Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
Add more filters

Publication year range
1.
Cell ; 181(6): 1410-1422.e27, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32413320

ABSTRACT

Tracing the lineage history of cells is key to answering diverse and fundamental questions in biology. Coupling of cell ancestry information with other molecular readouts represents an important goal in the field. Here, we describe the CRISPR array repair lineage tracing (CARLIN) mouse line and corresponding analysis tools that can be used to simultaneously interrogate the lineage and transcriptomic information of single cells inĀ vivo. This model exploits CRISPR technology to generate up to 44,000 transcribed barcodes in an inducible fashion at any point during development or adulthood, is compatible with sequential barcoding, and is fully genetically defined. We have used CARLIN to identify intrinsic biases in the activity of fetal liver hematopoietic stem cell (HSC) clones and to uncover a previously unappreciated clonal bottleneck in the response of HSCs to injury. CARLIN also allows the unbiased identification of transcriptional signatures associated with HSC activity without cell sorting.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Lineage/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Transcriptome/genetics , Animals , Cell Line , Female , Flow Cytometry/methods , Hematopoietic Stem Cells/physiology , Male , Mice , Transduction, Genetic/methods
2.
Cell ; 173(2): 430-442.e17, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29606353

ABSTRACT

Fetal hemoglobin (HbF, α2ƎĀ³2) level is genetically controlled and modifies severity of adult hemoglobin (HbA, α2Ɵ2) disorders, sickle cell disease, and Ɵ-thalassemia. Common genetic variation affects expression of BCL11A, a regulator of HbF silencing. To uncover how BCL11A supports the developmental switch from ƎĀ³- to Ɵ- globin, we use a functional assay and protein binding microarray to establish a requirement for a zinc-finger cluster in BCL11A in repression and identify a preferred DNA recognition sequence. This motif appears in embryonic and fetal-expressed globin promoters and is duplicated in ƎĀ³-globin promoters. The more distal of the duplicated motifs is mutated in individuals with hereditary persistence of HbF. Using the CUT&RUN approach to map protein binding sites in erythroid cells, we demonstrate BCL11A occupancy preferentially at the distal motif, which can be disrupted by editing the promoter. OurĀ findings reveal that direct ƎĀ³-globin gene promoter repression by BCL11A underlies hemoglobin switching.


Subject(s)
Carrier Proteins/metabolism , Fetal Hemoglobin/genetics , Nuclear Proteins/metabolism , Base Sequence , Binding Sites , Carrier Proteins/genetics , Cell Line , Chromatin/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Editing , Humans , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins , Zinc Fingers/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/pathology , gamma-Globins/genetics
3.
Cell ; 172(5): 1091-1107.e17, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474909

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) technologies are poised to reshape the current cell-type classification system. However, a transcriptome-based single-cell atlas has not been achieved for complex mammalian systems. Here, we developed Microwell-seq, a high-throughput and low-cost scRNA-seq platform using simple, inexpensive devices. Using Microwell-seq, we analyzed more than 400,000 single cells covering all of the major mouse organs and constructed a basic scheme for a mouse cell atlas (MCA). We reveal a single-cell hierarchy for many tissues that have not been well characterized previously. We built a web-based "single-cell MCA analysis" pipeline that accurately defines cell types based on single-cell digital expression. Our study demonstrates the wide applicability of the Microwell-seq technology and MCA resource.


Subject(s)
Sequence Analysis, RNA , Single-Cell Analysis , 3T3 Cells , Animals , Costs and Cost Analysis , Female , High-Throughput Nucleotide Sequencing/economics , Mice , Organ Specificity , Reproducibility of Results , Sequence Analysis, RNA/economics , Single-Cell Analysis/economics
4.
Cell ; 165(6): 1389-1400, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27212235

ABSTRACT

Bivalent promoters in embryonic stem cells (ESCs) carry methylation marks on two lysine residues, K4Ā and K27, in histone3 (H3). K4me2/3 is generally considered to promote transcription, and Polycomb Repressive Complex 2 (PRC2) places K27me3, which is erased at lineage-restricted genes when ESCs differentiate in culture. Molecular defects in various PRC2 null adult tissues lack a unifying explanation. We found that epigenomes in adult mouse intestine and other self-renewing tissues show fewer and distinct bivalent promoters compared to ESCs. Groups of tissue-specific genes that carry bivalent marks are repressed, despite the presence of promoter H3K4me2/3. These are the predominant genesĀ de-repressed in PRC2-deficient adult cells, where aberrant expression is proportional to the H3K4me2/3 levels observed at their promoters inĀ wild-type cells. Thus, in adult animals, PRC2 specifically represses genes with acquired, tissue-restricted promoter bivalency. These findings provide new insights into specificity in chromatin-based gene regulation.


Subject(s)
Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic , Animals , Cell Differentiation/genetics , DNA Methylation , Gene Expression Regulation , Histones/metabolism , Intestinal Mucosa/metabolism , Intestines/cytology , Lysine/metabolism , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 2/metabolism
6.
Genes Dev ; 36(1-2): 38-52, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34969824

ABSTRACT

Barrett's esophagus (BE) and gastric intestinal metaplasia are related premalignant conditions in which areas of human stomach epithelium express mixed gastric and intestinal features. Intestinal transcription factors (TFs) are expressed in both conditions, with unclear causal roles and cis-regulatory mechanisms. Ectopic CDX2 reprogrammed isogenic mouse stomach organoid lines to a hybrid stomach-intestinal state transcriptionally similar to clinical metaplasia; squamous esophageal organoids resisted this CDX2-mediated effect. Reprogramming was associated with induced activity at thousands of previously inaccessible intestine-restricted enhancers, where CDX2 occupied DNA directly. HNF4A, a TF recently implicated in BE pathogenesis, induced weaker intestinalization by binding a novel shadow Cdx2 enhancer and hence activating Cdx2 expression. CRISPR/Cas9-mediated germline deletion of that cis-element demonstrated its requirement in Cdx2 induction and in the resulting activation of intestinal genes in stomach cells. dCas9-conjugated KRAB repression mapped this activity to the shadow enhancer's HNF4A binding site. Altogether, we show extensive but selective recruitment of intestinal enhancers by CDX2 in gastric cells and that HNF4A-mediated ectopic CDX2 expression in the stomach occurs through a conserved shadow cis-element. These findings identify mechanisms for TF-driven intestinal metaplasia and a likely pathogenic TF hierarchy.


Subject(s)
Barrett Esophagus , Transcription Factors , Animals , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , CDX2 Transcription Factor/genetics , Homeodomain Proteins/genetics , Metaplasia/genetics , Mice , Transcription Factors/genetics
7.
Genes Dev ; 36(5-6): 368-389, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35301220

ABSTRACT

Acute myeloid leukemia with KMT2A (MLL) rearrangements is characterized by specific patterns of gene expression and enhancer architecture, implying unique core transcriptional regulatory circuitry. Here, we identified the transcription factors MEF2D and IRF8 as selective transcriptional dependencies of KMT2A-rearranged AML, where MEF2D displays partially redundant functions with its paralog, MEF2C. Rapid transcription factor degradation followed by measurements of genome-wide transcription rates and superresolution microscopy revealed that MEF2D and IRF8 form a distinct core regulatory module with a narrow direct transcriptional program that includes activation of the key oncogenes MYC, HOXA9, and BCL2. Our study illustrates a mechanism of context-specific transcriptional addiction whereby a specific AML subclass depends on a highly specialized core regulatory module to directly enforce expression of common leukemia oncogenes.


Subject(s)
Leukemia, Myeloid, Acute , Myeloid-Lymphoid Leukemia Protein , Gene Rearrangement , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogenes/genetics
8.
Nat Immunol ; 18(2): 184-195, 2017 02.
Article in English | MEDLINE | ID: mdl-27992400

ABSTRACT

Invariant natural killer T cells (iNKT cells) are innate-like lymphocytes that protect against infection, autoimmune disease and cancer. However, little is known about the epigenetic regulation of iNKT cell development. Here we found that the H3K27me3 histone demethylase UTX was an essential cell-intrinsic factor that controlled an iNKT-cell lineage-specific gene-expression program and epigenetic landscape in a demethylase-activity-dependent manner. UTX-deficient iNKT cells exhibited impaired expression of iNKT cell signature genes due to a decrease in activation-associated H3K4me3 marks and an increase in repressive H3K27me3 marks within the promoters occupied by UTX. We found that JunB regulated iNKT cell development and that the expression of genes that were targets of both JunB and the iNKT cell master transcription factor PLZF was UTX dependent. We identified iNKT cell super-enhancers and demonstrated that UTX-mediated regulation of super-enhancer accessibility was a key mechanism for commitment to the iNKT cell lineage. Our findings reveal how UTX regulates the development of iNKT cells through multiple epigenetic mechanisms.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Gene Expression Regulation , Histone Demethylases/metabolism , Natural Killer T-Cells/physiology , Animals , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Histone Demethylases/genetics , Immunity, Innate/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic/genetics , Promyelocytic Leukemia Zinc Finger Protein , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Cell ; 157(3): 549-64, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766805

ABSTRACT

Hematopoietic stem cells (HSCs) sustain blood formation throughout life and are the functional units of bone marrow transplantation. We show that transient expression of six transcription factors Run1t1, Hlf, Lmo2, Prdm5, Pbx1, and Zfp37 imparts multilineage transplantation potential onto otherwise committed lymphoid and myeloid progenitors and myeloid effector cells. Inclusion of Mycn and Meis1 and use of polycistronic viruses increase reprogramming efficacy. The reprogrammed cells, designated induced-HSCs (iHSCs), possess clonal multilineage differentiation potential, reconstitute stem/progenitor compartments, and are serially transplantable. Single-cell analysis revealed that iHSCs derived under optimal conditions exhibit a gene expression profile that is highly similar to endogenous HSCs. These findings demonstrate that expression of a set of defined factors is sufficient to activate the gene networks governing HSC functional identity in committed blood cells. Our results raise the prospect that blood cell reprogramming may be a strategy for derivation of transplantable stem cells for clinical application.


Subject(s)
Cellular Reprogramming , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Myeloid Ecotropic Viral Integration Site 1 Protein , N-Myc Proto-Oncogene Protein , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Single-Cell Analysis , Transcriptome
11.
Cell ; 150(3): 590-605, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863011

ABSTRACT

Endothelium in embryonic hematopoietic tissues generates hematopoietic stem/progenitor cells; however, it is unknown how its unique potential is specified. We show that transcription factor Scl/Tal1 is essential for both establishing the hematopoietic transcriptional program in hemogenic endothelium and preventing its misspecification to a cardiomyogenic fate. Scl(-/-) embryos activated a cardiac transcriptional program in yolk sac endothelium, leading to the emergence of CD31+Pdgfrα+ cardiogenic precursors that generated spontaneously beating cardiomyocytes. Ectopic cardiogenesis was also observed in Scl(-/-) hearts, where the disorganized endocardium precociously differentiated into cardiomyocytes. Induction of mosaic deletion of Scl in Scl(fl/fl)Rosa26Cre-ER(T2) embryos revealed a cell-intrinsic, temporal requirement for Scl to prevent cardiomyogenesis from endothelium. Scl(-/-) endothelium also upregulated the expression of Wnt antagonists, which promoted rapid cardiomyocyte differentiation of ectopic cardiogenic cells. These results reveal unexpected plasticity in embryonic endothelium such that loss of a single master regulator can induce ectopic cardiomyogenesis from endothelial cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelium, Vascular/embryology , Heart/embryology , Proto-Oncogene Proteins/metabolism , Animals , Cadherins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Gene Expression Regulation, Developmental , Hemangioblasts , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , LIM-Homeodomain Proteins/metabolism , Mesoderm/metabolism , Mice , Myocytes, Cardiac/cytology , Placenta/blood supply , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Receptor, Platelet-Derived Growth Factor alpha/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/metabolism , Yolk Sac/blood supply
12.
Mol Cell ; 74(3): 542-554.e5, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30905509

ABSTRACT

Developing and adult tissues use different cis-regulatory elements. Although DNA at some decommissioned embryonic enhancers is hypomethylated in adult cells, it is unknown whether this putative epigenetic memory is complete and recoverable. We find that, in adult mouse cells, hypomethylated CpG dinucleotides preserve a nearly complete archive of tissue-specific developmental enhancers. Sites that carry the active histone mark H3K4me1, and are therefore considered "primed," are mainly cis elements that act late in organogenesis. In contrast, sites decommissioned early in development retain hypomethylated DNA as a singular property. In adult intestinal and blood cells, sustained absence of polycomb repressive complex 2 indirectly reactivates most-and only-hypomethylated developmental enhancers. Embryonic and fetal transcriptional programs re-emerge as a result, in reverse chronology to cis element inactivation during development. Thus, hypomethylated DNA in adult cells preserves a "fossil record" of tissue-specific developmental enhancers, stably marking decommissioned sites and enabling recovery of this epigenetic memory.


Subject(s)
DNA Methylation/genetics , Enhancer Elements, Genetic/genetics , Epigenomics , Histones/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Mice
13.
Mol Cell ; 74(6): 1148-1163.e7, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31005419

ABSTRACT

Self-renewal and pluripotency of the embryonic stem cell (ESC) state are established and maintained by multiple regulatory networks that comprise transcription factors and epigenetic regulators. While much has been learned regarding transcription factors, the function of epigenetic regulators in theseĀ networks is less well defined. We conducted a CRISPR-Cas9-mediated loss-of-function genetic screen that identified two epigenetic regulators, TAF5L and TAF6L, components or co-activators of the GNAT-HAT complexes for the mouse ESC (mESC) state. Detailed molecular studies demonstrate that TAF5L/TAF6L transcriptionally activate c-Myc and Oct4 and their corresponding MYC and CORE regulatory networks. Besides, TAF5L/TAF6L predominantly regulate their target genes through H3K9ac deposition and c-MYC recruitment that eventually activate the MYC regulatory network for self-renewal of mESCs. Thus, our findings uncover a role of TAF5L/TAF6L in directing the MYC regulatory network that orchestrates gene expression programs to control self-renewal for the maintenance ofĀ mESC state.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , TATA-Binding Protein Associated Factors/genetics , Animals , CRISPR-Cas Systems , Cell Cycle/genetics , Cell Proliferation , Cellular Reprogramming , Embryo, Mammalian , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Editing , Gene Expression Regulation , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , TATA-Binding Protein Associated Factors/metabolism
14.
Cell ; 145(6): 835-50, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663790

ABSTRACT

The pluripotent state of embryonic stem cells (ESCs) provides a unique perspective on regulatory programs that govern self-renewal and differentiation and somatic cell reprogramming. Here, we review the highly connected protein and transcriptional networks that maintain pluripotency and how they are intertwined with factors that affect chromatin structure and function. The complex interrelationships between pluripotency and chromatin factors are illustrated by X chromosome inactivation, regulatory control by noncoding RNAs, and environmental influences on cell states. Manipulation of cell state through the process of transdifferentiation suggests that environmental cues may direct transcriptional programs as cells enter a transiently "plastic" state during reprogramming.


Subject(s)
Cellular Reprogramming , Chromatin/metabolism , Gene Regulatory Networks , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Cell Transdifferentiation , Embryonic Stem Cells/metabolism , Humans , X Chromosome Inactivation
15.
Nature ; 578(7794): 278-283, 2020 02.
Article in English | MEDLINE | ID: mdl-32025033

ABSTRACT

The biology of haematopoietic stem cells (HSCs) has predominantly been studied under transplantation conditions1,2. It has been particularly challenging to study dynamic HSC behaviour, given that the visualization of HSCs in the native niche in live animals has not, to our knowledge, been achieved. Here we describe a dual genetic strategy in mice that restricts reporter labelling to a subset of the most quiescent long-term HSCs (LT-HSCs) and that is compatible with current intravital imaging approaches in the calvarial bone marrow3-5. We show that this subset of LT-HSCs resides close to both sinusoidal blood vessels and the endosteal surface. By contrast, multipotent progenitor cells (MPPs) show greater variation in distance from the endosteum and are more likely to be associated with transition zone vessels. LT-HSCs are not found in bone marrow niches with the deepest hypoxia and instead are found in hypoxic environments similar to those of MPPs. In vivo time-lapse imaging revealed that LT-HSCs at steady-state show limited motility. Activated LT-HSCs show heterogeneous responses, with some cells becoming highly motile and a fraction of HSCs expanding clonally within spatially restricted domains. These domains have defined characteristics, as HSC expansion is found almost exclusively in a subset of bone marrow cavities with bone-remodelling activity. By contrast, cavities with low bone-resorbing activity do not harbour expanding HSCs. These findings point to previously unknown heterogeneity within the bone marrow microenvironment, imposed by the stages of bone turnover. Our approach enables the direct visualization of HSC behaviours and dissection of heterogeneity in HSC niches.


Subject(s)
Hematopoietic Stem Cells/metabolism , Molecular Imaging , Animals , Bone Remodeling , Cell Movement , Cell Proliferation , Cell Survival , Female , Genes, Reporter , Hypoxia/metabolism , MDS1 and EVI1 Complex Locus Protein/genetics , MDS1 and EVI1 Complex Locus Protein/metabolism , Male , Mice , Oxygen/metabolism , Skull/cytology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
16.
Proc Natl Acad Sci U S A ; 120(3): e2218959120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36626555

ABSTRACT

Transcription factors (TFs) control numerous genes that are directly relevant to many human disorders. However, developing specific reagents targeting TFs within intact cells is challenging due to the presence of highly disordered regions within these proteins. Intracellular antibodies offer opportunities to probe protein function and validate therapeutic targets. Here, we describe the optimization of nanobodies specific for BCL11A, a validated target for the treatment of hemoglobin disorders. We obtained first-generation nanobodies directed to a region of BCL11A comprising zinc fingers 4 to 6 (ZF456) from a synthetic yeast surface display library, and employed error-prone mutagenesis, structural determination, and molecular modeling to enhance binding affinity. Engineered nanobodies recognized ZF6 and mediated targeted protein degradation (TPD) of BCL11A protein in erythroid cells, leading to the anticipated reactivation of fetal hemoglobin (HbF) expression. Evolved nanobodies distinguished BCL11A from its close paralog BCL11B, which shares an identical DNA-binding specificity. Given the ease of manipulation of nanobodies and their exquisite specificity, nanobody-mediated TPD of TFs should be suitable for dissecting regulatory relationships of TFs and gene targets and validating therapeutic potential of proteins of interest.


Subject(s)
Single-Domain Antibodies , Humans , Repressor Proteins/genetics , Repressor Proteins/metabolism , Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Fetal Hemoglobin/metabolism
17.
Cell ; 143(2): 313-24, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20946988

ABSTRACT

c-Myc (Myc) is an important transcriptional regulator in embryonic stem (ES) cells, somatic cell reprogramming, and cancer. Here, we identify a Myc-centered regulatory network in ES cells by combining protein-protein and protein-DNA interaction studies and show that Myc interacts with the NuA4 complex, a regulator of ES cell identity. In combination with regulatory network information, we define three ES cell modules (Core, Polycomb, and Myc) and show that the modules are functionally separable, illustrating that the overall ES cell transcription program is composed of distinct units. With these modules as an analytical tool, we have reassessed the hypothesis linking an ES cell signature with cancer or cancer stem cells. We find that the Myc module, independent of the Core module, is active in various cancers and predicts cancer outcome. The apparent similarity of cancer and ES cell signatures reflects, in large part, the pervasive nature of Myc regulatory networks.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Acetylation , Animals , Cell Line , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Transcription Factors/metabolism , Transcription, Genetic
18.
Cell ; 142(1): 133-43, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20603019

ABSTRACT

Recent genome-wide studies have demonstrated that pausing of RNA polymerase II (Pol II) occurred on many vertebrate genes. By genetic studies in the zebrafish tif1gamma mutant moonshine we found that loss of function of Pol II-associated factors PAF or DSIF rescued erythroid gene transcription in tif1gamma-deficient animals. Biochemical analysis established physical interactions among TIF1gamma, the blood-specific SCL transcription complex, and the positive elongation factors p-TEFb and FACT. Chromatin immunoprecipitation assays in human CD34(+) cells supported a TIF1gamma-dependent recruitment of positive elongation factors to erythroid genes to promote transcription elongation by counteracting Pol II pausing. Our study establishes a mechanism for regulating tissue cell fate and differentiation through transcription elongation.


Subject(s)
Erythropoiesis , Transcription Factors/metabolism , Transcription, Genetic , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Line, Tumor , Cells, Cultured , Erythroid Cells/metabolism , Humans , RNA Polymerase II/metabolism , Zebrafish/metabolism
19.
Nature ; 572(7771): 676-680, 2019 08.
Article in English | MEDLINE | ID: mdl-31391581

ABSTRACT

The CCCTC-binding factor (CTCF), which anchors DNA loops that organize the genome into structural domains, has a central role in gene control by facilitating or constraining interactions between genes and their regulatory elements1,2. In cancer cells, the disruption of CTCF binding at specific loci by somatic mutation3,4 or DNA hypermethylation5 results in the loss of loop anchors and consequent activation of oncogenes. By contrast, the germ-cell-specific paralogue of CTCF, BORIS (brother of the regulator of imprinted sites, also known as CTCFL)6, is overexpressed in several cancers7-9, but its contributions to the malignant phenotype remain unclear. Here we show that aberrant upregulation of BORIS promotes chromatin interactions in ALK-mutated, MYCN-amplified neuroblastoma10 cells that develop resistance to ALK inhibition. These cells are reprogrammed to a distinct phenotypic state during the acquisition of resistance, a process defined by the initial loss of MYCN expression followed by subsequent overexpression of BORIS and a concomitant switch in cellular dependence from MYCN to BORIS. The resultant BORIS-regulated alterations in chromatin looping lead to the formation of super-enhancers that drive the ectopic expression of a subset of proneural transcription factors that ultimately define the resistance phenotype. These results identify a previously unrecognized role of BORIS-to promote regulatory chromatin interactions that support specific cancer phenotypes.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Anaplastic Lymphoma Kinase/antagonists & inhibitors , Anaplastic Lymphoma Kinase/genetics , Animals , CCCTC-Binding Factor/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Mice , Molecular Targeted Therapy , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/enzymology , Neuroblastoma/genetics , Phenotype , Protein Binding
20.
Blood ; 139(16): 2534-2546, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35030251

ABSTRACT

Master regulators, such as the hematopoietic transcription factor (TF) GATA1, play an essential role in orchestrating lineage commitment and differentiation. However, the precise mechanisms by which such TFs regulate transcription through interactions with specific cis-regulatory elements remain incompletely understood. Here, we describe a form of congenital hemolytic anemia caused by missense mutations in an intrinsically disordered region of GATA1, with a poorly understood role in transcriptional regulation. Through integrative functional approaches, we demonstrate that these mutations perturb GATA1 transcriptional activity by partially impairing nuclear localization and selectively altering precise chromatin occupancy by GATA1. These alterations in chromatin occupancy and concordant chromatin accessibility changes alter faithful gene expression, with failure to both effectively silence and activate select genes necessary for effective terminal red cell production. We demonstrate how disease-causing mutations can reveal regulatory mechanisms that enable the faithful genomic targeting of master TFs during cellular differentiation.


Subject(s)
Anemia , GATA1 Transcription Factor , Cell Differentiation/genetics , Chromatin/genetics , Chromatin Immunoprecipitation , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL