Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Cell Biol ; 8(11): 1246-54, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17013377

RESUMEN

Many proteins contain ubiquitin-binding domains or motifs (UBDs), such as the UIM (ubiquitin-interacting motif) and are referred to as ubiquitin receptors. Ubiquitin receptors themselves are frequently monoubiquitinated by a process that requires the presence of a UBD and is referred to as coupled monoubiquitination. Using a UIM-containing protein, eps15, as a model, we show here that coupled monoubiquitination strictly depends on the ability of the UIM to bind to monoubiquitin (mUb). We found that the underlying molecular mechanism is based on interaction between the UIM and a ubiquitin ligase (E3), which has itself been modified by ubiquitination. Furthermore, we demonstrate that the in vivo ubiquitination of members of the Nedd4 family of E3 ligases correlates with their ability to monoubiquitinate eps15. Thus, our results clarify the mechanism of coupled monoubiquitination and identify the ubiquitination of E3 ligases as a critical determinant in this process.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfoproteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sitios de Unión/genética , Proteínas de Unión al Calcio/genética , Catálisis , Complejos de Clasificación Endosomal Requeridos para el Transporte , Células HeLa , Humanos , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular/genética , Modelos Biológicos , Mutación/genética , Ubiquitina-Proteína Ligasas Nedd4 , Fosfoproteínas/genética , Unión Proteica , Transfección , Ubiquitina-Proteína Ligasas/genética
2.
Proc Natl Acad Sci U S A ; 107(14): 6459-64, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308550

RESUMEN

The phosphatase and tensin homolog (PTEN) is a tumor suppressor that is inactivated in many human cancers. PTEN loss has been associated with resistance to inhibitors of the epidermal growth factor receptor (EGFR), but the molecular basis of this resistance is unclear. It is believed that unopposed phosphatidylinositol-3-kinase (PI3K) activation through multiple receptor tyrosine kinases (RTKs) can relieve PTEN-deficient cancers from their "dependence" on EGFR or any other single RTK for survival. Here we report a distinct resistance mechanism whereby PTEN inactivation specifically raises EGFR activity by impairing the ligand-induced ubiquitylation and degradation of the activated receptor through destabilization of newly formed ubiquitin ligase Cbl complexes. PTEN-associated resistance to EGFR kinase inhibitors is phenocopied by expression of dominant negative Cbl and can be overcome by more complete EGFR kinase inhibition. PTEN inactivation does not confer resistance to inhibitors of the MET or PDGFRA kinase. Our study identifies a critical role for PTEN in EGFR signal termination and suggests that more potent EGFR inhibition should overcome resistance caused by PI3K pathway activation.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Fosfohidrolasa PTEN/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Apoptosis , Línea Celular , Activación Enzimática , Humanos , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Unión Proteica , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Ubiquitinación
3.
Mol Syst Biol ; 7: 462, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21245847

RESUMEN

The activity, localization and fate of many cellular proteins are regulated through ubiquitination, a process whereby one or more ubiquitin (Ub) monomers or chains are covalently attached to target proteins. While Ub-conjugated and Ub-associated proteomes have been described, we lack a high-resolution picture of the dynamics of ubiquitination in response to signaling. In this study, we describe the epidermal growth factor (EGF)-regulated Ubiproteome, as obtained by two complementary purification strategies coupled to quantitative proteomics. Our results unveil the complex impact of growth factor signaling on Ub-based intracellular networks to levels that extend well beyond what might have been expected. In addition to endocytic proteins, the EGF-regulated Ubiproteome includes a large number of signaling proteins, ubiquitinating and deubiquitinating enzymes, transporters and proteins involved in translation and transcription. The Ub-based signaling network appears to intersect both housekeeping and regulatory circuitries of cellular physiology. Finally, as proof of principle of the biological relevance of the EGF-Ubiproteome, we demonstrated that EphA2 is a novel, downstream ubiquitinated target of epidermal growth factor receptor (EGFR), critically involved in EGFR biological responses.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Biología de Sistemas/métodos , Ubiquitina/metabolismo , Animales , Western Blotting , Línea Celular , Análisis por Conglomerados , Factor de Crecimiento Epidérmico/química , Células HeLa , Humanos , Espectrometría de Masas , Ratones , Microscopía Fluorescente , Proteoma/química , Receptor EphA2/metabolismo , Transducción de Señal , Ubiquitina/química
4.
Elife ; 102021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34399888

RESUMEN

The molecular basis underlying glioblastoma (GBM) heterogeneity and plasticity is not fully understood. Using transcriptomic data of human patient-derived brain tumor stem cell lines (BTSCs), classified based on GBM-intrinsic signatures, we identify the AP-1 transcription factor FOSL1 as a key regulator of the mesenchymal (MES) subtype. We provide a mechanistic basis to the role of the neurofibromatosis type 1 gene (NF1), a negative regulator of the RAS/MAPK pathway, in GBM mesenchymal transformation through the modulation of FOSL1 expression. Depletion of FOSL1 in NF1-mutant human BTSCs and Kras-mutant mouse neural stem cells results in loss of the mesenchymal gene signature and reduction in stem cell properties and in vivo tumorigenic potential. Our data demonstrate that FOSL1 controls GBM plasticity and aggressiveness in response to NF1 alterations.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Células Madre Neoplásicas/patología , Neurofibromina 1/genética , Proteínas Proto-Oncogénicas c-fos/genética , Línea Celular Tumoral , Humanos , Neurofibromina 1/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo
5.
Nat Commun ; 11(1): 2977, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32532995

RESUMEN

Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Glicoproteínas de Membrana/genética , Oncogenes/genética , Isoformas de ARN/genética , Empalme del ARN , Receptor trkB/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Carcinogénesis/genética , Células Cultivadas , Perfilación de la Expresión Génica , Ontología de Genes , Glioma/metabolismo , Glioma/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones , Células 3T3 NIH , Células-Madre Neurales/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Isoformas de ARN/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/genética
6.
Nat Commun ; 11(1): 3883, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32753598

RESUMEN

Temozolomide (TMZ) is an oral alkylating agent used for the treatment of glioblastoma and is now becoming a chemotherapeutic option in patients diagnosed with high-risk low-grade gliomas. The O-6-methylguanine-DNA methyltransferase (MGMT) is responsible for the direct repair of the main TMZ-induced toxic DNA adduct, the O6-Methylguanine lesion. MGMT promoter hypermethylation is currently the only known biomarker for TMZ response in glioblastoma patients. Here we show that a subset of recurrent gliomas carries MGMT genomic rearrangements that lead to MGMT overexpression, independently from changes in its promoter methylation. By leveraging the CRISPR/Cas9 technology we generated some of these MGMT rearrangements in glioma cells and demonstrated that the MGMT genomic rearrangements contribute to TMZ resistance both in vitro and in vivo. Lastly, we showed that such fusions can be detected in tumor-derived exosomes and could potentially represent an early detection marker of tumor recurrence in a subset of patients treated with TMZ.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Resistencia a Antineoplásicos/genética , Reordenamiento Génico , Glioma/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Temozolomida/farmacología , Proteínas Supresoras de Tumor/genética , Adolescente , Adulto , Anciano , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Aductos de ADN/efectos de los fármacos , Aductos de ADN/metabolismo , Metilación de ADN , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Recurrencia Local de Neoplasia/prevención & control , Regiones Promotoras Genéticas/genética , RNA-Seq , Temozolomida/uso terapéutico , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba , Secuenciación Completa del Genoma , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
7.
Nat Commun ; 9(1): 1466, 2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29654229

RESUMEN

To accurately recapitulate the heterogeneity of human diseases, animal models require to recreate multiple complex genetic alterations. Here, we combine the RCAS-TVA system with the CRISPR-Cas9 genome editing tools for precise modeling of human tumors. We show that somatic deletion in neural stem cells of a variety of known tumor suppressor genes (Trp53, Cdkn2a, and Pten) leads to high-grade glioma formation. Moreover, by simultaneous delivery of pairs of guide RNAs we generate different gene fusions with oncogenic potential, either by chromosomal deletion (Bcan-Ntrk1) or by chromosomal translocation (Myb-Qk). Lastly, using homology-directed-repair, we also produce tumors carrying the homologous mutation to human BRAF V600E, frequently identified in a variety of tumors, including different types of gliomas. In summary, we have developed an extremely versatile mouse model for in vivo somatic genome editing, that will elicit the generation of more accurate cancer models particularly appropriate for pre-clinical testing.


Asunto(s)
Neoplasias Encefálicas/genética , Sistemas CRISPR-Cas , Edición Génica , ARN Guía de Kinetoplastida/genética , Animales , Antígenos de Neoplasias/genética , Benzamidas/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Brevicano/genética , Reparación del ADN , Reacciones Falso Positivas , Frecuencia de los Genes , Técnicas de Transferencia de Gen , Glioma/metabolismo , Humanos , Hibridación Fluorescente in Situ , Indazoles/farmacología , Ratones , Ratones SCID , Ratones Transgénicos , Mutación , Células 3T3 NIH , Receptor trkA/genética
8.
Nat Commun ; 8(1): 2035, 2017 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-29229958

RESUMEN

Transport of macromolecules through the nuclear pore by importins and exportins plays a critical role in the spatial regulation of protein activity. How cancer cells co-opt this process to promote tumorigenesis remains unclear. The epidermal growth factor receptor (EGFR) plays a critical role in normal development and in human cancer. Here we describe a mechanism of EGFR regulation through the importin ß family member RAN-binding protein 6 (RanBP6), a protein of hitherto unknown functions. We show that RanBP6 silencing impairs nuclear translocation of signal transducer and activator of transcription 3 (STAT3), reduces STAT3 binding to the EGFR promoter, results in transcriptional derepression of EGFR, and increased EGFR pathway output. Focal deletions of the RanBP6 locus on chromosome 9p were found in a subset of glioblastoma (GBM) and silencing of RanBP6 promoted glioma growth in vivo. Our results provide an example of EGFR deregulation in cancer through silencing of components of the nuclear import pathway.


Asunto(s)
Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , beta Carioferinas/genética , Proteína de Unión al GTP ran/genética , Transporte Activo de Núcleo Celular/genética , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Células Cultivadas , Doxorrubicina/farmacología , Receptores ErbB/metabolismo , Retroalimentación Fisiológica , Femenino , Técnicas de Silenciamiento del Gen , Glioma/tratamiento farmacológico , Glioma/metabolismo , Células HEK293 , Humanos , Ratones Noqueados , Ratones SCID , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , beta Carioferinas/metabolismo , Proteína de Unión al GTP ran/metabolismo
9.
Cancer Cell ; 28(6): 681-682, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26678333

RESUMEN

In this issue of Cancer Cell, Giachino and colleagues, employing various approaches, describe a tumor suppressor function for Notch signaling in forebrain tumors and suggest that decreased Notch activity could be a key molecular event in supratentorial primitive neuroectodermal tumors (sPNET).


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/metabolismo , Prosencéfalo/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Animales , Humanos
10.
Elife ; 32014 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-25551293

RESUMEN

The serine-threonine kinase AKT regulates proliferation and survival by phosphorylating a network of protein substrates. In this study, we describe a kinase-independent function of AKT. In cancer cells harboring gain-of-function alterations in MET, HER2, or Phosphatidyl-Inositol-3-Kinase (PI3K), catalytically inactive AKT (K179M) protected from drug induced cell death in a PH-domain dependent manner. An AKT kinase domain mutant found in human melanoma (G161V) lacked enzymatic activity in vitro and in AKT1/AKT2 double knockout cells, but promoted growth factor independent survival of primary human melanocytes. ATP-competitive AKT inhibitors failed to block the kinase-independent function of AKT, a liability that limits their effectiveness compared to allosteric AKT inhibitors. Our results broaden the current view of AKT function and have important implications for the development of AKT inhibitors for cancer.


Asunto(s)
Supervivencia Celular , Melanoma/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Línea Celular Tumoral , Humanos , Melanoma/enzimología
11.
Science ; 340(6132): 626-30, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23558169

RESUMEN

The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metabolism of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes associated with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant--but not IDH1-wild-type--glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.


Asunto(s)
Bencenoacetamidas/farmacología , Diferenciación Celular , Inhibidores Enzimáticos/farmacología , Glioma/enzimología , Glioma/patología , Imidazoles/farmacología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Isocitrato Deshidrogenasa/genética , Animales , Bencenoacetamidas/administración & dosificación , Bencenoacetamidas/toxicidad , Diferenciación Celular/efectos de los fármacos , Transformación Celular Neoplásica , Inhibidores Enzimáticos/toxicidad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/genética , Glutaratos/metabolismo , Histonas/metabolismo , Imidazoles/administración & dosificación , Imidazoles/toxicidad , Isocitrato Deshidrogenasa/química , Isocitrato Deshidrogenasa/metabolismo , Metilación , Ratones , Ratones SCID , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Multimerización de Proteína , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Discov ; 2(5): 458-71, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22588883

RESUMEN

UNLABELLED: Activation of the epidermal growth factor receptor (EGFR) in glioblastoma (GBM) occurs through mutations or deletions in the extracellular (EC) domain. Unlike lung cancers with EGFR kinase domain (KD) mutations, GBMs respond poorly to the EGFR inhibitor erlotinib. Using RNAi, we show that GBM cells carrying EGFR EC mutations display EGFR addiction. In contrast to KD mutants found in lung cancer, glioma-specific EGFR EC mutants are poorly inhibited by EGFR inhibitors that target the active kinase conformation (e.g., erlotinib). Inhibitors that bind to the inactive EGFR conformation, however, potently inhibit EGFR EC mutants and induce cell death in EGFR-mutant GBM cells. Our results provide first evidence for single kinase addiction in GBM and suggest that the disappointing clinical activity of first-generation EGFR inhibitors in GBM versus lung cancer may be attributed to the different conformational requirements of mutant EGFR in these 2 cancer types. SIGNIFICANCE: Approximately 40% of human glioblastomas harbor oncogenic EGFR alterations, but attempts to therapeutically target EGFR with first-generation EGFR kinase inhibitors have failed. Here, we demonstrate selective sensitivity of glioma-specific EGFR mutants to ATP-site competitive EGFR kinase inhibitors that target the inactive conformation of the catalytic domain.


Asunto(s)
Neoplasias Encefálicas/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Glioma/genética , Neoplasias Pulmonares/genética , Inhibidores de Proteínas Quinasas/farmacología , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Glioma/metabolismo , Humanos , Lapatinib , Neoplasias Pulmonares/metabolismo , Ratones , Mutación , Quinazolinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA