Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Genes Dev ; 36(1-2): 84-102, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34992147

ABSTRACT

The placenta is a highly evolved, specialized organ in mammals. It differs from other organs in that it functions only for fetal maintenance during gestation. Therefore, there must be intrinsic mechanisms that guarantee its unique functions. To address this question, we comprehensively analyzed epigenomic features of mouse trophoblast stem cells (TSCs). Our genome-wide, high-throughput analyses revealed that the TSC genome contains large-scale (>1-Mb) rigid heterochromatin architectures with a high degree of histone H3.1/3.2-H3K9me3 accumulation, which we termed TSC-defined highly heterochromatinized domains (THDs). Importantly, depletion of THDs by knockdown of CAF1, an H3.1/3.2 chaperone, resulted in down-regulation of TSC markers, such as Cdx2 and Elf5, and up-regulation of the pluripotent marker Oct3/4, indicating that THDs maintain the trophoblastic nature of TSCs. Furthermore, our nuclear transfer technique revealed that THDs are highly resistant to genomic reprogramming. However, when H3K9me3 was removed, the TSC genome was fully reprogrammed, giving rise to the first TSC cloned offspring. Interestingly, THD-like domains are also present in mouse and human placental cells in vivo, but not in other cell types. Thus, THDs are genomic architectures uniquely developed in placental lineage cells, which serve to protect them from fate reprogramming to stably maintain placental function.


Subject(s)
Histones , Trophoblasts , Animals , Cell Differentiation/genetics , Female , Histones/genetics , Histones/metabolism , Mammals , Mice , Placenta , Pregnancy , Stem Cells , Trophoblasts/metabolism
2.
Nucleic Acids Res ; 51(12): 6190-6207, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37178005

ABSTRACT

Heterochromatin is a key architectural feature of eukaryotic chromosomes critical for cell type-specific gene expression and genome stability. In the mammalian nucleus, heterochromatin segregates from transcriptionally active genomic regions and exists in large, condensed, and inactive nuclear compartments. However, the mechanisms underlying the spatial organization of heterochromatin need to be better understood. Histone H3 lysine 9 trimethylation (H3K9me3) and lysine 27 trimethylation (H3K27me3) are two major epigenetic modifications that enrich constitutive and facultative heterochromatin, respectively. Mammals have at least five H3K9 methyltransferases (SUV39H1, SUV39H2, SETDB1, G9a and GLP) and two H3K27 methyltransferases (EZH1 and EZH2). In this study, we addressed the role of H3K9 and H3K27 methylation in heterochromatin organization using a combination of mutant cells for five H3K9 methyltransferases and an EZH1/2 dual inhibitor, DS3201. We showed that H3K27me3, which is normally segregated from H3K9me3, was redistributed to regions targeted by H3K9me3 after the loss of H3K9 methylation and that the loss of both H3K9 and H3K27 methylation resulted in impaired condensation and spatial organization of heterochromatin. Our data demonstrate that the H3K27me3 pathway safeguards heterochromatin organization after the loss of H3K9 methylation in mammalian cells.


Subject(s)
Epigenesis, Genetic , Heterochromatin , Animals , Heterochromatin/genetics , Histones/metabolism , Lysine/metabolism , Mammals/genetics , Methylation , Histone Methyltransferases/metabolism
3.
J Cell Sci ; 135(2)2022 01 15.
Article in English | MEDLINE | ID: mdl-34888666

ABSTRACT

The organisation of chromatin is closely intertwined with biological activities of chromosome domains, including transcription and DNA replication status. Scaffold-attachment factor A (SAF-A), also known as heterogeneous nuclear ribonucleoprotein U (HNRNPU), contributes to the formation of open chromatin structure. Here, we demonstrate that SAF-A promotes the normal progression of DNA replication and enables resumption of replication after inhibition. We report that cells depleted of SAF-A show reduced origin licensing in G1 phase and, consequently, reduced origin activation frequency in S phase. Replication forks also progress less consistently in cells depleted of SAF-A, contributing to reduced DNA synthesis rate. Single-cell replication timing analysis revealed two distinct effects of SAF-A depletion: first, the boundaries between early- and late-replicating domains become more blurred; and second, SAF-A depletion causes replication timing changes that tend to bring regions of discordant domain compartmentalisation and replication timing into concordance. Associated with these defects, SAF-A-depleted cells show elevated formation of phosphorylated histone H2AX (γ-H2AX) and tend to enter quiescence. Overall, we find that SAF-A protein promotes robust DNA replication to ensure continuing cell proliferation.


Subject(s)
Chromosomes , DNA Replication , Chromatin/genetics , G1 Phase , Replication Origin/genetics , S Phase/genetics
4.
Biophys J ; 118(9): 2220-2228, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32191860

ABSTRACT

The one-dimensional information of genomic DNA is hierarchically packed inside the eukaryotic cell nucleus and organized in a three-dimensional (3D) space. Genome-wide chromosome conformation capture (Hi-C) methods have uncovered the 3D genome organization and revealed multiscale chromatin domains of compartments and topologically associating domains (TADs). Moreover, single-nucleosome live-cell imaging experiments have revealed the dynamic organization of chromatin domains caused by stochastic thermal fluctuations. However, the mechanism underlying the dynamic regulation of such hierarchical and structural chromatin units within the microscale thermal medium remains unclear. Microrheology is a way to measure dynamic viscoelastic properties coupling between thermal microenvironment and mechanical response. Here, we propose a new, to our knowledge, microrheology for Hi-C data to analyze the dynamic compliance property as a measure of rigidness and flexibility of genomic regions along with the time evolution. Our method allows the conversion of an Hi-C matrix into the spectrum of the dynamic rheological property along the genomic coordinate of a single chromosome. To demonstrate the power of the technique, we analyzed Hi-C data during the neural differentiation of mouse embryonic stem cells. We found that TAD boundaries behave as more rigid nodes than the intra-TAD regions. The spectrum clearly shows the dynamic viscoelasticity of chromatin domain formation at different timescales. Furthermore, we characterized the appearance of synchronous and liquid-like intercompartment interactions in differentiated cells. Together, our microrheology data derived from Hi-C data provide physical insights into the dynamics of the 3D genome organization.


Subject(s)
Chromatin , Chromosomes , Animals , Cell Nucleus , Chromatin/genetics , Chromosomes/genetics , DNA , Mice , Mouse Embryonic Stem Cells
5.
Nucleic Acids Res ; 42(3): 1524-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24174538

ABSTRACT

Three-dimensional genome organization is an important higher order transcription regulation mechanism that can be studied with the chromosome conformation capture techniques. Here, we combined chromatin organization analysis by chromosome conformation capture-carbon copy, computational modeling and epigenomics to achieve the first integrated view, through time, of a connection between chromatin state and its architecture. We used this approach to examine the chromatin dynamics of the HoxA cluster in a human myeloid leukemia cell line at various stages of differentiation. We found that cellular differentiation involves a transient activation of the 5'-end HoxA genes coinciding with a loss of contacts throughout the cluster, and by specific silencing at the 3'-end with H3K27 methylation. The 3D modeling of the data revealed an extensive reorganization of the cluster between the two previously reported topologically associated domains in differentiated cells. Our results support a model whereby silencing by polycomb group proteins and reconfiguration of CTCF interactions at a topologically associated domain boundary participate in changing the HoxA cluster topology, which compartmentalizes the genes following differentiation.


Subject(s)
Cell Differentiation/genetics , Chromatin/chemistry , Homeodomain Proteins/genetics , Multigene Family , Binding Sites , CCCTC-Binding Factor , Cell Line, Tumor , Chromatin/metabolism , Gene Expression Regulation , Histones/metabolism , Humans , Infant , Insulator Elements , Macrophages/cytology , Macrophages/metabolism , Male , Repressor Proteins/metabolism , Transcriptional Activation
6.
Biochim Biophys Acta ; 1819(5): 401-10, 2012 May.
Article in English | MEDLINE | ID: mdl-22207201

ABSTRACT

It has been known for some time that eukaryotic genomic DNA is packaged in the form of highly organized chromatin in vivo. This organization is important not only to reduce the length of chromosomes during interphase but also because it represents a type of higher-order genome regulation mechanism. Indeed, spatial chromatin architecture is known to be important for transcription, DNA replication and repair. Chromosome structure can be observed at different scales and studied with a variety of complementary techniques. For example, microscopy can provide single cell information while technologies such as the chromosome conformation capture (3C) method and its derivatives can yield higher-resolution data from cell populations. In this review, we report on the biological questions addressed with 3C and 3C-related techniques and what has been uncovered to date. We also explore what these methods may further reveal about the regulation of genomic DNA activities.


Subject(s)
Chromatin , Chromosomes , Microscopy/methods , Single-Cell Analysis/methods , Chromatin/metabolism , Chromatin/ultrastructure , Chromosomes/metabolism , Chromosomes/ultrastructure , DNA , DNA Repair/genetics , DNA Replication/genetics , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , Gene Expression Regulation , Genome , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure
7.
Nat Struct Mol Biol ; 30(8): 1224-1237, 2023 08.
Article in English | MEDLINE | ID: mdl-37563439

ABSTRACT

Chromosome-wide late replication is an enigmatic hallmark of the inactive X chromosome (Xi). How it is established and what it represents remains obscure. By single-cell DNA replication sequencing, here we show that the entire Xi is reorganized to replicate rapidly and uniformly in late S-phase during X-chromosome inactivation (XCI), reflecting its relatively uniform structure revealed by 4C-seq. Despite this uniformity, only a subset of the Xi became earlier replicating in SmcHD1-mutant cells. In the mutant, these domains protruded out of the Xi core, contacted each other and became transcriptionally reactivated. 4C-seq suggested that they constituted the outermost layer of the Xi even before XCI and were rich in escape genes. We propose that this default positioning forms the basis for their inherent heterochromatin instability in cells lacking the Xi-binding protein SmcHD1 or exhibiting XCI escape. These observations underscore the importance of 3D genome organization for heterochromatin stability and gene regulation.


Subject(s)
Heterochromatin , X Chromosome , Heterochromatin/genetics , X Chromosome/genetics , X Chromosome Inactivation , DNA Replication
8.
Curr Opin Genet Dev ; 73: 101898, 2022 04.
Article in English | MEDLINE | ID: mdl-35026526

ABSTRACT

In the mammalian cell nucleus, chromosomes are folded differently in interphase and mitosis. Interphase chromosomes are relatively decondensed and display at least two unique layers of higher-order organization: topologically associating domains (TADs) and cell-type-specific A/B compartments, which correlate well with early/late DNA replication timing (RT). In mitosis, these structures rapidly disappear but are gradually reconstructed during G1 phase, coincident with the establishment of the RT program. However, these structures also change dynamically during cell differentiation and reprogramming, and yet we are surprisingly ignorant about the relationship between their cell cycle dynamics and developmental dynamics. In this review, we summarize the recent findings on this topic, discuss how these two processes might be coordinated with each other and its potential significance.


Subject(s)
Chromosomes , Genome , Animals , Cell Cycle/genetics , Cell Nucleus/genetics , Chromatin/genetics , Chromosomes/genetics , Genome/genetics , Interphase/genetics , Mammals/genetics
9.
Nucleic Acids Res ; 37(3): 778-92, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074951

ABSTRACT

Mammalian tissue- and/or time-specific transcription is primarily regulated in a combinatorial fashion through interactions between a specific set of transcriptional regulatory factors (TRFs) and their cognate cis-regulatory elements located in the regulatory regions. In exploring the DNA regions and TRFs involved in combinatorial transcriptional regulation, we noted that individual knockdown of a set of human liver-enriched TRFs such as HNF1A, HNF3A, HNF3B, HNF3G and HNF4A resulted in perturbation of the expression of several single TRF genes, such as HNF1A, HNF3G and CEBPA genes. We thus searched the potential binding sites for these five TRFs in the highly conserved genomic regions around these three TRF genes and found several putative combinatorial regulatory regions. Chromatin immunoprecipitation analysis revealed that almost all of the putative regulatory DNA regions were bound by the TRFs as well as two coactivators (CBP and p300). The strong transcription-enhancing activity of the putative combinatorial regulatory region located downstream of the CEBPA gene was confirmed. EMSA demonstrated specific bindings of these HNFs to the target DNA region. Finally, co-transfection reporter assays with various combinations of expression vectors for these HNF genes demonstrated the transcriptional activation of the CEBPA gene in a combinatorial manner by these TRFs.


Subject(s)
Hepatocyte Nuclear Factors/metabolism , Regulatory Elements, Transcriptional , Transcriptional Activation , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Conserved Sequence , DNA/chemistry , DNA/metabolism , Electrophoretic Mobility Shift Assay , Hepatocyte Nuclear Factors/genetics , Humans , Liver/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , p300-CBP Transcription Factors/metabolism
10.
Nucleic Acids Res ; 37(4): 1049-60, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19129217

ABSTRACT

Transcriptional regulation by transcriptional regulatory factors (TRFs) of their target TRF genes is central to the control of gene expression. To study a static multi-tiered inter-TRF regulatory network in the human hepatoma cells, we have applied a Matrix RNAi approach in which siRNA knockdown and quantitative RT-PCR are used in combination on the same set of TRFs to determine their interdependencies. This approach focusing on several liver-enriched TRF families, each of which consists of structurally homologous members, revealed many significant regulatory relationships. These include the cross-talks between hepatocyte nuclear factors (HNFs) and the other TRF groups such as CCAAT/enhancer-binding proteins (CEBPs), retinoic acid receptors (RARs), retinoid receptors (RXRs) and RAR-related orphan receptors (RORs), which play key regulatory functions in human hepatocytes and liver. In addition, various multi-component regulatory motifs, which make up the complex inter-TRF regulatory network, were identified. A large part of the regulatory edges identified by the Matrix RNAi approach could be confirmed by chromatin immunoprecipitation. The resultant significant edges enabled us to depict the inter-TRF TRN forming an apparent regulatory hierarchy of (FOXA1, RXRA) --> TCF1 --> (HNF4A, ONECUT1) --> (RORC, CEBPA) as the main streamline.


Subject(s)
Gene Regulatory Networks , Liver/metabolism , RNA Interference , Transcription Factors/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
11.
Commun Biol ; 4(1): 571, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986449

ABSTRACT

Histone H3 lysine 9 dimethylation (H3K9me2) is a highly conserved silencing epigenetic mark. Chromatin marked with H3K9me2 forms large domains in mammalian cells and overlaps well with lamina-associated domains and the B compartment defined by Hi-C. However, the role of H3K9me2 in 3-dimensional (3D) genome organization remains unclear. Here, we investigated genome-wide H3K9me2 distribution, transcriptome, and 3D genome organization in mouse embryonic stem cells following the inhibition or depletion of H3K9 methyltransferases (MTases): G9a, GLP, SETDB1, SUV39H1, and SUV39H2. We show that H3K9me2 is regulated by all five MTases; however, H3K9me2 and transcription in the A and B compartments are regulated by different MTases. H3K9me2 in the A compartments is primarily regulated by G9a/GLP and SETDB1, while H3K9me2 in the B compartments is regulated by all five MTases. Furthermore, decreased H3K9me2 correlates with changes to more active compartmental state that accompanied transcriptional activation. Thus, H3K9me2 contributes to inactive compartment setting.


Subject(s)
Chromatin/metabolism , DNA Methylation , Fibroblasts/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Chromatin/chemistry , Chromatin/genetics , Fibroblasts/cytology , Genome , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/genetics , Lysine/chemistry , Lysine/genetics , Mice , Mouse Embryonic Stem Cells/cytology
12.
Nucleic Acids Res ; 36(10): 3443-54, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18445634

ABSTRACT

All-trans retinoic acid (ATRA) is a potent inducer of cell differentiation and growth arrest. Here, we investigated ATRA-induced regulatory cascades associated with growth arrest of the human hepatoma cell line HepG2. ATRA induced >2-fold changes in the expression of 402 genes including 55 linked to cell-cycle regulation, cell growth or apoptosis during 48 h treatment. Computational search predicted that 250 transcriptional regulatory factors (TRFs) could recognize the proximal upstream regions of any of the 55 genes. Expression of 61 TRF genes was significantly changed during ATRA incubation, providing many potential regulatory edges. We focused on six TRFs that could regulate many of the 55 genes and found a total of 160 potential edges in which the expression of each of the genes was changed later than the expression change of the corresponding regulator. RNAi knockdown of the selected TRFs caused perturbation of the respective potential targets. The genes showed an opposite regulation pattern by ATRA and specific siRNA treatments were selected as strong candidates for direct TRF targets. Finally, 36 transcriptional regulatory edges were validated by chromatin immunoprecipitation. These analyses enabled us to depict a part of the transcriptional regulatory cascades closely linked to ATRA-induced cell growth arrest.


Subject(s)
Gene Expression Regulation , Transcription, Genetic , Tretinoin/pharmacology , Binding Sites , CCAAT-Enhancer-Binding Protein-alpha/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Gene Regulatory Networks , Humans , Kinetics , Polymerase Chain Reaction , RNA Interference , Regulatory Elements, Transcriptional , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects
13.
Gigascience ; 9(1)2020 01 01.
Article in English | MEDLINE | ID: mdl-31919520

ABSTRACT

BACKGROUND: Hi-C is derived from chromosome conformation capture (3C) and targets chromatin contacts on a genomic scale. This method has also been used frequently in scaffolding nucleotide sequences obtained by de novo genome sequencing and assembly, in which the number of resultant sequences rarely converges to the chromosome number. Despite its prevalent use, the sample preparation methods for Hi-C have not been intensively discussed, especially from the standpoint of genome scaffolding. RESULTS: To gain insight into the best practice of Hi-C scaffolding, we performed a multifaceted methodological comparison using vertebrate samples and optimized various factors during sample preparation, sequencing, and computation. As a result, we identified several key factors that helped improve Hi-C scaffolding, including the choice and preparation of tissues, library preparation conditions, the choice of restriction enzyme(s), and the choice of scaffolding program and its usage. CONCLUSIONS: This study provides the first comparison of multiple sample preparation kits/protocols and computational programs for Hi-C scaffolding by an academic third party. We introduce a customized protocol designated "inexpensive and controllable Hi-C (iconHi-C) protocol," which incorporates the optimal conditions identified in this study, and demonstrate this technique on chromosome-scale genome sequences of the Chinese softshell turtle Pelodiscus sinensis.


Subject(s)
Chromatin/genetics , Chromosome Mapping , Chromosomes/genetics , Computational Biology/methods , Genomics/methods , Software , Animals , Chromosome Mapping/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , In Situ Hybridization , Turtles/genetics
14.
Nat Protoc ; 15(12): 4058-4100, 2020 12.
Article in English | MEDLINE | ID: mdl-33230331

ABSTRACT

Replication timing (RT) domains are stable units of chromosome structure that are regulated in the context of development and disease. Conventional genome-wide RT mapping methods require many S-phase cells for either the effective enrichment of replicating DNA through bromodeoxyuridine (BrdU) immunoprecipitation or the determination of copy-number differences during S-phase, which precludes their application to non-abundant cell types and single cells. Here, we provide a simple, cost-effective, and robust protocol for single-cell DNA replication sequencing (scRepli-seq). The scRepli-seq methodology relies on whole-genome amplification (WGA) of genomic DNA (gDNA) from single S-phase cells and next-generation sequencing (NGS)-based determination of copy-number differences that arise between replicated and unreplicated DNA. Haplotype-resolved scRepli-seq, which distinguishes pairs of homologous chromosomes within a single cell, is feasible by using single-nucleotide polymorphism (SNP)/indel information. We also provide computational pipelines for quality control, normalization, and binarization of the scRepli-seq data. The experimental portion of this protocol (before sequencing) takes 3 d.


Subject(s)
DNA Replication , Genomics/methods , Sequence Analysis, DNA/methods , Single-Cell Analysis/methods , Animals , Cell Line , Humans , S Phase/genetics
15.
Nat Genet ; 51(9): 1356-1368, 2019 09.
Article in English | MEDLINE | ID: mdl-31406346

ABSTRACT

In mammalian cells, chromosomes are partitioned into megabase-sized topologically associating domains (TADs). TADs can be in either A (active) or B (inactive) subnuclear compartments, which exhibit early and late replication timing (RT), respectively. Here, we show that A/B compartments change coordinately with RT changes genome wide during mouse embryonic stem cell (mESC) differentiation. While A to B compartment changes and early to late RT changes were temporally inseparable, B to A changes clearly preceded late to early RT changes and transcriptional activation. Compartments changed primarily by boundary shifting, altering the compartmentalization of TADs facing the A/B compartment interface, which was conserved during reprogramming and confirmed in individual cells by single-cell Repli-seq. Differentiating mESCs altered single-cell Repli-seq profiles gradually but uniformly, transiently resembling RT profiles of epiblast-derived stem cells (EpiSCs), suggesting that A/B compartments might also change gradually but uniformly toward a primed pluripotent state. These results provide insights into how megabase-scale chromosome organization changes in individual cells during differentiation.


Subject(s)
Cell Nucleus/metabolism , Chromatin Assembly and Disassembly , Chromosomes/genetics , DNA Replication , Mouse Embryonic Stem Cells/metabolism , Single-Cell Analysis/methods , Spatio-Temporal Analysis , Animals , Cell Differentiation , Cell Nucleus/genetics , Cells, Cultured , Cellular Reprogramming , Female , Genome , Male , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/cytology , Neurons/cytology , Neurons/metabolism
16.
Nat Genet ; 51(3): 529-540, 2019 03.
Article in English | MEDLINE | ID: mdl-30804559

ABSTRACT

Here, we report a single-cell DNA replication sequencing method, scRepli-seq, a genome-wide methodology that measures copy number differences between replicated and unreplicated DNA. Using scRepli-seq, we demonstrate that replication-domain organization is conserved among individual mouse embryonic stem cells (mESCs). Differentiated mESCs exhibited distinct profiles, which were also conserved among cells. Haplotype-resolved scRepli-seq revealed similar replication profiles of homologous autosomes, while the inactive X chromosome was clearly replicated later than its active counterpart. However, a small degree of cell-to-cell replication-timing heterogeneity was present, which was smallest at the beginning and the end of S phase. In addition, developmentally regulated domains were found to deviate from others and showed a higher degree of heterogeneity, thus suggesting a link to developmental plasticity. Moreover, allelic expression imbalance was found to strongly associate with replication-timing asynchrony. Our results form a foundation for single-cell-level understanding of DNA replication regulation and provide insights into three-dimensional genome organization.


Subject(s)
DNA Replication/genetics , DNA/genetics , Mammals/genetics , Animals , Cell Differentiation/genetics , Cell Line , DNA Copy Number Variations/genetics , DNA Replication Timing/genetics , Embryonic Stem Cells/physiology , Genome/genetics , Genome-Wide Association Study/methods , Genomic Instability/genetics , Humans , Mice , Mouse Embryonic Stem Cells/physiology , S Phase/genetics , X Chromosome/genetics
17.
Nat Commun ; 10(1): 3778, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439835

ABSTRACT

MCF7 cells acquire estrogen-independent proliferation after long-term estrogen deprivation (LTED), which recapitulates endocrine therapy resistance. LTED cells can become primed for apoptosis, but the underlying mechanism is largely unknown. We previously reported that Eleanor non-coding RNAs (ncRNAs) upregulate the ESR1 gene in LTED cells. Here, we show that Eleanors delineate the topologically associating domain (TAD) of the ESR1 locus in the active nuclear compartment of LTED cells. The TAD interacts with another transcriptionally active TAD, which is 42.9 Mb away from ESR1 and contains a gene encoding the apoptotic transcription factor FOXO3. Inhibition of a promoter-associated Eleanor suppresses all genes inside the Eleanor TAD and the long-range interaction between the two TADs, but keeps FOXO3 active to facilitate apoptosis in LTED cells. These data indicate a role of ncRNAs in chromatin domain regulation, which may underlie the apoptosis-prone nature of therapy-resistant breast cancer cells and could be good therapeutic targets.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , RNA, Untranslated/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Apoptosis/drug effects , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Binding Sites/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Chromatin/genetics , Chromatin/metabolism , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Genetic Loci/genetics , High-Throughput Nucleotide Sequencing , Humans , MCF-7 Cells , Promoter Regions, Genetic/genetics , Up-Regulation
18.
Methods Mol Biol ; 1861: 221-245, 2018.
Article in English | MEDLINE | ID: mdl-30218370

ABSTRACT

Recent advances in next-generation sequencing (NGS) and chromosome conformation capture (3C) analysis have led to the development of Hi-C, a genome-wide version of the 3C method. Hi-C has identified new levels of chromosome organization such as A/B compartments, topologically associating domains (TADs) as well as large megadomains on the inactive X chromosome, while allowing the identification of chromatin loops at the genome scale. Despite its powerfulness, Hi-C data analysis is much more involved compared to conventional NGS applications such as RNA-seq or ChIP-seq and requires many more steps. This presents a significant hurdle for those who wish to implement Hi-C technology into their laboratory. On the other hand, genomics data repository sites sometimes contain processed Hi-C data sets, allowing researchers to perform further analysis without the need for high-spec workstations and servers. In this chapter, we provide a detailed description on how to calculate A/B compartment profiles from processed Hi-C data on the autosomes and the active/inactive X chromosomes.


Subject(s)
Chromatin/ultrastructure , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Nucleic Acid Conformation , Software , Animals , Chromatin/metabolism , Computational Biology , DNA/chemistry , DNA/metabolism , Humans , Mice , Sequence Analysis, DNA/methods
19.
Nat Commun ; 9(1): 1387, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643333

ABSTRACT

Multicellular organisms consist of multiple cell types. The identity of these cells is primarily maintained by cell-type-specific gene expression programs; however, mechanisms that suppress these programs are poorly defined. Here we show that serum response factor (Srf), a transcription factor that is activated by various extracellular stimuli, can repress cell-type-specific genes and promote cellular reprogramming to pluripotency. Manipulations that decrease ß-actin monomer quantity result in the nuclear accumulation of Mkl1 and the activation of Srf, which downregulate cell-type-specific genes and alter the epigenetics of regulatory regions and chromatin organization. Mice overexpressing Srf exhibit various pathologies including an ulcerative colitis-like symptom and a metaplasia-like phenotype in the pancreas. Our results demonstrate an unexpected function of Srf via a mechanism by which extracellular stimuli actively destabilize cell identity and suggest Srf involvement in a wide range of diseases.


Subject(s)
Chromatin/chemistry , Colitis, Ulcerative/genetics , Induced Pluripotent Stem Cells/metabolism , Metaplasia/genetics , Neural Stem Cells/metabolism , Pancreas/metabolism , Serum Response Factor/genetics , Actins/genetics , Actins/metabolism , Animals , Cell Differentiation , Cellular Reprogramming/genetics , Chromatin/metabolism , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Induced Pluripotent Stem Cells/cytology , Male , Metaplasia/metabolism , Metaplasia/pathology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Pancreas/pathology , Serum Response Factor/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism
20.
Nucleic Acids Res ; 32(9): 2740-50, 2004.
Article in English | MEDLINE | ID: mdl-15148361

ABSTRACT

The identification of genes targeted by a specific transcription regulatory factor (TRF) is essential to our understanding of the regulatory mechanism of gene expression. We constructed a system for the comprehensive identification of genes directly regulated by a TRF. It includes a combination of perturbation of gene expression by RNA interference (RNAi) of the TRF, cDNA microarray analysis, computer searches for the putative TRF recognition sequences, and in vivo and in vitro TRF-DNA binding assays. Endogenous hepatocyte nuclear factor-1beta (HNF-1beta) mRNA was efficiently degraded by transfection of mouse hepatoma cells with short interfering RNAs. Expression profile analysis with 20 K mouse cDNA microarrays detected 243 genes whose expression levels were decreased by >50% upon RNAi of HNF-1beta. The upstream regions of the top 26 downregulated genes were searched for the HNF-1beta consensus recognition sequences leading to the extraction of 13 candidate genes. Finally, TRF-DNA binding assays identified five novel as well as three known HNF-1beta-regulated genes. In combination with quantitative real-time RT-PCR, the present system revealed the existence of a more expanded regulatory network among seven HNF family members, demonstrating its practicability to identify the TRF network as well as genes directly regulated by a specific TRF.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , RNA Interference , Transcription Factors/genetics , Transcription Factors/metabolism , 5' Flanking Region/genetics , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Consensus Sequence/genetics , DNA/genetics , DNA/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Hepatocyte Nuclear Factor 1-beta , Humans , Mice , Oligonucleotide Array Sequence Analysis , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Response Elements/genetics , Substrate Specificity , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL