Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Blood ; 143(21): 2166-2177, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38437728

RESUMEN

ABSTRACT: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy. Current treatments, based on intensive chemotherapy regimens provide overall survival rates of ∼85% in children and <50% in adults, calling the search of new therapeutic options. We previously reported that targeting the T-cell receptor (TCR) in T-ALL with anti-CD3 (αCD3) monoclonal antibodies (mAbs) enforces a molecular program akin to thymic negative selection, a major developmental checkpoint in normal T-cell development; induces leukemic cell death; and impairs leukemia progression to ultimately improve host survival. However, αCD3 monotherapy resulted in relapse. To find out actionable targets able to re-enforce leukemic cells' vulnerability to αCD3 mAbs, including the clinically relevant teplizumab, we identified the molecular program induced by αCD3 mAbs in patient-derived xenografts derived from T-ALL cases. Using large-scale transcriptomic analysis, we found prominent expression of tumor necrosis factor α (TNFα), lymphotoxin α (LTα), and multiple components of the "TNFα via NF-κB signaling" pathway in anti-CD3-treated T-ALL. We show in vivo that etanercept, a sink for TNFα/LTα, enhances αCD3 antileukemic properties, indicating that TNF/TNF receptor (TNFR) survival pathways interferes with TCR-induced leukemic cell death. However, suppression of TNF-mediated survival and switch to TNFR-mediated cell death through inhibition of cellular inhibitor of apoptosis protein-1/2 (cIAP1/2) with the second mitochondrial-derived activator of caspases (SMAC) mimetic birinapant synergizes with αCD3 to impair leukemia expansion in a receptor-interacting serine/threonine-protein kinase 1-dependent manner and improve mice survival. Thus, our results advocate the use of either TNFα/LTα inhibitors, or birinapant/other SMAC mimetics to improve anti-CD3 immunotherapy in T-ALL.


Asunto(s)
Complejo CD3 , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Factor de Necrosis Tumoral alfa , Humanos , Animales , Ratones , Complejo CD3/inmunología , Complejo CD3/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Inmunoterapia/métodos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico
2.
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
3.
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
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 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.

9.
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
10.
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
11.
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
12.
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).

13.
J Clin Invest ; 120(10): 3663-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20941850

RESUMEN

Accumulating evidence points to inflammation as a promoter of carcinogenesis. MyD88 is an adaptor molecule in TLR and IL-1R signaling that was recently implicated in tumorigenesis through proinflammatory mechanisms. Here we have shown that MyD88 is also required in a cell-autonomous fashion for RAS-mediated carcinogenesis in mice in vivo and for MAPK activation and transformation in vitro. Mechanistically, MyD88 bound to the key MAPK, Erk, and prevented its inactivation by its phosphatase, MKP3, thereby amplifying the activation of the canonical RAS pathway. The relevance of this mechanism to human neoplasia was suggested by the finding that MyD88 was overexpressed and interacted with activated Erk in primary human cancer tissues. Collectively, these results show that in addition to its role in inflammation, MyD88 plays what we believe to be a crucial direct role in RAS signaling, cell-cycle control, and cell transformation.


Asunto(s)
Transformación Celular Neoplásica , Inflamación/complicaciones , Factor 88 de Diferenciación Mieloide/fisiología , Transducción de Señal , Proteínas ras/fisiología , 9,10-Dimetil-1,2-benzantraceno , Animales , Ciclo Celular , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Fosforilación , Acetato de Tetradecanoilforbol
14.
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
15.
Mol Oncol ; 1(4): 425-30, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19383316

RESUMEN

With the aim to correlate BRAF mutation status with gene expression in human primary cutaneous melanomas, and thus to get more insight on the consequences of BRAF mutation on cell biology, we analyzed all expression data obtained in melanomas from which DNA was extracted from the same tissue slides that were used for the expression study. A cohort of 69 frozen primary melanoma whose oligonucleotide micro-array expression data were available, were genotyped for BRAF and NRAS genes. The expression data from these melanomas were re-analyzed according to BRAF mutational status. A set of 250 probes representing 209 genes that were significantly (raw P< or =0.001) associated with BRAF mutation status was identified and 17 of these were previously shown to be implicated in cutaneous melanoma progression or pigmentation pathway-associated genes driven by the microphthalmia transcription factor (MITF). The list of 34 top probes contained no more than 1% of false discoveries with a probability of 0.95. Among the genes that differentiated most strongly between BRAF mutated and non-mutated melanomas, there were those involved in melanoma immune response such as MAGE-D2, CD63, and HSP70. These findings support the immunogenicity of BRAF(V600E), eliciting patients T-cell responses in various in vitro assays. The genes whose expression is associated with BRAF mutations are not simply restricted to the MAPK/ERK signaling but also converge to enhanced immune responsiveness, cell motility and melanosomes processing involved in the adaptative UV response.


Asunto(s)
Perfilación de la Expresión Génica , Melanoma/genética , Mutación Missense , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Movimiento Celular/genética , Niño , Preescolar , Análisis Mutacional de ADN , Sondas de ADN/normas , Femenino , Regulación Neoplásica de la Expresión Génica , Genes ras/genética , Humanos , Inmunidad/genética , Lactante , Masculino , Melanoma/patología , Melanosomas , Persona de Mediana Edad , Rayos Ultravioleta , Adulto Joven
16.
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
17.
AIDS Res Hum Retroviruses ; 22(7): 630-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16831087

RESUMEN

HIV-1 Vpr is a 96-amino acid auxiliary protein that performs numerous activities during viral infection. In the present study, 10 antibodies were generated after mice immunization with either the N- or the C-terminus domain of Vpr, respectively, Vpr(1-51) and Vpr(52-96). ELISA and immunoblot experiments using pure synthetic overlapping Vpr peptides suggested that these anti-Vpr antibodies could be classified into five groups and that they recognized conformational or linear Vpr epitopes. Further analysis revealed the effect of C-terminal arginine mutations on the antibody binding. Two of the antibodies precipitated Vpr expressed after transfection of a Vpr-encoding vector in human cells. More importantly, one of them was able to detect Vpr in HIV-1-infected U1 cells and in HIV-1-infected human PBMC. Surface plasmon resonance experiments demonstrated that some of these antibodies prevented the interaction between Vpr and one of its cellular partners, the adenine nucleotide translocator. Thus, these anti-Vpr monoclonal antibodies may be useful to any laboratory working on the molecular mechanism of HIV-1 infection.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Productos del Gen vpr/inmunología , VIH-1/inmunología , Leucocitos Mononucleares/virología , Animales , Anticuerpos Monoclonales/inmunología , Mapeo Epitopo/métodos , Femenino , Humanos , Inmunoprecipitación/métodos , Ratones , Ratones Endogámicos BALB C , Resonancia por Plasmón de Superficie , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
18.
Protein Sci ; 14(2): 375-86, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15659370

RESUMEN

Gag protein oligomerization, an essential step during virus assembly, results in budding of spherical virus particles. This process is critically dependent on the spacer p2, located between the capsid and the nucleocapsid proteins. P2 contributes also, in association with NCp7, to specific recognition of the HIV-1 packaging signal resulting in viral genome encapsidation. There is no structural information about the 20 last amino acids of the C-terminal part of capsid (CA[CTD]) and p2, in the molecular mechanism of Gag assembly. In this study the structure of a peptide encompassing the 14 residues of p2 with the upstream 21 residues and the downstream 13 residues was determined by (1)H NMR in 30% trifluoroethanol (TFE). The main structural motif is a well-defined amphipathic alpha-helix including p2, the seven last residues of the CA(CTD), and the two first residues of NCp7. Peptides containing the p2 domain have a strong tendency to aggregate in solution, as shown by gel filtration analyses in pure H(2)O. To take into account the aggregation phenomena, models of dimer and trimer formed through hydrophobic or hydrophilic interfaces were constructed by molecular dynamic simulations. Gel shift experiments demonstrate that the presence of at least p2 and the 13 first residues of NCp7 is required for RNA binding. A computer-generated model of the Gag polyprotein segment (282-434)Gag interacting with the packaging element SL3 is proposed, illustrating the importance of p2 and NCp7 in genomic encapsidation.


Asunto(s)
Productos del Gen gag/química , VIH-1/química , Espectroscopía de Resonancia Magnética/métodos , Fragmentos de Péptidos/química , ARN Viral/química , Ensamble de Virus , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Cápside , Proteínas de la Cápside/química , Cromatografía en Gel , Bases de Datos de Proteínas , Dimerización , Genes gag , Genoma Viral , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , ARN/química , Programas Informáticos , Agua/química , Productos del Gen gag del Virus de la Inmunodeficiencia Humana
19.
EMBO J ; 21(15): 4070-80, 2002 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12145207

RESUMEN

Syncytia arising from the fusion of cells expressing the HIV-1-encoded Env gene with cells expressing the CD4/CXCR4 complex undergo apoptosis following the nuclear translocation of mammalian target of rapamycin (mTOR), mTOR-mediated phosphorylation of p53 on Ser15 (p53(S15)), p53-dependent upregulation of Bax and activation of the mitochondrial death pathway. p53(S15) phosphorylation is only detected in syncytia in which nuclear fusion (karyogamy) has occurred. Karyogamy is secondary to a transient upregulation of cyclin B and a mitotic prophase-like dismantling of the nuclear envelope. Inhibition of cyclin-dependent kinase-1 (Cdk1) prevents karyogamy, mTOR activation, p53(S15) phosphorylation and apoptosis. Neutralization of p53 fails to prevent karyogamy, yet suppresses apoptosis. Peripheral blood mononuclear cells from HIV-1-infected patients exhibit an increase in cyclin B and mTOR expression, correlating with p53(S15) phosphorylation and viral load. Cdk1 inhibition prevents the death of syncytia elicited by HIV-1 infection of primary CD4 lymphoblasts. Thus, HIV-1 elicits a pro-apoptotic signal transduction pathway relying on the sequential action of cyclin B-Cdk1, mTOR and p53.


Asunto(s)
Apoptosis/fisiología , Antígenos CD4/fisiología , Proteína Quinasa CDC2/fisiología , Núcleo Celular/fisiología , Productos del Gen env/fisiología , VIH-1/fisiología , Proteínas Quinasas/fisiología , Proteínas Proto-Oncogénicas c-bcl-2 , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/fisiología , Adulto , Terapia Antirretroviral Altamente Activa , Antígenos CD4/genética , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Núcleo Celular/ultraestructura , Perfilación de la Expresión Génica , Células Gigantes/citología , Infecciones por VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Células HeLa/citología , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Sustancias Macromoleculares , Fusión de Membrana , Mitocondrias/fisiología , Proteínas de Neoplasias/fisiología , Membrana Nuclear/fisiología , Membrana Nuclear/ultraestructura , Fosforilación , Fosfoserina/química , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/fisiología , Proteínas Recombinantes de Fusión/fisiología , Serina-Treonina Quinasas TOR , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Carga Viral , Proteína X Asociada a bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...