Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 588, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35102191

RESUMEN

High-grade diffuse glioma (HGG) is the leading cause of brain tumour death. While the genetic drivers of HGG have been well described, targeting these has thus far had little impact on survival suggesting other mechanisms are at play. Here we interrogate the alternative splicing landscape of pediatric and adult HGG through multi-omic analyses, uncovering an increased splicing burden compared with normal brain. The rate of recurrent alternative splicing in cancer drivers exceeds their mutation rate, a pattern that is recapitulated in pan-cancer analyses, and is associated with worse prognosis in HGG. We investigate potential oncogenicity by interrogating cancer pathways affected by alternative splicing in HGG; spliced cancer drivers include members of the RAS/MAPK pathway. RAS suppressor neurofibromin 1 is differentially spliced to a less active isoform in >80% of HGG downstream from REST upregulation, activating the RAS/MAPK pathway and reducing glioblastoma patient survival. Overall, our results identify non-mutagenic mechanisms by which cancers activate oncogenic pathways which need to accounted for in personalized medicine approaches.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Oncogenes/genética , Empalme del ARN/genética , Adulto , Empalme Alternativo/genética , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Niño , Cromatina/metabolismo , Exones/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Glioma/patología , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Mutación/genética , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Represoras/metabolismo , Empalmosomas/genética , Factores de Transcripción/metabolismo , Proteínas ras/metabolismo
2.
Methods Mol Biol ; 2423: 39-50, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34978686

RESUMEN

Studies of DNA-protein interactions have revealed regulatory mechanisms of DNA replication, repair, remodeling, and transcription. Perturbation of any or all of these processes result in differential gene expression that can lead to tumor development. Chromatin immunoprecipitation assay (ChIP), currently the only method available to explore DNA-binding in vivo, has become a vastly utilized tool for cancer research. In this article we discuss an assay specified for a pediatric medulloblastoma (MB) cell line DAOY used to determine binding of transcription factors, to detect histone modifications, and to identify novel therapeutic targets.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Niño , Cromatina/genética , Inmunoprecipitación de Cromatina/métodos , Humanos , Meduloblastoma/genética , Meduloblastoma/patología , Factores de Transcripción/metabolismo
3.
Mol Oncol ; 15(5): 1486-1506, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33469989

RESUMEN

Expression of the RE1-silencing transcription factor (REST), a master regulator of neurogenesis, is elevated in medulloblastoma (MB) tumors. A cell-intrinsic function for REST in MB tumorigenesis is known. However, a role for REST in the regulation of MB tumor microenvironment has not been investigated. Here, we implicate REST in remodeling of the MB vasculature and describe underlying mechanisms. Using RESTTG mice, we demonstrate that elevated REST expression in cerebellar granule cell progenitors, the cells of origin of sonic hedgehog (SHH) MBs, increased vascular growth. This was recapitulated in MB xenograft models and validated by transcriptomic analyses of human MB samples. REST upregulation was associated with enhanced secretion of proangiogenic factors. Surprisingly, a REST-dependent increase in the expression of the proangiogenic transcription factor E26 oncogene homolog 1, and its target gene encoding the vascular endothelial growth factor receptor-1, was observed in MB cells, which coincided with their localization at the tumor vasculature. These observations were confirmed by RNA-Seq and microarray analyses of MB cells and SHH-MB tumors. Thus, our data suggest that REST elevation promotes vascular growth by autocrine and paracrine mechanisms.


Asunto(s)
Neoplasias Cerebelosas/irrigación sanguínea , Meduloblastoma/irrigación sanguínea , Neovascularización Patológica/genética , Proteína Proto-Oncogénica c-ets-1/fisiología , Proteínas Represoras/fisiología , Animales , Proliferación Celular/genética , Células Cultivadas , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Neovascularización Patológica/patología , Microambiente Tumoral/genética
4.
Sci Signal ; 12(565)2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670636

RESUMEN

In medulloblastomas (MBs), the expression and activity of RE1-silencing transcription factor (REST) is increased in tumors driven by the sonic hedgehog (SHH) pathway, specifically the SHH-α (children 3 to 16 years) and SHH-ß (infants) subgroups. Neuronal maturation is greater in SHH-ß than SHH-α tumors, but both correlate with poor overall patient survival. We studied the contribution of REST to MB using a transgenic mouse model (RESTTG ) wherein conditional NeuroD2-controlled REST transgene expression in lineage-committed Ptch1 +/- cerebellar granule neuron progenitors (CGNPs) accelerated tumorigenesis and increased penetrance and infiltrative disease. This model revealed a neuronal maturation context-specific antagonistic interplay between the transcriptional repressor REST and the activator GLI1 at Ptch1 Expression of Arrb1, which encodes ß-arrestin1 (a GLI1 inhibitor), was substantially reduced in proliferating and, to a lesser extent, lineage-committed RESTTG cells compared with wild-type proliferating CGNPs. Lineage-committed RESTTG cells also had decreased GLI1 activity and increased histone H3K9 methylation at the Ptch1 locus, which correlated with premature silencing of Ptch1 These cells also had decreased expression of Pten, which encodes a negative regulator of the kinase AKT. Expression of PTCH1 and GLI1 were less, and ARRB1 was somewhat greater, in patient SHH-ß than SHH-α MBs, whereas that of PTEN was similarly lower in both subtypes than in others. Inhibition of histone modifiers or AKT reduced proliferation and induced apoptosis, respectively, in cultured REST-high MB cells. Our findings linking REST to differentiation-specific chromatin remodeling, PTCH1 silencing, and AKT activation in MB tissues reveal potential subgroup-specific therapeutic targets for MB patients.


Asunto(s)
Neoplasias Cerebelosas/genética , Cromatina/genética , Proteínas Hedgehog/genética , Meduloblastoma/genética , Receptor Patched-1/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Represoras/genética , Adulto , Animales , Línea Celular Tumoral , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Niño , Cromatina/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Lactante , Masculino , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Estadificación de Neoplasias , Receptor Patched-1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Trasplante Heterólogo
5.
Bioengineering (Basel) ; 5(4)2018 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-30279402

RESUMEN

Approximately five out of 100,000 children from 0 to 19 years old are diagnosed with a brain tumor. These children are treated with medication designed for adults that are highly toxic to a developing brain. Those that survive are at high risk for a lifetime of limited physical, psychological, and cognitive abilities. Despite much effort, not one drug exists that was designed specifically for pediatric patients. Stagnant government funding and the lack of economic incentives for the pharmaceutical industry greatly limits preclinical research and the development of clinically applicable pediatric brain tumor models. As more data are collected, the recognition of disease sub-groups based on molecular heterogeneity increases the need for designing specific models suitable for predictive drug screening. To overcome these challenges, preclinical approaches will need continual enhancement. In this review, we examine the advantages and shortcomings of in vitro and in vivo preclinical pediatric brain tumor models and explore potential solutions based on past, present, and future strategies for improving their clinical relevancy.

6.
Mol Cancer Res ; 15(8): 1073-1084, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28483947

RESUMEN

The deubiquitylase (DUB) USP37 is a component of the ubiquitin system and controls cell proliferation by regulating the stability of the cyclin-dependent kinase inhibitor 1B, (CDKN1B/p27Kip1). The expression of USP37 is downregulated in human medulloblastoma tumor specimens. In the current study, we show that USP37 prevents medulloblastoma growth in mouse orthotopic models, suggesting that it has tumor-suppressive properties in this neural cancer. Here, we also report on the mechanism underlying USP37 loss in medulloblastoma. Previously, we observed that the expression of USP37 is transcriptionally repressed by the RE1 silencing transcription factor (REST), which requires chromatin remodeling factors for its activity. Genetic and pharmacologic approaches were employed to identify a specific role for G9a, a histone methyltransferase (HMT), in promoting methylation of histone H3 lysine-9 (H3K9) mono- and dimethylation, and surprisingly trimethylation, at the USP37 promoter to repress its gene expression. G9a inhibition also blocked the tumorigenic potential of medulloblastoma cells in vivo Using isogenic low- and high-REST medulloblastoma cells, we further showed a REST-dependent elevation in G9a activity, which further increased mono- and trimethylation of histone H3K9, accompanied by downregulation of USP37 expression. Together, these findings reveal a role for REST-associated G9a and histone H3K9 methylation in the repression of USP37 expression in medulloblastoma.Implications: Reactivation of USP37 by G9a inhibition has the potential for therapeutic applications in REST-expressing medulloblastomas. Mol Cancer Res; 15(8); 1073-84. ©2017 AACR.


Asunto(s)
Endopeptidasas/genética , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/genética , Meduloblastoma/genética , Proteínas Represoras/genética , Animales , Carcinogénesis/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Histonas/genética , Humanos , Meduloblastoma/patología , Metilación , Metiltransferasas/genética , Ratones , Ubiquitina/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
CNS Oncol ; 4(2): 79-89, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25768332

RESUMEN

Medulloblastoma is the most common malignant pediatric brain tumor. Current treatments including surgery, craniospinal radiation and high-dose chemotherapy have led to improvement in survival. However, the risk for recurrence as well as significant long-term neurocognitive and endocrine sequelae associated with current treatment modalities underscore the urgent need for novel tumor-specific, normal brain-sparing therapies. It has also provided the impetus for research focused on providing a better understanding of medulloblastoma biology. The expectation is that such studies will lead to the identification of new therapeutic targets and eventually to an increase in personalized treatment approaches.


Asunto(s)
Neoplasias Encefálicas/terapia , Meduloblastoma/terapia , Animales , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Toma de Decisiones Clínicas , Ensayos Clínicos como Asunto , Humanos , Meduloblastoma/diagnóstico , Meduloblastoma/genética , Meduloblastoma/metabolismo
8.
Mol Cancer Res ; 11(8): 887-900, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23661421

RESUMEN

UNLABELLED: Overexpression of the oncoprotein Aurora A kinase occurs in multiple types of cancer, often early during cell transformation. To identify the mechanism(s) contributing to enhanced Aurora A protein expression, a comparison between normal human lung fibroblast and breast epithelial cells to nontumorigenic breast (MCF10A and MCF12A) and tumorigenic breast (MCF-7) and cervical cell lines (HeLa S3) was performed. A subset of these immortalized lines (MCF10A, MCF12A, and HeLa S3) exhibited increased levels of Aurora A protein, independent of tumorigenicity. The increase in Aurora A protein in these immortalized cells was not due to increased transcription/RNA stability, protein half-life, or cap-dependent translation. Assays utilizing monocistronic and dicistronic RNA constructs revealed that the 5'-leader sequence of Aurora A contains an internal ribosomal entry site (IRES), which is regulated in a cell cycle-dependent manner, peaking in G2/M phase. Moreover, IRES activity was increased in the immortalized cell lines in which Aurora A protein expression was also enhanced. Additional studies indicated that the increased internal initiation is specific to the IRES of Aurora A and may be an early event during cancer progression. These results identify a novel mechanism contributing to Aurora A kinase overexpression. IMPLICATIONS: The current study indicates that Aurora A kinase contributes to immortalization and tumorigenesis.


Asunto(s)
Regiones no Traducidas 5' , Aurora Quinasa A/genética , Aurora Quinasa A/metabolismo , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Células MCF-7 , Iniciación de la Cadena Peptídica Traduccional , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
9.
BMC Mol Biol ; 9: 89, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18922172

RESUMEN

BACKGROUND: Regulating synthesis of the Fragile X gene (FMR1) product, FMRP alters neural plasticity potentially through its role in the microRNA pathway. Cap-dependent translation of the FMR1 mRNA, a process requiring ribosomal scanning through the 5' leader, is likely impeded by the extensive secondary structure generated by the high guanosine/cytosine nucleotide content including the CGG triplet nucleotide repeats in the 5' leader. An alternative mechanism to initiate translation - internal initiation often utilizes secondary structure to recruit the translational machinery. Consequently, studies were undertaken to confirm and extend a previous observation that the FMR1 5' leader contains an internal ribosomal entry site (IRES). RESULTS: Cellular transfection of a dicistronic DNA construct containing the FMR1 5' leader inserted into the intercistronic region yielded significant translation of the second cistron, but the FMR1 5' leader was also found to contain a cryptic promoter possibly confounding interpretation of these results. However, transfection of dicistronic and monocistronic RNA ex vivo or in vitro confirmed that the FMR1 5' leader contains an IRES. Moreover, inhibiting cap-dependent translation ex vivo did not affect the expression level of endogenous FMRP indicating a role for IRES-dependent translation of FMR1 mRNA. Analysis of the FMR1 5' leader revealed that the CGG repeats and the 5' end of the leader were vital for internal initiation. Functionally, exposure to potassium chloride or intracellular acidification and addition of polyinosinic:polycytidylic acid as mimics of neural activity and double stranded RNA, respectively, differentially affected FMR1 IRES activity. CONCLUSION: Our results indicate that multiple stimuli influence IRES-dependent translation of the FMR1 mRNA and suggest a functional role for the CGG nucleotide repeats.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación de la Expresión Génica , Iniciación de la Cadena Peptídica Traduccional , Biosíntesis de Proteínas/genética , Regiones no Traducidas 5' , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/biosíntesis , Genes , Humanos , Conformación de Ácido Nucleico , Caperuzas de ARN , ARN Mensajero , Repeticiones de Trinucleótidos
10.
Nucleic Acids Res ; 33(9): 2929-41, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15908588

RESUMEN

Translational regulation of the dendritically localized mRNA encoding for the neurotrophin receptor TrkB has important ramifications for synaptic function. We examined whether the TrkB mRNA is translated through an internal initiation entry site (IRES). The human TrkB 5' leaders are derived from the use of alternative promoters and alternative splicing, but all 5' leaders share a common exon. Insertion of a full-length 5' leader, as well as the common exon into the intercistronic region of a dicistronic luciferase construct, yielded luciferase activity generated from the second cistron that was either equivalent or higher than that observed from the encephalomyocarditis virus IRES. Moreover, inhibiting cap-dependent translation ex vivo and in in vitro lysates had only a minimal effect on the translation of mRNA containing the TrkB 5' leader. Dissecting the 5' leader showed that the IRES is located in the exon common to all TrkB 5' leaders. Moreover, six regions ranging from 2 to 25 nt were identified that either promoted or inhibited IRES activity. Taken together, these results suggest that the 5' leader of the human TrkB mRNA contains multiple cis-elements that regulate internal initiation of translation and that this mechanism may contribute significantly to the translation of the TrkB mRNA in neuronal dendrites.


Asunto(s)
Regiones no Traducidas 5'/química , Iniciación de la Cadena Peptídica Traduccional , Receptor trkB/genética , Secuencia de Bases , Línea Celular , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Mensajero/biosíntesis , ARN Mensajero/química , Receptor trkB/biosíntesis , Eliminación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...