Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Protoc ; 19(1): 83-112, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37935964

ABSTRACT

Cleavage under targets and tagmentation (CUT&Tag) is an antibody-directed in situ chromatin profiling strategy that is rapidly replacing immune precipitation-based methods, such as chromatin immunoprecipitation-sequencing. The efficiency of the method enables chromatin profiling in single cells but is limited by the numbers of cells that can be profiled. Here, we describe a combinatorial barcoding strategy for CUT&Tag that harnesses a nanowell dispenser for simple, high-resolution, high-throughput, single-cell chromatin profiling. In this single-cell combinatorial indexing CUT&Tag (sciCUT&Tag) protocol, lightly cross-linked nuclei are bound to magnetic beads and incubated with primary and secondary antibodies in bulk and then arrayed in a 96-well plate for a first round of cellular indexing by antibody-directed Tn5 tagmentation. The sample is then repooled, mixed and arrayed across 5,184 nanowells at a density of 12-24 nuclei per well for a second round of cellular indexing during PCR amplification of the sequencing-ready library. This protocol can be completed in 1.5 days by a research technician, and we illustrate the optimized protocol by profiling histone modifications associated with developmental gene repression (H3K27me3) as well as transcriptional activation (H3K4me1-2-3) in human peripheral blood mononuclear cells and use single-nucleotide polymorphisms to facilitate collision removal. We have also used sciCUT&Tag for simultaneous profiling of multiple chromatin epitopes in single cells. The reduced cost, improved resolution and scalability of sciCUT&Tag make it an attractive platform to profile chromatin features in single cells.


Subject(s)
Histones , Leukocytes, Mononuclear , Humans , Histones/genetics , Histones/metabolism , Leukocytes, Mononuclear/metabolism , Chromatin/genetics , Protein Processing, Post-Translational , Histone Code , Single-Cell Analysis/methods
2.
Nat Commun ; 14(1): 5930, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37739938

ABSTRACT

For more than a century, formalin-fixed paraffin-embedded (FFPE) sample preparation has been the preferred method for long-term preservation of biological material. However, the use of FFPE samples for epigenomic studies has been difficult because of chromatin damage from long exposure to high concentrations of formaldehyde. Previously, we introduced Cleavage Under Targeted Accessible Chromatin (CUTAC), an antibody-targeted chromatin accessibility mapping protocol based on CUT&Tag. Here we show that simple modifications of our CUTAC protocol either in single tubes or directly on slides produce high-resolution maps of paused RNA Polymerase II at enhancers and promoters using FFPE samples. We find that transcriptional regulatory element differences produced by FFPE-CUTAC distinguish between mouse brain tumors and identify and map regulatory element markers with high confidence and precision, including microRNAs not detectable by RNA-seq. Our simple workflows make possible affordable epigenomic profiling of archived biological samples for biomarker identification, clinical applications and retrospective studies.


Subject(s)
Chromatin , Epigenomics , Animals , Mice , Paraffin Embedding , Retrospective Studies , Chromatin/genetics , Formaldehyde
3.
PLoS Pathog ; 19(1): e1011101, 2023 01.
Article in English | MEDLINE | ID: mdl-36706161

ABSTRACT

Transcriptional silencing of latent HIV-1 proviruses entails complex and overlapping mechanisms that pose a major barrier to in vivo elimination of HIV-1. We developed a new latency CRISPR screening strategy, called Latency HIV-CRISPR which uses the packaging of guideRNA-encoding lentiviral vector genomes into the supernatant of budding virions as a direct readout of factors involved in the maintenance of HIV-1 latency. We developed a custom guideRNA library targeting epigenetic regulatory genes and paired the screen with and without a latency reversal agent-AZD5582, an activator of the non-canonical NFκB pathway-to examine a combination of mechanisms controlling HIV-1 latency. A component of the Nucleosome Acetyltransferase of H4 histone acetylation (NuA4 HAT) complex, ING3, acts in concert with AZD5582 to activate proviruses in J-Lat cell lines and in a primary CD4+ T cell model of HIV-1 latency. We found that the knockout of ING3 reduces acetylation of the H4 histone tail and BRD4 occupancy on the HIV-1 LTR. However, the combination of ING3 knockout accompanied with the activation of the non-canonical NFκB pathway via AZD5582 resulted in a dramatic increase in initiation and elongation of RNA Polymerase II on the HIV-1 provirus in a manner that is nearly unique among all cellular promoters.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Humans , Histones/metabolism , Nuclear Proteins/metabolism , HIV-1/physiology , Transcription Factors/metabolism , Virus Latency/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , HIV Seropositivity/genetics , Proviruses/genetics , CD4-Positive T-Lymphocytes , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle Proteins/metabolism
4.
Cancer Discov ; 13(3): 632-653, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36399432

ABSTRACT

Advanced prostate cancers comprise distinct phenotypes, but tumor classification remains clinically challenging. Here, we harnessed circulating tumor DNA (ctDNA) to study tumor phenotypes by ascertaining nucleosome positioning patterns associated with transcription regulation. We sequenced plasma ctDNA whole genomes from patient-derived xenografts representing a spectrum of androgen receptor active (ARPC) and neuroendocrine (NEPC) prostate cancers. Nucleosome patterns associated with transcriptional activity were reflected in ctDNA at regions of genes, promoters, histone modifications, transcription factor binding, and accessible chromatin. We identified the activity of key phenotype-defining transcriptional regulators from ctDNA, including AR, ASCL1, HOXB13, HNF4G, and GATA2. To distinguish NEPC and ARPC in patient plasma samples, we developed prediction models that achieved accuracies of 97% for dominant phenotypes and 87% for mixed clinical phenotypes. Although phenotype classification is typically assessed by IHC or transcriptome profiling from tumor biopsies, we demonstrate that ctDNA provides comparable results with diagnostic advantages for precision oncology. SIGNIFICANCE: This study provides insights into the dynamics of nucleosome positioning and gene regulation associated with cancer phenotypes that can be ascertained from ctDNA. New methods for classification in phenotype mixtures extend the utility of ctDNA beyond assessments of somatic DNA alterations with important implications for molecular classification and precision oncology. This article is highlighted in the In This Issue feature, p. 517.


Subject(s)
Circulating Tumor DNA , Prostatic Neoplasms , Male , Humans , Circulating Tumor DNA/genetics , Nucleosomes/genetics , Precision Medicine , Prostatic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Phenotype
5.
Nat Biotechnol ; 41(5): 708-716, 2023 05.
Article in English | MEDLINE | ID: mdl-36316484

ABSTRACT

Chromatin profiling at locus resolution uncovers gene regulatory features that define cell types and developmental trajectories, but it remains challenging to map and compare different chromatin-associated proteins in the same sample. Here we describe Multiple Target Identification by Tagmentation (MulTI-Tag), an antibody barcoding approach for profiling multiple chromatin features simultaneously in single cells. We optimized MulTI-Tag to retain high sensitivity and specificity, and we demonstrate detection of up to three histone modifications in the same cell: H3K27me3, H3K4me1/2 and H3K36me3. We apply MulTI-Tag to resolve distinct cell types and developmental trajectories; to distinguish unique, coordinated patterns of active and repressive element regulatory usage associated with differentiation outcomes; and to uncover associations between histone marks. Multifactorial epigenetic profiling holds promise for comprehensively characterizing cell-specific gene regulatory landscapes in development and disease.


Subject(s)
Chromatin , Histones , Histones/genetics , Histones/metabolism , Chromatin/genetics , Gene Expression Regulation , Protein Processing, Post-Translational , Epigenesis, Genetic/genetics
6.
bioRxiv ; 2023 Dec 30.
Article in English | MEDLINE | ID: mdl-38234854

ABSTRACT

Chromosomal translocations involving the Lysine-Methyl-Tansferase-2A ( KMT2A ) locus generate potent oncogenes that cause highly aggressive acute leukemias 1 . KMT2A and the most frequent translocation partners encode proteins that interact with DNA to regulate developmental gene expression 2 . KMT2A-oncogenic fusion proteins (oncoproteins) contribute to the epigenetic mechanisms that allow KMT2A -rearranged leukemias to evade targeted therapies. By profiling the oncoprotein-target sites of 34 KMT2A -rearranged leukemia samples, we find that the genomic enrichment of oncoprotein binding is highly variable between samples. At high levels of expression, the oncoproteins preferentially activate either the lymphoid or myeloid lineage program depending on the fusion partner. These fusion-partner-dependent binding sites correspond to the frequencies of each mutation in acute lymphoid leukemia versus acute myeloid leukemia. By profiling a sample that underwent a lymphoid-to-myeloid lineage switching event in response to lymphoid-directed treatment, we find the global oncoprotein levels are reduced and the oncoprotein-target gene network changes. At lower levels of expression, the oncoprotein shifts to a non-canonical regulatory program that favors the myeloid lineage, and in a subset of resistant patients, the Menin inhibitor Revumenib induces a similar response. The dynamic shifts in KMT2A oncoproteins we describe likely contribute to epigenetic resistance of KMT2A -rearranged leukemias to targeted therapies.

7.
Genome Biol ; 23(1): 81, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35300717

ABSTRACT

Cleavage Under Targets and Tagmentation (CUT&Tag) is an antibody-directed transposase tethering strategy for in situ chromatin profiling in small samples and single cells. We describe a modified CUT&Tag protocol using a mixture of an antibody to the initiation form of RNA polymerase II (Pol2 Serine-5 phosphate) and an antibody to repressive Polycomb domains (H3K27me3) followed by computational signal deconvolution to produce high-resolution maps of both the active and repressive regulomes in single cells. The ability to seamlessly map active promoters, enhancers, and repressive regulatory elements using a single workflow provides a complete regulome profiling strategy suitable for high-throughput single-cell platforms.


Subject(s)
Chromatin , Histones , Chromatin/genetics , Histones/metabolism , RNA Polymerase II/genetics , Regulatory Sequences, Nucleic Acid , Transposases/metabolism
8.
Elife ; 112022 02 28.
Article in English | MEDLINE | ID: mdl-35225231

ABSTRACT

The Fbw7 ubiquitin ligase targets many proteins for proteasomal degradation, which include oncogenic transcription factors (TFs) (e.g., c-Myc, c-Jun, and Notch). Fbw7 is a tumor suppressor and tumors often contain mutations in FBXW7, the gene that encodes Fbw7. The complexity of its substrate network has obscured the mechanisms of Fbw7-associated tumorigenesis, yet this understanding is needed for developing therapies. We used an integrated approach employing RNA-Seq and high-resolution mapping (cleavage under target and release using nuclease) of histone modifications and TF occupancy (c-Jun and c-Myc) to examine the combinatorial effects of misregulated Fbw7 substrates in colorectal cancer (CRC) cells with engineered tumor-associated FBXW7 null or missense mutations. Both Fbw7 mutations caused widespread transcriptional changes associated with active chromatin and altered TF occupancy: some were common to both Fbw7 mutant cell lines, whereas others were mutation specific. We identified loci where both Jun and Myc were coregulated by Fbw7, suggesting that substrates may have synergistic effects. One coregulated gene was CIITA, the master regulator of MHC Class II gene expression. Fbw7 loss increased MHC Class II expression and Fbw7 mutations were correlated with increased CIITA expression in TCGA colorectal tumors and cell lines, which may have immunotherapeutic implications for Fbw7-associated cancers. Analogous studies in neural stem cells in which FBXW7 had been acutely deleted closely mirrored the results in CRC cells. Gene set enrichment analyses revealed Fbw7-associated pathways that were conserved across both cell types that may reflect fundamental Fbw7 functions. These analyses provide a framework for understanding normal and neoplastic context-specific Fbw7 functions.


Subject(s)
Colorectal Neoplasms , F-Box Proteins , F-Box-WD Repeat-Containing Protein 7/genetics , Cell Cycle Proteins/metabolism , Colorectal Neoplasms/pathology , F-Box Proteins/genetics , F-Box Proteins/metabolism , Humans , Mutation , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Nat Genet ; 53(11): 1586-1596, 2021 11.
Article in English | MEDLINE | ID: mdl-34663924

ABSTRACT

Acute myeloid and lymphoid leukemias often harbor chromosomal translocations involving the KMT2A gene, encoding the KMT2A lysine methyltransferase (also known as mixed-lineage leukemia-1), and produce in-frame fusions of KMT2A to other chromatin-regulatory proteins. Here we map fusion-specific targets across the genome for diverse KMT2A oncofusion proteins in cell lines and patient samples. By modifying CUT&Tag chromatin profiling for full automation, we identify common and tumor-subtype-specific sites of aberrant chromatin regulation induced by KMT2A oncofusion proteins. A subset of KMT2A oncofusion-binding sites are marked by bivalent (H3K4me3 and H3K27me3) chromatin signatures, and single-cell CUT&Tag profiling reveals that these sites display cell-to-cell heterogeneity suggestive of lineage plasticity. In addition, we find that aberrant enrichment of H3K4me3 in gene bodies is sensitive to Menin inhibitors, demonstrating the utility of automated chromatin profiling for identifying therapeutic vulnerabilities. Thus, integration of automated and single-cell CUT&Tag can uncover epigenomic heterogeneity within patient samples and predict sensitivity to therapeutic agents.


Subject(s)
Chromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Leukemia/genetics , Leukemia/pathology , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/metabolism , Antineoplastic Agents/pharmacology , Automation, Laboratory , Benzamides/pharmacology , Benzimidazoles/pharmacology , Binding Sites , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing/methods , Gene Expression Regulation, Leukemic/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histones , Humans , Leukemia/drug therapy , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrimidines/pharmacology , Single-Cell Analysis/methods , Transcriptional Elongation Factors/genetics
11.
J Clin Invest ; 131(10)2021 05 17.
Article in English | MEDLINE | ID: mdl-33998604

ABSTRACT

Prostate cancer (PC) is driven by androgen receptor (AR) activity, a master regulator of prostate development and homeostasis. Frontline therapies for metastatic PC deprive the AR of the activating ligands testosterone (T) and dihydrotestosterone (DHT) by limiting their biosynthesis or blocking AR binding. Notably, AR signaling is dichotomous, inducing growth at lower activity levels, while suppressing growth at higher levels. Recent clinical studies have exploited this effect by administration of supraphysiological concentrations of T, resulting in clinical responses and improvements in quality of life. However, the use of T as a therapeutic agent in oncology is limited by poor drug-like properties as well as rapid and variable metabolism. Here, we investigated the antitumor effects of selective AR modulators (SARMs), which are small-molecule nonsteroidal AR agonists developed to treat muscle wasting and cachexia. Several orally administered SARMs activated the AR program in PC models. AR cistromes regulated by steroidal androgens and SARMs were superimposable. Coregulatory proteins including HOXB13 and GRHL2 comprised AR complexes assembled by both androgens and SARMs. At bioavailable concentrations, SARMs repressed MYC oncoprotein expression and inhibited the growth of castration-sensitive and castration-resistant PC in vitro and in vivo. These results support further clinical investigation of SARMs for treating advanced PC.


Subject(s)
Androgens/pharmacology , Neoplasm Proteins/agonists , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Dihydrotestosterone/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Signal Transduction/genetics
12.
PLoS Biol ; 18(12): e3001001, 2020 12.
Article in English | MEDLINE | ID: mdl-33362208

ABSTRACT

Histone variants expand chromatin functions in eukaryote genomes. H2A.B genes are testis-expressed short histone H2A variants that arose in placental mammals. Their biological functions remain largely unknown. To investigate their function, we generated a knockout (KO) model that disrupts all 3 H2A.B genes in mice. We show that H2A.B KO males have globally altered chromatin structure in postmeiotic germ cells. Yet, they do not show impaired spermatogenesis or testis function. Instead, we find that H2A.B plays a crucial role postfertilization. Crosses between H2A.B KO males and females yield embryos with lower viability and reduced size. Using a series of genetic crosses that separate parental and zygotic contributions, we show that the H2A.B status of both the father and mother, but not of the zygote, affects embryonic viability and growth during gestation. We conclude that H2A.B is a novel parental-effect gene, establishing a role for short H2A histone variants in mammalian development. We posit that parental antagonism over embryonic growth drove the origin and ongoing diversification of short histone H2A variants in placental mammals.


Subject(s)
Embryonic Development/genetics , Histones/genetics , Animals , Chromatin/genetics , Female , Gene Expression Regulation, Developmental/genetics , Genetic Variation , Genome/genetics , Histones/metabolism , Infertility, Male/genetics , Male , Mice/embryology , Mice, Knockout , Testis/embryology , Testis/metabolism
13.
Elife ; 92020 11 26.
Article in English | MEDLINE | ID: mdl-33241994

ABSTRACT

Stem cells that indirectly generate differentiated cells through intermediate progenitors drives vertebrate brain evolution. Due to a lack of lineage information, how stem cell functionality, including the competency to generate intermediate progenitors, becomes extinguished during progenitor commitment remains unclear. Type II neuroblasts in fly larval brains divide asymmetrically to generate a neuroblast and a progeny that commits to an intermediate progenitor (INP) identity. We identified Tailless (Tll) as a master regulator of type II neuroblast functional identity, including the competency to generate INPs. Successive expression of transcriptional repressors functions through Hdac3 to silence tll during INP commitment. Reducing repressor activity allows re-activation of Notch in INPs to ectopically induce tll expression driving supernumerary neuroblast formation. Knocking-down hdac3 function prevents downregulation of tll during INP commitment. We propose that continual inactivation of stem cell identity genes allows intermediate progenitors to stably commit to generating diverse differentiated cells during indirect neurogenesis.


Subject(s)
Brain/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Silencing , Neural Stem Cells/metabolism , Neurogenesis , Transcription Factors/genetics , Transcriptional Activation , Animals , Animals, Genetically Modified , Brain/embryology , Cell Lineage , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases , Larva/genetics , Larva/metabolism , Phenotype , Receptors, Notch , Repressor Proteins , Transcription Factors/metabolism
14.
Nat Protoc ; 15(10): 3264-3283, 2020 10.
Article in English | MEDLINE | ID: mdl-32913232

ABSTRACT

We recently introduced Cleavage Under Targets & Tagmentation (CUT&Tag), an epigenomic profiling strategy in which antibodies are bound to chromatin proteins in situ in permeabilized nuclei. These antibodies are then used to tether the cut-and-paste transposase Tn5. Activation of the transposase simultaneously cleaves DNA and adds adapters ('tagmentation') for paired-end DNA sequencing. Here, we introduce a streamlined CUT&Tag protocol that suppresses DNA accessibility artefacts to ensure high-fidelity mapping of the antibody-targeted protein and improves the signal-to-noise ratio over current chromatin profiling methods. Streamlined CUT&Tag can be performed in a single PCR tube, from cells to amplified libraries, providing low-cost genome-wide chromatin maps. By simplifying library preparation CUT&Tag requires less than a day at the bench, from live cells to sequencing-ready barcoded libraries. As a result of low background levels, barcoded and pooled CUT&Tag libraries can be sequenced for as little as $25 per sample. This enables routine genome-wide profiling of chromatin proteins and modifications and requires no special skills or equipment.


Subject(s)
Chromatin/genetics , Chromosome Mapping/methods , Epigenomics/methods , Base Sequence , DNA/genetics , Gene Library , High-Throughput Nucleotide Sequencing/methods , Histones/metabolism , Sequence Analysis, DNA/methods , Single-Cell Analysis/methods , Transposases/genetics , Transposases/metabolism
15.
Elife ; 92020 09 09.
Article in English | MEDLINE | ID: mdl-32902381

ABSTRACT

Lysine 27-to-methionine (K27M) mutations in the H3.1 or H3.3 histone genes are characteristic of pediatric diffuse midline gliomas (DMGs). These oncohistone mutations dominantly inhibit histone H3K27 trimethylation and silencing, but it is unknown how oncohistone type affects gliomagenesis. We show that the genomic distributions of H3.1 and H3.3 oncohistones in human patient-derived DMG cells are consistent with the DNAreplication-coupled deposition of histone H3.1 and the predominant replication-independent deposition of histone H3.3. Although H3K27 trimethylation is reduced for both oncohistone types, H3.3K27M-bearing cells retain some domains, and only H3.1K27M-bearing cells lack H3K27 trimethylation. Neither oncohistone interferes with PRC2 binding. Using Drosophila as a model, we demonstrate that inhibition of H3K27 trimethylation occurs only when H3K27M oncohistones are deposited into chromatin and only when expressed in cycling cells. We propose that oncohistones inhibit the H3K27 methyltransferase as chromatin patterns are being duplicated in proliferating cells, predisposing them to tumorigenesis.


Subject(s)
Chromatin , Gene Expression Regulation, Neoplastic/genetics , Histones , Mutation/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Drosophila/genetics , Glioma/genetics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Larva/genetics , Larva/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism
16.
Genes Dev ; 33(17-18): 1252-1264, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31395740

ABSTRACT

Although MAX is regarded as an obligate dimerization partner for MYC, its function in normal development and neoplasia is poorly defined. We show that B-cell-specific deletion of Max has a modest effect on B-cell development but completely abrogates Eµ-Myc-driven lymphomagenesis. While Max loss affects only a few hundred genes in normal B cells, it leads to the global down-regulation of Myc-activated genes in premalignant Eµ-Myc cells. We show that the balance between MYC-MAX and MNT-MAX interactions in B cells shifts in premalignant B cells toward a MYC-driven transcriptional program. Moreover, we found that MAX loss leads to a significant reduction in MYC protein levels and down-regulation of direct transcriptional targets, including regulators of MYC stability. This phenomenon is also observed in multiple cell lines treated with MYC-MAX dimerization inhibitors. Our work uncovers a layer of Myc autoregulation critical for lymphomagenesis yet partly dispensable for normal development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Active Transport, Cell Nucleus , Animals , Carcinogenesis/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Kynurenine/genetics , Kynurenine/metabolism , Lymphoma/physiopathology , Mice , Organoids/growth & development , Organoids/physiopathology , Oximes/pharmacology , Sulfonamides/pharmacology
17.
Mol Cell ; 75(3): 562-575.e5, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31253573

ABSTRACT

Although the in vitro structural and in vivo spatial characteristics of transcription factor (TF) binding are well defined, TF interactions with chromatin and other companion TFs during development are poorly understood. To analyze such interactions in vivo, we profiled several TFs across a time course of human embryonic stem cell differentiation and studied their interactions with nucleosomes and co-occurring TFs by enhanced chromatin occupancy (EChO), a computational strategy for classifying TF interactions with chromatin. EChO shows that multiple individual TFs can employ either direct DNA binding or "pioneer" nucleosome binding at different enhancer targets. Nucleosome binding is not exclusively confined to inaccessible chromatin but rather correlated with local binding of other TFs and degeneracy at key bases in the pioneer factor target motif responsible for direct DNA binding. Our strategy reveals a dynamic exchange of TFs at enhancers across developmental time that is aided by pioneer nucleosome binding.


Subject(s)
Cell Differentiation/genetics , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Transcription Factors/genetics , Binding Sites , DNA-Binding Proteins/genetics , Humans , Nucleosomes/genetics
18.
Epigenetics Chromatin ; 11(1): 74, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30577869

ABSTRACT

BACKGROUND: Our understanding of eukaryotic gene regulation is limited by the complexity of protein-DNA interactions that comprise the chromatin landscape and by inefficient methods for characterizing these interactions. We recently introduced CUT&RUN, an antibody-targeted nuclease cleavage method that profiles DNA-binding proteins, histones and chromatin-modifying proteins in situ with exceptional sensitivity and resolution. RESULTS: Here, we describe an automated CUT&RUN platform and apply it to characterize the chromatin landscapes of human cells. We find that automated CUT&RUN profiles of histone modifications crisply demarcate active and repressed chromatin regions, and we develop a continuous metric to identify cell-type-specific promoter and enhancer activities. We test the ability of automated CUT&RUN to profile frozen tumor samples and find that our method readily distinguishes two pediatric glioma xenografts by their subtype-specific gene expression programs. CONCLUSIONS: The easy, cost-effective workflow makes automated CUT&RUN an attractive tool for high-throughput characterization of cell types and patient samples.


Subject(s)
Chromatin Immunoprecipitation/methods , Gene Expression Profiling/methods , Binding Sites , Chromatin/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/classification , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , High-Throughput Screening Assays/methods , Histone Code/genetics , Histones/genetics , Humans , In Situ Hybridization/methods , K562 Cells , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Software , Transcription Factors/genetics
19.
Dev Cell ; 40(4): 367-380.e7, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28245922

ABSTRACT

How the developmental potential of differentiating stem cell progeny becomes rapidly and stably restricted following asymmetric stem cell division is unclear. In the fly larval brain, earmuff (erm) uniquely functions to restrict the developmental potential of intermediate neural progenitors (INPs) generated by asymmetrically dividing neural stem cells (neuroblasts). Here we demonstrate that the histone deacetylase Hdac1/Rpd3 functions together with self-renewal transcriptional repressors to maintain the erm immature INP enhancer in an inactive but poised state in neuroblasts. Within 2 hr of immature INP birth, downregulation of repressor activities alleviates Rpd3-mediated repression on the erm enhancer, enabling acetylation of multiple histone proteins and activating Erm expression. Erm restricts the developmental potential in immature INPs by repressing genes encoding neuroblast transcriptional activators. We propose that poising the fast-activating enhancers of master regulators of differentiation through continual histone deacetylation in stem cells enables self-renewal and rapid restriction of developmental potential following asymmetric division.


Subject(s)
Asymmetric Cell Division , Cell Self Renewal , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Histone Deacetylase 1/metabolism , Acetylation , Animals , Base Sequence , Binding Sites/genetics , Cell Differentiation/genetics , Consensus Sequence/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic/genetics , Feedback, Physiological , Histone-Lysine N-Methyltransferase , Histones/metabolism , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Protein Binding/genetics , Repressor Proteins/metabolism , Reproducibility of Results , Transcription, Genetic
20.
Elife ; 32014 Oct 06.
Article in English | MEDLINE | ID: mdl-25285447

ABSTRACT

The mechanisms that maintain the functional heterogeneity of stem cells, which generates diverse differentiated cell types required for organogenesis, are not understood. In this study, we report that Trithorax (Trx) actively maintains the heterogeneity of neural stem cells (neuroblasts) in the developing Drosophila larval brain. trx mutant type II neuroblasts gradually adopt a type I neuroblast functional identity, losing the competence to generate intermediate neural progenitors (INPs) and directly generating differentiated cells. Trx regulates a type II neuroblast functional identity in part by maintaining chromatin in the buttonhead (btd) locus in an active state through the histone methyltransferase activity of the SET1/MLL complex. Consistently, btd is necessary and sufficient for eliciting a type II neuroblast functional identity. Furthermore, over-expression of btd restores the competence to generate INPs in trx mutant type II neuroblasts. Thus, Trx instructs a type II neuroblast functional identity by epigenetically promoting Btd expression, thereby maintaining neuroblast functional heterogeneity.


Subject(s)
Brain/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Transcription Factors/genetics , Animals , Brain/cytology , Brain/growth & development , Cell Differentiation , Cell Lineage/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Genetic Loci , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Larva/cytology , Larva/genetics , Larva/growth & development , Larva/metabolism , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Neural Stem Cells/cytology , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...