Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 158(3): 593-606, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083870

RESUMEN

Notch signaling is a key developmental pathway that is subject to frequent genetic and epigenetic perturbations in many different human tumors. Here we investigate whether long noncoding RNA (lncRNA) genes, in addition to mRNAs, are key downstream targets of oncogenic Notch1 in human T cell acute lymphoblastic leukemia (T-ALL). By integrating transcriptome profiles with chromatin state maps, we have uncovered many previously unreported T-ALL-specific lncRNA genes, a fraction of which are directly controlled by the Notch1/Rpbjκ activator complex. Finally we have shown that one specific Notch-regulated lncRNA, LUNAR1, is required for efficient T-ALL growth in vitro and in vivo due to its ability to enhance IGF1R mRNA expression and sustain IGF1 signaling. These results confirm that lncRNAs are important downstream targets of the Notch signaling pathway, and additionally they are key regulators of the oncogenic state in T-ALL.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , ARN Largo no Codificante/análisis , Receptor Notch1/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , ARN Largo no Codificante/genética , Transducción de Señal , Timo/patología
2.
Cell ; 153(7): 1552-66, 2013 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-23791182

RESUMEN

Sequencing efforts led to the identification of somatic mutations that could affect the self-renewal and differentiation of cancer-initiating cells. One such recurrent mutation targets the binding pocket of the ubiquitin ligase Fbxw7. Missense FBXW7 mutations are prevalent in various tumors, including T cell acute lymphoblastic leukemia (T-ALL). To study the effects of such lesions, we generated animals carrying regulatable Fbxw7 mutant alleles. Here, we show that these mutations specifically bolster cancer-initiating cell activity in collaboration with Notch1 oncogenes but spare normal hematopoietic stem cell function. We were also able to show that FBXW7 mutations specifically affect the ubiquitylation and half-life of c-Myc protein, a key T-ALL oncogene. Using animals carrying c-Myc fusion alleles, we connected Fbxw7 function to c-Myc abundance and correlated c-Myc expression to leukemia-initiating activity. Finally, we demonstrated that small-molecule-mediated suppression of MYC activity leads to T-ALL remission, suggesting an effective therapeutic strategy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Modelos Animales de Enfermedad , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Noqueados , Mutación Missense , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Receptor Notch1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
3.
Nat Immunol ; 16(6): 653-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25867473

RESUMEN

The methylcytosine dioxygenase TET1 ('ten-eleven translocation 1') is an important regulator of 5-hydroxymethylcytosine (5hmC) in embryonic stem cells. The diminished expression of TET proteins and loss of 5hmC in many tumors suggests a critical role for the maintenance of this epigenetic modification. Here we found that deletion of Tet1 promoted the development of B cell lymphoma in mice. TET1 was required for maintenance of the normal abundance and distribution of 5hmC, which prevented hypermethylation of DNA, and for regulation of the B cell lineage and of genes encoding molecules involved in chromosome maintenance and DNA repair. Whole-exome sequencing of TET1-deficient tumors revealed mutations frequently found in non-Hodgkin B cell lymphoma (B-NHL), in which TET1 was hypermethylated and transcriptionally silenced. Our findings provide in vivo evidence of a function for TET1 as a tumor suppressor of hematopoietic malignancy.


Asunto(s)
Linfocitos B/fisiología , Citosina/análogos & derivados , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/fisiología , Linfoma de Células B/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Inestabilidad Cromosómica , Citosina/metabolismo , Metilación de ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Epigénesis Genética , Exoma/genética , Perfilación de la Expresión Génica , Humanos , Ratones , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Supresoras de Tumor/genética
5.
Nature ; 514(7523): 513-7, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25132549

RESUMEN

T-cell acute lymphoblastic leukaemia (T-ALL) is a haematological malignancy with a dismal overall prognosis, including a relapse rate of up to 25%, mainly because of the lack of non-cytotoxic targeted therapy options. Drugs that target the function of key epigenetic factors have been approved in the context of haematopoietic disorders, and mutations that affect chromatin modulators in a variety of leukaemias have recently been identified; however, 'epigenetic' drugs are not currently used for T-ALL treatment. Recently, we described that the polycomb repressive complex 2 (PRC2) has a tumour-suppressor role in T-ALL. Here we delineated the role of the histone 3 lysine 27 (H3K27) demethylases JMJD3 and UTX in T-ALL. We show that JMJD3 is essential for the initiation and maintenance of T-ALL, as it controls important oncogenic gene targets by modulating H3K27 methylation. By contrast, we found that UTX functions as a tumour suppressor and is frequently genetically inactivated in T-ALL. Moreover, we demonstrated that the small molecule inhibitor GSKJ4 (ref. 5) affects T-ALL growth, by targeting JMJD3 activity. These findings show that two proteins with a similar enzymatic function can have opposing roles in the context of the same disease, paving the way for treating haematopoietic malignancies with a new category of epigenetic inhibitors.


Asunto(s)
Histona Demetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Animales , Benzazepinas/farmacología , Epigénesis Genética/efectos de los fármacos , Histona Demetilasas/genética , Histonas/química , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Lisina/metabolismo , Metilación/efectos de los fármacos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pirimidinas/farmacología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
6.
Mol Cell ; 47(6): 873-85, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22864115

RESUMEN

Class switch recombination (CSR) has the potential to generate genomic instability in B cells as activation-induced cytidine deaminase (AID), which mediates this process, is known to target many sites outside Igh. Nonetheless we do not fully understand what factors influence AID targeting genome-wide. Given that errors in CSR can lead to dangerous, oncogenic chromosomal translocations it is important to identify the elements that determine which genes are at risk of being "hit" and could be involved in aberrant rearrangements. Here we have investigated the influence of nuclear organization in determining "off-target" activity and the choice of fusion partners. Our studies indicate that the vast majority of known AID-mediated Igh translocation partners are found in chromosomal domains that contact this locus during class switching. Further, these interaction domains can be used to identify other genes that are hit by AID.


Asunto(s)
Linfocitos B/citología , Citidina Desaminasa/metabolismo , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Cambio de Clase de Inmunoglobulina , Translocación Genética , Animales , Linfocitos B/metabolismo , Citidina Desaminasa/genética , Inestabilidad Genómica , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Hipermutación Somática de Inmunoglobulina
7.
BMC Genomics ; 18(1): 434, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28583068

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) have emerged as a class of factors that are important for regulating development and cancer. Computational prediction of lncRNAs from ultra-deep RNA sequencing has been successful in identifying candidate lncRNAs. However, the complexity of handling and integrating different types of genomics data poses significant challenges to experimental laboratories that lack extensive genomics expertise. RESULT: To address this issue, we have developed lncRNA-screen, a comprehensive pipeline for computationally screening putative lncRNA transcripts over large multimodal datasets. The main objective of this work is to facilitate the computational discovery of lncRNA candidates to be further examined by functional experiments. lncRNA-screen provides a fully automated easy-to-run pipeline which performs data download, RNA-seq alignment, assembly, quality assessment, transcript filtration, novel lncRNA identification, coding potential estimation, expression level quantification, histone mark enrichment profile integration, differential expression analysis, annotation with other type of segmented data (CNVs, SNPs, Hi-C, etc.) and visualization. Importantly, lncRNA-screen generates an interactive report summarizing all interesting lncRNA features including genome browser snapshots and lncRNA-mRNA interactions based on Hi-C data. CONCLUSION: lncRNA-screen provides a comprehensive solution for lncRNA discovery and an intuitive interactive report for identifying promising lncRNA candidates. lncRNA-screen is available as open-source software on GitHub.


Asunto(s)
Bases de Datos Genéticas , Genómica/métodos , ARN Largo no Codificante/genética , Interfaz Usuario-Computador , Minería de Datos , Epigenómica , Perfilación de la Expresión Génica , Histonas/metabolismo , Alineación de Secuencia , Programas Informáticos
8.
Nature ; 473(7346): 230-3, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21562564

RESUMEN

Notch signalling is a central regulator of differentiation in a variety of organisms and tissue types. Its activity is controlled by the multi-subunit γ-secretase (γSE) complex. Although Notch signalling can play both oncogenic and tumour-suppressor roles in solid tumours, in the haematopoietic system it is exclusively oncogenic, notably in T-cell acute lymphoblastic leukaemia, a disease characterized by Notch1-activating mutations. Here we identify novel somatic-inactivating Notch pathway mutations in a fraction of patients with chronic myelomonocytic leukaemia (CMML). Inactivation of Notch signalling in mouse haematopoietic stem cells (HSCs) results in an aberrant accumulation of granulocyte/monocyte progenitors (GMPs), extramedullary haematopoieisis and the induction of CMML-like disease. Transcriptome analysis revealed that Notch signalling regulates an extensive myelomonocytic-specific gene signature, through the direct suppression of gene transcription by the Notch target Hes1. Our studies identify a novel role for Notch signalling during early haematopoietic stem cell differentiation and suggest that the Notch pathway can play both tumour-promoting and -suppressive roles within the same tissue.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/patología , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Silenciador del Gen , Células Progenitoras de Granulocitos y Macrófagos/citología , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Receptores Notch/deficiencia , Factor de Transcripción HES-1 , Células Tumorales Cultivadas
9.
Nature ; 459(7249): 1000-4, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19536265

RESUMEN

T-cell acute lymphoblastic leukaemia (T-ALL) is a blood malignancy afflicting mainly children and adolescents. T-ALL patients present at diagnosis with increased white cell counts and hepatosplenomegaly, and are at an increased risk of central nervous system (CNS) relapse. For that reason, T-ALL patients usually receive cranial irradiation in addition to intensified intrathecal chemotherapy. The marked increase in survival is thought to be worth the considerable side-effects associated with this therapy. Such complications include secondary tumours, neurocognitive deficits, endocrine disorders and growth impairment. Little is known about the mechanism of leukaemic cell infiltration of the CNS, despite its clinical importance. Here we show, using T-ALL animal modelling and gene-expression profiling, that the chemokine receptor CCR7 (ref. 5) is the essential adhesion signal required for the targeting of leukaemic T-cells into the CNS. Ccr7 gene expression is controlled by the activity of the T-ALL oncogene Notch1 and is expressed in human tumours carrying Notch1-activating mutations. Silencing of either CCR7 or its chemokine ligand CCL19 (ref. 6) in an animal model of T-ALL specifically inhibits CNS infiltration. Furthermore, murine CNS-targeting by human T-ALL cells depends on their ability to express CCR7. These studies identify a single chemokine-receptor interaction as a CNS 'entry' signal, and open the way for future pharmacological targeting. Targeted inhibition of CNS involvement in T-ALL could potentially decrease the intensity of CNS-targeted therapy, thus reducing its associated short- and long-term complications.


Asunto(s)
Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Receptores CCR7/metabolismo , Transducción de Señal , Animales , Adhesión Celular , Línea Celular Tumoral , Quimiocina CCL19/deficiencia , Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores CCR7/deficiencia
10.
Theranostics ; 11(4): 1594-1608, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33408769

RESUMEN

The Notch pathway is highly active in almost all patients with T-cell acute lymphoblastic leukemia (T-ALL), but the implication of Notch ligands in T-ALL remains underexplored. Methods: We used a genetic mouse model of Notch ligand delta like 4 (DLL4)-driven T-ALL and performed thymectomies and splenectomies in those animals. We also used several patient-derived T-ALL (PDTALL) models, including one with DLL4 expression on the membrane and we treated PDTALL cells in vitro and in vivo with demcizumab, a blocking antibody against human DLL4 currently being tested in clinical trials in patients with solid cancer. Results: We show that surgical removal of the spleen abrogated T-ALL development in our preclinical DLL4-driven T-ALL mouse model. Mechanistically, we found that the spleen, and not the thymus, promoted the accumulation of circulating CD4+CD8+ T cells before T-ALL onset, suggesting that DLL4-driven T-ALL derives from these cells. Then, we identified a small subset of T-ALL patients showing higher levels of DLL4 expression. Moreover, in mice xenografted with a DLL4-positive PDTALL model, treatment with demcizumab had the same therapeutic effect as global Notch pathway inhibition using the potent γ-secretase inhibitor dibenzazepine. This result demonstrates that, in this PDTALL model, Notch pathway activity depends on DLL4 signaling, thus validating our preclinical mouse model. Conclusion: DLL4 expression in human leukemic cells can be a source of Notch activity in T-ALL, and the spleen plays a major role in a genetic mouse model of DLL4-driven T-ALL.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptores Notch/metabolismo , Bazo/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptores Notch/genética , Bazo/metabolismo , Bazo/cirugía , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Nat Genet ; 52(4): 388-400, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32203470

RESUMEN

Differences in three-dimensional (3D) chromatin architecture can influence the integrity of topologically associating domains (TADs) and rewire specific enhancer-promoter interactions, impacting gene expression and leading to human disease. Here we investigate the 3D chromatin architecture in T cell acute lymphoblastic leukemia (T-ALL) by using primary human leukemia specimens and examine the dynamic responses of this architecture to pharmacological agents. Systematic integration of matched in situ Hi-C, RNA-seq and CTCF ChIP-seq datasets revealed widespread differences in intra-TAD chromatin interactions and TAD boundary insulation in T-ALL. Our studies identify and focus on a TAD 'fusion' event associated with absence of CTCF-mediated insulation, enabling direct interactions between the MYC promoter and a distal super-enhancer. Moreover, our data also demonstrate that small-molecule inhibitors targeting either oncogenic signal transduction or epigenetic regulation can alter specific 3D interactions found in leukemia. Overall, our study highlights the impact, complexity and dynamic nature of 3D chromatin architecture in human acute leukemia.


Asunto(s)
Cromatina/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Linfocitos T/fisiología , Animales , Factor de Unión a CCCTC/genética , Carcinogénesis/genética , Línea Celular Tumoral , Elementos de Facilitación Genéticos/genética , Epigénesis Genética/genética , Humanos , Células Jurkat , Ratones , Regiones Promotoras Genéticas/genética
12.
J Neurochem ; 108(1): 246-59, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19046323

RESUMEN

Culture of chicken nodose neurons with CNTF but not BDNF causes a significant increase in T-type Ca(2+) channel expression. CNTF-induced channel expression requires 12 h stimulation to reach maximal expression and is not affected by inhibition of protein synthesis, suggesting the involvement of a post-translational mechanism. In this study, we have investigated the biochemical mechanism responsible for the CNTF-dependent stimulation of T-type channel expression in nodose neurons. Stimulation of nodose neurons with CNTF evoked a considerable increase in signal transducer and activator of transcription (STAT3) and extracellular signal-regulated kinase (ERK) phosphorylation. CNTF-evoked ERK phosphorylation was transient whereas BDNF-evoked activation of ERK was sustained. Pre-treatment of nodose neurons with the Janus tyrosine kinase (JAK) inhibitor P6 blocked STAT3 and ERK phosphorylation, whereas the ERK inhibitor U0126 prevented ERK activation but not STAT3 phosphorylation. Both P6 and U0126 inhibited the stimulatory effect of CNTF on T-type channel expression. Inhibition of STAT3 activation by the selective blocker stattic has no effect on ERK phosphorylation and T-type channel expression. These results indicate that CNTF-evoked stimulation of T-type Ca(2+) channel expression in chicken nodose neurons requires JAK-dependent ERK signaling. A cardiac tissue extract derived from E20 chicken heart was also effective in promoting T-type Ca(2+) channel expression and STAT3 and ERK phosphorylation. The ability of the heart extract to stimulate JAK/STAT and ERK activation was developmentally regulated. These findings provide further support to the idea that CNTF or a CNTF-like factor mediates normal expression of T-type channels.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Factor Neurotrófico Ciliar/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas Janus/metabolismo , Ganglio Nudoso/citología , Células Receptoras Sensoriales/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Calcio/farmacología , Células Cultivadas , Embrión de Pollo , Subunidad alfa del Receptor del Factor Neurotrófico Ciliar/genética , Subunidad alfa del Receptor del Factor Neurotrófico Ciliar/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Factores de Tiempo
13.
Crit Care Med ; 37(4): 1322-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19242319

RESUMEN

OBJECTIVES: To evaluate the association between plasma granulocyte colony-stimulating factor (G-CSF) levels and clinical outcomes including mortality in patients with acute lung injury (ALI), and to determine whether lower tidal volume ventilation was associated with a more rapid decrease in plasma G-CSF over time in patients with ALI. DESIGN: Retrospective measurement of G-CSF levels in plasma samples that were collected prospectively as part of a large multicenter clinical trial. SETTING: Intensive care units in ten university centers. PATIENTS: The study included 645 patients enrolled in the National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome Clinical Network trial of lower tidal volumes compared with traditional tidal volumes for ALI. MEASUREMENTS AND MAIN RESULTS: Baseline plasma levels of G-CSF were associated with an increased risk of death and a decrease in ventilator-free days and organ failure-free days in multivariate analyses controlling for ventilation strategy, age, and sex (Odds ratio death 1.2/log10 increment G-CSF, 95% confidence interval 1.01 to 1.4). Stratification of G-CSF levels into quartiles revealed a strong association between the highest levels of G-CSF and an increased risk of death and decreased ventilator-free days and organ failure-free days in multivariate analyses controlling for ventilation strategy, Acute Physiology and Chronic Health Evaluation III score, Pao2/Fio2 ratio, creatinine, and platelet count (p < 0.05). Subgroup multivariate analysis of patients with sepsis as their risk factor for ALI revealed a U-shaped association between mortality and G-CSF levels such that risk increased linearly from the second through fourth (highest) quartiles, yet also increased in the first (lowest) quartile. G-CSF levels decreased over time in both tidal volume groups, and there was no statistical difference in the extent of decrease between ventilator strategies. CONCLUSIONS: In patients with ALI, plasma G-CSF levels are associated with morbidity and mortality, but these levels are not influenced by tidal volume strategy. In patients with sepsis-related ALI, a bimodal association between baseline plasma G-CSF levels and subsequent morbidity and mortality from this disease was found.


Asunto(s)
Lesión Pulmonar Aguda/sangre , Factor Estimulante de Colonias de Granulocitos/sangre , Síndrome de Dificultad Respiratoria/sangre , Lesión Pulmonar Aguda/mortalidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Síndrome de Dificultad Respiratoria/mortalidad , Estudios Retrospectivos
14.
Nat Med ; 24(8): 1157-1166, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30038221

RESUMEN

Cellular transformation is accompanied by extensive rewiring of many biological processes leading to augmented levels of distinct types of cellular stress, including proteotoxic stress. Cancer cells critically depend on stress-relief pathways for their survival. However, the mechanisms underlying the transcriptional initiation and maintenance of the oncogenic stress response remain elusive. Here, we show that the expression of heat shock transcription factor 1 (HSF1) and the downstream mediators of the heat shock response is transcriptionally upregulated in T cell acute lymphoblastic leukemia (T-ALL). Hsf1 ablation suppresses the growth of human T-ALL and eradicates leukemia in mouse models of T-ALL, while sparing normal hematopoiesis. HSF1 drives a compact transcriptional program and among the direct HSF1 targets, specific chaperones and co-chaperones mediate its critical role in T-ALL. Notably, we demonstrate that the central T-ALL oncogene NOTCH1 hijacks the cellular stress response machinery by inducing the expression of HSF1 and its downstream effectors. The NOTCH1 signaling status controls the levels of chaperone/co-chaperone complexes and predicts the response of T-ALL patient samples to HSP90 inhibition. Our data demonstrate an integral crosstalk between mediators of oncogene and non-oncogene addiction and reveal critical nodes of the heat shock response pathway that can be targeted therapeutically.


Asunto(s)
Oncogenes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Estrés Fisiológico , Animales , Línea Celular Tumoral , Regulación Leucémica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico/genética , Factores de Transcripción del Choque Térmico/metabolismo , Respuesta al Choque Térmico , Hematopoyesis , Humanos , Ratones Endogámicos C57BL , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptores Notch/metabolismo , Transducción de Señal
15.
Cell Rep ; 15(10): 2159-2169, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27239026

RESUMEN

V(D)J recombination relies on the presence of proximal enhancers that activate the antigen receptor (AgR) loci in a lineage- and stage-specific manner. Unexpectedly, we find that both active and inactive AgR enhancers cooperate to disseminate their effects in a localized and long-range manner. Here, we demonstrate the importance of short-range contacts between active enhancers that constitute an Igk super-enhancer in B cells. Deletion of one element reduces the interaction frequency between other enhancers in the hub, which compromises the transcriptional output of each component. Furthermore, we establish that, in T cells, long-range contact and cooperation between the inactive Igk enhancer MiEκ and the active Tcrb enhancer Eß alters enrichment of CBFß binding in a manner that impacts Tcrb recombination. These findings underline the complexities of enhancer regulation and point to a role for localized and long-range enhancer-sharing between active and inactive elements in lineage- and stage-specific control.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Animales , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Ratones Endogámicos C57BL , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/inmunología
16.
Nat Cell Biol ; 18(11): 1127-1138, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27749823

RESUMEN

Pluripotent embryonic stem cells (ESCs) self-renew or differentiate into all tissues of the developing embryo and cell-specification factors are necessary to balance gene expression. Here we delineate the function of the PHD-finger protein 5a (Phf5a) in ESC self-renewal and ascribe its role in regulating pluripotency, cellular reprogramming and myoblast specification. We demonstrate that Phf5a is essential for maintaining pluripotency, since depleted ESCs exhibit hallmarks of differentiation. Mechanistically, we attribute Phf5a function to the stabilization of the Paf1 transcriptional complex and control of RNA polymerase II elongation on pluripotency loci. Apart from an ESC-specific factor, we demonstrate that Phf5a controls differentiation of adult myoblasts. Our findings suggest a potent mode of regulation by Phf5a in stem cells, which directs their transcriptional programme, ultimately regulating maintenance of pluripotency and cellular reprogramming.


Asunto(s)
Proteínas Portadoras/genética , Diferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Embrionarias de Ratones/citología , Mioblastos/citología , Células Madre Pluripotentes/citología , Transcripción Genética , Envejecimiento , Animales , Línea Celular , Proliferación Celular/genética , Proteínas de Unión al ADN , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al ARN , Transactivadores
17.
Cancer Cell ; 27(3): 321-3, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25759017

RESUMEN

Pre B-ALL is an aggressive cancer of the blood for which treatment of patients with relapsed and refractory disease remains a challenge. In this issue of Cancer Cell, Geng and colleagues surveyed the activation status of the pre-B cell receptor and comprehensively investigated downstream signaling mechanisms currently targetable with small molecule inhibitors.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-bcl-6
18.
Nat Cell Biol ; 17(3): 322-332, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25720964

RESUMEN

​Heat-shock factor 1 (​HSF1) orchestrates the heat-shock response in eukaryotes. Although this pathway has evolved to help cells adapt in the presence of challenging conditions, it is co-opted in cancer to support malignancy. However, the mechanisms that regulate ​HSF1 and thus cellular stress response are poorly understood. Here we show that the ubiquitin ligase ​FBXW7α interacts with ​HSF1 through a conserved motif phosphorylated by ​GSK3ß and ​ERK1. ​FBXW7α ubiquitylates ​HSF1 and loss of ​FBXW7α results in impaired degradation of nuclear ​HSF1 and defective heat-shock response attenuation. ​FBXW7α is either mutated or transcriptionally downregulated in melanoma and ​HSF1 nuclear stabilization correlates with increased metastatic potential and disease progression. ​FBXW7α deficiency and subsequent ​HSF1 accumulation activates an invasion-supportive transcriptional program and enhances the metastatic potential of human melanoma cells. These findings identify a post-translational mechanism of regulation of the ​HSF1 transcriptional program both in the presence of exogenous stress and in cancer.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas F-Box/genética , Glucógeno Sintasa Quinasa 3/genética , Melanoma/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Procesamiento Proteico-Postraduccional , Neoplasias Cutáneas/genética , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Factores de Transcripción del Choque Térmico , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Trasplante de Neoplasias , Alineación de Secuencia , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
19.
Cancer Cell ; 27(6): 755-68, 2015 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-26058075

RESUMEN

The role of the microenvironment in T cell acute lymphoblastic leukemia (T-ALL), or any acute leukemia, is poorly understood. Here we demonstrate that T-ALL cells are in direct, stable contact with CXCL12-producing bone marrow stroma. Cxcl12 deletion from vascular endothelial, but not perivascular, cells impeded tumor growth, suggesting a vascular niche for T-ALL. Moreover, genetic targeting of Cxcr4 in murine T-ALL after disease onset led to rapid, sustained disease remission, and CXCR4 antagonism suppressed human T-ALL in primary xenografts. Loss of CXCR4 targeted key T-ALL regulators, including the MYC pathway, and decreased leukemia initiating cell activity in vivo. Our data identify a T-ALL niche and suggest targeting CXCL12/CXCR4 signaling as a powerful therapeutic approach for T-ALL.


Asunto(s)
Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/biosíntesis , Endotelio Vascular/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Piridinas/farmacología , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Quimiocina CXCL12/genética , Endotelio Vascular/patología , Femenino , Eliminación de Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Nat Genet ; 45(8): 846-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23892662

RESUMEN

Recent studies have identified recurrent mutations in SETBP1, the gene that encodes SET-binding protein 1, in several types of myeloid malignancies, including chronic myeloid and acute myeloid leukemias. The identified mutations frequently target the SKI-homologous domain, although the exact pathogenic mechanisms remain unknown.


Asunto(s)
Proteínas Portadoras/genética , Exoma , Janus Quinasa 3/genética , Leucemia Mieloide/genética , Leucemia Mielomonocítica Juvenil/genética , Mutación , Trastornos Mieloproliferativos/genética , Proteínas Nucleares/genética , Animales , Femenino , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA