Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 140(13): 1482-1495, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-35820055

RESUMEN

Molecular programs initiating cell fate divergence (CFD) are difficult to identify. Current approaches usually compare cells long after CFD initiation, therefore missing molecular changes at its start. Ideally, single cells that differ in their CFD molecular program but are otherwise identical are compared early in CFD. This is possible in diverging sister cells, which were identical until their mother's division and thus differ mainly in CFD properties. In asymmetrically dividing cells, divergent daughter fates are prospectively committed during division, and diverging sisters can thus be identified at the start of CFD. Using asymmetrically dividing blood stem cells, we developed a pipeline (ie, trackSeq) for imaging, tracking, isolating, and transcriptome sequencing of single cells. Their identities, kinship, and histories are maintained throughout, massively improving molecular noise filtering and candidate identification. In addition to many identified blood stem CFD regulators, we offer here this pipeline for use in CFDs other than asymmetric division.


Asunto(s)
Rastreo Celular , Células Madre , Diferenciación Celular , División Celular
2.
Nat Commun ; 14(1): 267, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36650172

RESUMEN

Developmental control of gene expression critically depends on distal cis-regulatory elements including enhancers which interact with promoters to activate gene expression. To date no global experiments have been conducted that identify their cell type and cell stage-specific activity within one developmental pathway and in a chromatin context. Here, we describe a high-throughput method that identifies thousands of differentially active cis-elements able to stimulate a minimal promoter at five stages of hematopoietic progenitor development from embryonic stem (ES) cells, which can be adapted to any ES cell derived cell type. We show that blood cell-specific gene expression is controlled by the concerted action of thousands of differentiation stage-specific sets of cis-elements which respond to cytokine signals terminating at signalling responsive transcription factors. Our work provides an important resource for studies of hematopoietic specification and highlights the mechanisms of how and where extrinsic signals program a cell type-specific chromatin landscape driving hematopoietic differentiation.


Asunto(s)
Cromatina , Secuencias Reguladoras de Ácidos Nucleicos , Cromatina/genética , Diferenciación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regiones Promotoras Genéticas/genética , Elementos de Facilitación Genéticos/genética
3.
Nat Biotechnol ; 40(1): 74-85, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34489600

RESUMEN

Molecular profiling of single cells has advanced our knowledge of the molecular basis of development. However, current approaches mostly rely on dissociating cells from tissues, thereby losing the crucial spatial context of regulatory processes. Here, we apply an image-based single-cell transcriptomics method, sequential fluorescence in situ hybridization (seqFISH), to detect mRNAs for 387 target genes in tissue sections of mouse embryos at the 8-12 somite stage. By integrating spatial context and multiplexed transcriptional measurements with two single-cell transcriptome atlases, we characterize cell types across the embryo and demonstrate that spatially resolved expression of genes not profiled by seqFISH can be imputed. We use this high-resolution spatial map to characterize fundamental steps in the patterning of the midbrain-hindbrain boundary (MHB) and the developing gut tube. We uncover axes of cell differentiation that are not apparent from single-cell RNA-sequencing (scRNA-seq) data, such as early dorsal-ventral separation of esophageal and tracheal progenitor populations in the gut tube. Our method provides an approach for studying cell fate decisions in complex tissues and development.


Asunto(s)
Análisis de la Célula Individual , Transcriptoma , Animales , Hibridación Fluorescente in Situ/métodos , Ratones , Organogénesis/genética , ARN Mensajero/genética , Análisis de la Célula Individual/métodos , Transcriptoma/genética
4.
Mol Cell Biol ; 27(20): 7206-19, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17709394

RESUMEN

The SCL/Tal-1 gene encodes a basic helix-loop-helix transcription factor with key roles in hematopoietic and neural development. SCL is expressed in, and required for, both primitive and definitive erythropoiesis. Thus far, we have identified only one erythroid SCL enhancer. Located 40 kb downstream of exon 1a, the +40 enhancer displays activity in primitive erythroblasts. We demonstrate here that a 3.7-kb fragment containing this element also targets expression to the midbrain, a known site of endogenous SCL expression. Although the 3.7-kb construct was active in primitive, but not definitive, erythroblasts, a larger 5.0-kb fragment, encompassing the 3.7-kb region, was active in both fetal and adult definitive hematopoietic cells. This included Ter119+ erythroid cells along with fetal liver erythroid and myeloid progenitors. Unlike two other SCL hematopoietic enhancers (+18/19 and -4), +40 enhancer transgenes were inactive in the endothelium. A conserved 400-bp core region, essential for both hematopoietic and midbrain +40 enhancer activity in embryos, relied on two GATA/E-box motifs and was bound in vivo by GATA-1 and SCL in erythroid cells. These results suggest a model in which the SCL +18/19 and/or -4 enhancers initiate SCL expression in early mesodermal derivatives capable of generating blood and endothelium, with subsequent activation of the +40 enhancer via an autoregulatory loop.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Elementos de Facilitación Genéticos , Factores de Transcripción GATA/metabolismo , Hematopoyesis/fisiología , Mesencéfalo/fisiología , Proteínas Proto-Oncogénicas , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células de la Médula Ósea/fisiología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Factores de Transcripción GATA/genética , Humanos , Hígado/citología , Hígado/embriología , Hígado/fisiología , Mesencéfalo/citología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Alineación de Secuencia , Homología de Secuencia , Células Madre/fisiología , Proteína 1 de la Leucemia Linfocítica T Aguda , Transgenes
5.
Mol Cell Biol ; 25(12): 5215-25, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15923636

RESUMEN

The stem cell leukemia (SCL) gene, also known as TAL-1, encodes a basic helix-loop-helix protein that is essential for the formation of all hematopoietic lineages, including primitive erythropoiesis. Appropriate transcriptional regulation is essential for the biological functions of SCL, and we have previously identified five distinct enhancers which target different subdomains of the normal SCL expression pattern. However, it is not known whether these SCL enhancers also regulate neighboring genes within the SCL locus, and the erythroid expression of SCL remains unexplained. Here, we have quantitated transcripts from SCL and neighboring genes in multiple hematopoietic cell types. Our results show striking coexpression of SCL and its immediate downstream neighbor, MAP17, suggesting that they share regulatory elements. A systematic survey of histone H3 and H4 acetylation throughout the SCL locus in different hematopoietic cell types identified several peaks of histone acetylation between SIL and MAP17, all of which corresponded to previously characterized SCL enhancers or to the MAP17 promoter. Downstream of MAP17 (and 40 kb downstream of SCL exon 1a), an additional peak of acetylation was identified in hematopoietic cells and was found to correlate with expression of SCL but not other neighboring genes. This +40 region is conserved in human-dog-mouse-rat sequence comparisons, functions as an erythroid cell-restricted enhancer in vitro, and directs beta-galactosidase expression to primitive, but not definitive, erythroblasts in transgenic mice. The SCL +40 enhancer provides a powerful tool for studying the molecular and cellular biology of the primitive erythroid lineage.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Línea Celular , Linaje de la Célula , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Células Madre Hematopoyéticas/citología , Histonas/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Neoplasias , Regiones Promotoras Genéticas , Proteína 1 de la Leucemia Linfocítica T Aguda
6.
Nat Biotechnol ; 18(2): 181-6, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10657125

RESUMEN

The SCL gene encodes a highly conserved bHLH transcription factor with a pivotal role in hemopoiesis and vasculogenesis. We have sequenced and analyzed 320 kb of genomic DNA composing the SCL loci from human, mouse, and chicken. Long-range sequence comparisons demonstrated multiple peaks of human/mouse homology, a subset of which corresponded precisely with known SCL enhancers. Comparisons between mammalian and chicken sequences identified some, but not all, SCL enhancers. Moreover, one peak of human/mouse homology (+23 region), which did not correspond to a known enhancer, showed significant homology to an analogous region of the chicken SCL locus. A transgenic Xenopus reporter assay was established and demonstrated that the +23 region contained a new neural enhancer. This combination of long-range comparative sequence analysis with a high-throughput transgenic bioassay provides a powerful strategy for identifying and characterizing developmentally important enhancers.


Asunto(s)
Secuencia Conservada , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Proteínas Proto-Oncogénicas , Factores de Transcripción/genética , Vertebrados/genética , Proteínas de Xenopus , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Pollos , Secuencias Hélice-Asa-Hélice , Humanos , Ratones , Datos de Secuencia Molecular , Rombencéfalo/embriología , Homología de Secuencia de Aminoácido , Proteína 1 de la Leucemia Linfocítica T Aguda , Xenopus
7.
Leukemia ; 30(1): 14-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26126967

RESUMEN

Transcriptional dysregulation is associated with haematological malignancy. Although mutations of the key haematopoietic transcription factor PU.1 are rare in human acute myeloid leukaemia (AML), they are common in murine models of radiation-induced AML, and PU.1 downregulation and/or dysfunction has been described in human AML patients carrying the fusion oncogenes RUNX1-ETO and PML-RARA. To study the transcriptional programmes associated with compromised PU.1 activity, we adapted a Pu.1-mutated murine AML cell line with an inducible wild-type PU.1. PU.1 induction caused transition from leukaemia phenotype to monocytic differentiation. Global binding maps for PU.1, CEBPA and the histone mark H3K27Ac with and without PU.1 induction showed that mutant PU.1 retains DNA-binding ability, but the induction of wild-type protein dramatically increases both the number and the height of PU.1-binding peaks. Correlating chromatin immunoprecipitation (ChIP) Seq with gene expression data, we found that PU.1 recruitment coupled with increased histone acetylation induces gene expression and activates a monocyte/macrophage transcriptional programme. PU.1 induction also caused the reorganisation of a subgroup of CEBPA binding peaks. Finally, we show that the PU.1 target gene set defined in our model allows the stratification of primary human AML samples, shedding light on both known and novel AML subtypes that may be driven by PU.1 dysfunction.


Asunto(s)
Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Transcripción Genética , Acetilación , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Línea Celular Tumoral , ADN/metabolismo , Genoma Humano , Histonas/metabolismo , Humanos , Monocitos/citología , Monocitos/metabolismo
8.
Oncogene ; 35(3): 279-89, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25893291

RESUMEN

Growing evidence links abnormal epigenetic control to the development of hematological malignancies. Accordingly, inhibition of epigenetic regulators is emerging as a promising therapeutic strategy. The acetylation status of lysine residues in histone tails is one of a number of epigenetic post-translational modifications that alter DNA-templated processes, such as transcription, to facilitate malignant transformation. Although histone deacetylases are already being clinically targeted, the role of histone lysine acetyltransferases (KAT) in malignancy is less well characterized. We chose to study this question in the context of acute myeloid leukemia (AML), where, using in vitro and in vivo genetic ablation and knockdown experiments in murine models, we demonstrate a role for the epigenetic regulators CBP and p300 in the induction and maintenance of AML. Furthermore, using selective small molecule inhibitors of their lysine acetyltransferase activity, we validate CBP/p300 as therapeutic targets in vitro across a wide range of human AML subtypes. We proceed to show that growth retardation occurs through the induction of transcriptional changes that induce apoptosis and cell-cycle arrest in leukemia cells and finally demonstrate the efficacy of the KAT inhibitors in decreasing clonogenic growth of primary AML patient samples. Taken together, these data suggest that CBP/p300 are promising therapeutic targets across multiple subtypes in AML.


Asunto(s)
Proteína p300 Asociada a E1A/genética , Epigénesis Genética , Leucemia Mieloide Aguda/genética , Fragmentos de Péptidos/genética , Sialoglicoproteínas/genética , Animales , Apoptosis/efectos de los fármacos , Benzoatos/administración & dosificación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proteína p300 Asociada a E1A/biosíntesis , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Acetiltransferasas/genética , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Nitrobencenos , Fragmentos de Péptidos/biosíntesis , Pirazoles/administración & dosificación , Pirazolonas , Sialoglicoproteínas/biosíntesis
9.
Oncogene ; 11(1): 131-9, 1995 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-7624120

RESUMEN

The SCL/TAL1 gene was originally identified by virtue of its rearrangement and transcriptional activation in patients with T cell acute lymphoblastic leukaemia. It encodes a helix-loop-helix transcription factor, is not normally expressed in T cells, but is expressed in erythroid, mast, megakaryocytic and progenitor cells. Over-expression of sense and antisense constructs have implicated SCL as a positive regulator of erythroid differentiation. In addition we have previously shown that SCL mRNA levels undergo biphasic modulation during induced erythroid differentiation of murine erythroleukaemia (MEL) cells with a transient early fall followed by a late rise. In this paper we have studied expression of the SCL protein during erythroid differentiation and also the molecular basis for the raised SCL mRNA levels that accompany erythroid differentiation. We have generated an anti-SCL antiserum and used it to demonstrate that an early transient fall in SCL protein does not occur during induced differentiation of MEL cells. Furthermore SCL protein levels underwent a late fall in three different models of erythroid differentiation and in two models of myeloid differentiation. The fall in SCL protein levels during induced erythroid differentiation contrasted with the concomitant marked rise in SCL mRNA levels. These observations have significant implications for the mechanism by which SCL may regulate erythropoiesis. In addition we have demonstrated that the stability of SCL mRNA was only marginally enhanced during erythroid differentiation of MEL cells, whereas the activity of a luciferase reporter construct driven by the SCL promoter was increased 11- to 17-fold. Up-regulation of transcription therefore accounted for most of the increase in SCL mRNA levels during erythroid differentiation.


Asunto(s)
Proteínas de Unión al ADN/genética , Eritropoyesis/genética , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas , Factores de Transcripción , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Línea Celular , Proteínas de Unión al ADN/inmunología , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Transcripción Genética
10.
Oncogene ; 15(20): 2419-28, 1997 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-9395238

RESUMEN

The SCL gene (also known as TAL-1) encodes a basic helix-loop-helix transcription factor that is essential for the development of all haematopoietic lineages, and ectopic expression of which results in T cell leukaemia. SCL is expressed in normal pluripotent haematopoietic stem cells and its expression is maintained during differentiation along erythroid, mast and megakaryocytic lineages, but is extinguished following commitment to other cell types. The mechanisms responsible for this pattern of expression are poorly understood, but are likely to illuminate the molecular basis for stem cell development and lineage commitment. We have identified multiple lineage-restricted DNase I hypersensitive sites in a 45 kb region spanning the murine SCL locus. Committed erythroid cells and CD34 positive primitive myeloid cells exhibited both shared and unique DNase I hypersensitive sites whereas none were found in T cells. The function of each hypersensitive site was studied using both transient and stable reporter assays in erythroid, primitive myeloid and T cells. Multiple positive and negative regulatory elements were characterised and found to display lineage-specificity, promoter-specificity and/or chromatin-dependence. These results represent the first description of key components of a complex network of regulatory elements controlling SCL expression during haematopoiesis.


Asunto(s)
Cromatina/fisiología , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción , Animales , Antígenos CD34/análisis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/genética , Linaje de la Célula/genética , Cromatina/ultraestructura , Proteínas de Unión al ADN/biosíntesis , Desoxirribonucleasa I/metabolismo , Elementos de Facilitación Genéticos , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patología , Ratones , Regiones Promotoras Genéticas , Proteína 1 de la Leucemia Linfocítica T Aguda , Transcripción Genética , Células Tumorales Cultivadas
11.
Oncogene ; 14(10): 1215-22, 1997 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-9121771

RESUMEN

Polycythemia vera (PV) is an acquired clonal disorder characterized by increased production of mature red cells and growth of erythroid colonies in the absence of erythropoietin. Mutation of the erythropoietin receptor has been demonstrated to cause familial polycythemia, but no mutations have been found in PV. Moreover, both erythroid and myeloid progenitors from patients with PV have been reported to be hypersensitive to a number of different growth factors. Attention has therefore focused on post-receptor signal transduction pathways. The SHP-1 gene is an especially attractive candidate gene. Firstly, SHP-1 binds to and negatively regulates signalling from the erythropoietin receptor and is likely to regulate other cytokine receptors in a similar manner. Secondly, absence of SHP-1 protein in the motheaten mouse is accompanied by increased sensitivity of hematopoietic progenitors to a number of cytokines including erythropoietin. Thirdly, familial or sporadic polycythemia in man may result from mutations of the SHP-1 binding domain of the erythropoietin receptor. We have therefore searched for mutations of the SHP-1 gene in genomic DNA from patients with PV. In this disease the majority of peripheral blood lymphocytes are not part of the malignant clone and a variable proportion of myeloid cells may arise from normal progenitors. We have therefore chosen to study DNA from purified peripheral blood granulocytes obtained from nine women in whom the granulocytes were clonally derived. Southern analysis was used to show that the gene was not rearranged and densitometry confirmed the presence of two copies of the gene in each DNA sample. Sequencing of the entire coding region and all splice junctions revealed no mutations. Hematopoietic transcription factor binding sites in the SHP-1 promoter region were intact and the methylation status of the two SHP-1 promoters in PV patients was identical to that in three normal controls. Finally, we showed that levels of SHP-1 protein in granulocytes from patients was similar to those from normal controls. These results demonstrate that the SHP-1 gene is structurally and transcriptionally intact in patients with PV.


Asunto(s)
Policitemia Vera/enzimología , Policitemia Vera/genética , Proteínas Tirosina Fosfatasas/sangre , Proteínas Tirosina Fosfatasas/genética , Anciano , Anciano de 80 o más Años , Southern Blotting , ADN/sangre , ADN/genética , ADN/metabolismo , Metilación de ADN , Exones , Femenino , Reordenamiento Génico , Granulocitos/enzimología , Células Madre Hematopoyéticas/enzimología , Heterocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Persona de Mediana Edad , Policitemia Vera/sangre , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Empalme del ARN , Linfocitos T/enzimología , Transcripción Genética
12.
Leukemia ; 13(5): 750-9, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10374880

RESUMEN

The stem cell leukaemia (SCL) gene is a member of the basic helix-loop-helix family of transcription factors and is essential for the development of all haematopoietic lineages. SCL is expressed in pluripotent haematopoietic stem cells and also following commitment to the erythroid, mast and megakaryocytic lineages. The mechanisms responsible for this pattern of expression are poorly understood, but are likely to illuminate the molecular basis for stem cell development and lineage commitment. Here we present the first description of the regulation of the SCL gene in mast cells. In this study we systematically analysed the chromatin structure of a 45 kb region of the murine SCL locus in mast cells. The pattern of DNase 1 and restriction endonuclease hypersensitive sites in mast cells was distinct from, but overlapped with, the pattern previously described in erythroid and primitive myeloid cells. Each potential regulatory element was tested using transient reporter assays to assess their functional significance in mast cells. These studies identified two potent enhancers, one of which was downstream of the SCL gene. Further characterisation of this 3' enhancer demonstrated that it required the presence of two distinct DNase 1 hypersensitive sites for full activity, and that it was capable of stimulating transcription from both promoter 1a and 1b. Since the 3' enhancer is active in both erythroid and mast cells, it will now be important to see whether it is independently activated in these lineages, or whether it is also active in haematopoietic stem cells.


Asunto(s)
Cromatina/química , Proteínas de Unión al ADN/genética , Mastocitos/metabolismo , Proteínas Proto-Oncogénicas , Factores de Transcripción/genética , Transcripción Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Línea Celular , ADN/análisis , Desoxirribonucleasa I/farmacología , Elementos de Facilitación Genéticos , Ratones , Regiones Promotoras Genéticas , Proteína 1 de la Leucemia Linfocítica T Aguda
13.
Gene ; 239(2): 373-9, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10548740

RESUMEN

We describe here Tdr2, a new class of Tc1-like transposons in zebrafish. Tdr2 was identified from the genomic sequence of a zebrafish PAC (P1 artificial chromosome) clone, and fragments of Tdr2 were found in several zebrafish EST (expressed sequence tag) sequences. Predicted translation of the Tdr2 transposase gene showed that it was most closely related to Caenorhabditis elegans Tc3A, suggesting an ancient origin of the Tdr2 transposon. Tdr2 spans 1. 1kb and is flanked by inverted repeats of approx. 100bp. The 5' repeat is itself composed of an inverted repeat, raising the possibility of the formation of a cruciform DNA structure. Tdr2 transposons may facilitate the development of novel transposon-based tools for the genetic analysis of zebrafish.


Asunto(s)
Elementos Transponibles de ADN/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN/química , ADN/genética , Datos de Secuencia Molecular , Filogenia , Secuencias Repetitivas de Ácidos Nucleicos/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Transposasas/genética
14.
Leukemia ; 28(1): 88-97, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23929215

RESUMEN

Small molecule inhibition of the BET family of proteins, which bind acetylated lysines within histones, has been shown to have a marked therapeutic benefit in pre-clinical models of mixed lineage leukemia (MLL) fusion protein-driven leukemias. Here, we report that I-BET151, a highly specific BET family bromodomain inhibitor, leads to growth inhibition in a human erythroleukemic (HEL) cell line as well as in erythroid precursors isolated from polycythemia vera patients. One of the genes most highly downregulated by I-BET151 was LMO2, an important oncogenic regulator of hematopoietic stem cell development and erythropoiesis. We previously reported that LMO2 transcription is dependent upon Janus kinase 2 (JAK2) kinase activity in HEL cells. Here, we show that the transcriptional changes induced by a JAK2 inhibitor (TG101209) and I-BET151 in HEL cells are significantly over-lapping, suggesting a common pathway of action. We generated JAK2 inhibitor resistant HEL cells and showed that these retain sensitivity to I-BET151. These data highlight I-BET151 as a potential alternative treatment against myeloproliferative neoplasms driven by constitutively active JAK2 kinase.


Asunto(s)
Neoplasias Hematológicas/patología , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/patología , Proteínas Oncogénicas/antagonistas & inhibidores , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Neoplasias Hematológicas/enzimología , Neoplasias Hematológicas/metabolismo , Humanos , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Leukemia ; 27(6): 1348-57, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23302769

RESUMEN

LMO1 is a transcriptional regulator and a T-acute lymphoblastic leukaemia (T-ALL) oncogene. Although first identified in association with a chromosomal translocation in T-ALL, the ectopic expression of LMO1 occurs far more frequently in the absence of any known mutation involving its locus. Given that LMO1 is barely expressed in any haematopoietic lineage, and activation of transcriptional drivers in leukaemic cells is not well described, we investigated the regulation of this gene in normal haematopoietic and leukaemic cells. We show that LMO1 has two promoters that drive reporter gene expression in transgenic mice to neural tissues known to express endogenous LMO1. The LMO1 promoters display bivalent histone marks in multiple blood lineages including T-cells, and a 3' flanking region at LMO1 +57 contains a transcriptional enhancer that is active in developing blood cells in transgenic mouse embryos. The LMO1 promoters become activated in T-ALL together with the 3' enhancer, which is bound in primary T-ALL cells by SCL/TAL1 and GATA3. Taken together, our results show that LMO1 is poised for expression in normal progenitors, where activation of SCL/TAL1 together with a breakdown of epigenetic repression of LMO1 regulatory elements induces ectopic LMO1 expression that contributes to the development and maintenance of T-ALL.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Proteínas con Dominio LIM/genética , Oncogenes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Animales , Inmunoprecipitación de Cromatina , Humanos , Ratones , Ratones Transgénicos
16.
Oncogene ; 32(48): 5471-80, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23708655

RESUMEN

The Lim Domain Only 2 (LMO2) leukaemia oncogene encodes an LIM domain transcriptional cofactor required for early haematopoiesis. During embryogenesis, LMO2 is also expressed in developing tail and limb buds, an expression pattern we now show to be recapitulated in transgenic mice by an enhancer in LMO2 intron 4. Limb bud expression depended on a cluster of HOX binding sites, while posterior tail expression required the HOX sites and two E-boxes. Given the importance of both LMO2 and HOX genes in acute leukaemias, we further demonstrated that the regulatory hierarchy of HOX control of LMO2 is activated in leukaemia mouse models as well as in patient samples. Moreover, Lmo2 knock-down impaired the growth of leukaemic cells, and high LMO2 expression at diagnosis correlated with poor survival in cytogenetically normal AML patients. Taken together, these results establish a regulatory hierarchy of HOX control of LMO2 in normal development, which can be resurrected during leukaemia development. Redeployment of embryonic regulatory hierarchies in an aberrant context is likely to be relevant in human pathologies beyond the specific example of ectopic activation of LMO2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox , Proteínas con Dominio LIM/genética , Mesodermo/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/embriología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Secuencia de Bases , Línea Celular Tumoral , Proliferación Celular , Cromatina/genética , Secuencia Conservada , Elementos E-Box , Extremidades/embriología , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/metabolismo , Humanos , Intrones/genética , Proteínas con Dominio LIM/deficiencia , Ratones , Datos de Secuencia Molecular , Fenotipo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Proto-Oncogénicas/deficiencia , Activación Transcripcional/genética
17.
Oncogene ; 29(43): 5796-808, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-20676125

RESUMEN

The T-cell oncogene Lim-only 2 (LMO2) critically influences both normal and malignant haematopoiesis. LMO2 is not normally expressed in T cells, yet ectopic expression is seen in the majority of T-acute lymphoblastic leukaemia (T-ALL) patients with specific translocations involving LMO2 in only a subset of these patients. Ectopic lmo2 expression in thymocytes of transgenic mice causes T-ALL, and retroviral vector integration into the LMO2 locus was implicated in the development of clonal T-cell disease in patients undergoing gene therapy. Using array-based chromatin immunoprecipitation, we now demonstrate that in contrast to B-acute lymphoblastic leukaemia, human T-ALL samples largely use promoter elements with little influence from distal enhancers. Active LMO2 promoter elements in T-ALL included a previously unrecognized third promoter, which we demonstrate to be active in cell lines, primary T-ALL patients and transgenic mice. The ETS factors ERG and FLI1 previously implicated in lmo2-dependent mouse models of T-ALL bind to the novel LMO2 promoter in human T-ALL samples, while in return LMO2 binds to blood stem/progenitor enhancers in the FLI1 and ERG gene loci. Moreover, LMO2, ERG and FLI1 all regulate the +1 enhancer of HHEX/PRH, which was recently implicated as a key mediator of early progenitor expansion in LMO2-driven T-ALL. Our data therefore suggest that a self-sustaining triad of LMO2/ERG/FLI1 stabilizes the expression of important mediators of the leukaemic phenotype such as HHEX/PRH.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , Metaloproteínas/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Regiones Promotoras Genéticas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Inmunoprecipitación de Cromatina , Expresión Génica , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Proteínas con Dominio LIM , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Proto-Oncogénica c-fli-1/genética , Proteínas Proto-Oncogénicas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética , Regulador Transcripcional ERG
20.
Bioessays ; 17(3): 187-9, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7748171

RESUMEN

The study of haemopoiesis enables us to address one of the central questions of developmental biology, concerning the molecular mechanisms by which a multipotent cell develops into distinct differentiated progeny. Recent work suggests specific roles for retinoic acid receptors at two distinct stages of haemopoiesis. Continuous cell lines of lymphohaemopoietic progenitors were established by infection with a retrovirus containing a dominant negative retinoic acid receptor. The cell lines depend on stem cell factor for their proliferation and can be induced to diffentiate into B-lymphocytes, erythrocytes, neutrophils, monocytes, mast cells and megakaryocytes. Since lymphohaemopoietic progenitors represent less than 0.01% of nucleated marrow cells, immortalised progenitors provide a valuable system with which to study haemopoiesis on a molecular level.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Tretinoina/metabolismo , Animales , Diferenciación Celular , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA