Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2226: 285-302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33326110

RESUMEN

Ewing sarcoma is a highly malignant tumor characterized by a chromosomal translocation that modifies the activity of an ETS family transcription factor. The most prevalent translocation product, EWSR1-FLI1, exploits a permissive and unique chromatin environment of stem cells, and transforms them into an oncogenic state through alterations to gene expression and gene regulatory programs. Though the transformation ability of, and subsequent reliance on EWSR1-FLI1 had been previously described, the advent of genome-wide sequencing technologies allowed for the specific identification of genomic loci and genes targeted by EWSR1-FLI1. Furthermore, the characterization of the chromatin environment in these, and other, cell types could not have been accomplished without the computational and statistical methods that enable large-scale data analysis. Here, we outline in detail the tools and steps needed to analyze genome-wide transcription factor binding and histone modification data (chromatin immunoprecipitation, ChIP-seq), as well as chromatin accessibility data (assay for transposase-accessible chromatin, ATAC-seq) from Ewing sarcoma cells. Our protocol includes a compilation of data quality control metrics, trimming of adapter sequences, reference genome alignment, identification of enriched sites ("peaks") and motifs, as well as annotation and visualization, using real-world data. These steps should provide a platform on which molecular biologists can build their own analytical pipelines to aid in data processing, analysis, and interpretation.


Asunto(s)
Neoplasias Óseas/genética , Epigénesis Genética , Epigenómica , Regulación Neoplásica de la Expresión Génica , Sarcoma de Ewing/genética , Biomarcadores de Tumor , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Biología Computacional/métodos , Bases de Datos Genéticas , Epigenómica/métodos , Humanos , Programas Informáticos , Navegador Web
2.
Genes Dev ; 34(23-24): 1713-1734, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33184221

RESUMEN

Through recurrent bouts synchronous with the hair cycle, quiescent melanocyte stem cells (McSCs) become activated to generate proliferative progeny that differentiate into pigment-producing melanocytes. The signaling factors orchestrating these events remain incompletely understood. Here, we use single-cell RNA sequencing with comparative gene expression analysis to elucidate the transcriptional dynamics of McSCs through quiescence, activation, and melanocyte maturation. Unearthing converging signs of increased WNT and BMP signaling along this progression, we endeavored to understand how these pathways are integrated. Employing conditional lineage-specific genetic ablation studies in mice, we found that loss of BMP signaling in the lineage leads to hair graying due to a block in melanocyte maturation. We show that interestingly, BMP signaling functions downstream from activated McSCs and maintains WNT effector, transcription factor LEF1. Employing pseudotime analysis, genetics, and chromatin landscaping, we show that following WNT-mediated activation of McSCs, BMP and WNT pathways collaborate to trigger the commitment of proliferative progeny by fueling LEF1- and MITF-dependent differentiation. Our findings shed light upon the signaling interplay and timing of cues that orchestrate melanocyte lineage progression in the hair follicle and underscore a key role for BMP signaling in driving complete differentiation.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Diferenciación Celular/genética , Melanocitos/citología , Transducción de Señal/genética , Células Madre/citología , Animales , Linaje de la Célula/genética , Perfilación de la Expresión Génica , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Factor de Transcripción Asociado a Microftalmía/metabolismo , Análisis de la Célula Individual
3.
Elife ; 82019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31556874

RESUMEN

During mammalian embryogenesis, extensive cellular remodeling is needed for tissue morphogenesis. As effectors of cytoskeletal dynamics, Rho GTPases and their regulators are likely involved, but their daunting complexity has hindered progress in dissecting their functions. We overcome this hurdle by employing high throughput in utero RNAi-mediated screening to identify key Rho regulators of skin morphogenesis. Our screen unveiled hitherto unrecognized roles for Rho-mediated cytoskeletal remodeling events that impact hair follicle specification, differentiation, downgrowth and planar cell polarity. Coupling our top hit with gain/loss-of-function genetics, interactome proteomics and tissue imaging, we show that RHOU, an atypical Rho, governs the cytoskeletal-junction dynamics that establish columnar shape and planar cell polarity in epidermal progenitors. Conversely, RHOU downregulation is required to remodel to a conical cellular shape that enables hair bud invagination and downgrowth. Our findings underscore the power of coupling screens with proteomics to unravel the physiological significance of complex gene families.


Asunto(s)
Regulación de la Expresión Génica , Morfogénesis , Piel/embriología , Proteínas de Unión al GTP rho/metabolismo , Animales , Silenciador del Gen , Pruebas Genéticas , Ratones , Interferencia de ARN
4.
Mol Cell Biol ; 39(14)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31036566

RESUMEN

Ewing sarcoma is characterized by a pathognomonic chromosomal translocation that generates the EWSR1-FLI1 chimeric transcription factor. The transcriptional targets of EWSR1-FLI1 that are essential for tumorigenicity are incompletely defined. Here, we found that EWSR1-FLI1 modulates the expression of cancer/testis (CT) antigen genes, whose expression is biased to the testes but is also activated in cancer. Among these CT antigens, fetal and adult testis expressed 1 (FATE1) is most robustly induced. EWSR1-FLI1 associates with the GGAA repeats in the proximal promoter of FATE1, which exhibits accessible chromatin exclusively in mesenchymal progenitor cells (MPCs) and Ewing sarcoma cells. Expression of EWSR1-FLI1 in non-Ewing sarcoma cells and in MPCs enhances FATE1 mRNA and protein expression. Conversely, depletion of EWSR1-FLI1 in Ewing sarcoma cells leads to a loss of FATE1 expression. Importantly, we found that FATE1 is required for survival and anchorage-independent growth in Ewing sarcoma cells via attenuating the accumulation of BNIP3L, a BH3-only protein that is toxic when stabilized. This action appears to be mediated by the E3 ligase RNF183. We propose that engaging FATE1 function can permit the bypass of cell death mechanisms that would otherwise inhibit tumor progression.


Asunto(s)
Neoplasias Óseas/genética , Proteínas de Unión al ADN/genética , Proteínas de Fusión Oncogénica/genética , Sarcoma de Ewing/genética , Factores de Transcripción/genética , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HeLa , Células Hep G2 , Humanos , Proteínas de la Membrana/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Sarcoma de Ewing/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
5.
Nature ; 569(7757): 497-502, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31092920

RESUMEN

Cell competition-the sensing and elimination of less fit 'loser' cells by neighbouring 'winner' cells-was first described in Drosophila. Although cell competition has been proposed as a selection mechanism to optimize tissue and organ development, its evolutionary generality remains unclear. Here, by using live imaging, lineage tracing, single-cell transcriptomics and genetics, we identify two cell competition mechanisms that sequentially shape and maintain the architecture of stratified tissue during skin development in mice. In the single-layered epithelium of the early embryonic epidermis, winner progenitors kill and subsequently clear neighbouring loser cells by engulfment. Later, as the tissue begins to stratify, the basal layer instead expels losers through upward flux of differentiating progeny. This cell competition switch is physiologically relevant: when it is perturbed, so too is barrier formation. Our findings show that cell competition is a selective force that optimizes vertebrate tissue function, and illuminate how a tissue dynamically adjusts cell competition strategies to preserve fitness as its architectural complexity increases during morphogenesis.


Asunto(s)
Comunicación Celular , Células Epidérmicas/citología , Epidermis/embriología , Morfogénesis , Animales , Apoptosis , Células Clonales/citología , Drosophila melanogaster/citología , Drosophila melanogaster/embriología , Células Epidérmicas/metabolismo , Femenino , Masculino , Ratones , Fagocitosis , RNA-Seq , Análisis de la Célula Individual
6.
Nature ; 560(7716): E2, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29973713

RESUMEN

In Fig. 2g of this Article, a panel was inadvertently duplicated. The 'D30 IMQ' image was a duplicate of the 'D6 Ctrl' image. Fig. 2g has been corrected online to show the correct 'D30 IMQ' image (showing skin inflammation induced by the NALP3 agonist imiquimod, IMQ). The Supplementary Information to this Amendment contains the old, incorrect Fig. 2 for transparency.

7.
Nature ; 550(7677): 475-480, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29045388

RESUMEN

The skin barrier is the body's first line of defence against environmental assaults, and is maintained by epithelial stem cells (EpSCs). Despite the vulnerability of EpSCs to inflammatory pressures, neither the primary response to inflammation nor its enduring consequences are well understood. Here we report a prolonged memory to acute inflammation that enables mouse EpSCs to hasten barrier restoration after subsequent tissue damage. This functional adaptation does not require skin-resident macrophages or T cells. Instead, EpSCs maintain chromosomal accessibility at key stress response genes that are activated by the primary stimulus. Upon a secondary challenge, genes governed by these domains are transcribed rapidly. Fuelling this memory is Aim2, which encodes an activator of the inflammasome. The absence of AIM2 or its downstream effectors, caspase-1 and interleukin-1ß, erases the ability of EpSCs to recollect inflammation. Although EpSCs benefit from inflammatory tuning by heightening their responsiveness to subsequent stressors, this enhanced sensitivity probably increases their susceptibility to autoimmune and hyperproliferative disorders, including cancer.


Asunto(s)
Células Epiteliales/citología , Inflamación/genética , Inflamación/patología , Piel/citología , Piel/patología , Células Madre/citología , Cicatrización de Heridas/fisiología , Aminoquinolinas/farmacología , Animales , Enfermedades Autoinmunes/patología , Caspasa 1/metabolismo , Linaje de la Célula , Cromatina/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Imiquimod , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Interleucina-1beta/metabolismo , Macrófagos , Ratones , Neoplasias/patología , Regeneración/efectos de los fármacos , Regeneración/genética , Piel/efectos de los fármacos , Piel/inmunología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Estrés Fisiológico/genética , Linfocitos T , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
8.
Cell ; 169(4): 636-650.e14, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28434617

RESUMEN

Tissue stem cells contribute to tissue regeneration and wound repair through cellular programs that can be hijacked by cancer cells. Here, we investigate such a phenomenon in skin, where during homeostasis, stem cells of the epidermis and hair follicle fuel their respective tissues. We find that breakdown of stem cell lineage confinement-granting privileges associated with both fates-is not only hallmark but also functional in cancer development. We show that lineage plasticity is critical in wound repair, where it operates transiently to redirect fates. Investigating mechanism, we discover that irrespective of cellular origin, lineage infidelity occurs in wounding when stress-responsive enhancers become activated and override homeostatic enhancers that govern lineage specificity. In cancer, stress-responsive transcription factor levels rise, causing lineage commanders to reach excess. When lineage and stress factors collaborate, they activate oncogenic enhancers that distinguish cancers from wounds.


Asunto(s)
Carcinoma de Células Escamosas/patología , Linaje de la Célula , Células Epidérmicas , Folículo Piloso/citología , Neoplasias Cutáneas/patología , Piel/citología , Células Madre/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Epidermis/metabolismo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Cutáneas/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Trasplante Heterólogo , Cicatrización de Heridas
9.
Nature ; 541(7638): 494-499, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28077873

RESUMEN

We are just beginning to understand how translational control affects tumour initiation and malignancy. Here we use an epidermis-specific, in vivo ribosome profiling strategy to investigate the translational landscape during the transition from normal homeostasis to malignancy. Using a mouse model of inducible SOX2, which is broadly expressed in oncogenic RAS-associated cancers, we show that despite widespread reductions in translation and protein synthesis, certain oncogenic mRNAs are spared. During tumour initiation, the translational apparatus is redirected towards unconventional upstream initiation sites, enhancing the translational efficiency of oncogenic mRNAs. An in vivo RNA interference screen of translational regulators revealed that depletion of conventional eIF2 complexes has adverse effects on normal but not oncogenic growth. Conversely, the alternative initiation factor eIF2A is essential for cancer progression, during which it mediates initiation at these upstream sites, differentially skewing translation and protein expression. Our findings unveil a role for the translation of 5' untranslated regions in cancer, and expose new targets for therapeutic intervention.


Asunto(s)
Regiones no Traducidas 5'/genética , Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Sistemas de Lectura Abierta/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Animales , Carcinogénesis/patología , Carcinoma de Células Escamosas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epidermis/embriología , Epidermis/metabolismo , Epidermis/patología , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Queratinocitos , Masculino , Ratones , Oncogenes/genética , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Pronóstico , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Neoplasias Cutáneas/metabolismo
10.
Cell Rep ; 17(6): 1607-1620, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27806299

RESUMEN

Chromatin regulation is critical for differentiation and disease. However, features linking the chromatin environment of stem cells with disease remain largely unknown. We explored chromatin accessibility in embryonic and multipotent stem cells and unexpectedly identified widespread chromatin accessibility at repetitive elements. Integrating genomic and biochemical approaches, we demonstrate that these sites of increased accessibility are associated with well-positioned nucleosomes marked by distinct histone modifications. Differentiation is accompanied by chromatin remodeling at repetitive elements associated with altered expression of genes in relevant developmental pathways. Remarkably, we found that the chromatin environment of Ewing sarcoma, a mesenchymally derived tumor, is shared with primary mesenchymal stem cells (MSCs). Accessibility at repetitive elements in MSCs offers a permissive environment that is exploited by the critical oncogene responsible for this cancer. Our data demonstrate that stem cells harbor a unique chromatin landscape characterized by accessibility at repetitive elements, a feature associated with differentiation and oncogenesis.


Asunto(s)
Cromatina/metabolismo , Neoplasias/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Células Madre/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Ensamble y Desensamble de Cromatina , Desoxirribonucleasas/metabolismo , Histonas/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Neoplasias/patología , Nucleosomas/metabolismo , Oncogenes , Procesamiento Proteico-Postraduccional , Elementos de Nucleótido Esparcido Corto/genética , Transcripción Genética
11.
Dev Cell ; 36(3): 262-75, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26859351

RESUMEN

Human mutations in the cardiac transcription factor gene TBX5 cause congenital heart disease (CHD), although the underlying mechanism is unknown. We report characterization of the endogenous TBX5 cardiac interactome and demonstrate that TBX5, long considered a transcriptional activator, interacts biochemically and genetically with the nucleosome remodeling and deacetylase (NuRD) repressor complex. Incompatible gene programs are repressed by TBX5 in the developing heart. CHD mis-sense mutations that disrupt the TBX5-NuRD interaction cause depression of a subset of repressed genes. Furthermore, the TBX5-NuRD interaction is required for heart development. Phylogenetic analysis showed that the TBX5-NuRD interaction domain evolved during early diversification of vertebrates, simultaneous with the evolution of cardiac septation. Collectively, this work defines a TBX5-NuRD interaction essential to cardiac development and the evolution of the mammalian heart, and when altered may contribute to human CHD.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Regulación del Desarrollo de la Expresión Génica/genética , Corazón/embriología , Miocardio/metabolismo , Proteínas de Dominio T Box/genética , Animales , Humanos , Ratones Transgénicos , Organogénesis/genética , Organogénesis/fisiología , Proteínas de Dominio T Box/metabolismo , Transcripción Genética/genética
12.
Nat Genet ; 47(9): 964-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26313223

RESUMEN

The identification of gene-regulatory polymorphisms that influence cancer susceptibility can identify key oncogenic pathways. A new study links a germline variant to Ewing sarcoma disease susceptibility and EWSR1-FLI1-mediated gene activation.


Asunto(s)
Neoplasias Óseas/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Animales , Humanos
13.
Mol Cancer Res ; 12(11): 1610-20, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24994750

RESUMEN

UNLABELLED: Recent evidence implicates the insulin-like growth factor (IGF) pathway in development of Ewing sarcoma, a highly malignant bone and soft-tissue tumor that primarily affects children and young adults. Despite promising results from preclinical studies of therapies that target this pathway, early-phase clinical trials have shown that a significant fraction of patients do not benefit, suggesting that cellular factors determine tumor sensitivity. Using FAIRE-seq, a chromosomal deletion of the PTEN locus in a Ewing sarcoma cell line was identified. In primary tumors, PTEN deficiency was observed in a large subset of cases, although not mediated by large chromosomal deletions. PTEN loss resulted in hyperactivation of the AKT signaling pathway. PTEN rescue led to decreased proliferation, inhibition of colony formation, and increased apoptosis. Strikingly, PTEN loss decreased sensitivity to IGF1R inhibitors but increased responsiveness to temsirolimus, a potent mTOR inhibitor, as marked by induction of autophagy. These results suggest that PTEN is lost in a significant fraction of primary tumors, and this deficiency may have therapeutic consequences by concurrently attenuating responsiveness to IGF1R inhibition while increasing activity of mTOR inhibitors. The identification of PTEN status in the tumors of patients with recurrent disease could help guide the selection of therapies. IMPLICATIONS: PTEN status in Ewing sarcoma affects cellular responses to IGFI and mTOR-directed therapy, thus justifying its consideration as a biomarker in future clinical trials.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/farmacología , Fosfohidrolasa PTEN/deficiencia , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Eliminación de Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/patología , Transducción de Señal , Sirolimus/análogos & derivados , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
14.
Genes Cancer ; 4(5-6): 213-23, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-24069508

RESUMEN

The gene encoding EWS (EWSR1) is involved in various chromosomal translocations that cause the production of oncoproteins responsible for multiple cancers including Ewing sarcoma, myxoid liposarcoma, soft tissue clear cell sarcoma, and desmoplastic small round cell sarcoma. It is well known that EWS fuses to FLI to create EWS/FLI, which is the abnormal transcription factor that drives tumor development in Ewing sarcoma. However, the role of wild-type EWS in Ewing sarcoma pathogenesis remains unclear. In the current study, we identified EWS-regulated genes and cellular processes through RNA interference combined with RNA sequencing and functional annotation analyses. Interestingly, we found that EWS and EWS/FLI co-regulate a significant cluster of genes, indicating an interplay between the 2 proteins in regulating cellular functions. We found that among the EWS-down-regulated genes are a subset of neuronal genes that contain binding sites for the RE1-silencing transcription factor (REST or neuron-restrictive silencer factor [NRSF]), neuron-restrictive silencer element (NRSE), suggesting a cooperative interaction between REST and EWS in gene regulation. Co-immunoprecipitation analysis demonstrated that EWS interacts directly with REST. Genome-wide binding analysis showed that EWS binds chromatin at or near NRSE. Furthermore, functional studies revealed that both EWS and REST inhibit neuronal phenotype development and oncogenic transformation in Ewing sarcoma cells. Our data implicate an important role of EWS in the development of Ewing sarcoma phenotype and highlight a potential value in modulating EWS function in the treatment of Ewing sarcoma and other EWS translocation-based cancers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...