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










Publication year range
2.
Nat Commun ; 15(1): 1768, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38409079

ABSTRACT

Extrachromosomal circular DNAs (eccDNAs) have emerged as important intra-cellular mobile genetic elements that affect gene copy number and exert in trans regulatory roles within the cell nucleus. Here, we describe scCircle-seq, a method for profiling eccDNAs and unraveling their diversity and complexity in single cells. We implement and validate scCircle-seq in normal and cancer cell lines, demonstrating that most eccDNAs vary largely between cells and are stochastically inherited during cell division, although their genomic landscape is cell type-specific and can be used to accurately cluster cells of the same origin. eccDNAs are preferentially produced from chromatin regions enriched in H3K9me3 and H3K27me3 histone marks and are induced during replication stress conditions. Concomitant sequencing of eccDNAs and RNA from the same cell uncovers the absence of correlation between eccDNA copy number and gene expression levels, except for a few oncogenes, including MYC, contained within a large eccDNA in colorectal cancer cells. Lastly, we apply scCircle-seq to one prostate cancer and two breast cancer specimens, revealing cancer-specific eccDNA landscapes and a higher propensity of eccDNAs to form in amplified genomic regions. scCircle-seq is a scalable tool that can be used to dissect the complexity of eccDNAs across different cell and tissue types, and further expands the potential of eccDNAs for cancer diagnostics.


Subject(s)
DNA, Circular , DNA , Male , Humans , DNA, Circular/genetics , Chromosomes , Cell Line , Oncogenes
3.
Sci Adv ; 9(45): eadi2095, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37939182

ABSTRACT

Co-transcriptional RNA-DNA hybrids can not only cause DNA damage threatening genome integrity but also regulate gene activity in a mechanism that remains unclear. Here, we show that the nucleotide excision repair factor XPF interacts with the insulator binding protein CTCF and the cohesin subunits SMC1A and SMC3, leading to R-loop-dependent DNA looping upon transcription activation. To facilitate R-loop processing, XPF interacts and recruits with TOP2B on active gene promoters, leading to double-strand break accumulation and the activation of a DNA damage response. Abrogation of TOP2B leads to the diminished recruitment of XPF, CTCF, and the cohesin subunits to promoters of actively transcribed genes and R-loops and the concurrent impairment of CTCF-mediated DNA looping. Together, our findings disclose an essential role for XPF with TOP2B and the CTCF/cohesin complex in R-loop processing for transcription activation with important ramifications for DNA repair-deficient syndromes associated with transcription-associated DNA damage.


Subject(s)
DNA-Binding Proteins , R-Loop Structures , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Chromosomes , DNA Repair , Chromatin
4.
Sci Data ; 9(1): 400, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35821502

ABSTRACT

Endogenous DNA double-strand breaks (DSBs) occurring in neural cells have been implicated in the pathogenesis of neurodevelopmental disorders (NDDs). Currently, a genomic map of endogenous DSBs arising during human neurogenesis is missing. Here, we applied in-suspension Breaks Labeling In Situ and Sequencing (sBLISS), RNA-Seq, and Hi-C to chart the genomic landscape of DSBs and relate it to gene expression and genome architecture in 2D cultures of human neuroepithelial stem cells (NES), neural progenitor cells (NPC), and post-mitotic neural cells (NEU). Endogenous DSBs were enriched at the promoter and along the gene body of transcriptionally active genes, at the borders of topologically associating domains (TADs), and around chromatin loop anchors. NDD risk genes harbored significantly more DSBs in comparison to other protein-coding genes, especially in NEU cells. We provide sBLISS, RNA-Seq, and Hi-C datasets for each differentiation stage, and all the scripts needed to reproduce our analyses. Our datasets and tools represent a unique resource that can be harnessed to investigate the role of genome fragility in the pathogenesis of NDDs.


Subject(s)
DNA Breaks, Double-Stranded , Neurogenesis , Cell Line, Tumor , DNA/metabolism , Genomics , Humans
5.
Trends Genet ; 38(10): 1062-1075, 2022 10.
Article in English | MEDLINE | ID: mdl-35680466

ABSTRACT

Over a decade ago the advent of high-throughput chromosome conformation capture (Hi-C) sparked a new era of 3D genomics. Since then the number of methods for mapping the 3D genome has flourished, enabling an ever-increasing understanding of how DNA is packaged in the nucleus and how the spatiotemporal organization of the genome orchestrates its vital functions. More recently, the next generation of spatial genomics technologies has begun to reveal how genome sequence and 3D genome organization vary between cells in their tissue context. We summarize how the toolkit for charting genome topology has evolved over the past decade and discuss how new technological developments are advancing the field of 3D and spatial genomics.


Subject(s)
Genome , Genomics , Cell Nucleus , Chromatin/genetics , Chromosomes/genetics , Genome/genetics , Genomics/methods , Molecular Conformation
6.
Curr Opin Cell Biol ; 74: 7-12, 2022 02.
Article in English | MEDLINE | ID: mdl-34998095

ABSTRACT

A growing body of evidence points to a role of nuclear RNAs (nucRNAs) in shaping the three-dimensional (3D) architecture of the genome within the nucleus of a eukaryotic cell. nucRNAs are non-homogeneously distributed within the nucleus where they can form global and local gradients that might contribute to instructing the formation and coordinating the function of different types of 3D genome structures. In this article, we highlight the available literature supporting a role of nucRNAs as 3D genome shapers and propose that nucRNA gradients are key mediators of genome structure and function.


Subject(s)
Cell Nucleus , RNA , Cell Nucleus/metabolism , Chromatin/metabolism , Eukaryotic Cells , Genome/genetics , RNA/genetics , RNA/metabolism
7.
Methods Mol Biol ; 2162: 261-281, 2021.
Article in English | MEDLINE | ID: mdl-32926388

ABSTRACT

Clustered regularly interspaced palindromic repeat (CRISPR) systems are revolutionizing many areas of biology and medicine, where they are increasingly utilized as therapeutic tools for correcting disease-causing mutations. From a clinical perspective, unintended off-target (OT) DNA double-strand break (DSB) induction by CRISPR nucleases represents a major concern. Therefore, in recent years considerable effort has been dedicated to developing methods for assessing the OT activity of CRISPR nucleases, which in turn can be used to guide engineering of nucleases with minimal OT activity. Here we describe a detailed protocol for quantifying OT DSBs genome-wide in cultured cells transfected with CRISPR enzymes, based on the breaks labeling in situ and sequencing (BLISS) method that we have previously developed. CRISPR-BLISS is versatile and scalable, and allows assessment of multiple guide RNAs in different cell types and time points following cell transfection or transduction.


Subject(s)
CRISPR-Cas Systems , DNA Breaks, Double-Stranded , Gene Editing/methods , Gene Targeting/methods , HEK293 Cells , Humans
8.
Nat Protoc ; 15(12): 3894-3941, 2020 12.
Article in English | MEDLINE | ID: mdl-33139954

ABSTRACT

sBLISS (in-suspension breaks labeling in situ and sequencing) is a versatile and widely applicable method for identification of endogenous and induced DNA double-strand breaks (DSBs) in any cell type that can be brought into suspension. sBLISS provides genome-wide profiles of the most consequential DNA lesion implicated in a variety of pathological, but also physiological, processes. In sBLISS, after in situ labeling, DSB ends are linearly amplified, followed by next-generation sequencing and DSB landscape analysis. Here, we present a step-by-step experimental protocol for sBLISS, as well as a basic computational analysis. The main advantages of sBLISS are (i) the suspension setup, which renders the protocol user-friendly and easily scalable; (ii) the possibility of adapting it to a high-throughput or single-cell workflow; and (iii) its flexibility and its applicability to virtually every cell type, including patient-derived cells, organoids, and isolated nuclei. The wet-lab protocol can be completed in 1.5 weeks and is suitable for researchers with intermediate expertise in molecular biology and genomics. For the computational analyses, basic-to-intermediate bioinformatics expertise is required.


Subject(s)
DNA Breaks, Double-Stranded , Genomics/methods , Base Sequence , Cell Line , Suspensions
9.
Nat Med ; 26(7): 1063-1069, 2020 07.
Article in English | MEDLINE | ID: mdl-32483361

ABSTRACT

The mucosal epithelium is a common target of damage by chronic bacterial infections and the accompanying toxins, and most cancers originate from this tissue. We investigated whether colibactin, a potent genotoxin1 associated with certain strains of Escherichia coli2, creates a specific DNA-damage signature in infected human colorectal cells. Notably, the genomic contexts of colibactin-induced DNA double-strand breaks were enriched for an AT-rich hexameric sequence motif, associated with distinct DNA-shape characteristics. A survey of somatic mutations at colibactin target sites of several thousand cancer genomes revealed notable enrichment of this motif in colorectal cancers. Moreover, the exact double-strand-break loci corresponded with mutational hot spots in cancer genomes, reminiscent of a trinucleotide signature previously identified in healthy colorectal epithelial cells3. The present study provides evidence for the etiological role of colibactin in human cancer.


Subject(s)
Colorectal Neoplasms/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , Peptides/pharmacology , Polyketides/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Epithelial Cells/drug effects , Escherichia coli/pathogenicity , Humans , Mutation/drug effects , Nucleotide Motifs/drug effects
10.
Nat Protoc ; 15(2): 364-397, 2020 02.
Article in English | MEDLINE | ID: mdl-31932773

ABSTRACT

We present the experimental protocol and data analysis toolbox for multi-contact 4C (MC-4C), a new proximity ligation method tailored to study the higher-order chromatin contact patterns of selected genomic sites. Conventional chromatin conformation capture (3C) methods fragment proximity ligation products for efficient analysis of pairwise DNA contacts. By contrast, MC-4C is designed to preserve and collect large concatemers of proximity ligated fragments for long-molecule sequencing on an Oxford Nanopore or Pacific Biosciences platform. Each concatemer of proximity ligation products represents a snapshot topology of a different individual allele, revealing its multi-way chromatin interactions. By inverse PCR with primers specific for a fragment of interest (the viewpoint) and DNA size selection, sequencing is selectively targeted to thousands of different complex interactions containing this viewpoint. A tailored statistical analysis toolbox is able to generate background models and three-way interaction profiles from the same dataset. These profiles can be used to distinguish whether contacts between more than two regulatory sequences are mutually exclusive or, conversely, simultaneously occurring at chromatin hubs. The entire procedure can be completed in 2 w, and requires standard molecular biology and data analysis skills and equipment, plus access to a third-generation sequencing platform.


Subject(s)
Chromatin/chemistry , Chromatin/genetics , Sequence Analysis, DNA/methods , Humans , K562 Cells , Molecular Conformation
11.
Cell Rep ; 29(3): 560-572.e4, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618627

ABSTRACT

DNA double-strand breaks (DSBs) are deleterious and tumorigenic but could also be essential for DNA-based processes. Yet the landscape of physiological DSBs and their role and repair are still elusive. Here, we mapped DSBs at high resolution in cancer and non-tumorigenic cells and found a transcription-coupled repair mechanism at oncogenic super-enhancers. At these super-enhancers the transcription factor TEAD4, together with various transcription factors and co-factors, co-localizes with the repair factor RAD51 of the homologous recombination pathway. Depletion of TEAD4 or RAD51 increases DSBs at RAD51/TEAD4 common binding sites within super-enhancers and decreases expression of related genes, which are mostly oncogenes. Co-localization of RAD51 with transcription factors at super-enhancers occurs in various cell types, suggesting a broad phenomenon. Together, our findings uncover a coupling between transcription and repair mechanisms at oncogenic super-enhancers, to control the hyper-transcription of multiple cancer drivers.


Subject(s)
DNA Repair , Rad51 Recombinase/metabolism , Amino Acid Motifs , Binding Sites , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Replication , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Estradiol/pharmacology , Humans , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rad51 Recombinase/antagonists & inhibitors , Rad51 Recombinase/genetics , Radiation, Ionizing , TEA Domain Transcription Factors , Transcription Factor AP-1/chemistry , Transcription Factor AP-1/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Nat Genet ; 51(6): 1011-1023, 2019 06.
Article in English | MEDLINE | ID: mdl-31110352

ABSTRACT

It is not clear how spontaneous DNA double-strand breaks (DSBs) form and are processed in normal cells, and whether they predispose to cancer-associated translocations. We show that DSBs in normal mammary cells form upon release of paused RNA polymerase II (Pol II) at promoters, 5' splice sites and active enhancers, and are processed by end-joining in the absence of a canonical DNA-damage response. Logistic and causal-association models showed that Pol II pausing at long genes is the main predictor and determinant of DSBs. Damaged introns with paused Pol II-pS5, TOP2B and XRCC4 are enriched in translocation breakpoints, and map at topologically associating domain boundary-flanking regions showing high interaction frequencies with distal loci. Thus, in unperturbed growth conditions, release of paused Pol II at specific loci and chromatin territories favors DSB formation, leading to chromosomal translocations.


Subject(s)
DNA Breaks, Double-Stranded , Genetic Loci , Neoplasms/genetics , Neoplasms/metabolism , RNA Polymerase II/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , DNA Repair , Enhancer Elements, Genetic , Etoposide/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Genomics/methods , Introns , Neoplasms/pathology , Promoter Regions, Genetic , RNA Splice Sites , Topoisomerase Inhibitors/pharmacology , Transcription Initiation Site
13.
Genome Biol ; 20(1): 28, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30736820

ABSTRACT

BACKGROUND: Structural variants (SVs) are known to play important roles in a variety of cancers, but their origins and functional consequences are still poorly understood. Many SVs are thought to emerge from errors in the repair processes following DNA double strand breaks (DSBs). RESULTS: We used experimentally quantified DSB frequencies in cell lines with matched chromatin and sequence features to derive the first quantitative genome-wide models of DSB susceptibility. These models are accurate and provide novel insights into the mutational mechanisms generating DSBs. Models trained in one cell type can be successfully applied to others, but a substantial proportion of DSBs appear to reflect cell type-specific processes. Using model predictions as a proxy for susceptibility to DSBs in tumors, many SV-enriched regions appear to be poorly explained by selectively neutral mutational bias alone. A substantial number of these regions show unexpectedly high SV breakpoint frequencies given their predicted susceptibility to mutation and are therefore credible targets of positive selection in tumors. These putatively positively selected SV hotspots are enriched for genes previously shown to be oncogenic. In contrast, several hundred regions across the genome show unexpectedly low levels of SVs, given their relatively high susceptibility to mutation. These novel coldspot regions appear to be subject to purifying selection in tumors and are enriched for active promoters and enhancers. CONCLUSIONS: We conclude that models of DSB susceptibility offer a rigorous approach to the inference of SVs putatively subject to selection in tumors.


Subject(s)
DNA Breaks, Double-Stranded , Genomic Structural Variation , Models, Genetic , Neoplasms/genetics , Humans , K562 Cells , MCF-7 Cells , Regression Analysis
14.
Genes (Basel) ; 9(12)2018 Dec 14.
Article in English | MEDLINE | ID: mdl-30558210

ABSTRACT

DNA double-strand breaks (DSBs) jeopardize genome integrity and can-when repaired unfaithfully-give rise to structural rearrangements associated with cancer. Exogenous agents such as ionizing radiation or chemotherapy can invoke DSBs, but a vast amount of breakage arises during vital endogenous DNA transactions, such as replication and transcription. Additionally, chromatin looping involved in 3D genome organization and gene regulation is increasingly recognized as a possible contributor to DSB events. In this review, we first discuss insights into the mechanisms of endogenous DSB formation, showcasing the trade-off between essential DNA transactions and the intrinsic challenges that these processes impose on genomic integrity. In the second part, we highlight emerging methods for genome-wide profiling of DSBs, and discuss future directions of research that will help advance our understanding of genome-wide DSB formation and repair.

15.
Nat Genet ; 50(8): 1151-1160, 2018 08.
Article in English | MEDLINE | ID: mdl-29988121

ABSTRACT

Chromatin folding contributes to the regulation of genomic processes such as gene activity. Existing conformation capture methods characterize genome topology through analysis of pairwise chromatin contacts in populations of cells but cannot discern whether individual interactions occur simultaneously or competitively. Here we present multi-contact 4C (MC-4C), which applies Nanopore sequencing to study multi-way DNA conformations of individual alleles. MC-4C distinguishes cooperative from random and competing interactions and identifies previously missed structures in subpopulations of cells. We show that individual elements of the ß-globin superenhancer can aggregate into an enhancer hub that can simultaneously accommodate two genes. Neighboring chromatin domain loops can form rosette-like structures through collision of their CTCF-bound anchors, as seen most prominently in cells lacking the cohesin-unloading factor WAPL. Here, massive collision of CTCF-anchored chromatin loops is believed to reflect 'cohesin traffic jams'. Single-allele topology studies thus help us understand the mechanisms underlying genome folding and functioning.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic/genetics , Alleles , Animals , CCCTC-Binding Factor/genetics , Mice , Nucleic Acid Conformation , Regulatory Sequences, Nucleic Acid/genetics , beta-Globins/genetics
16.
Mol Cell ; 60(3): 460-74, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26593720

ABSTRACT

Upon recruitment to active enhancers and promoters, RNA polymerase II (Pol II) generates short non-coding transcripts of unclear function. The mechanisms that control the length and the amount of ncRNAs generated by cis-regulatory elements are largely unknown. Here, we show that the adaptor protein WDR82 and its associated complexes actively limit such non-coding transcription. WDR82 targets the SET1 H3K4 methyltransferases and the nuclear protein phosphatase 1 (PP1) complexes to the initiating Pol II. WDR82 and PP1 also interact with components of the transcriptional termination and RNA processing machineries. Depletion of WDR82, SET1, or the PP1 subunit required for its nuclear import caused distinct but overlapping transcription termination defects at highly expressed genes and active enhancers and promoters, thus enabling the increased synthesis of unusually long ncRNAs. These data indicate that transcription initiated from cis-regulatory elements is tightly coordinated with termination mechanisms that impose the synthesis of short RNAs.


Subject(s)
Cell Nucleus/metabolism , Enhancer Elements, Genetic/physiology , Promoter Regions, Genetic/physiology , RNA Polymerase II/metabolism , RNA, Untranslated/biosynthesis , Transcription Termination, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , Cell Nucleus/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , RNA Polymerase II/genetics , RNA, Untranslated/genetics
17.
Genome Biol ; 16: 154, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26257189

ABSTRACT

The hierarchical levels of genome architecture exert transcriptional control by tuning the accessibility and proximity of genes and regulatory elements. Here, we review current insights into the trans-acting factors that enable the genome to flexibly adopt different functionally relevant conformations.


Subject(s)
Chromatin/chemistry , Genome , Transcription, Genetic , Enhancer Elements, Genetic , Gene Expression Regulation , Promoter Regions, Genetic , Transcription Factors/metabolism
18.
FEBS Lett ; 589(20 Pt A): 2905-13, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-25957773

ABSTRACT

Pluripotent stem cells (PSCs) have the ability to self-renew and are capable of generating all embryonic germ layers (Evans and Kaufman, 1981; Thomson et al., 1998). PSCs can be isolated from early embryos or may be induced via overexpression of pluripotency transcription factors in differentiated cells (Takahashi and Yamanaka, 2006). As PSCs hold great promise for regenerative medicine, the mechanisms underlying pluripotency and induction thereof are studied intensively. Pluripotency is characterized by a unique transcriptional program that is in part controlled by an exceptionally plastic regulatory chromatin landscape. In recent years, 3D genome configuration has emerged as an important regulator of transcriptional control and cellular identity (Taddei et al., 2004 [4]; Lanctot et al., 2007 [5]; Gibcus and Dekker, 2013; Misteli, 2009 [7]). Here we provide an overview of recent findings on the 3D genome organization in PSCs and discuss its putative functional role in regulation of the pluripotent state.


Subject(s)
Genome, Human , Pluripotent Stem Cells/physiology , Animals , Cellular Reprogramming , Chromosomes/genetics , Epigenesis, Genetic , Humans , Nucleic Acid Conformation , Regulatory Sequences, Nucleic Acid
19.
Cell ; 157(2): 369-381, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24703711

ABSTRACT

Chromosomal rearrangements without gene fusions have been implicated in leukemogenesis by causing deregulation of proto-oncogenes via relocation of cryptic regulatory DNA elements. AML with inv(3)/t(3;3) is associated with aberrant expression of the stem-cell regulator EVI1. Applying functional genomics and genome-engineering, we demonstrate that both 3q rearrangements reposition a distal GATA2 enhancer to ectopically activate EVI1 and simultaneously confer GATA2 functional haploinsufficiency, previously identified as the cause of sporadic familial AML/MDS and MonoMac/Emberger syndromes. Genomic excision of the ectopic enhancer restored EVI1 silencing and led to growth inhibition and differentiation of AML cells, which could be replicated by pharmacologic BET inhibition. Our data show that structural rearrangements involving the chromosomal repositioning of a single enhancer can cause deregulation of two unrelated distal genes, with cancer as the outcome.


Subject(s)
Chromosomes, Human, Pair 3 , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Cell Line, Tumor , Chromosome Inversion , Humans , MDS1 and EVI1 Complex Locus Protein , Promoter Regions, Genetic , Transcriptional Activation , Translocation, Genetic
20.
Nature ; 501(7466): 227-31, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-23883933

ABSTRACT

It is becoming increasingly clear that the shape of the genome importantly influences transcription regulation. Pluripotent stem cells such as embryonic stem cells were recently shown to organize their chromosomes into topological domains that are largely invariant between cell types. Here we combine chromatin conformation capture technologies with chromatin factor binding data to demonstrate that inactive chromatin is unusually disorganized in pluripotent stem-cell nuclei. We show that gene promoters engage in contacts between topological domains in a largely tissue-independent manner, whereas enhancers have a more tissue-restricted interaction profile. Notably, genomic clusters of pluripotency factor binding sites find each other very efficiently, in a manner that is strictly pluripotent-stem-cell-specific, dependent on the presence of Oct4 and Nanog protein and inducible after artificial recruitment of Nanog to a selected chromosomal site. We conclude that pluripotent stem cells have a unique higher-order genome structure shaped by pluripotency factors. We speculate that this interactome enhances the robustness of the pluripotent state.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Chromosome Positioning , Genome/genetics , Imaging, Three-Dimensional , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Binding Sites , Cell Line , Chromatin/genetics , Chromatin Immunoprecipitation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Molecular Imaging , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Organ Specificity , Promoter Regions, Genetic , SOXB1 Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...