Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(17)2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39273194

RESUMEN

The nuclear hormone family of receptors regulates gene expression. The androgen receptor (AR), upon ligand binding and homodimerization, shuttles from the cytosol into the nucleus to activate gene expression. Thyroid hormone receptors (TRs), retinoic acid receptors (RARs), and the vitamin D receptor (VDR) are present in the nucleus bound to chromatin as a heterodimer with the retinoid X receptors (RXRs) and repress gene expression. Ligand binding leads to transcription activation. The hormonal ligands for these receptors play crucial roles to ensure the proper conduct of very many tissues and exert effects on prostate cancer (PCa) cells. Androgens support PCa proliferation and androgen deprivation alone or with chemotherapy is the standard therapy for PCa. RARγ activation and 3,5,3'-triiodo-L-thyronine (T3) stimulation of TRß support the growth of PCa cells. Ligand stimulation of VDR drives growth arrest, differentiation, and apoptosis of PCa cells. Often these receptors are explored as separate avenues to find treatments for PCa and other cancers. However, there is accumulating evidence to support receptor interactions and crosstalk of regulatory events whereby a better understanding might lead to new combinatorial treatments.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Receptores de Calcitriol , Receptores de Hormona Tiroidea , Humanos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Masculino , Receptores de Calcitriol/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Animales , Hormonas Tiroideas/metabolismo , Terapia Molecular Dirigida
2.
Haematologica ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38813748

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is a cancer of the immune system. Approximately 20% of paediatric and 50% of adult T-ALL patients have refractory disease or relapse and die from the disease. To improve patient outcome new therapeutics are needed. With the aim to identify new therapeutic targets, we combined the analysis of T-ALL gene expression and metabolism to identify the metabolic adaptations that T-ALL cells exhibit. We found that glutamine uptake is essential for T-ALL proliferation. Isotope tracing experiments showed that glutamine fuels aspartate synthesis through the TCA cycle and that glutamine and glutamine-derived aspartate together supply three nitrogen atoms in purines and all but one atom in pyrimidine rings. We show that the glutamate-aspartate transporter EAAT1 (SLC1A3), which is normally expressed in the central nervous system, is crucial for glutamine conversion to aspartate and nucleotides and that T-ALL cell proliferation depends on EAAT1 function. Through this work, we identify EAAT1 as a novel therapeutic target for T-ALL treatment.

3.
Stem Cell Reports ; 18(5): 1090-1106, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37163979

RESUMEN

Mitochondrial dysfunction involving mitochondria-associated ER membrane (MAM) dysregulation is implicated in the pathogenesis of late-onset neurodegenerative diseases, but understanding is limited for rare early-onset conditions. Loss of the MAM-resident protein WFS1 causes Wolfram syndrome (WS), a rare early-onset neurodegenerative disease that has been linked to mitochondrial abnormalities. Here we demonstrate mitochondrial dysfunction in human induced pluripotent stem cell-derived neuronal cells of WS patients. VDAC1 is identified to interact with WFS1, whereas loss of this interaction in WS cells could compromise mitochondrial function. Restoring WFS1 levels in WS cells reinstates WFS1-VDAC1 interaction, which correlates with an increase in MAMs and mitochondrial network that could positively affect mitochondrial function. Genetic rescue by WFS1 overexpression or pharmacological agents modulating mitochondrial function improves the viability and bioenergetics of WS neurons. Our data implicate a role of WFS1 in regulating mitochondrial functionality and highlight a therapeutic intervention for WS and related rare diseases with mitochondrial defects.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Síndrome de Wolfram , Humanos , Síndrome de Wolfram/genética , Síndrome de Wolfram/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Mitocondrias/metabolismo , Mutación
4.
Nucleic Acids Res ; 45(17): 9874-9888, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-28973433

RESUMEN

LMO2 is a bridging factor within a DNA binding complex and is required for definitive haematopoiesis to occur. The developmental stage of the block in haematopoietic specification is not known. We show that Lmo2-/- mouse embryonic stem cells differentiated to Flk-1+ haemangioblasts, but less efficiently to haemogenic endothelium, which only produced primitive haematopoietic progenitors. Genome-wide approaches indicated that LMO2 is required at the haemangioblast stage to position the TAL1/LMO2/LDB1 complex to regulatory elements that are important for the establishment of the haematopoietic developmental program. In the absence of LMO2, the target site recognition of TAL1 is impaired. The lack of LMO2 resulted in altered gene expression levels already at the haemangioblast stage, with transcription factor genes accounting for ∼15% of affected genes. Comparison of Lmo2-/- with Tal1-/- Flk-1+ cells further showed that TAL1 was required to initiate or sustain Lmo2 expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al ADN/genética , ADN/genética , Genoma , Hemangioblastos/metabolismo , Proteínas con Dominio LIM/genética , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Diferenciación Celular , Línea Celular , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos/citología , Hematopoyesis/genética , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Unión Proteica , Proteínas Proto-Oncogénicas/deficiencia , Elementos Reguladores de la Transcripción , Transducción de Señal , Proteína 1 de la Leucemia Linfocítica T Aguda , Transcripción Genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
5.
Development ; 143(23): 4324-4340, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27802171

RESUMEN

The transmission of extracellular signals into the nucleus involves inducible transcription factors, but how different signalling pathways act in a cell type-specific fashion is poorly understood. Here, we studied the regulatory role of the AP-1 transcription factor family in blood development using embryonic stem cell differentiation coupled with genome-wide transcription factor binding and gene expression analyses. AP-1 factors respond to MAP kinase signalling and comprise dimers of FOS, ATF and JUN proteins. To examine genes regulated by AP-1 and to examine how it interacts with other inducible transcription factors, we abrogated its global DNA-binding activity using a dominant-negative FOS peptide. We show that FOS and JUN bind to and activate a specific set of vascular genes and that AP-1 inhibition shifts the balance between smooth muscle and hematopoietic differentiation towards blood. Furthermore, AP-1 is required for de novo binding of TEAD4, a transcription factor connected to Hippo signalling. Our bottom-up approach demonstrates that AP-1- and TEAD4-associated cis-regulatory elements form hubs for multiple signalling-responsive transcription factors and define the cistrome that regulates vascular and hematopoietic development by extrinsic signals.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción Activadores/metabolismo , Animales , Sitios de Unión/genética , Línea Celular , Proteínas de Unión al ADN/genética , Expresión Génica/genética , Perfilación de la Expresión Génica , Ratones , Músculo Liso Vascular/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción de Dominio TEA , Factor de Transcripción AP-1/antagonistas & inhibidores
6.
Dev Cell ; 36(5): 572-87, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26923725

RESUMEN

Metazoan development involves the successive activation and silencing of specific gene expression programs and is driven by tissue-specific transcription factors programming the chromatin landscape. To understand how this process executes an entire developmental pathway, we generated global gene expression, chromatin accessibility, histone modification, and transcription factor binding data from purified embryonic stem cell-derived cells representing six sequential stages of hematopoietic specification and differentiation. Our data reveal the nature of regulatory elements driving differential gene expression and inform how transcription factor binding impacts on promoter activity. We present a dynamic core regulatory network model for hematopoietic specification and demonstrate its utility for the design of reprogramming experiments. Functional studies motivated by our genome-wide data uncovered a stage-specific role for TEAD/YAP factors in mammalian hematopoietic specification. Our study presents a powerful resource for studying hematopoiesis and demonstrates how such data advance our understanding of mammalian development.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Animales , Linaje de la Célula/fisiología , Proteínas de Homeodominio/metabolismo , Ratones , Unión Proteica/genética , Factores de Transcripción/metabolismo
7.
Stem Cell Reports ; 5(2): 232-47, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26235892

RESUMEN

Transcription-factor-induced somatic cell conversions are highly relevant for both basic and clinical research yet their mechanism is not fully understood and it is unclear whether they reflect normal differentiation processes. Here we show that during pre-B-cell-to-macrophage transdifferentiation, C/EBPα binds to two types of myeloid enhancers in B cells: pre-existing enhancers that are bound by PU.1, providing a platform for incoming C/EBPα; and de novo enhancers that are targeted by C/EBPα, acting as a pioneer factor for subsequent binding by PU.1. The order of factor binding dictates the upregulation kinetics of nearby genes. Pre-existing enhancers are broadly active throughout the hematopoietic lineage tree, including B cells. In contrast, de novo enhancers are silent in most cell types except in myeloid cells where they become activated by C/EBP factors. Our data suggest that C/EBPα recapitulates physiological developmental processes by short-circuiting two macrophage enhancer pathways in pre-B cells.


Asunto(s)
Linfocitos B/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Transdiferenciación Celular , Células Mieloides/metabolismo , Mielopoyesis , Proteínas Proto-Oncogénicas c-ets/metabolismo , Linfocitos B/citología , Proteína alfa Potenciadora de Unión a CCAAT/genética , Línea Celular , Humanos , Células Mieloides/citología , Proteínas Proto-Oncogénicas c-ets/genética
8.
Cell Rep ; 12(5): 821-36, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212328

RESUMEN

Acute myeloid leukemia (AML) is characterized by recurrent mutations that affect the epigenetic regulatory machinery and signaling molecules, leading to a block in hematopoietic differentiation. Constitutive signaling from mutated growth factor receptors is a major driver of leukemic growth, but how aberrant signaling affects the epigenome in AML is less understood. Furthermore, AML cells undergo extensive clonal evolution, and the mutations in signaling genes are often secondary events. To elucidate how chronic growth factor signaling alters the transcriptional network in AML, we performed a system-wide multi-omics study of primary cells from patients suffering from AML with internal tandem duplications in the FLT3 transmembrane domain (FLT3-ITD). This strategy revealed cooperation between the MAP kinase (MAPK) inducible transcription factor AP-1 and RUNX1 as a major driver of a common, FLT3-ITD-specific gene expression and chromatin signature, demonstrating a major impact of MAPK signaling pathways in shaping the epigenome of FLT3-ITD AML.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/enzimología , Sistema de Señalización de MAP Quinasas , Mutación , Tirosina Quinasa 3 Similar a fms/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Estructura Terciaria de Proteína , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
9.
Cell Rep ; 8(6): 1974-1988, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25242324

RESUMEN

Oncogenic transcription factors such as RUNX1/ETO, which is generated by the chromosomal translocation t(8;21), subvert normal blood cell development by impairing differentiation and driving malignant self-renewal. Here, we use digital footprinting and chromatin immunoprecipitation sequencing (ChIP-seq) to identify the core RUNX1/ETO-responsive transcriptional network of t(8;21) cells. We show that the transcriptional program underlying leukemic propagation is regulated by a dynamic equilibrium between RUNX1/ETO and RUNX1 complexes, which bind to identical DNA sites in a mutually exclusive fashion. Perturbation of this equilibrium in t(8;21) cells by RUNX1/ETO depletion leads to a global redistribution of transcription factor complexes within preexisting open chromatin, resulting in the formation of a transcriptional network that drives myeloid differentiation. Our work demonstrates on a genome-wide level that the extent of impaired myeloid differentiation in t(8;21) is controlled by the dynamic balance between RUNX1/ETO and RUNX1 activities through the repression of transcription factors that drive differentiation.


Asunto(s)
Leucemia Mieloide Aguda/patología , Translocación Genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Redes Reguladoras de Genes , Humanos , Proteínas con Dominio LIM/metabolismo , Leucemia Mieloide Aguda/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Análisis de Secuencia de ARN , Transactivadores/metabolismo
10.
Cancer Cell ; 24(5): 575-88, 2013 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24183681

RESUMEN

Mutation or epigenetic silencing of the transcription factor C/EBPα is observed in ∼10% of patients with acute myeloid leukemia (AML). In both cases, a common global gene expression profile is observed, but downstream targets relevant for leukemogenesis are not known. Here, we identify Sox4 as a direct target of C/EBPα whereby its expression is inversely correlated with C/EBPα activity. Downregulation of Sox4 abrogated increased self-renewal of leukemic cells and restored their differentiation. Gene expression profiles of leukemia-initiating cells (LICs) from both Sox4 overexpression and murine C/EBPα mutant AML models clustered together but differed from other types of AML. Our data demonstrate that Sox4 overexpression resulting from C/EBPα inactivation contributes to the development of leukemia with a distinct LIC phenotype.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Factores de Transcripción SOXC/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Noqueados , Mutación , Células Mieloides/metabolismo , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Oncogenes , Factores de Transcripción SOXC/metabolismo , Transcriptoma
11.
Mol Cell Biol ; 33(7): 1383-93, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23358417

RESUMEN

KMT2B (MLL2/WBP7) is a member of the MLL subfamily of H3K4-specific histone lysine methyltransferases (KMT2) and is vital for normal embryonic development in the mouse. To gain insight into the molecular mechanism underlying KMT2B function, we focused on MagohB, which is controlled by a CpG island promoter. We show that in cells lacking Mll2-the gene encoding KMT2B-the MagohB promoter resides in inaccessible chromatin and is methylated. To dissect the molecular events leading to the establishment of silencing, we performed kinetic studies in Mll2-conditional-knockout embryonic stem cells. KMT2B depletion was followed by the loss of the active chromatin marks and progressive loss of RNA polymerase II binding with a concomitant downregulation of MagohB expression. Once the active chromatin marks were lost, the MagohB promoter was rapidly methylated. We demonstrate that in the presence of KMT2B, neither transcription elongation nor RNA polymerase II binding is required to maintain H3K4 trimethylation at the MagohB promoter and protect it from DNA methylation. Reexpression of KMT2B was sufficient to reinstate an active MagohB promoter. Our study provides a paradigm for the idea that KMT2 proteins are crucial components for establishing and maintaining the transcriptionally active and unmethylated state of CpG island promoters.


Asunto(s)
Islas de CpG , Metilación de ADN , N-Metiltransferasa de Histona-Lisina/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/genética , ARN Polimerasa II/genética , Animales , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Regulación hacia Abajo , Células Madre Embrionarias/metabolismo , Silenciador del Gen , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Cinética , Ratones , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo
12.
Blood ; 118(8): 2275-84, 2011 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-21730352

RESUMEN

The differentiation of HSCs into myeloid lineages requires the transcription factor PU.1. Whereas PU.1-dependent induction of myeloid-specific target genes has been intensively studied, negative regulation of stem cell or alternate lineage programs remains incompletely characterized. To test for such negative regulatory events, we searched for PU.1-controlled microRNAs (miRs) by expression profiling using a PU.1-inducible myeloid progenitor cell line model. We provide evidence that PU.1 directly controls expression of at least 4 of these miRs (miR-146a, miR-342, miR-338, and miR-155) through temporally dynamic occupation of binding sites within regulatory chromatin regions adjacent to their genomic coding loci. Ectopic expression of the most robustly induced PU.1 target miR, miR-146a, directed the selective differentiation of HSCs into functional peritoneal macrophages in mouse transplantation assays. In agreement with this observation, disruption of Dicer expression or specific antagonization of miR-146a function inhibited the formation of macrophages during early zebrafish (Danio rerio) development. In the present study, we describe a PU.1-orchestrated miR program that mediates key functions of PU.1 during myeloid differentiation.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Mielopoyesis/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Transactivadores/antagonistas & inhibidores , Pez Cebra/embriología , Pez Cebra/genética
13.
Blood ; 117(10): 2827-38, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21239694

RESUMEN

The transcription factor PU.1 occupies a central role in controlling myeloid and early B-cell development, and its correct lineage-specific expression is critical for the differentiation choice of hematopoietic progenitors. However, little is known of how this tissue-specific pattern is established. We previously identified an upstream regulatory cis element whose targeted deletion in mice decreases PU.1 expression and causes leukemia. We show here that the upstream regulatory cis element alone is insufficient to confer physiologic PU.1 expression in mice but requires the cooperation with other, previously unidentified elements. Using a combination of transgenic studies, global chromatin assays, and detailed molecular analyses we present evidence that PU.1 is regulated by a novel mechanism involving cross talk between different cis elements together with lineage-restricted autoregulation. In this model, PU.1 regulates its expression in B cells and macrophages by differentially associating with cell type-specific transcription factors at one of its cis-regulatory elements to establish differential activity patterns at other elements.


Asunto(s)
Linfocitos B/metabolismo , Regulación de la Expresión Génica/genética , Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas/genética , Elementos Reguladores de la Transcripción/genética , Transactivadores/genética , Animales , Southern Blotting , Western Blotting , Separación Celular , Retroalimentación Fisiológica/fisiología , Citometría de Flujo , Expresión Génica , Hematopoyesis/genética , Humanos , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/metabolismo
14.
Mol Cell Biol ; 30(24): 5741-51, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20956563

RESUMEN

Development progresses through a sequence of cellular identities which are determined by the activities of networks of transcription factor genes. Alterations in cis-regulatory elements of these genes play a major role in evolutionary change, but little is known about the mechanisms responsible for maintaining conserved patterns of gene expression. We have studied the evolution of cis-regulatory mechanisms controlling the SCL gene, which encodes a key transcriptional regulator of blood, vasculature, and brain development and exhibits conserved function and pattern of expression throughout vertebrate evolution. SCL cis-regulatory elements are conserved between frog and chicken but accrued alterations at an accelerated rate between 310 and 200 million years ago, with subsequent fixation of a new cis-regulatory pattern at the beginning of the mammalian radiation. As a consequence, orthologous elements shared by mammals and lower vertebrates exhibit functional differences and binding site turnover between widely separated cis-regulatory modules. However, the net effect of these alterations is constancy of overall regulatory inputs and of expression pattern. Our data demonstrate remarkable cis-regulatory remodelling across the SCL locus and indicate that stable patterns of expression can mask extensive regulatory change. These insights illuminate our understanding of vertebrate evolution.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Evolución Biológica , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Proto-Oncogénicas/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Vertebrados/genética , Proteínas de Xenopus/genética , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Pollos , Secuencia Conservada/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Embrión no Mamífero/anatomía & histología , Embrión no Mamífero/fisiología , Genes Reporteros , Hematopoyesis/genética , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/metabolismo , Alineación de Secuencia , Proteína 1 de la Leucemia Linfocítica T Aguda , Distribución Tisular , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo
15.
Blood Cells Mol Dis ; 44(4): 287-90, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20194037

RESUMEN

The transcription factor RUNX1 is essential for definitive hematopoiesis and is required for the expression of a number of important hematopoietic regulator genes. It was recently shown that RUNX1 acts within a narrow developmental window during which it cannot be replaced by other members of the RUNX transcription factor family. Studies of the molecular basis of this phenomenon revealed that RUNX1 is required for the opening of chromatin of important hematopoietic regulator genes and for the formation, but not the maintenance of stable transcription factor complexes on these genes. However, the chromatin opening activity of RUNX1 is context dependent, indicating that it cooperates with alternate transcription factors at different stages of hematopoietic development. This review summarizes recent results on the regulation of chromatin structure by RUNX1 in developing hematopoietic cells.


Asunto(s)
Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hematopoyesis/genética , Animales , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Subunidad beta del Factor de Unión al Sitio Principal/fisiología , Endotelio/citología , Epigénesis Genética/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Complejos Multiproteicos , Proteínas Proto-Oncogénicas/fisiología , Receptor de Factor Estimulante de Colonias de Macrófagos/fisiología , Transactivadores/fisiología , Factores de Transcripción/metabolismo
16.
J Exp Med ; 206(11): 2329-37, 2009 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-19841090

RESUMEN

Runx proteins are essential for hematopoiesis and play an important role in T cell development by regulating key target genes, such as CD4 and CD8 as well as lymphokine genes, during the specialization of naive CD4 T cells into distinct T helper subsets. In regulatory T (T reg) cells, the signature transcription factor Foxp3 interacts with and modulates the function of several other DNA binding proteins, including Runx family members, at the protein level. We show that Runx proteins also regulate the initiation and the maintenance of Foxp3 gene expression in CD4 T cells. Full-length Runx promoted the de novo expression of Foxp3 during inducible T reg cell differentiation, whereas the isolated dominant-negative Runt DNA binding domain antagonized de novo Foxp3 expression. Foxp3 expression in natural T reg cells remained dependent on Runx proteins and correlated with the binding of Runx/core-binding factor beta to regulatory elements within the Foxp3 locus. Our data show that Runx and Foxp3 are components of a feed-forward loop in which Runx proteins contribute to the expression of Foxp3 and cooperate with Foxp3 proteins to regulate the expression of downstream target genes.


Asunto(s)
Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Factores de Transcripción Forkhead/metabolismo , Animales , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Subunidades alfa del Factor de Unión al Sitio Principal/química , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Retroalimentación Fisiológica , Genes Dominantes , Ratones , Estructura Terciaria de Proteína , Linfocitos T Reguladores/metabolismo
17.
Blood ; 114(2): 299-309, 2009 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-19339695

RESUMEN

At the cellular level, development progresses through successive regulatory states, each characterized by their specific gene expression profile. However, the molecular mechanisms regulating first the priming and then maintenance of gene expression within one developmental pathway are essentially unknown. The hematopoietic system represents a powerful experimental model to address these questions and here we have focused on a regulatory circuit playing a central role in myelopoiesis: the transcription factor PU.1, its target gene colony-stimulating-factor 1 receptor (Csf1r), and key upstream regulators such as RUNX1. We find that during ontogeny, chromatin unfolding precedes the establishment of active histone marks and the formation of stable transcription factor complexes at the Pu.1 locus and we show that chromatin remodeling is mediated by the transient binding of RUNX1 to Pu.1 cis-elements. By contrast, chromatin reorganization of Csf1r requires prior expression of PU.1 together with RUNX1 binding. Once the full hematopoietic program is established, stable transcription factor complexes and active chromatin can be maintained without RUNX1. Our experiments therefore demonstrate how individual transcription factors function in a differentiation stage-specific manner to differentially affect the initiation versus maintenance of a developmental program.


Asunto(s)
Células Sanguíneas/metabolismo , Cromatina/genética , Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Animales , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Metilación de ADN , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , Factores de Tiempo
18.
Genes Dev ; 22(15): 2085-92, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18676813

RESUMEN

The transcription factor PU.1 is an important regulator of hematopoiesis; precise expression levels are critical for normal hematopoietic development and suppression of leukemia. We show here that noncoding antisense RNAs are important modulators of proper dosages of PU.1. Antisense and sense RNAs are regulated by shared evolutionarily conserved cis-regulatory elements, and we can show that antisense RNAs inhibit PU.1 expression by modulating mRNA translation. We propose that such antisense RNAs will likely be important in the regulation of many genes and may be the reason for the large number of overlapping complementary transcripts with so far unknown function.


Asunto(s)
Expresión Génica , Proteínas Proto-Oncogénicas/genética , ARN sin Sentido/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transactivadores/genética , Animales , Línea Celular , Electroporación , Granulocitos/citología , Granulocitos/metabolismo , Células HL-60 , Humanos , Separación Inmunomagnética , Células Jurkat , Macrófagos/metabolismo , Ratones , Modelos Genéticos , Biosíntesis de Proteínas , Interferencia de ARN , ARN sin Sentido/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Linfocitos T/metabolismo , Transcripción Genética , Células U937
19.
Semin Immunol ; 20(4): 257-63, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18579409

RESUMEN

Hematopoietic stem cells exhibit a multi-lineage gene expression program, and this expression program is either maintained when these cells self-renew, or re-programmed when they differentiate. Both processes require the regulated expression of sequence-specific transcription factors and their interaction with the epigenetic regulatory machinery which programs the chromatin of hematopoietic genes in a cell type specific fashion. This article describes recent findings on the complexity of these molecular interactions and their consequences with respect to the regulation of cell fate decisions. We also describe recent findings from studies of genes expressed in the myeloid lineage (Pu.1 and csf1r) which highlight some of the molecular principles governing cell fate decisions at the epigenetic level.


Asunto(s)
Diferenciación Celular/fisiología , Cromatina/fisiología , Regulación de la Expresión Génica/fisiología , Células Mieloides/fisiología , Factores de Transcripción/fisiología , Animales , Linaje de la Célula , Silenciador del Gen/fisiología , Humanos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
20.
Mol Cell Biol ; 27(21): 7425-38, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17785440

RESUMEN

The Ets family transcription factor PU.1 is crucial for the regulation of hematopoietic development. Pu.1 is activated in hematopoietic stem cells and is expressed in mast cells, B cells, granulocytes, and macrophages but is switched off in T cells. Many of the transcription factors regulating Pu.1 have been identified, but little is known about how they organize Pu.1 chromatin in development. We analyzed the Pu.1 promoter and the upstream regulatory element (URE) using in vivo footprinting and chromatin immunoprecipitation assays. In B cells, Pu.1 was bound by a set of transcription factors different from that in myeloid cells and adopted alternative chromatin architectures. In T cells, Pu.1 chromatin at the URE was open and the same transcription factor binding sites were occupied as in B cells. The transcription factor RUNX1 was bound to the URE in precursor cells, but binding was down-regulated in maturing cells. In PU.1 knockout precursor cells, the Ets factor Fli-1 compensated for the lack of PU.1, and both proteins could occupy a subset of Pu.1 cis elements in PU.1-expressing cells. In addition, we identified novel URE-derived noncoding transcripts subject to tissue-specific regulation. Our results provide important insights into how overlapping, but different, sets of transcription factors program tissue-specific chromatin structures in the hematopoietic system.


Asunto(s)
Cromatina/química , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Proteínas Proto-Oncogénicas/genética , ARN no Traducido/genética , Transactivadores/genética , Transcripción Genética , Animales , Linfocitos B/enzimología , Linfocitos B/metabolismo , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Macrófagos/enzimología , Macrófagos/metabolismo , Ratones , Datos de Secuencia Molecular , Células Mieloides/citología , Células Mieloides/metabolismo , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Linfocitos T/enzimología , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...