medulloblastoma cell lines and determined cytotoxic response to the DNA damaging agents cisplatin and topoisomerase I inhibitor SN-38 in vitro and in vivo. RESULTS: High SLFN11 expressing cases exhibited significantly longer survival than low expressing cases. SLFN11 was highly expressed in the WNT-activated subgroup and in a proportion of the SHH-activated subgroup. While WNT activation was not a direct cause of the high expression of SLFN11, a specific hypomethylation locus on the SLFN11 promoter was significantly correlated with high SLFN11 expression. Overexpression or deletion of SLFN11 made medulloblastoma cells sensitive and resistant to cisplatin and SN-38, respectively. Pharmacological upregulation of SLFN11 by the brain-penetrant histone deacetylase-inhibitor RG2833 markedly increased sensitivity to cisplatin and SN-38 in SLFN11-negative medulloblastoma cells. Intracranial xenograft studies also showed marked sensitivity to cisplatin by SLFN11-overexpression in medulloblastoma cells. CONCLUSIONS: High SLFN11 expression is one factor which renders favorable outcomes in WNT-activated and a subset of SHH-activated medulloblastoma possibly through enhancing response to cisplatin.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Humanos , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/genética , Cisplatino/farmacología , Regulación hacia Arriba , Irinotecán , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/genética , Epigénesis Genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas Nucleares/metabolismo
6.
Nat Commun ; 14(1): 6669, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37863903

RESUMEN

Atypical teratoid rhabdoid tumors (ATRT) are divided into MYC, TYR and SHH subgroups, suggesting diverse lineages of origin. Here, we investigate the imaging of human ATRT at diagnosis and the precise anatomic origin of brain tumors in the Rosa26-CreERT2::Smarcb1flox/flox model. This cross-species analysis points to an extra-cerebral origin for MYC tumors. Additionally, we clearly distinguish SHH ATRT emerging from the cerebellar anterior lobe (CAL) from those emerging from the basal ganglia (BG) and intra-ventricular (IV) regions. Molecular characteristics point to the midbrain-hindbrain boundary as the origin of CAL SHH ATRT, and to the ganglionic eminence as the origin of BG/IV SHH ATRT. Single-cell RNA sequencing on SHH ATRT supports these hypotheses. Trajectory analyses suggest that SMARCB1 loss induces a de-differentiation process mediated by repressors of the neuronal program such as REST, ID and the NOTCH pathway.


Asunto(s)
Neoplasias Encefálicas , Tumor Rabdoide , Teratoma , Humanos , Tumor Rabdoide/genética , Multiómica , Proteína SMARCB1/genética , Factores de Transcripción/genética , Neoplasias Encefálicas/genética , Diagnóstico por Imagen , Teratoma/patología , Proteínas Hedgehog/genética
7.
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
8.
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
9.
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
10.
Clin Cancer Res ; 26(21): 5735-5746, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32900798

RESUMEN

PURPOSE: Medulloblastoma is an important cause of mortality and morbidity in pediatric oncology. Here, we investigated whether the DNA repair inhibitor, AsiDNA, could help address a significant unmet clinical need in medulloblastoma care, by improving radiotherapy efficacy without increasing radiation-associated toxicity. EXPERIMENTAL DESIGN: To evaluate the brain permeability of AsiDNA upon systemic delivery, we intraperitoneally injected a fluorescence form of AsiDNA in models harboring brain tumors and in models still in development. Studies evaluated toxicity associated with combination of AsiDNA with radiation in the treatment of young developing animals at subacute levels, related to growth and development, and at chronic levels, related to brain organization and cognitive skills. Efficacy of the combination of AsiDNA with radiation was tested in two different preclinical xenografted models of high-risk medulloblastoma and in a panel of medulloblastoma cell lines from different molecular subgroups and TP53 status. Role of TP53 on the AsiDNA-mediated radiosensitization was analyzed by RNA-sequencing, DNA repair recruitment, and cell death assays. RESULTS: Capable of penetrating young brain tissues, AsiDNA showed no added toxicity to radiation. Combination of AsiDNA with radiotherapy improved the survival of animal models more efficiently than increasing radiation doses. Medulloblastoma radiosensitization by AsiDNA was not restricted to a specific molecular group or status of TP53. Molecular mechanisms of AsiDNA, previously observed in adult malignancies, were conserved in pediatric models and resembled dose increase when combined with irradiation. CONCLUSIONS: Our results suggest that AsiDNA is an attractive candidate to improve radiotherapy in medulloblastoma, with no indication of additional toxicity in developing brain tissues.


Asunto(s)
ADN/farmacología , Meduloblastoma/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/farmacología , Proteína p53 Supresora de Tumor/genética , Adulto , Animales , Línea Celular Tumoral , Niño , ADN/efectos adversos , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Xenoinjertos , Humanos , Masculino , Meduloblastoma/genética , Meduloblastoma/patología , Meduloblastoma/radioterapia , Pediatría , RNA-Seq , Fármacos Sensibilizantes a Radiaciones/efectos adversos
11.
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
12.
Cancer Cell ; 36(6): 597-612.e8, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31708437

RESUMEN

Rhabdoid tumors (RTs) are genomically simple pediatric cancers driven by the biallelic inactivation of SMARCB1, leading to SWI/SNF chromatin remodeler complex deficiency. Comprehensive evaluation of the immune infiltrates of human and mice RTs, including immunohistochemistry, bulk RNA sequencing and DNA methylation profiling studies showed a high rate of tumors infiltrated by T and myeloid cells. Single-cell RNA (scRNA) and T cell receptor sequencing highlighted the heterogeneity of these cells and revealed therapeutically targetable exhausted effector and clonally expanded tissue resident memory CD8+ T subpopulations, likely representing tumor-specific cells. Checkpoint blockade therapy in an experimental RT model induced the regression of established tumors and durable immune responses. Finally, we show that one mechanism mediating RTs immunogenicity involves SMARCB1-dependent re-expression of endogenous retroviruses and interferon-signaling activation.


Asunto(s)
Ensamble y Desensamble de Cromatina/inmunología , Tumor Rabdoide/genética , Tumor Rabdoide/inmunología , Linfocitos T/inmunología , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Humanos , Inmunohistoquímica/métodos , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología
13.
EMBO Mol Med ; 10(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29463565

RESUMEN

FGFR3 alterations (mutations or translocation) are among the most frequent genetic events in bladder carcinoma. They lead to an aberrant activation of FGFR3 signaling, conferring an oncogenic dependence, which we studied here. We discovered a positive feedback loop, in which the activation of p38 and AKT downstream from the altered FGFR3 upregulates MYC mRNA levels and stabilizes MYC protein, respectively, leading to the accumulation of MYC, which directly upregulates FGFR3 expression by binding to active enhancers upstream from FGFR3 Disruption of this FGFR3/MYC loop in bladder cancer cell lines by treatment with FGFR3, p38, AKT, or BET bromodomain inhibitors (JQ1) preventing MYC transcription decreased cell viability in vitro and tumor growth in vivo A relevance of this loop to human bladder tumors was supported by the positive correlation between FGFR3 and MYC levels in tumors bearing FGFR3 mutations, and the decrease in FGFR3 and MYC levels following anti-FGFR treatment in a PDX model bearing an FGFR3 mutation. These findings open up new possibilities for the treatment of bladder tumors displaying aberrant FGFR3 activation.


Asunto(s)
Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Azepinas/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Triazoles/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
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
15.
Cancer Cell ; 34(3): 379-395.e7, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30205043

RESUMEN

The current consensus recognizes four main medulloblastoma subgroups (wingless, Sonic hedgehog, group 3 and group 4). While medulloblastoma subgroups have been characterized extensively at the (epi-)genomic and transcriptomic levels, the proteome and phosphoproteome landscape remain to be comprehensively elucidated. Using quantitative (phospho)-proteomics in primary human medulloblastomas, we unravel distinct posttranscriptional regulation leading to highly divergent oncogenic signaling and kinase activity profiles in groups 3 and 4 medulloblastomas. Specifically, proteomic and phosphoproteomic analyses identify aberrant ERBB4-SRC signaling in group 4. Hence, enforced expression of an activated SRC combined with p53 inactivation induces murine tumors that resemble group 4 medulloblastoma. Therefore, our integrative proteogenomics approach unveils an oncogenic pathway and potential therapeutic vulnerability in the most common medulloblastoma subgroup.


Asunto(s)
Neoplasias Cerebelosas/patología , Meduloblastoma/patología , Receptor ErbB-4/metabolismo , Familia-src Quinasas/metabolismo , Adolescente , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Cerebelo/patología , Niño , Preescolar , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Lactante , Masculino , Meduloblastoma/genética , Ratones , Ratones Transgénicos , Fosforilación , Proteoma/metabolismo , Proteómica/métodos , Transducción de Señal , Familia-src Quinasas/genética
17.
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.

18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA