Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Hemasphere ; 8(6): e90, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38903535

RESUMEN

Transcriptional cofactors of the ETO family are recurrent fusion partners in acute leukemia. We characterized the ETO2 regulome by integrating transcriptomic and chromatin binding analyses in human erythroleukemia xenografts and controlled ETO2 depletion models. We demonstrate that beyond its well-established repressive activity, ETO2 directly activates transcription of MYB, among other genes. The ETO2-activated signature is associated with a poorer prognosis in erythroleukemia but also in other acute myeloid and lymphoid leukemia subtypes. Mechanistically, ETO2 colocalizes with EP300 and MYB at enhancers supporting the existence of an ETO2/MYB feedforward transcription activation loop (e.g., on MYB itself). Both small-molecule and PROTAC-mediated inhibition of EP300 acetyltransferases strongly reduced ETO2 protein, chromatin binding, and ETO2-activated transcripts. Taken together, our data show that ETO2 positively enforces a leukemia maintenance program that is mediated in part by the MYB transcription factor and that relies on acetyltransferase cofactors to stabilize ETO2 scaffolding activity.

2.
STAR Protoc ; 5(2): 103016, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38640065

RESUMEN

Precise insertion of fluorescent tags by CRISPR-Cas9-mediated homologous recombination (HR) in mammalian genes is a powerful tool allowing to study gene function and protein gene products. Here, we present a protocol for efficient HR-mediated targeted insertion of fluorescent markers in the genome of hard-to-transfect erythroid cell lines MEL (mouse erythroleukemic) and MEDEP (mouse ES cell-derived erythroid progenitor line). We describe steps for plasmid construction, electroporation, amplification, and verification of genome editing. We then detail procedures for isolating positive clones and validating knockin clones. For complete details on the use and execution of this protocol, please refer to Deleuze et al.1.


Asunto(s)
Sistemas CRISPR-Cas , Células Eritroides , Edición Génica , Técnicas de Sustitución del Gen , Sistemas CRISPR-Cas/genética , Animales , Ratones , Células Eritroides/metabolismo , Células Eritroides/citología , Técnicas de Sustitución del Gen/métodos , Edición Génica/métodos , Transfección/métodos , Línea Celular , Recombinación Homóloga/genética , Electroporación/métodos
3.
iScience ; 26(9): 107641, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37670779

RESUMEN

Targeted genome editing holds great promise in biology. However, efficient genome modification, including gene knock-in (KI), remains an unattained goal in multiple cell types and loci due to poor transfection efficiencies and low target genes expression, impeding the positive selection of recombined cells. Here, we describe a genome editing approach to achieve efficient gene targeting using hard to transfect erythroid cell lines. We demonstrate robust fluorescent protein KI efficiency in low expressed transcription factor (TF) genes (e.g., Myb or Zeb1). We further show the ability to target two independent loci in individual cells, exemplified by MYB-GFP and NuMA-Cherry double KI, allowing multicolor labeling of regulatory factors at physiological endogenous levels. Our KI tagging approach allowed us to perform genome-wide TF analysis at increased signal-to-noise ratios, and highlighted previously unidentified MYB target genes and pathways. Overall, we establish a versatile CRISPR-Cas9-based platform, offering attractive opportunities for the dissection of the erythroid differentiation process.

4.
Nucleic Acids Res ; 51(16): 8413-8433, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37462077

RESUMEN

Genotoxicants have been used for decades as front-line therapies against cancer on the basis of their DNA-damaging actions. However, some of their non-DNA-damaging effects are also instrumental for killing dividing cells. We report here that the anthracycline Daunorubicin (DNR), one of the main drugs used to treat Acute Myeloid Leukemia (AML), induces rapid (3 h) and broad transcriptional changes in AML cells. The regulated genes are particularly enriched in genes controlling cell proliferation and death, as well as inflammation and immunity. These transcriptional changes are preceded by DNR-dependent deSUMOylation of chromatin proteins, in particular at active promoters and enhancers. Surprisingly, inhibition of SUMOylation with ML-792 (SUMO E1 inhibitor), dampens DNR-induced transcriptional reprogramming. Quantitative proteomics shows that the proteins deSUMOylated in response to DNR are mostly transcription factors, transcriptional co-regulators and chromatin organizers. Among them, the CCCTC-binding factor CTCF is highly enriched at SUMO-binding sites found in cis-regulatory regions. This is notably the case at the promoter of the DNR-induced NFKB2 gene. DNR leads to a reconfiguration of chromatin loops engaging CTCF- and SUMO-bound NFKB2 promoter with a distal cis-regulatory region and inhibition of SUMOylation with ML-792 prevents these changes.


Asunto(s)
Daunorrubicina , Leucemia Mieloide Aguda , Humanos , Daunorrubicina/farmacología , Daunorrubicina/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ésteres/uso terapéutico , Cromatina/genética
5.
Epigenetics Chromatin ; 16(1): 23, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37312221

RESUMEN

BACKGROUND: High-throughput sequencing (HTS) offers unprecedented opportunities for the discovery of causative gene variants in multiple human disorders including cancers, and has revolutionized clinical diagnostics. However, despite more than a decade of use of HTS-based assays, extracting relevant functional information from whole-exome sequencing (WES) data remains challenging, especially for non-specialists lacking in-depth bioinformatic skills. RESULTS: To address this limitation, we developed Var∣Decrypt, a web-based tool designed to greatly facilitate WES data browsing and analysis. Var∣Decrypt offers a wide range of gene and variant filtering possibilities, clustering and enrichment tools, providing an efficient way to derive patient-specific functional information and to prioritize gene variants for functional analyses. We applied Var∣Decrypt on WES datasets of 10 acute erythroid leukemia patients, a rare and aggressive form of leukemia, and recovered known disease oncogenes in addition to novel putative drivers. We additionally validated the performance of Var∣Decrypt using an independent dataset of ~ 90 multiple myeloma WES, recapitulating the identified deregulated genes and pathways, showing the general applicability and versatility of Var∣Decrypt for WES analysis. CONCLUSION: Despite years of use of WES in human health for diagnosis and discovery of disease drivers, WES data analysis still remains a complex task requiring advanced bioinformatic skills. In that context, there is a need for user-friendly all-in-one dedicated tools for data analysis, to allow biologists and clinicians to extract relevant biological information from patient datasets. Here, we provide Var∣Decrypt (trial version accessible here: https://vardecrypt.com/app/vardecrypt ), a simple and intuitive Rshiny application created to fill this gap. Source code and detailed user tutorial are available at https://gitlab.com/mohammadsalma/vardecrypt .


Asunto(s)
Biología Computacional , Leucemia , Humanos , Secuenciación del Exoma , Análisis por Conglomerados , Secuenciación de Nucleótidos de Alto Rendimiento
6.
Blood Cells Mol Dis ; 101: 102745, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37121019

RESUMEN

Genome-wide analysis of transcription factors and epigenomic features is instrumental to shed light on DNA-templated regulatory processes such as transcription, cellular differentiation or to monitor cellular responses to environmental cues. Two decades of technological developments have led to a rich set of approaches progressively pushing the limits of epigenetic profiling towards single cells. More recently, disruptive technologies using innovative biochemistry came into play. Assays such as CUT&RUN, CUT&Tag and variations thereof show considerable potential to survey multiple TFs or histone modifications in parallel from a single experiment and in native conditions. These are in the path to become the dominant assays for genome-wide analysis of TFs and chromatin modifications in bulk, single-cell, and spatial genomic applications. The principles together with pros and cons are discussed.


Asunto(s)
Cromatina , Histonas , Humanos , Cromatina/genética , Factores de Transcripción/genética , Genómica , Epigenómica/métodos
7.
Int J Mol Sci ; 23(11)2022 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-35682828

RESUMEN

Studies of the regulatory networks and signals controlling erythropoiesis have brought important insights in several research fields of biology and have been a rich source of discoveries with far-reaching implications beyond erythroid cells biology. The aim of this review is to highlight key recent discoveries and show how studies of erythroid cells bring forward novel concepts and refine current models related to genome and 3D chromatin organization, signaling and disease, with broad interest in life sciences.


Asunto(s)
Cromatina , Factores de Transcripción , Cromatina/genética , Células Eritroides , Eritropoyesis/genética , Factores de Transcripción/genética
8.
J Clin Invest ; 132(14)2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35587378

RESUMEN

Acute megakaryoblastic leukemia of Down syndrome (DS-AMKL) is a model of clonal evolution from a preleukemic transient myeloproliferative disorder requiring both a trisomy 21 (T21) and a GATA1s mutation to a leukemia driven by additional driver mutations. We modeled the megakaryocyte differentiation defect through stepwise gene editing of GATA1s, SMC3+/-, and MPLW515K, providing 20 different T21 or disomy 21 (D21) induced pluripotent stem cell (iPSC) clones. GATA1s profoundly reshaped iPSC-derived hematopoietic architecture with gradual myeloid-to-megakaryocyte shift and megakaryocyte differentiation alteration upon addition of SMC3 and MPL mutations. Transcriptional, chromatin accessibility, and GATA1-binding data showed alteration of essential megakaryocyte differentiation genes, including NFE2 downregulation that was associated with loss of GATA1s binding and functionally involved in megakaryocyte differentiation blockage. T21 enhanced the proliferative phenotype, reproducing the cellular and molecular abnormalities of DS-AMKL. Our study provides an array of human cell-based models revealing individual contributions of different mutations to DS-AMKL differentiation blockage, a major determinant of leukemic progression.


Asunto(s)
Síndrome de Down , Leucemia Megacarioblástica Aguda , Proteínas de Ciclo Celular/genética , Niño , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteínas Cromosómicas no Histona/genética , Síndrome de Down/genética , Factor de Transcripción GATA1/genética , Hematopoyesis , Humanos , Leucemia Megacarioblástica Aguda/complicaciones , Leucemia Megacarioblástica Aguda/genética , Leucemia Megacarioblástica Aguda/metabolismo , Megacariocitos/metabolismo , Mutación , Trisomía
9.
J Clin Invest ; 131(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33914703

RESUMEN

Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Proteínas de Neoplasias/agonistas , Neoplasias Experimentales/tratamiento farmacológico , PPAR gamma/agonistas , Mielofibrosis Primaria/tratamiento farmacológico , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Modelos Animales de Enfermedad , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , PPAR gamma/genética , PPAR gamma/metabolismo , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
10.
Blood ; 137(1): 89-102, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-32818241

RESUMEN

The role of ribosome biogenesis in erythroid development is supported by the recognition of erythroid defects in ribosomopathies in both Diamond-Blackfan anemia and 5q- syndrome. Whether ribosome biogenesis exerts a regulatory function on normal erythroid development is still unknown. In the present study, a detailed characterization of ribosome biogenesis dynamics during human and murine erythropoiesis showed that ribosome biogenesis is abruptly interrupted by the decline in ribosomal DNA transcription and the collapse of ribosomal protein neosynthesis. Its premature arrest by the RNA Pol I inhibitor CX-5461 targeted the proliferation of immature erythroblasts. p53 was activated spontaneously or in response to CX-5461, concomitant to ribosome biogenesis arrest, and drove a transcriptional program in which genes involved in cell cycle-arrested, negative regulation of apoptosis, and DNA damage response were upregulated. RNA Pol I transcriptional stress resulted in nucleolar disruption and activation of the ATR-CHK1-p53 pathway. Our results imply that the timing of ribosome biogenesis extinction and p53 activation is crucial for erythroid development. In ribosomopathies in which ribosome availability is altered by unbalanced production of ribosomal proteins, the threshold downregulation of ribosome biogenesis could be prematurely reached and, together with pathological p53 activation, prevents a normal expansion of erythroid progenitors.


Asunto(s)
Diferenciación Celular/fisiología , Células Eritroides/citología , Eritropoyesis/fisiología , Ribosomas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Madre Hematopoyéticas , Humanos , Ratones , Biogénesis de Organelos
11.
Phys Rev Lett ; 124(23): 238003, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32603179

RESUMEN

Recovery of ground-state degeneracy in two-dimensional square ice is a significant challenge in the field of geometric frustration with far-reaching fundamental implications, such as realization of vertex models and understanding the effect of dimensionality reduction. We combine experiments, theory, and numerical simulations to demonstrate that sheared square colloidal ice partially recovers the ground-state degeneracy for a wide range of field strengths and lattice shear angles. Our method could inspire engineering a novel class of frustrated microstructures and nanostructures based on sheared magnetic lattices in a wide range of soft- and condensed-matter systems.

12.
Blood ; 136(6): 698-714, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32350520

RESUMEN

Acute erythroleukemia (AEL or acute myeloid leukemia [AML]-M6) is a rare but aggressive hematologic malignancy. Previous studies showed that AEL leukemic cells often carry complex karyotypes and mutations in known AML-associated oncogenes. To better define the underlying molecular mechanisms driving the erythroid phenotype, we studied a series of 33 AEL samples representing 3 genetic AEL subgroups including TP53-mutated, epigenetic regulator-mutated (eg, DNMT3A, TET2, or IDH2), and undefined cases with low mutational burden. We established an erythroid vs myeloid transcriptome-based space in which, independently of the molecular subgroup, the majority of the AEL samples exhibited a unique mapping different from both non-M6 AML and myelodysplastic syndrome samples. Notably, >25% of AEL patients, including in the genetically undefined subgroup, showed aberrant expression of key transcriptional regulators, including SKI, ERG, and ETO2. Ectopic expression of these factors in murine erythroid progenitors blocked in vitro erythroid differentiation and led to immortalization associated with decreased chromatin accessibility at GATA1-binding sites and functional interference with GATA1 activity. In vivo models showed development of lethal erythroid, mixed erythroid/myeloid, or other malignancies depending on the cell population in which AEL-associated alterations were expressed. Collectively, our data indicate that AEL is a molecularly heterogeneous disease with an erythroid identity that results in part from the aberrant activity of key erythroid transcription factors in hematopoietic stem or progenitor cells.


Asunto(s)
Leucemia Eritroblástica Aguda/genética , Proteínas de Neoplasias/fisiología , Factores de Transcripción/fisiología , Transcriptoma , Adulto , Animales , Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Eritroblastos/metabolismo , Eritropoyesis/genética , Femenino , Factor de Transcripción GATA1/deficiencia , Factor de Transcripción GATA1/genética , Técnicas de Sustitución del Gen , Heterogeneidad Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos , Persona de Mediana Edad , Mutación , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , RNA-Seq , Quimera por Radiación , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Factores de Transcripción/genética , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/fisiología , Secuenciación del Exoma , Adulto Joven
13.
Curr Opin Hematol ; 27(3): 141-148, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32167946

RESUMEN

PURPOSE OF REVIEW: ß-hemoglobinopathies, such as ß-Thalassemias (ß-Thal) and sickle cell disease (SCD) are among the most common inherited genetic disorders in humans worldwide. These disorders are characterized by a quantitative (ß-Thal) or qualitative (SCD) defects in adult hemoglobin production, leading to anemia, ineffective erythropoiesis and severe secondary complications. Reactivation of the fetal globin genes (γ-globin), making-up fetal hemoglobin (HbF), which are normally silenced in adults, represents a major strategy to ameliorate anemia and disease severity. RECENT FINDINGS: Following the identification of the first 'switching factors' for the reactivation of fetal globin gene expression more than 10 years ago, a multitude of novel leads have recently been uncovered. SUMMARY: Recent findings provided invaluable functional insights into the genetic and molecular networks controlling globin genes expression, revealing that complex repression systems evolved in erythroid cells to maintain HbF silencing in adults. This review summarizes these unique and exciting discoveries of the regulatory factors controlling the globin switch. New insights and novel leads for therapeutic strategies based on the pharmacological induction of HbF are discussed. This represents a major breakthrough for rational drug design in the treatment of ß-Thal and SCD.


Asunto(s)
Anemia de Células Falciformes , Diseño de Fármacos , Hemoglobina Fetal , Regulación de la Expresión Génica , Talasemia beta , gamma-Globinas , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Anemia de Células Falciformes/patología , Células Eritroides/metabolismo , Células Eritroides/patología , Hemoglobina Fetal/biosíntesis , Hemoglobina Fetal/genética , Humanos , Talasemia beta/tratamiento farmacológico , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/patología , gamma-Globinas/biosíntesis , gamma-Globinas/genética
14.
Nat Genet ; 49(7): 1073-1081, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28581502

RESUMEN

Gene expression in mammals is precisely regulated by the combination of promoters and gene-distal regulatory regions, known as enhancers. Several studies have suggested that some promoters might have enhancer functions. However, the extent of this type of promoters and whether they actually function to regulate the expression of distal genes have remained elusive. Here, by exploiting a high-throughput enhancer reporter assay, we unravel a set of mammalian promoters displaying enhancer activity. These promoters have distinct genomic and epigenomic features and frequently interact with other gene promoters. Extensive CRISPR-Cas9 genomic manipulation demonstrated the involvement of these promoters in the cis regulation of expression of distal genes in their natural loci. Our results have important implications for the understanding of complex gene regulation in normal development and disease.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Células 3T3 , Animales , Sistemas CRISPR-Cas , Epigenómica , Ontología de Genes , Células HeLa , Humanos , Interferón-alfa/farmacología , Células K562 , Mamíferos/genética , Ratones
15.
Cancer Cell ; 31(3): 452-465, 2017 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-28292442

RESUMEN

Chimeric transcription factors are a hallmark of human leukemia, but the molecular mechanisms by which they block differentiation and promote aberrant self-renewal remain unclear. Here, we demonstrate that the ETO2-GLIS2 fusion oncoprotein, which is found in aggressive acute megakaryoblastic leukemia, confers megakaryocytic identity via the GLIS2 moiety while both ETO2 and GLIS2 domains are required to drive increased self-renewal properties. ETO2-GLIS2 directly binds DNA to control transcription of associated genes by upregulation of expression and interaction with the ETS-related ERG protein at enhancer elements. Importantly, specific interference with ETO2-GLIS2 oligomerization reverses the transcriptional activation at enhancers and promotes megakaryocytic differentiation, providing a relevant interface to target in this poor-prognosis pediatric leukemia.


Asunto(s)
Leucemia Megacarioblástica Aguda/patología , Proteínas de Fusión Oncogénica/fisiología , Activación Transcripcional , Animales , Diferenciación Celular , Niño , Elementos de Facilitación Genéticos , Factor de Transcripción GATA1/genética , Humanos , Ratones , Proteínas de Fusión Oncogénica/química , Regulador Transcripcional ERG/fisiología
16.
Oncotarget ; 8(3): 5111-5122, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-27974684

RESUMEN

The tripartite motif (TRIM) family of proteins plays important roles in innate immunity and antimicrobial infection. None of these proteins has been shown to directly regulate transcription of genes in monocyte/macrophage except TRIM33 that we have recently shown to be a macrophage specific transcriptional inhibitor of Ifnb1. Using ChIP-seq analyses, we now report that TRIM33 is bound to two fold more genes in immature than in mature myeloid cell lines. When located near the same genes, TRIM33 is bound to different sequences in the two cell lines suggesting a role of TRIM33 in both immature and mature myeloid cells. Accordingly, expression of TRIM33 in immature myeloid cells is necessary for efficient production of small peritoneal macrophages, monocytes and bone marrow derived macrophage (BMDM) and TRIM33 targets a subset of genes involved in the inflammatory response only in mature myeloid cells. Functionally, this targeting is associated with impaired repression of pathways regulating the late phases of lipopolysaccharide (LPS) activation of BMDM and a high sensitivity to LPS in vivo when the trim33 gene is inactivated in mature myeloid cells. These findings pinpoint TRIM33 as an important transcriptional actor of monocyte/macrophage mediated inflammation.


Asunto(s)
Cromatina/metabolismo , Activación de Macrófagos , Macrófagos/citología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Cromatina/genética , Inmunoprecipitación de Cromatina , ADN/metabolismo , Redes Reguladoras de Genes , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Células Mieloides/citología , Células Mieloides/inmunología , Células RAW 264.7 , Análisis de Secuencia de ADN , Factores de Transcripción/genética
17.
Methods Mol Biol ; 1507: 199-220, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27832542

RESUMEN

The development and widespread implementation of chromosome conformation capture (3C) technology has allowed unprecedented new insight into how chromosomes are folded in three-dimensional (3D) space. 3C and its derivatives have contributed tremendously to the now widely accepted view that genome topology plays an important role in many major cellular processes, at a chromosome-wide scale, but certainly also at the level of individual genetic loci. A particularly popular application of 3C technology is to study transcriptional regulation, allowing researchers to draw maps of gene regulatory connections beyond the linear genome through addition of the third dimension. In this chapter, we provide a highly detailed protocol describing 3C coupled to high-throughput sequencing (referred to as 3C-Seq or more commonly 4C-Seq), allowing the unbiased interrogation of genome-wide chromatin interactions with specific genomic regions of interest. Interactions between spatially clustered DNA fragments are revealed by crosslinking the cells with formaldehyde, digesting the genome with a restriction endonuclease and performing a proximity ligation step to link interacting genomic fragments. Next, interactions with a selected DNA fragment are extracted from the 3C library through a second round of digestion and ligation followed by an inverse PCR. The generated products are immediately compatible with high-throughput sequencing, and amplicons from different PCR reactions can easily be multiplexed to dramatically increase throughput. Finally, we provide suggestions for data analysis and visualization.


Asunto(s)
Cromosomas de los Mamíferos/ultraestructura , ADN/aislamiento & purificación , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ADN , Animales , Células Cultivadas , Cromosomas de los Mamíferos/genética , ADN/genética , Genoma , Humanos , Reacción en Cadena de la Polimerasa
18.
FEBS Lett ; 590(22): 4084-4104, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27645909

RESUMEN

Cellular differentiation requires precisely regulated tissue-specific and developmental stage-specific gene expression patterns. Numerous studies have highlighted the predictive power of enhancers on lineage-specific gene expression programs and have started to unravel their mechanisms of action. We review here the dynamics of the enhancer landscape during hematopoietic differentiation and how enhancers function in the context of the 3D organization of the genome. We further discuss the involvement of aberrant enhancer activity in human diseases and emerging strategies aiming at controlling enhancer activity and chromosome topology for therapeutic purposes.


Asunto(s)
Diferenciación Celular/genética , Elementos de Facilitación Genéticos , Enfermedades Genéticas Congénitas/genética , Células Madre Hematopoyéticas , Linaje de la Célula/genética , Cromosomas/genética , Regulación del Desarrollo de la Expresión Génica , Enfermedades Genéticas Congénitas/patología , Genoma Humano , Humanos , Especificidad de Órganos/genética
19.
Nat Commun ; 6: 8893, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26593974

RESUMEN

How transcription factors (TFs) cooperate within large protein complexes to allow rapid modulation of gene expression during development is still largely unknown. Here we show that the key haematopoietic LIM-domain-binding protein-1 (LDB1) TF complex contains several activator and repressor components that together maintain an erythroid-specific gene expression programme primed for rapid activation until differentiation is induced. A combination of proteomics, functional genomics and in vivo studies presented here identifies known and novel co-repressors, most notably the ETO2 and IRF2BP2 proteins, involved in maintaining this primed state. The ETO2-IRF2BP2 axis, interacting with the NCOR1/SMRT co-repressor complex, suppresses the expression of the vast majority of archetypical erythroid genes and pathways until its decommissioning at the onset of terminal erythroid differentiation. Our experiments demonstrate that multimeric regulatory complexes feature a dynamic interplay between activating and repressing components that determines lineage-specific gene expression and cellular differentiation.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Eritroides/citología , Eritropoyesis , Humanos , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética
20.
Nat Commun ; 6: 8900, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26592194

RESUMEN

Despite its importance during viral or bacterial infections, transcriptional regulation of the interferon-ß gene (Ifnb1) in activated macrophages is only partially understood. Here we report that TRIM33 deficiency results in high, sustained expression of Ifnb1 at late stages of toll-like receptor-mediated activation in macrophages but not in fibroblasts. In macrophages, TRIM33 is recruited by PU.1 to a conserved region, the Ifnb1 Control Element (ICE), located 15 kb upstream of the Ifnb1 transcription start site. ICE constitutively interacts with Ifnb1 through a TRIM33-independent chromatin loop. At late phases of lipopolysaccharide activation of macrophages, TRIM33 is bound to ICE, regulates Ifnb1 enhanceosome loading, controls Ifnb1 chromatin structure and represses Ifnb1 gene transcription by preventing recruitment of CBP/p300. These results characterize a previously unknown mechanism of macrophage-specific regulation of Ifnb1 transcription whereby TRIM33 is critical for Ifnb1 gene transcription shutdown.


Asunto(s)
Regulación de la Expresión Génica , Interferón beta/genética , Macrófagos/citología , Macrófagos/metabolismo , Factores de Transcripción/metabolismo , Animales , Femenino , Interferón beta/metabolismo , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA