Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Nat Genet ; 56(8): 1542-1543, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112687
2.
Nat Commun ; 14(1): 5208, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37626123

ABSTRACT

Aberrant enhancer activation is a key mechanism driving oncogene expression in many cancers. While much is known about the regulation of larger chromosome domains in eukaryotes, the details of enhancer-promoter interactions remain poorly understood. Recent work suggests co-activators like BRD4 and Mediator have little impact on enhancer-promoter interactions. In leukemias controlled by the MLL-AF4 fusion protein, we use the ultra-high resolution technique Micro-Capture-C (MCC) to show that MLL-AF4 binding promotes broad, high-density regions of enhancer-promoter interactions at a subset of key targets. These enhancers are enriched for transcription elongation factors like PAF1C and FACT, and the loss of these factors abolishes enhancer-promoter contact. This work not only provides an additional model for how MLL-AF4 is able to drive high levels of transcription at key genes in leukemia but also suggests a more general model linking enhancer-promoter crosstalk and transcription elongation.


Subject(s)
Leukemia , Nuclear Proteins , Humans , Nuclear Proteins/genetics , Transcription Factors/genetics , Regulatory Sequences, Nucleic Acid , Leukemia/genetics , Promoter Regions, Genetic/genetics , Cell Cycle Proteins , Oncogene Proteins, Fusion/genetics , Myeloid-Lymphoid Leukemia Protein/genetics
4.
Cell Stem Cell ; 30(5): 722-740.e11, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37146586

ABSTRACT

Understanding clonal evolution and cancer development requires experimental approaches for characterizing the consequences of somatic mutations on gene regulation. However, no methods currently exist that efficiently link high-content chromatin accessibility with high-confidence genotyping in single cells. To address this, we developed Genotyping with the Assay for Transposase-Accessible Chromatin (GTAC), enabling accurate mutation detection at multiple amplified loci, coupled with robust chromatin accessibility readout. We applied GTAC to primary acute myeloid leukemia, obtaining high-quality chromatin accessibility profiles and clonal identities for multiple mutations in 88% of cells. We traced chromatin variation throughout clonal evolution, showing the restriction of different clones to distinct differentiation stages. Furthermore, we identified switches in transcription factor motif accessibility associated with a specific combination of driver mutations, which biased transformed progenitors toward a leukemia stem cell-like chromatin state. GTAC is a powerful tool to study clonal heterogeneity across a wide spectrum of pre-malignant and neoplastic conditions.


Subject(s)
Chromatin , Leukemia, Myeloid, Acute , Humans , Transposases/genetics , Transposases/metabolism , Genotype , Genomics , Gene Expression Regulation
5.
Nat Commun ; 14(1): 2238, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37076455

ABSTRACT

Haemoglobin E (HbE) ß-thalassaemia causes approximately 50% of all severe thalassaemia worldwide; equating to around 30,000 births per year. HbE ß-thalassaemia is due to a point mutation in codon 26 of the human HBB gene on one allele (GAG; glutamatic acid → AAG; lysine, E26K), and any mutation causing severe ß-thalassaemia on the other. When inherited together in compound heterozygosity these mutations can cause a severe thalassaemic phenotype. However, if only one allele is mutated individuals are carriers for the respective mutation and have an asymptomatic phenotype (ß-thalassaemia trait). Here we describe a base editing strategy which corrects the HbE mutation either to wildtype (WT) or a normal variant haemoglobin (E26G) known as Hb Aubenas and thereby recreates the asymptomatic trait phenotype. We have achieved editing efficiencies in excess of 90% in primary human CD34 + cells. We demonstrate editing of long-term repopulating haematopoietic stem cells (LT-HSCs) using serial xenotransplantation in NSG mice. We have profiled the off-target effects using a combination of circularization for in vitro reporting of cleavage effects by sequencing (CIRCLE-seq) and deep targeted capture and have developed machine-learning based methods to predict functional effects of candidate off-target mutations.


Subject(s)
Hemoglobin E , Thalassemia , beta-Thalassemia , Humans , Animals , Mice , beta-Thalassemia/genetics , Hemoglobin E/genetics , Thalassemia/genetics , Mutation , Point Mutation
6.
Nat Protoc ; 18(6): 1687-1711, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36991220

ABSTRACT

Micro Capture-C (MCC) is a chromatin conformation capture (3C) method for visualizing reproducible three-dimensional contacts of specified regions of the genome at base pair resolution. These methods are an established family of techniques that use proximity ligation to assay the topology of chromatin. MCC can generate data at substantially higher resolution than previous techniques through multiple refinements of the 3C method. Using a sequence agnostic nuclease, the maintenance of cellular integrity and full sequencing of the ligation junctions, MCC achieves subnucleosomal levels of resolution, which can be used to reveal transcription factor binding sites analogous to DNAse I footprinting. Gene dense regions, close-range enhancer-promoter contacts, individual enhancers within super-enhancers and multiple other types of loci or regulatory regions that were previously challenging to assay with conventional 3C techniques, are readily observed using MCC. MCC requires training in common molecular biology techniques and bioinformatics to perform the experiment and analyze the data. The protocol can be expected to be completed in a 3 week timeframe for experienced molecular biologists.


Subject(s)
Chromatin , Chromosomes , Chromatin/genetics , Genome , Computational Biology/methods , Regulatory Sequences, Nucleic Acid
7.
Nat Commun ; 13(1): 2139, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440598

ABSTRACT

Enhancers and promoters predominantly interact within large-scale topologically associating domains (TADs), which are formed by loop extrusion mediated by cohesin and CTCF. However, it is unclear whether complex chromatin structures exist at sub-kilobase-scale and to what extent fine-scale regulatory interactions depend on loop extrusion. To address these questions, we present an MNase-based chromosome conformation capture (3C) approach, which has enabled us to generate the most detailed local interaction data to date (20 bp resolution) and precisely investigate the effects of cohesin and CTCF depletion on chromatin architecture. Our data reveal that cis-regulatory elements have distinct internal nano-scale structures, within which local insulation is dependent on CTCF, but which are independent of cohesin. In contrast, we find that depletion of cohesin causes a subtle reduction in longer-range enhancer-promoter interactions and that CTCF depletion can cause rewiring of regulatory contacts. Together, our data show that loop extrusion is not essential for enhancer-promoter interactions, but contributes to their robustness and specificity and to precise regulation of gene expression.


Subject(s)
Chromatin , Chromosomal Proteins, Non-Histone , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , Cohesins
8.
Nat Protoc ; 17(2): 445-475, 2022 02.
Article in English | MEDLINE | ID: mdl-35121852

ABSTRACT

Chromosome conformation capture (3C) methods measure the spatial proximity between DNA elements in the cell nucleus. Many methods have been developed to sample 3C material, including the Capture-C family of protocols. Capture-C methods use oligonucleotides to enrich for interactions of interest from sequencing-ready 3C libraries. This approach is modular and has been adapted and optimized to work for sampling of disperse DNA elements (NuTi Capture-C), including from low cell inputs (LI Capture-C), as well as to generate Hi-C like maps for specific regions of interest (Tiled-C) and to interrogate multiway interactions (Tri-C). We present the design, experimental protocol and analysis pipeline for NuTi Capture-C in addition to the variations for generation of LI Capture-C, Tiled-C and Tri-C data. The entire procedure can be performed in 3 weeks and requires standard molecular biology skills and equipment, access to a next-generation sequencing platform, and basic bioinformatic skills. Implemented with other sequencing technologies, these methods can be used to identify regulatory interactions and to compare the structural organization of the genome in different cell types and genetic models.


Subject(s)
Chromosomes
9.
Nat Genet ; 53(11): 1606-1615, 2021 11.
Article in English | MEDLINE | ID: mdl-34737427

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS­CoV­2) disease (COVID-19) pandemic has caused millions of deaths worldwide. Genome-wide association studies identified the 3p21.31 region as conferring a twofold increased risk of respiratory failure. Here, using a combined multiomics and machine learning approach, we identify the gain-of-function risk A allele of an SNP, rs17713054G>A, as a probable causative variant. We show with chromosome conformation capture and gene-expression analysis that the rs17713054-affected enhancer upregulates the interacting gene, leucine zipper transcription factor like 1 (LZTFL1). Selective spatial transcriptomic analysis of lung biopsies from patients with COVID-19 shows the presence of signals associated with epithelial-mesenchymal transition (EMT), a viral response pathway that is regulated by LZTFL1. We conclude that pulmonary epithelial cells undergoing EMT, rather than immune cells, are likely responsible for the 3p21.31-associated risk. Since the 3p21.31 effect is conferred by a gain-of-function, LZTFL1 may represent a therapeutic target.


Subject(s)
COVID-19/complications , Chromosomes, Human, Pair 3/genetics , Epithelial-Mesenchymal Transition , Lung/virology , Polymorphism, Single Nucleotide , SARS-CoV-2/isolation & purification , Transcription Factors/genetics , COVID-19/transmission , COVID-19/virology , Case-Control Studies , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Genome-Wide Association Study , Humans , Lung/metabolism , Lung/pathology , Male , Transcription Factors/metabolism
10.
Nat Commun ; 12(1): 4439, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290235

ABSTRACT

The α- and ß-globin loci harbor developmentally expressed genes, which are silenced throughout post-natal life. Reactivation of these genes may offer therapeutic approaches for the hemoglobinopathies, the most common single gene disorders. Here, we address mechanisms regulating the embryonically expressed α-like globin, termed ζ-globin. We show that in embryonic erythroid cells, the ζ-gene lies within a ~65 kb sub-TAD (topologically associating domain) of open, acetylated chromatin and interacts with the α-globin super-enhancer. By contrast, in adult erythroid cells, the ζ-gene is packaged within a small (~10 kb) sub-domain of hypoacetylated, facultative heterochromatin within the acetylated sub-TAD and that it no longer interacts with its enhancers. The ζ-gene can be partially re-activated by acetylation and inhibition of histone de-acetylases. In addition to suggesting therapies for severe α-thalassemia, these findings illustrate the general principles by which reactivation of developmental genes may rescue abnormalities arising from mutations in their adult paralogues.


Subject(s)
Gene Expression Regulation, Developmental , Gene Silencing , Transcriptional Activation , zeta-Globins/genetics , Acetylation , Animals , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Silencing/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation/drug effects , alpha-Globins/genetics
11.
Nature ; 595(7865): 125-129, 2021 07.
Article in English | MEDLINE | ID: mdl-34108683

ABSTRACT

In higher eukaryotes, many genes are regulated by enhancers that are 104-106 base pairs (bp) away from the promoter. Enhancers contain transcription-factor-binding sites (which are typically around 7-22 bp), and physical contact between the promoters and enhancers is thought to be required to modulate gene expression. Although chromatin architecture has been mapped extensively at resolutions of 1 kilobase and above; it has not been possible to define physical contacts at the scale of the proteins that determine gene expression. Here we define these interactions in detail using a chromosome conformation capture method (Micro-Capture-C) that enables the physical contacts between different classes of regulatory elements to be determined at base-pair resolution. We find that highly punctate contacts occur between enhancers, promoters and CCCTC-binding factor (CTCF) sites and we show that transcription factors have an important role in the maintenance of the contacts between enhancers and promoters. Our data show that interactions between CTCF sites are increased when active promoters and enhancers are located within the intervening chromatin. This supports a model in which chromatin loop extrusion1 is dependent on cohesin loading at active promoters and enhancers, which explains the formation of tissue-specific chromatin domains without changes in CTCF binding.


Subject(s)
Base Pairing/genetics , Genome/genetics , Animals , Binding Sites , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic/genetics , alpha-Globins/genetics , Cohesins
12.
Nat Commun ; 12(1): 531, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483495

ABSTRACT

Chromosome conformation capture (3C) provides an adaptable tool for studying diverse biological questions. Current 3C methods generally provide either low-resolution interaction profiles across the entire genome, or high-resolution interaction profiles at limited numbers of loci. Due to technical limitations, generation of reproducible high-resolution interaction profiles has not been achieved at genome-wide scale. Here, to overcome this barrier, we systematically test each step of 3C and report two improvements over current methods. We show that up to 30% of reporter events generated using the popular in situ 3C method arise from ligations between two individual nuclei, but this noise can be almost entirely eliminated by isolating intact nuclei after ligation. Using Nuclear-Titrated Capture-C, we generate reproducible high-resolution genome-wide 3C interaction profiles by targeting 8055 gene promoters in erythroid cells. By pairing high-resolution 3C interaction calls with nascent gene expression we interrogate the role of promoter hubs and super-enhancers in gene regulation.


Subject(s)
Cell Nucleus/genetics , Chromatin/genetics , Erythroid Cells/metabolism , Genome, Human/genetics , Genome-Wide Association Study/methods , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cells, Cultured , Chromosome Mapping/methods , Computational Biology/methods , Gene Expression Regulation , Genomics/methods , Humans , Mice, Inbred C57BL , Mice, Inbred CBA
13.
Nat Commun ; 12(1): 223, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431820

ABSTRACT

Enhancers are DNA sequences that enable complex temporal and tissue-specific regulation of genes in higher eukaryotes. Although it is not entirely clear how enhancer-promoter interactions can increase gene expression, this proximity has been observed in multiple systems at multiple loci and is thought to be essential for the maintenance of gene expression. Bromodomain and Extra-Terminal domain (BET) and Mediator proteins have been shown capable of forming phase condensates and are thought to be essential for super-enhancer function. Here, we show that targeting of cells with inhibitors of BET proteins or pharmacological degradation of BET protein Bromodomain-containing protein 4 (BRD4) has a strong impact on transcription but very little impact on enhancer-promoter interactions. Dissolving phase condensates reduces BRD4 and Mediator binding at enhancers and can also strongly affect gene transcription, without disrupting enhancer-promoter interactions. These results suggest that activation of transcription and maintenance of enhancer-promoter interactions are separable events. Our findings further indicate that enhancer-promoter interactions are not dependent on high levels of BRD4 and Mediator, and are likely maintained by a complex set of factors including additional activator complexes and, at some sites, CTCF and cohesin.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , Transcription, Genetic , CCCTC-Binding Factor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Glycols/pharmacology , Histones/metabolism , Humans , Leukemia/genetics , Leukemia/pathology , Models, Genetic , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic/drug effects , Cohesins
15.
Cell Stem Cell ; 27(5): 765-783.e14, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32991838

ABSTRACT

Non-coding mutations at the far end of a large gene desert surrounding the SOX9 gene result in a human craniofacial disorder called Pierre Robin sequence (PRS). Leveraging a human stem cell differentiation model, we identify two clusters of enhancers within the PRS-associated region that regulate SOX9 expression during a restricted window of facial progenitor development at distances up to 1.45 Mb. Enhancers within the 1.45 Mb cluster exhibit highly synergistic activity that is dependent on the Coordinator motif. Using mouse models, we demonstrate that PRS phenotypic specificity arises from the convergence of two mechanisms: confinement of Sox9 dosage perturbation to developing facial structures through context-specific enhancer activity and heightened sensitivity of the lower jaw to Sox9 expression reduction. Overall, we characterize the longest-range human enhancers involved in congenital malformations, directly demonstrate that PRS is an enhanceropathy, and illustrate how small changes in gene expression can lead to morphological variation.


Subject(s)
Neural Crest , Pierre Robin Syndrome , Cell Differentiation , Humans , Mutation/genetics , Regulatory Sequences, Nucleic Acid , SOX9 Transcription Factor/genetics
17.
Nat Genet ; 50(12): 1744-1751, 2018 12.
Article in English | MEDLINE | ID: mdl-30374068

ABSTRACT

The promoters of mammalian genes are commonly regulated by multiple distal enhancers, which physically interact within discrete chromatin domains. How such domains form and how the regulatory elements within them interact in single cells is not understood. To address this we developed Tri-C, a new chromosome conformation capture (3C) approach, to characterize concurrent chromatin interactions at individual alleles. Analysis by Tri-C identifies heterogeneous patterns of single-allele interactions between CTCF boundary elements, indicating that the formation of chromatin domains likely results from a dynamic process. Within these domains, we observe specific higher-order structures that involve simultaneous interactions between multiple enhancers and promoters. Such regulatory hubs provide a structural basis for understanding how multiple cis-regulatory elements act together to establish robust regulation of gene expression.


Subject(s)
Alleles , Chromatin , Genetic Loci , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Female , Gene Expression Regulation, Developmental , Globins/genetics , Linkage Disequilibrium , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Transcription Factors/metabolism
18.
Nat Commun ; 9(1): 100, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311615

ABSTRACT

The repression of telomerase activity during cellular differentiation promotes replicative aging and functions as a physiological barrier for tumorigenesis in long-lived mammals, including humans. However, the underlying mechanisms remain largely unclear. Here we describe how miR-615-3p represses hTERT expression. mir-615-3p is located in an intron of the HOXC5 gene, a member of the highly conserved homeobox family of transcription factors controlling embryogenesis and development. Unexpectedly, we found that HoxC5 also represses hTERT expression by disrupting the long-range interaction between hTERT promoter and its distal enhancer. The 3'UTR of hTERT and its upstream enhancer region are well conserved in long-lived primates. Both mir-615-3p and HOXC5 are activated upon differentiation, which constitute a feed-forward loop that coordinates transcriptional and post-transcriptional repression of hTERT during cellular differentiation. Deregulation of HOXC5 and mir-615-3p expression may contribute to the activation of hTERT in human cancers.


Subject(s)
Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , MicroRNAs/genetics , Telomerase/biosynthesis , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Cell Line, Tumor , Enhancer Elements, Genetic/genetics , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Neoplasms/genetics , Neoplasms/pathology , Promoter Regions, Genetic/genetics
20.
Bio Protoc ; 7(23)2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29270442

ABSTRACT

Chromosome conformation capture (3C) techniques are crucial to understanding tissue-specific regulation of gene expression, but current methods generally require large numbers of cells. This protocol describes two new low-input Capture-C approaches that can generate high-quality 3C interaction profiles from 10,000-20,000 cells, depending on the resolution used for analysis.

SELECTION OF CITATIONS
SEARCH DETAIL