Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Blood ; 139(7): 1080-1097, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34695195

RESUMEN

In an effort to identify novel drugs targeting fusion-oncogene-induced acute myeloid leukemia (AML), we performed high-resolution proteomic analysis. In AML1-ETO (AE)-driven AML, we uncovered a deregulation of phospholipase C (PLC) signaling. We identified PLCgamma 1 (PLCG1) as a specific target of the AE fusion protein that is induced after AE binding to intergenic regulatory DNA elements. Genetic inactivation of PLCG1 in murine and human AML inhibited AML1-ETO dependent self-renewal programs, leukemic proliferation, and leukemia maintenance in vivo. In contrast, PLCG1 was dispensable for normal hematopoietic stem and progenitor cell function. These findings are extended to and confirmed by pharmacologic perturbation of Ca++-signaling in AML1-ETO AML cells, indicating that the PLCG1 pathway poses an important therapeutic target for AML1-ETO+ leukemic stem cells.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/patología , Proteínas de Fusión Oncogénica/metabolismo , Fosfolipasa C gamma/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Animales , Autorrenovación de las Células , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/genética , Fosfolipasa C gamma/genética , Proteoma , Proteína 1 Compañera de Translocación de RUNX1/genética , Transcriptoma , Translocación Genética
2.
Haematologica ; 2024 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-38841800

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common malignancy that develops in patients with ataxia-telangiectasia, a cancer-predisposing inherited syndrome characterized by inactivating germline ATM mutations. ATM is also frequently mutated in sporadic DLBCL. To investigate lymphomagenic mechanisms and lymphoma-specific dependencies underlying defective ATM, we applied ribonucleic acid (RNA)-seq and genome-scale loss-offunction clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screens to systematically interrogate B-cell lymphomas arising in a novel murine model (Atm-/-nu-/-) with constitutional Atm loss, thymic aplasia but residual T-cell populations. Atm-/-nu-/-lymphomas, which phenotypically resemble either activated B-cell-like or germinal center Bcell-like DLBCL, harbor a complex karyotype, and are characterized by MYC pathway activation. In Atm-/-nu-/-lymphomas, we discovered nucleotide biosynthesis as a MYCdependent cellular vulnerability that can be targeted through the synergistic nucleotidedepleting actions of mycophenolate mofetil (MMF) and the WEE1 inhibitor, adavosertib (AZD1775). The latter is mediated through a synthetically lethal interaction between RRM2 suppression and MYC dysregulation that results in replication stress overload in Atm-/-nu-/-lymphoma cells. Validation in cell line models of human DLBCL confirmed the broad applicability of nucleotide depletion as a therapeutic strategy for MYC-driven DLBCL independent of ATM mutation status. Our findings extend current understanding of lymphomagenic mechanisms underpinning ATM loss and highlight nucleotide metabolism as a targetable therapeutic vulnerability in MYC-driven DLBCL.

3.
Haematologica ; 106(6): 1569-1580, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32299907

RESUMEN

Hematological malignancies are characterised by a block in differentiation, which in many cases is caused by recurrent mutations affecting the activity of hematopoietic transcription factors. RUNX1-EVI1 is a fusion protein formed by the t(3;21) translocation linking two transcription factors required for normal hematopoiesis. RUNX1-EVI1 expression is found in myelodysplastic syndrome, secondary acute myeloid leukemia, and blast crisis of chronic myeloid leukemia; with clinical outcomes being worse than in patients with RUNX1-ETO, RUNX1 or EVI1 mutations alone. RUNX1-EVI1 is usually found as a secondary mutation, therefore the molecular mechanisms underlying how RUNX1-EVI1 alone contributes to poor prognosis are unknown. To address this question, we induced expression of RUNX1-EVI1 in hematopoietic cells derived from an embryonic stem cell differentiation model. Induction resulted in disruption of the RUNX1-dependent endothelial-hematopoietic transition, blocked the cell cycle and undermined cell fate decisions in multipotent hematopoietic progenitor cells. Integrative analyses of gene expression with chromatin and transcription factor binding data demonstrated that RUNX1-EVI1 binding caused the re-distribution of endogenous RUNX1 within the genome and interfered with both RUNX1 and EVI1 regulated gene expression programs. In summary, RUNX1-EVI1 expression alone leads to extensive epigenetic reprogramming which is incompatible with healthy blood production.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Leucemia Mieloide Aguda , Ciclo Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Redes Reguladoras de Genes , Humanos , Leucemia Mieloide Aguda/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Translocación Genética
4.
EMBO J ; 35(5): 515-35, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26796577

RESUMEN

Immunological memory is a defining feature of vertebrate physiology, allowing rapid responses to repeat infections. However, the molecular mechanisms required for its establishment and maintenance remain poorly understood. Here, we demonstrated that the first steps in the acquisition of T-cell memory occurred during the initial activation phase of naïve T cells by an antigenic stimulus. This event initiated extensive chromatin remodeling that reprogrammed immune response genes toward a stably maintained primed state, prior to terminal differentiation. Activation induced the transcription factors NFAT and AP-1 which created thousands of new DNase I-hypersensitive sites (DHSs), enabling ETS-1 and RUNX1 recruitment to previously inaccessible sites. Significantly, these DHSs remained stable long after activation ceased, were preserved following replication, and were maintained in memory-phenotype cells. We show that primed DHSs maintain regions of active chromatin in the vicinity of inducible genes and enhancers that regulate immune responses. We suggest that this priming mechanism may contribute to immunological memory in T cells by facilitating the induction of nearby inducible regulatory elements in previously activated T cells.


Asunto(s)
Cromatina/metabolismo , Memoria Inmunológica , Linfocitos T/metabolismo , Animales , Células Cultivadas , Quimiocina CCL1/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Desoxirribonucleasa I/metabolismo , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Interleucina-3/genética , Células Jurkat , Ratones Transgénicos , Factores de Transcripción NFATC/genética , Proteína Proto-Oncogénica c-ets-1/genética , ARN Mensajero/metabolismo , Bazo/inmunología , Linfocitos T/inmunología , Factor de Transcripción AP-1/genética
5.
PLoS Comput Biol ; 15(11): e1007337, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31682597

RESUMEN

Gene expression governs cell fate, and is regulated via a complex interplay of transcription factors and molecules that change chromatin structure. Advances in sequencing-based assays have enabled investigation of these processes genome-wide, leading to large datasets that combine information on the dynamics of gene expression, transcription factor binding and chromatin structure as cells differentiate. While numerous studies focus on the effects of these features on broader gene regulation, less work has been done on the mechanisms of gene-specific transcriptional control. In this study, we have focussed on the latter by integrating gene expression data for the in vitro differentiation of murine ES cells to macrophages and cardiomyocytes, with dynamic data on chromatin structure, epigenetics and transcription factor binding. Combining a novel strategy to identify communities of related control elements with a penalized regression approach, we developed individual models to identify the potential control elements predictive of the expression of each gene. Our models were compared to an existing method and evaluated using the existing literature and new experimental data from embryonic stem cell differentiation reporter assays. Our method is able to identify transcriptional control elements in a gene specific manner that reflect known regulatory relationships and to generate useful hypotheses for further testing.


Asunto(s)
Diferenciación Celular/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Elementos Reguladores de la Transcripción/genética , Animales , Diferenciación Celular/fisiología , Cromatina/metabolismo , Bases de Datos Genéticas , Epigénesis Genética , Epigenómica , Regulación de la Expresión Génica/genética , Genoma , Macrófagos/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo
6.
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
7.
Blood ; 130(10): 1213-1222, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28710059

RESUMEN

Understanding and blocking the self-renewal pathway of preleukemia stem cells could prevent acute myeloid leukemia (AML) relapse. In this study, we show that increased FOXO1 represents a critical mechanism driving aberrant self-renewal in preleukemic cells expressing the t(8;21)-associated oncogene AML1-ETO (AE). Although generally considered as a tumor suppressor, FOXO1 is consistently upregulated in t(8;21) AML. Expression of FOXO1 in human CD34+ cells promotes a preleukemic state with enhanced self-renewal and dysregulated differentiation. The DNA binding domain of FOXO1 is essential for these functions. FOXO1 activates a stem cell molecular signature that is also present in AE preleukemia cells and preserved in t(8;21) patient samples. Genome-wide binding studies show that AE and FOXO1 share the majority of their binding sites, whereby FOXO1 binds to multiple crucial self-renewal genes and is required for their activation. In agreement with this observation, genetic and pharmacological ablation of FOXO1 inhibited the long-term proliferation and clonogenicity of AE cells and t(8;21) AML cell lines. Targeting of FOXO1 therefore provides a potential therapeutic strategy for elimination of stem cells at both preleukemic and leukemic stages.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteína Forkhead Box O1/metabolismo , Redes Reguladoras de Genes , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/metabolismo , Lesiones Precancerosas/genética , Animales , Antígenos CD34/metabolismo , Línea Celular Tumoral , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Genoma Humano , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Ratones SCID , Proteínas de Fusión Oncogénica/genética , Lesiones Precancerosas/patología , Proteína 1 Compañera de Translocación de RUNX1 , Regulación hacia Arriba/genética
8.
Immunity ; 30(4): 508-20, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19345119

RESUMEN

Pax5 is an essential regulator of B cell identity and function. Here, we used transgenesis and deletion mapping to identify a potent enhancer in intron 5 of the Pax5 locus. This enhancer in combination with the promoter region was sufficient to recapitulate the B lymphoid expression of Pax5. The enhancer was silenced by DNA methylation in embryonic stem cells, but became activated in multipotent hematopoietic progenitors. It contained functional binding sites for the transcription factors PU.1, IRF4, IRF8, and NF-kappaB, suggesting that these regulators contribute to sequential enhancer activation in hematopoietic progenitors and during B cell development. In contrast, the promoter region was repressed by Polycomb group proteins in non-B cells and was activated only at the onset of pro-B cell development through induction of chromatin remodeling by the transcription factor EBF1. These experiments demonstrate a stepwise activation of Pax5 in early lymphopoiesis and provide mechanistic insights into the process of B cell commitment.


Asunto(s)
Linfocitos B/inmunología , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Linfopoyesis/fisiología , Factor de Transcripción PAX5 , Regiones Promotoras Genéticas , Transgenes/genética , Animales , Linfocitos B/citología , Secuencia de Bases , Cromosomas Artificiales Bacterianos/genética , Citometría de Flujo , Humanos , Ratones , Datos de Secuencia Molecular , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Transactivadores/genética , Regulación hacia Arriba
9.
Eur J Immunol ; 46(3): 634-46, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26631626

RESUMEN

The link between the extensive usage of calcineurin (CN) inhibitors cyclosporin A and tacrolimus (FK506) in transplantation medicine and the increasing rate of opportunistic infections within this segment of patients is alarming. Currently, how peritoneal infections are favored by these drugs, which impair the activity of several signaling pathways including the Ca(++) /CN/NFAT, Ca(++) /CN/cofilin, Ca(++) /CN/BAD, and NF-κB networks, is unknown. Here, we show that Saccharomyces cerevisiae infection of peritoneal resident macrophages triggers the transient nuclear translocation of NFATc1ß isoforms, resulting in a coordinated, CN-dependent induction of the Ccl2, Ccl7, and Ccl12 genes, all encoding CCR2 agonists. CN inhibitors block the CCR2-dependent recruitment of inflammatory monocytes (IM) to the peritoneal cavities of S. cerevisiae infected mice. In myeloid cells, NFATc1/ß proteins represent the most prominent NFATc1 isoforms. NFATc1/ß ablation leads to a decrease of CCR2 chemokines, impaired mobilization of IMs, and delayed clearance of infection. We show that, upon binding to a composite NFAT/BCL6 regulatory element within the Ccl2 promoter, NFATc1/ß proteins release the BCL6-dependent repression of Ccl2 gene in macrophages. These findings suggest a novel CN-dependent cross-talk between NFAT and BCL6 transcription factors, which may affect the outcome of opportunistic fungal infections in immunocompromised patients.


Asunto(s)
Macrófagos Peritoneales/metabolismo , Factores de Transcripción NFATC/inmunología , Factores de Transcripción NFATC/fisiología , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores CCR2/agonistas , Receptores CCR2/inmunología , Saccharomyces cerevisiae/inmunología , Animales , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Quimiocina CCL2/genética , Quimiocina CCL7/genética , Macrófagos Peritoneales/microbiología , Ratones , Proteínas Quimioatrayentes de Monocitos/genética , Monocitos/inmunología , FN-kappa B/metabolismo , Factores de Transcripción NFATC/deficiencia , Factores de Transcripción NFATC/genética , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/virología , Regiones Promotoras Genéticas , Isoformas de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-6/genética
10.
Development ; 141(12): 2391-401, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24850855

RESUMEN

Mammalian development is regulated by the interplay of tissue-specific and ubiquitously expressed transcription factors, such as Sp1. Sp1 knockout mice die in utero with multiple phenotypic aberrations, but the underlying molecular mechanism of this differentiation failure has been elusive. Here, we have used conditional knockout mice as well as the differentiation of mouse ES cells as a model with which to address this issue. To this end, we examined differentiation potential, global gene expression patterns and Sp1 target regions in Sp1 wild-type and Sp1-deficient cells representing different stages of hematopoiesis. Sp1(-/-) cells progress through most embryonic stages of blood cell development but cannot complete terminal differentiation. This failure to fully differentiate is not seen when Sp1 is knocked out at later developmental stages. For most Sp1 target and non-target genes, gene expression is unaffected by Sp1 inactivation. However, Cdx genes and multiple Hox genes are stage-specific targets of Sp1 and are downregulated at an early stage. As a consequence, expression of genes involved in hematopoietic specification is progressively deregulated. Our work demonstrates that the early absence of active Sp1 sets a cascade in motion that culminates in a failure of terminal hematopoietic differentiation and emphasizes the role of ubiquitously expressed transcription factors for tissue-specific gene regulation. In addition, our global side-by-side analysis of the response of the transcriptional network to perturbation sheds a new light on the regulatory hierarchy of hematopoietic specification.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/citología , Factor de Transcripción Sp1/fisiología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Macrófagos/citología , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Unión Proteica , Células Madre/citología
11.
Adv Exp Med Biol ; 962: 65-81, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28299651

RESUMEN

RUNX transcription factors belong to a highly conserved class of transcriptional regulators which play various roles in the development of the majority of metazoans. In this review we focus on the founding member of the family, RUNX1, and its role in the transcriptional control of blood cell development in mammals. We summarize data showing that RUNX1 functions both as activator and repressor within a chromatin environment, a feature that requires its interaction with multiple other transcription factors and co-factors. Furthermore, we outline how RUNX1 works together with other factors to reshape the epigenetic landscape and the three-dimensional structure of gene loci within the nucleus. Finally, we review how aberrant forms of RUNX1 deregulate blood cell development and cause hematopoietic malignancies.


Asunto(s)
Células Sanguíneas/metabolismo , Células Sanguíneas/fisiología , Diferenciación Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Animales , Cromatina/metabolismo , Neoplasias Hematológicas/metabolismo , Humanos , Factores de Transcripción/metabolismo , Transcripción Genética/genética
12.
Proc Natl Acad Sci U S A ; 111(42): E4513-22, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25288773

RESUMEN

Deregulated transcription factor (TF) activities are commonly observed in hematopoietic malignancies. Understanding tumorigenesis therefore requires determining the function and hierarchical role of individual TFs. To identify TFs central to lymphomagenesis, we identified lymphoma type-specific accessible chromatin by global mapping of DNaseI hypersensitive sites and analyzed enriched TF-binding motifs in these regions. Applying this unbiased approach to classical Hodgkin lymphoma (HL), a common B-cell-derived lymphoma with a complex pattern of deregulated TFs, we discovered interferon regulatory factor (IRF) sites among the top enriched motifs. High-level expression of the proinflammatory TF IRF5 was specific to HL cells and crucial for their survival. Furthermore, IRF5 initiated a regulatory cascade in human non-Hodgkin B-cell lines and primary murine B cells by inducing the TF AP-1 and cooperating with NF-κB to activate essential characteristic features of HL. Our strategy efficiently identified a lymphoma type-specific key regulator and uncovered a tumor promoting role of IRF5.


Asunto(s)
Cromatina/metabolismo , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/metabolismo , Factores Reguladores del Interferón/metabolismo , Factor de Transcripción AP-1/metabolismo , Secuencias de Aminoácidos , Animales , Linfocitos B/citología , Línea Celular Tumoral , Linaje de la Célula , Quimiocinas/metabolismo , Quimiotaxis , Citocinas/metabolismo , Desoxirribonucleasa I/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación , Leucocitos Mononucleares/citología , Linfoma/metabolismo , Linfoma no Hodgkin/metabolismo , Ratones , FN-kappa B/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Plásmidos/metabolismo , Bazo/citología
13.
EMBO J ; 31(22): 4318-33, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-23064151

RESUMEN

Cell fate decisions during haematopoiesis are governed by lineage-specific transcription factors, such as RUNX1, SCL/TAL1, FLI1 and C/EBP family members. To gain insight into how these transcription factors regulate the activation of haematopoietic genes during embryonic development, we measured the genome-wide dynamics of transcription factor assembly on their target genes during the RUNX1-dependent transition from haemogenic endothelium (HE) to haematopoietic progenitors. Using a Runx1-/- embryonic stem cell differentiation model expressing an inducible Runx1 gene, we show that in the absence of RUNX1, haematopoietic genes bind SCL/TAL1, FLI1 and C/EBPß and that this early priming is required for correct temporal expression of the myeloid master regulator PU.1 and its downstream targets. After induction, RUNX1 binds to numerous de novo sites, initiating a local increase in histone acetylation and rapid global alterations in the binding patterns of SCL/TAL1 and FLI1. The acquisition of haematopoietic fate controlled by Runx1 therefore does not represent the establishment of a new regulatory layer on top of a pre-existing HE program but instead entails global reorganization of lineage-specific transcription factor assemblies.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Epigénesis Genética/fisiología , Hematopoyesis/fisiología , Acetilación , Animales , Secuencia de Bases , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Embrionarias/fisiología , Epigénesis Genética/genética , Hematopoyesis/genética , Histonas/metabolismo , Ratones , Datos de Secuencia Molecular , Unión Proteica , Factores de Transcripción/fisiología
14.
Blood ; 124(11): e11-20, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25082880

RESUMEN

During ontogeny, the transcription factor RUNX1 governs the emergence of definitive hematopoietic cells from specialized endothelial cells called hemogenic endothelium (HE). The ultimate consequence of this endothelial-to-hematopoietic transition is the concomitant activation of the hematopoietic program and downregulation of the endothelial program. However, due to the rare and transient nature of the HE, little is known about the initial role of RUNX1 within this population. We, therefore, developed and implemented a highly sensitive DNA adenine methyltransferase identification-based methodology, including a novel data analysis pipeline, to map early RUNX1 transcriptional targets in HE cells. This novel transcription factor binding site identification protocol should be widely applicable to other low abundance cell types and factors. Integration of the RUNX1 binding profile with gene expression data revealed an unexpected early role for RUNX1 as a positive regulator of cell adhesion- and migration-associated genes within the HE. This suggests that RUNX1 orchestrates HE cell positioning and integration prior to the release of hematopoietic cells. Overall, our genome-wide analysis of the RUNX1 binding and transcriptional profile in the HE provides a novel comprehensive resource of target genes that will facilitate the precise dissection of the role of RUNX1 in early blood development.


Asunto(s)
Movimiento Celular/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Hematopoyesis/fisiología , Animales , Adhesión Celular/fisiología , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Endoteliales/citología , Endotelio Vascular/citología , Ratones , Ratones Noqueados
15.
Mol Cell ; 32(1): 129-39, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18851839

RESUMEN

Transcription of the lysozyme gene is rapidly induced by proinflammatory stimuli such as treatment with bacterial lipopolysaccharide (LPS). Here we show that this induction involves both the relief of repression mediated by the enhancer-blocking protein CTCF that binds to a negative regulatory element at -2.4 kb, and the activation of two flanking enhancer elements. The downstream enhancer has promoter activity, and LPS stimulation initiates the transient synthesis of a noncoding RNA (LINoCR) transcribed through the -2.4 kb element. Expression of LINoCR is correlated with IKKalpha recruitment, histone H3 phosphoacetylation in the transcribed region, the repositioning of a nucleosome over the CTCF binding site, and, eventually, CTCF eviction. Each of these events requires transcription elongation. Our data reveal a transcription-dependent mechanism of chromatin remodeling that switches a cis-regulatory region from a repressive to an active conformation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Muramidasa/genética , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Factor de Unión a CCCTC , Línea Celular , Pollos , Cartilla de ADN/genética , Elementos de Facilitación Genéticos , Histonas/metabolismo , Lipopolisacáridos/farmacología , Nucleosomas/efectos de los fármacos , Nucleosomas/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética , Elementos Reguladores de la Transcripción , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
16.
Yale J Biol Med ; 89(4): 513-525, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28018142

RESUMEN

Sp1 belongs to the 26 member strong Sp/KLF family of transcription factors. It is a paradigm for a ubiquitously expressed transcription factor and is involved in regulating the expression of genes associated with a wide range of cellular processes in mammalian cells. Sp1 can interact with a range of proteins, including other transcription factors, members of the transcription initiation complex and epigenetic regulators, enabling tight regulation of its target genes. In this review, we discuss the mechanisms involved in Sp1-mediated transcriptional regulation, as well as how a ubiquitous transcription factor can be involved in establishing a tissue-specific pattern of gene expression and mechanisms by which its activity may be regulated. We also consider the role of Sp1 in human diseases, such as cancer.


Asunto(s)
Factor de Transcripción Sp1/metabolismo , Animales , Regulación de la Expresión Génica/genética , Humanos , Factor de Transcripción Sp1/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética
17.
BMC Genomics ; 16: 1000, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26608661

RESUMEN

BACKGROUND: The analysis of differential gene expression is a fundamental tool to relate gene regulation with specific biological processes. Differential binding of transcription factors (TFs) can drive differential gene expression. While DNase-seq data can provide global snapshots of TF binding, tools for detecting differential binding from pairs of DNase-seq data sets are lacking. RESULTS: In order to link expression changes with changes in TF binding we introduce the concept of differential footprinting alongside a computational tool. We demonstrate that differential footprinting is associated with differential gene expression and can be used to define cell types by their specific TF occupancy patterns. CONCLUSIONS: Our new tool, Wellington-bootstrap, will enable the detection of differential TF binding facilitating the study of gene regulatory systems.


Asunto(s)
Sitios de Unión , Biología Computacional/métodos , Huella de ADN , Desoxirribonucleasas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Factores de Transcripción/metabolismo , Antígenos CD19/metabolismo , Subgrupos de Linfocitos B/metabolismo , Linfocitos T CD8-positivos/metabolismo , Análisis por Conglomerados , Huella de ADN/métodos , Regulación de la Expresión Génica , Humanos , Especificidad de Órganos/genética , Unión Proteica
18.
EMBO J ; 30(19): 4059-70, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21873977

RESUMEN

The transcription factor RUNX1 is essential to establish the haematopoietic gene expression programme; however, the mechanism of how it activates transcription of haematopoietic stem cell (HSC) genes is still elusive. Here, we obtained novel insights into RUNX1 function by studying regulation of the human CD34 gene, which is expressed in HSCs. Using transgenic mice carrying human CD34 PAC constructs, we identified a novel downstream regulatory element (DRE), which is bound by RUNX1 and is necessary for human CD34 expression in long-term (LT)-HSCs. Conditional deletion of Runx1 in mice harbouring human CD34 promoter-DRE constructs abrogates human CD34 expression. We demonstrate by chromosome conformation capture assays in LT-HSCs that the DRE physically interacts with the human CD34 promoter. Targeted mutagenesis of RUNX binding sites leads to perturbation of this interaction and decreased human CD34 expression in LT-HSCs. Overall, our in vivo data provide novel evidence about the role of RUNX1 in mediating interactions between distal and proximal elements of the HSC gene CD34.


Asunto(s)
Antígenos CD34/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Animales , Trasplante de Médula Ósea , Cromatina/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Sangre Fetal/citología , Genotipo , Células HL-60 , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Secuencias Reguladoras de Ácidos Nucleicos/genética
19.
Blood ; 122(5): 759-69, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23616623

RESUMEN

The activation of B-cell-specific genes, such as CD19 and PAX5, is a hallmark of t(8;21) acute myeloid leukemia (AML) which expresses the translocation product RUNX1/ETO. PAX5 is an important regulator of B-lymphoid development and blocks myeloid differentiation when ectopically expressed. To understand the molecular mechanism of PAX5 deregulation, we examined its chromatin structure and regulation in t(8;21) AML cells, non-t(8;21) myeloid precursor control cells, and pre-B cells. In non-t(8;21) myeloid precursors, PAX5 is poised for transcription, but is repressed by polycomb complexes. In t(8;21) AML, PAX5 is not directly activated by RUNX1/ETO, but expression requires constitutive mitogen-activated protein (MAP) kinase signaling. Using a model of t(8;21) carrying an activating KIT mutation, we demonstrate that deregulated MAP kinase signaling in t(8;21) AML abrogates the association of polycomb complexes to PAX5 and leads to aberrant gene activation. Our findings therefore suggest a novel role of activating tyrosine kinase mutations in lineage-inappropriate gene expression in AML.


Asunto(s)
Linaje de la Célula/genética , Leucemia Mieloide Aguda/genética , Factor de Transcripción PAX5/genética , Proteínas del Grupo Polycomb/fisiología , Línea Celular Tumoral , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Regulación hacia Abajo/fisiología , Regulación Leucémica de la Expresión Génica , Células HL-60 , Células HeLa , Humanos , Leucemia Mieloide Aguda/patología , Sistema de Señalización de MAP Quinasas/fisiología , Modelos Biológicos , Factor de Transcripción PAX5/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Unión Proteica , Transducción de Señal/genética , Transducción de Señal/fisiología , Translocación Genética
20.
Nucleic Acids Res ; 41(21): e201, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24071585

RESUMEN

The expression of eukaryotic genes is regulated by cis-regulatory elements such as promoters and enhancers, which bind sequence-specific DNA-binding proteins. One of the great challenges in the gene regulation field is to characterise these elements. This involves the identification of transcription factor (TF) binding sites within regulatory elements that are occupied in a defined regulatory context. Digestion with DNase and the subsequent analysis of regions protected from cleavage (DNase footprinting) has for many years been used to identify specific binding sites occupied by TFs at individual cis-elements with high resolution. This methodology has recently been adapted for high-throughput sequencing (DNase-seq). In this study, we describe an imbalance in the DNA strand-specific alignment information of DNase-seq data surrounding protein-DNA interactions that allows accurate prediction of occupied TF binding sites. Our study introduces a novel algorithm, Wellington, which considers the imbalance in this strand-specific information to efficiently identify DNA footprints. This algorithm significantly enhances specificity by reducing the proportion of false positives and requires significantly fewer predictions than previously reported methods to recapitulate an equal amount of ChIP-seq data. We also provide an open-source software package, pyDNase, which implements the Wellington algorithm to interface with DNase-seq data and expedite analyses.


Asunto(s)
Algoritmos , Huella de ADN/métodos , Proteínas de Unión al ADN/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Sitios de Unión , Desoxirribonucleasas , Genómica/métodos , Humanos , Programas Informáticos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda