Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Mol Cell ; 83(5): 715-730.e6, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36868189

RESUMEN

Transcriptional enhancers have been extensively characterized, but cis-regulatory elements involved in acute gene repression have received less attention. Transcription factor GATA1 promotes erythroid differentiation by activating and repressing distinct gene sets. Here, we study the mechanism by which GATA1 silences the proliferative gene Kit during murine erythroid cell maturation and define stages from initial loss of activation to heterochromatinization. We find that GATA1 inactivates a potent upstream enhancer but concomitantly creates a discrete intronic regulatory region marked by H3K27ac, short noncoding RNAs, and de novo chromatin looping. This enhancer-like element forms transiently and serves to delay Kit silencing. The element is ultimately erased via the FOG1/NuRD deacetylase complex, as revealed by the study of a disease-associated GATA1 variant. Hence, regulatory sites can be self-limiting by dynamic co-factor usage. Genome-wide analyses across cell types and species uncover transiently active elements at numerous genes during repression, suggesting that modulation of silencing kinetics is widespread.


Asunto(s)
Estudio de Asociación del Genoma Completo , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Ratones , Intrones , Diferenciación Celular , Silenciador del Gen , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2
2.
Mol Cell ; 81(2): 239-254.e8, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33301730

RESUMEN

Metazoan transcription factors typically regulate large numbers of genes. Here we identify via a CRISPR-Cas9 genetic screen ZNF410, a pentadactyl DNA-binding protein that in human erythroid cells directly activates only a single gene, the NuRD component CHD4. Specificity is conveyed by two highly evolutionarily conserved clusters of ZNF410 binding sites near the CHD4 gene with no counterparts elsewhere in the genome. Loss of ZNF410 in adult-type human erythroid cell culture systems and xenotransplantation settings diminishes CHD4 levels and derepresses the fetal hemoglobin genes. While previously known to be silenced by CHD4, the fetal globin genes are exposed here as among the most sensitive to reduced CHD4 levels.. In vitro DNA binding assays and crystallographic studies reveal the ZNF410-DNA binding mode. ZNF410 is a remarkably selective transcriptional activator in erythroid cells, and its perturbation might offer new opportunities for treatment of hemoglobinopathies.


Asunto(s)
ADN/genética , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Factores de Transcripción/genética , Animales , Sitios de Unión , Células COS , Sistemas CRISPR-Cas , Chlorocebus aethiops , ADN/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/trasplante , Sangre Fetal/citología , Sangre Fetal/metabolismo , Hemoglobina Fetal/metabolismo , Feto , Edición Génica , Células HEK293 , Xenoinjertos , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones , Modelos Moleculares , Células Madre Embrionarias de Ratones/citología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Activación Transcripcional
3.
Genome Res ; 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38951027

RESUMEN

Knowledge of locations and activities of cis-regulatory elements (CREs) is needed to decipher basic mechanisms of gene regulation and to understand the impact of genetic variants on complex traits. Previous studies identified candidate CREs (cCREs) using epigenetic features in one species, making comparisons difficult between species. In contrast, we conducted an interspecies study defining epigenetic states and identifying cCREs in blood cell types to generate regulatory maps that are comparable between species, using integrative modeling of eight epigenetic features jointly in human and mouse in our Validated Systematic Integration (VISION) Project. The resulting catalogs of cCREs are useful resources for further studies of gene regulation in blood cells, indicated by high overlap with known functional elements and strong enrichment for human genetic variants associated with blood cell phenotypes. The contribution of each epigenetic state in cCREs to gene regulation, inferred from a multivariate regression, was used to estimate epigenetic state regulatory potential (esRP) scores for each cCRE in each cell type, which were used to categorize dynamic changes in cCREs. Groups of cCREs displaying similar patterns of regulatory activity in human and mouse cell types, obtained by joint clustering on esRP scores, harbor distinctive transcription factor binding motifs that are similar between species. An interspecies comparison of cCREs revealed both conserved and species-specific patterns of epigenetic evolution. Finally, we show that comparisons of the epigenetic landscape between species can reveal elements with similar roles in regulation, even in the absence of genomic sequence alignment.

4.
Mol Cell ; 73(3): 519-532.e4, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30554946

RESUMEN

Transcriptional regulation occurs via changes to rates of different biochemical steps of transcription, but it remains unclear which rates are subject to change upon biological perturbation. Biochemical studies have suggested that stimuli predominantly affect the rates of RNA polymerase II (Pol II) recruitment and polymerase release from promoter-proximal pausing. Single-cell studies revealed that transcription occurs in discontinuous bursts, suggesting that features of such bursts like frequency and intensity could also be regulated. We combined Pol II chromatin immunoprecipitation sequencing (ChIP-seq) and single-cell transcriptional measurements to show that an independently regulated burst initiation step is required before polymerase recruitment can occur. Using a number of global and targeted transcriptional regulatory perturbations, we showed that biological perturbations regulated both burst initiation and polymerase pause release rates but seemed not to regulate polymerase recruitment rate. Our results suggest that transcriptional regulation primarily acts by changing the rates of burst initiation and polymerase pause release.


Asunto(s)
Células Madre Embrionarias de Ratones/enzimología , ARN Polimerasa II/metabolismo , ARN/biosíntesis , Sitio de Iniciación de la Transcripción , Iniciación de la Transcripción Genética , Activación Transcripcional , Animales , Sitios de Unión , Línea Celular , Simulación por Computador , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Modelos Genéticos , Unión Proteica , ARN/genética , ARN Polimerasa II/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Factores de Tiempo
5.
Blood ; 143(19): 1980-1991, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38364109

RESUMEN

ABSTRACT: The switch from fetal hemoglobin (γ-globin, HBG) to adult hemoglobin (ß-globin, HBB) gene transcription in erythroid cells serves as a paradigm for a complex and clinically relevant developmental gene regulatory program. We previously identified HIC2 as a regulator of the switch by inhibiting the transcription of BCL11A, a key repressor of HBG production. HIC2 is highly expressed in fetal cells, but the mechanism of its regulation is unclear. Here we report that HIC2 developmental expression is controlled by microRNAs (miRNAs), as loss of global miRNA biogenesis through DICER1 depletion leads to upregulation of HIC2 and HBG messenger RNA. We identified the adult-expressed let-7 miRNA family as a direct posttranscriptional regulator of HIC2. Ectopic expression of let-7 in fetal cells lowered HIC2 levels, whereas inhibition of let-7 in adult erythroblasts increased HIC2 production, culminating in decommissioning of a BCL11A erythroid enhancer and reduced BCL11A transcription. HIC2 depletion in let-7-inhibited cells restored BCL11A-mediated repression of HBG. Together, these data establish that fetal hemoglobin silencing in adult erythroid cells is under the control of a miRNA-mediated inhibitory pathway (let-7 ⊣ HIC2 ⊣ BCL11A ⊣ HBG).


Asunto(s)
Hemoglobina Fetal , Factores de Transcripción de Tipo Kruppel , MicroARNs , Proteínas Represoras , Humanos , Globinas beta/genética , Globinas beta/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Eritroblastos/metabolismo , Eritroblastos/citología , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , gamma-Globinas/genética , gamma-Globinas/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Transcripción Genética
6.
Nature ; 583(7818): 699-710, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728249

RESUMEN

The human and mouse genomes contain instructions that specify RNAs and proteins and govern the timing, magnitude, and cellular context of their production. To better delineate these elements, phase III of the Encyclopedia of DNA Elements (ENCODE) Project has expanded analysis of the cell and tissue repertoires of RNA transcription, chromatin structure and modification, DNA methylation, chromatin looping, and occupancy by transcription factors and RNA-binding proteins. Here we summarize these efforts, which have produced 5,992 new experimental datasets, including systematic determinations across mouse fetal development. All data are available through the ENCODE data portal (https://www.encodeproject.org), including phase II ENCODE1 and Roadmap Epigenomics2 data. We have developed a registry of 926,535 human and 339,815 mouse candidate cis-regulatory elements, covering 7.9 and 3.4% of their respective genomes, by integrating selected datatypes associated with gene regulation, and constructed a web-based server (SCREEN; http://screen.encodeproject.org) to provide flexible, user-defined access to this resource. Collectively, the ENCODE data and registry provide an expansive resource for the scientific community to build a better understanding of the organization and function of the human and mouse genomes.


Asunto(s)
ADN/genética , Bases de Datos Genéticas , Genoma/genética , Genómica , Anotación de Secuencia Molecular , Sistema de Registros , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Cromatina/genética , Cromatina/metabolismo , ADN/química , Huella de ADN , Metilación de ADN/genética , Momento de Replicación del ADN , Desoxirribonucleasa I/metabolismo , Genoma Humano , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Transcripción Genética/genética , Transposasas/metabolismo
7.
Blood ; 141(22): 2756-2770, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36893455

RESUMEN

The switch from fetal hemoglobin (HbF) to adult hemoglobin (HbA) is a paradigm for developmental gene expression control with relevance to sickle cell disease and ß-thalassemia. Polycomb repressive complex (PRC) proteins regulate this switch, and an inhibitor of PRC2 has entered a clinical trial for HbF activation. Yet, how PRC complexes function in this process, their target genes, and relevant subunit composition are unknown. Here, we identified the PRC1 subunit BMI1 as a novel HbF repressor. We uncovered the RNA binding proteins LIN28B, IGF2BP1, and IGF2BP3 genes as direct BMI1 targets, and demonstrate that they account for the entirety of BMI1's effect on HbF regulation. BMI1 functions as part of the canonical PRC1 (cPRC1) subcomplex as revealed by the physical and functional dissection of BMI1 protein partners. Lastly, we demonstrate that BMI1/cPRC1 acts in concert with PRC2 to repress HbF through the same target genes. Our study illuminates how PRC silences HbF, highlighting an epigenetic mechanism involved in hemoglobin switching.


Asunto(s)
Hemoglobina Fetal , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Hemoglobina Fetal/genética , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo
8.
Nature ; 576(7785): 158-162, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31776509

RESUMEN

Features of higher-order chromatin organization-such as A/B compartments, topologically associating domains and chromatin loops-are temporarily disrupted during mitosis1,2. Because these structures are thought to influence gene regulation, it is important to understand how they are re-established after mitosis. Here we examine the dynamics of chromosome reorganization by Hi-C after mitosis in highly purified, synchronous mouse erythroid cell populations. We observed rapid establishment of A/B compartments, followed by their gradual intensification and expansion. Contact domains form from the 'bottom up'-smaller subTADs are formed initially, followed by convergence into multi-domain TAD structures. CTCF is partially retained on mitotic chromosomes and immediately resumes full binding in ana/telophase. By contrast, cohesin is completely evicted from mitotic chromosomes and regains focal binding at a slower rate. The formation of CTCF/cohesin co-anchored structural loops follows the kinetics of cohesin positioning. Stripe-shaped contact patterns-anchored by CTCF-grow in length, which is consistent with a loop-extrusion process after mitosis. Interactions between cis-regulatory elements can form rapidly, with rates exceeding those of CTCF/cohesin-anchored contacts. Notably, we identified a group of rapidly emerging transient contacts between cis-regulatory elements in ana/telophase that are dissolved upon G1 entry, co-incident with the establishment of inner boundaries or nearby interfering chromatin loops. We also describe the relationship between transcription reactivation and architectural features. Our findings indicate that distinct but mutually influential forces drive post-mitotic chromatin reconfiguration.


Asunto(s)
Cromatina , Fase G1 , Mitosis , Animales , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Ratones , Cohesinas
9.
Mol Cell ; 66(1): 102-116.e7, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388437

RESUMEN

Bromodomain and extraterminal motif (BET) proteins are pharmacologic targets for the treatment of diverse diseases, yet the roles of individual BET family members remain unclear. We find that BRD2, but not BRD4, co-localizes with the architectural/insulator protein CCCTC-binding factor (CTCF) genome-wide. CTCF recruits BRD2 to co-bound sites whereas BRD2 is dispensable for CTCF occupancy. Disruption of a CTCF/BRD2-occupied element positioned between two unrelated genes enables regulatory influence to spread from one gene to another, suggesting that CTCF and BRD2 form a transcriptional boundary. Accordingly, single-molecule mRNA fluorescence in situ hybridization (FISH) reveals that, upon site-specific CTCF disruption or BRD2 depletion, expression of the two genes becomes increasingly correlated. HiC shows that BRD2 depletion weakens boundaries co-occupied by CTCF and BRD2, but not those that lack BRD2. These findings indicate that BRD2 supports boundary activity, and they raise the possibility that pharmacologic BET inhibitors can influence gene expression in part by perturbing domain boundary function.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Factor de Unión a CCCTC , Sistemas CRISPR-Cas , Línea Celular , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Edición Génica/métodos , Hibridación Fluorescente in Situ , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Imagen Individual de Molécula/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
10.
Genes Dev ; 31(16): 1704-1713, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28916711

RESUMEN

Chromatin structure is tightly intertwined with transcription regulation. Here we compared the chromosomal architectures of fetal and adult human erythroblasts and found that, globally, chromatin structures and compartments A/B are highly similar at both developmental stages. At a finer scale, we detected distinct folding patterns at the developmentally controlled ß-globin locus. Specifically, new fetal stage-specific contacts were uncovered between a region separating the fetal (γ) and adult (δ and ß) globin genes (encompassing the HBBP1 and BGLT3 noncoding genes) and two distal chromosomal sites (HS5 and 3'HS1) that flank the locus. In contrast, in adult cells, the HBBP1-BGLT3 region contacts the embryonic ε-globin gene, physically separating the fetal globin genes from the enhancer (locus control region [LCR]). Deletion of the HBBP1 region in adult cells alters contact landscapes in ways more closely resembling those of fetal cells, including increased LCR-γ-globin contacts. These changes are accompanied by strong increases in γ-globin transcription. Notably, the effects of HBBP1 removal on chromatin architecture and gene expression closely mimic those of deleting the fetal globin repressor BCL11A, implicating BCL11A in the function of the HBBP1 region. Our results uncover a new critical regulatory region as a potential target for therapeutic genome editing for hemoglobinopathies and highlight the power of chromosome conformation analysis in discovering new cis control elements.


Asunto(s)
Cromatina/química , Eritroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Elementos Reguladores de la Transcripción , Globinas beta/genética , Adulto , Proteínas Portadoras/genética , Feto , Silenciador del Gen , Humanos , Región de Control de Posición , Proteínas Nucleares/genética , Seudogenes , Proteínas Represoras , Transcriptoma , gamma-Globinas/genética
11.
Genes Dev ; 30(12): 1423-39, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27340175

RESUMEN

During mitosis, RNA polymerase II (Pol II) and many transcription factors dissociate from chromatin, and transcription ceases globally. Transcription is known to restart in bulk by telophase, but whether de novo transcription at the mitosis-G1 transition is in any way distinct from later in interphase remains unknown. We tracked Pol II occupancy genome-wide in mammalian cells progressing from mitosis through late G1. Unexpectedly, during the earliest rounds of transcription at the mitosis-G1 transition, ∼50% of active genes and distal enhancers exhibit a spike in transcription, exceeding levels observed later in G1 phase. Enhancer-promoter chromatin contacts are depleted during mitosis and restored rapidly upon G1 entry but do not spike. Of the chromatin-associated features examined, histone H3 Lys27 acetylation levels at individual loci in mitosis best predict the mitosis-G1 transcriptional spike. Single-molecule RNA imaging supports that the mitosis-G1 transcriptional spike can constitute the maximum transcriptional activity per DNA copy throughout the cell division cycle. The transcriptional spike occurs heterogeneously and propagates to cell-to-cell differences in mature mRNA expression. Our results raise the possibility that passage through the mitosis-G1 transition might predispose cells to diverge in gene expression states.


Asunto(s)
Ciclo Celular/genética , Fase G1/genética , Genoma/genética , Mitosis/genética , Activación Transcripcional/genética , Animales , Línea Celular , Células Cultivadas , Cromatina/metabolismo , ADN Intergénico/genética , Elementos de Facilitación Genéticos/genética , Eritroblastos/citología , Ratones , Regiones Promotoras Genéticas/genética , Regulación hacia Arriba
12.
BMC Bioinformatics ; 24(1): 102, 2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36941541

RESUMEN

BACKGROUND: Epigenetic modification of chromatin plays a pivotal role in regulating gene expression during cell differentiation. The scale and complexity of epigenetic data pose significant challenges for biologists to identify the regulatory events controlling cell differentiation. RESULTS: To reduce the complexity, we developed a package, called Snapshot, for clustering and visualizing candidate cis-regulatory elements (cCREs) based on their epigenetic signals during cell differentiation. This package first introduces a binarized indexing strategy for clustering the cCREs. It then provides a series of easily interpretable figures for visualizing the signal and epigenetic state patterns of the cCREs clusters during the cell differentiation. It can also use different hierarchies of cell types to highlight the epigenetic history specific to any particular cell lineage. We demonstrate the utility of Snapshot using data from a consortium project for ValIdated Systematic IntegratiON (VISION) of epigenomic data in hematopoiesis. CONCLUSION: The package Snapshot can identify all distinct clusters of genomic locations with unique epigenetic signal patterns during cell differentiation. It outperforms other methods in terms of interpreting and reproducing the identified cCREs clusters. The package of Snapshot is available at GitHub: https://github.com/guanjue/Snapshot .


Asunto(s)
Cromatina , Epigenómica , Epigenómica/métodos , Diferenciación Celular/genética , Epigénesis Genética , Análisis por Conglomerados
13.
Genome Res ; 30(3): 472-484, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32132109

RESUMEN

Thousands of epigenomic data sets have been generated in the past decade, but it is difficult for researchers to effectively use all the data relevant to their projects. Systematic integrative analysis can help meet this need, and the VISION project was established for validated systematic integration of epigenomic data in hematopoiesis. Here, we systematically integrated extensive data recording epigenetic features and transcriptomes from many sources, including individual laboratories and consortia, to produce a comprehensive view of the regulatory landscape of differentiating hematopoietic cell types in mouse. By using IDEAS as our integrative and discriminative epigenome annotation system, we identified and assigned epigenetic states simultaneously along chromosomes and across cell types, precisely and comprehensively. Combining nuclease accessibility and epigenetic states produced a set of more than 200,000 candidate cis-regulatory elements (cCREs) that efficiently capture enhancers and promoters. The transitions in epigenetic states of these cCREs across cell types provided insights into mechanisms of regulation, including decreases in numbers of active cCREs during differentiation of most lineages, transitions from poised to active or inactive states, and shifts in nuclease accessibility of CTCF-bound elements. Regression modeling of epigenetic states at cCREs and gene expression produced a versatile resource to improve selection of cCREs potentially regulating target genes. These resources are available from our VISION website to aid research in genomics and hematopoiesis.


Asunto(s)
Epigénesis Genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Ratones , Elementos Reguladores de la Transcripción , Transcriptoma
14.
Blood ; 138(8): 662-673, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-33786584

RESUMEN

Chronic natural killer large granular lymphocyte (NK-LGL) leukemia, also referred to as chronic lymphoproliferative disorder of NK cells, is a rare disorder defined by prolonged expansion of clonal NK cells. Similar prevalence of STAT3 mutations in chronic T-LGL and NK-LGL leukemia is suggestive of common pathogenesis. We undertook whole-genome sequencing to identify mutations unique to NK-LGL leukemia. The results were analyzed to develop a resequencing panel that was applied to 58 patients. Phosphatidylinositol 3-kinase pathway gene mutations (PIK3CD/PIK3AP1) and TNFAIP3 mutations were seen in 5% and 10% of patients, respectively. TET2 was exceptional in that mutations were present in 16 (28%) of 58 patient samples, with evidence that TET2 mutations can be dominant and exclusive to the NK compartment. Reduced-representation bisulfite sequencing revealed that methylation patterns were significantly altered in TET2 mutant samples. The promoter of TET2 and that of PTPRD, a negative regulator of STAT3, were found to be methylated in additional cohort samples, largely confined to the TET2 mutant group. Mutations in STAT3 were observed in 19 (33%) of 58 patient samples, 7 of which had concurrent TET2 mutations. Thrombocytopenia and resistance to immunosuppressive agents were uniquely observed in those patients with only TET2 mutation (Games-Howell post hoc test, P = .0074; Fisher's exact test, P = .00466). Patients with STAT3 mutation, inclusive of those with TET2 comutation, had lower hematocrit, hemoglobin, and absolute neutrophil count compared with STAT3 wild-type patients (Welch's t test, P ≤ .015). We present the discovery of TET2 mutations in chronic NK-LGL leukemia and evidence that it identifies a unique molecular subtype.


Asunto(s)
Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Leucemia Linfocítica Granular Grande/genética , Mutación , Proteínas de Neoplasias/genética , Sistema de Registros , Enfermedad Crónica , Proteínas de Unión al ADN/sangre , Dioxigenasas/sangre , Femenino , Humanos , Leucemia Linfocítica Granular Grande/sangre , Masculino , Proteínas de Neoplasias/sangre
16.
Blood ; 135(24): 2121-2132, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32299090

RESUMEN

Reactivation of fetal hemoglobin remains a critical goal in the treatment of patients with sickle cell disease and ß-thalassemia. Previously, we discovered that silencing of the fetal γ-globin gene requires the erythroid-specific eIF2α kinase heme-regulated inhibitor (HRI), suggesting that HRI might present a pharmacologic target for raising fetal hemoglobin levels. Here, via a CRISPR-Cas9-guided loss-of-function screen in human erythroblasts, we identify transcription factor ATF4, a known HRI-regulated protein, as a novel γ-globin regulator. ATF4 directly stimulates transcription of BCL11A, a repressor of γ-globin transcription, by binding to its enhancer and fostering enhancer-promoter contacts. Notably, HRI-deficient mice display normal Bcl11a levels, suggesting species-selective regulation, which we explain here by demonstrating that the analogous ATF4 motif at the murine Bcl11a enhancer is largely dispensable. Our studies uncover a linear signaling pathway from HRI to ATF4 to BCL11A to γ-globin and illustrate potential limits of murine models of globin gene regulation.


Asunto(s)
Factor de Transcripción Activador 4/genética , Hemoglobina Fetal/genética , Proteínas Represoras/genética , eIF-2 Quinasa/genética , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Animales , Sistemas CRISPR-Cas , Células Cultivadas , Elementos de Facilitación Genéticos , Eritroblastos/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Especificidad de la Especie , Talasemia beta/sangre , Talasemia beta/genética , Talasemia beta/terapia , gamma-Globinas/biosíntesis , gamma-Globinas/genética
17.
Blood ; 135(22): 1957-1968, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32268371

RESUMEN

Reversing the developmental switch from fetal hemoglobin (HbF, α2γ2) to adult hemoglobin (HbA, α2ß2) is an important therapeutic approach in sickle cell disease (SCD) and ß-thalassemia. In healthy individuals, SCD patients, and patients treated with pharmacologic HbF inducers, HbF is present only in a subset of red blood cells known as F cells. Despite more than 50 years of observations, the cause for this heterocellular HbF expression pattern, even among genetically identical cells, remains unknown. Adult F cells might represent a reversion of a given cell to a fetal-like epigenetic and transcriptional state. Alternatively, isolated transcriptional or posttranscriptional events at the γ-globin genes might underlie heterocellularity. Here, we set out to understand the heterogeneity of HbF activation by developing techniques to purify and profile differentiation stage-matched late erythroblast F cells and non-F cells (A cells) from the human HUDEP2 erythroid cell line and primary human erythroid cultures. Transcriptional and proteomic profiling of these cells demonstrated very few differences between F and A cells at the RNA level either under baseline conditions or after treatment with HbF inducers hydroxyurea or pomalidomide. Surprisingly, we did not find differences in expression of any known HbF regulators, including BCL11A or LRF, that would account for HbF activation. Our analysis shows that F erythroblasts are not significantly different from non-HbF-expressing cells and that the primary differences likely occur at the transcriptional level at the ß-globin locus.


Asunto(s)
Eritroblastos/metabolismo , Hemoglobina Fetal/biosíntesis , Hemoglobina A/metabolismo , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/genética , Línea Celular , Separación Celular/métodos , Células Cultivadas , Eritroblastos/clasificación , Eritroblastos/efectos de los fármacos , Células Eritroides/clasificación , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Perfilación de la Expresión Génica , Hemoglobina A/genética , Humanos , Hidroxiurea/farmacología , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Talidomida/análogos & derivados , Talidomida/farmacología
18.
Nucleic Acids Res ; 48(8): e43, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32086521

RESUMEN

Quantitative comparison of epigenomic data across multiple cell types or experimental conditions is a promising way to understand the biological functions of epigenetic modifications. However, differences in sequencing depth and signal-to-noise ratios in the data from different experiments can hinder our ability to identify real biological variation from raw epigenomic data. Proper normalization is required prior to data analysis to gain meaningful insights. Most existing methods for data normalization standardize signals by rescaling either background regions or peak regions, assuming that the same scale factor is applicable to both background and peak regions. While such methods adjust for differences in sequencing depths, they do not address differences in the signal-to-noise ratios across different experiments. We developed a new data normalization method, called S3norm, that normalizes the sequencing depths and signal-to-noise ratios across different data sets simultaneously by a monotonic nonlinear transformation. We show empirically that the epigenomic data normalized by our method, compared to existing methods, can better capture real biological variation, such as impact on gene expression regulation.


Asunto(s)
Epigenómica/métodos , Análisis de Secuencia de ADN/métodos , Expresión Génica , Código de Histonas , RNA-Seq , Programas Informáticos
19.
IUBMB Life ; 72(1): 27-38, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769130

RESUMEN

Members of the GATA family of transcription factors play key roles in the differentiation of specific cell lineages by regulating the expression of target genes. Three GATA factors play distinct roles in hematopoietic differentiation. In order to better understand how these GATA factors function to regulate genes throughout the genome, we are studying the epigenomic and transcriptional landscapes of hematopoietic cells in a model-driven, integrative fashion. We have formed the collaborative multi-lab VISION project to conduct ValIdated Systematic IntegratiON of epigenomic data in mouse and human hematopoiesis. The epigenomic data included nuclease accessibility in chromatin, CTCF occupancy, and histone H3 modifications for 20 cell types covering hematopoietic stem cells, multilineage progenitor cells, and mature cells across the blood cell lineages of mouse. The analysis used the Integrative and Discriminative Epigenome Annotation System (IDEAS), which learns all common combinations of features (epigenetic states) simultaneously in two dimensions-along chromosomes and across cell types. The result is a segmentation that effectively paints the regulatory landscape in readily interpretable views, revealing constitutively active or silent loci as well as the loci specifically induced or repressed in each stage and lineage. Nuclease accessible DNA segments in active chromatin states were designated candidate cis-regulatory elements in each cell type, providing one of the most comprehensive registries of candidate hematopoietic regulatory elements to date. Applications of VISION resources are illustrated for the regulation of genes encoding GATA1, GATA2, GATA3, and Ikaros. VISION resources are freely available from our website http://usevision.org.


Asunto(s)
Cromatina/metabolismo , Epigenoma , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Diferenciación Celular , Cromatina/genética , Factores de Transcripción GATA/genética , Humanos
20.
Nature ; 515(7527): 371-375, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25409826

RESUMEN

To broaden our understanding of the evolution of gene regulation mechanisms, we generated occupancy profiles for 34 orthologous transcription factors (TFs) in human-mouse erythroid progenitor, lymphoblast and embryonic stem-cell lines. By combining the genome-wide transcription factor occupancy repertoires, associated epigenetic signals, and co-association patterns, here we deduce several evolutionary principles of gene regulatory features operating since the mouse and human lineages diverged. The genomic distribution profiles, primary binding motifs, chromatin states, and DNA methylation preferences are well conserved for TF-occupied sequences. However, the extent to which orthologous DNA segments are bound by orthologous TFs varies both among TFs and with genomic location: binding at promoters is more highly conserved than binding at distal elements. Notably, occupancy-conserved TF-occupied sequences tend to be pleiotropic; they function in several tissues and also co-associate with many TFs. Single nucleotide variants at sites with potential regulatory functions are enriched in occupancy-conserved TF-occupied sequences.


Asunto(s)
Secuencia Conservada/genética , Genoma/genética , Genómica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Humanos , Ratones , Polimorfismo de Nucleótido Simple/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA