Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Proc Natl Acad Sci U S A ; 117(4): 2020-2031, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31937660

ABSTRACT

The DNA-binding protein CCCTC-binding factor (CTCF) and the cohesin complex function together to shape chromatin architecture in mammalian cells, but the molecular details of this process remain unclear. Here, we demonstrate that a 79-aa region within the CTCF N terminus is essential for cohesin positioning at CTCF binding sites and chromatin loop formation. However, the N terminus of CTCF fused to artificial zinc fingers was not sufficient to redirect cohesin to non-CTCF binding sites, indicating a lack of an autonomously functioning domain in CTCF responsible for cohesin positioning. BORIS (CTCFL), a germline-specific paralog of CTCF, was unable to anchor cohesin to CTCF DNA binding sites. Furthermore, CTCF-BORIS chimeric constructs provided evidence that, besides the N terminus of CTCF, the first two CTCF zinc fingers, and likely the 3D geometry of CTCF-DNA complexes, are also involved in cohesin retention. Based on this knowledge, we were able to convert BORIS into CTCF with respect to cohesin positioning, thus providing additional molecular details of the ability of CTCF to retain cohesin. Taken together, our data provide insight into the process by which DNA-bound CTCF constrains cohesin movement to shape spatiotemporal genome organization.


Subject(s)
Breast Neoplasms/metabolism , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA, Neoplasm/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CCCTC-Binding Factor/genetics , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genome, Human , Humans , Protein Binding , Protein Domains , Tumor Cells, Cultured , Cohesins
2.
Nature ; 518(7539): 331-6, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25693564

ABSTRACT

Higher-order chromatin structure is emerging as an important regulator of gene expression. Although dynamic chromatin structures have been identified in the genome, the full scope of chromatin dynamics during mammalian development and lineage specification remains to be determined. By mapping genome-wide chromatin interactions in human embryonic stem (ES) cells and four human ES-cell-derived lineages, we uncover extensive chromatin reorganization during lineage specification. We observe that although self-associating chromatin domains are stable during differentiation, chromatin interactions both within and between domains change in a striking manner, altering 36% of active and inactive chromosomal compartments throughout the genome. By integrating chromatin interaction maps with haplotype-resolved epigenome and transcriptome data sets, we find widespread allelic bias in gene expression correlated with allele-biased chromatin states of linked promoters and distal enhancers. Our results therefore provide a global view of chromatin dynamics and a resource for studying long-range control of gene expression in distinct human cell lineages.


Subject(s)
Cell Differentiation , Chromatin Assembly and Disassembly , Chromatin/chemistry , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/genetics , Alleles , Allelic Imbalance/genetics , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Enhancer Elements, Genetic/genetics , Epigenomics , Gene Regulatory Networks , Humans , Promoter Regions, Genetic/genetics , Reproducibility of Results
3.
Nature ; 488(7409): 116-20, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22763441

ABSTRACT

The laboratory mouse is the most widely used mammalian model organism in biomedical research. The 2.6 × 10(9) bases of the mouse genome possess a high degree of conservation with the human genome, so a thorough annotation of the mouse genome will be of significant value to understanding the function of the human genome. So far, most of the functional sequences in the mouse genome have yet to be found, and the cis-regulatory sequences in particular are still poorly annotated. Comparative genomics has been a powerful tool for the discovery of these sequences, but on its own it cannot resolve their temporal and spatial functions. Recently, ChIP-Seq has been developed to identify cis-regulatory elements in the genomes of several organisms including humans, Drosophila melanogaster and Caenorhabditis elegans. Here we apply the same experimental approach to a diverse set of 19 tissues and cell types in the mouse to produce a map of nearly 300,000 murine cis-regulatory sequences. The annotated sequences add up to 11% of the mouse genome, and include more than 70% of conserved non-coding sequences. We define tissue-specific enhancers and identify potential transcription factors regulating gene expression in each tissue or cell type. Finally, we show that much of the mouse genome is organized into domains of coordinately regulated enhancers and promoters. Our results provide a resource for the annotation of functional elements in the mammalian genome and for the study of mechanisms regulating tissue-specific gene expression.


Subject(s)
Gene Expression Regulation/genetics , Genome/genetics , Mice/genetics , Physical Chromosome Mapping , Regulatory Sequences, Nucleic Acid/genetics , Acetylation , Animals , Chromatin/metabolism , Chromatin Immunoprecipitation , Conserved Sequence , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Male , Methylation , Mice, Inbred C57BL , Molecular Sequence Annotation , Nucleotide Motifs , Organ Specificity , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , Transcription Factors/metabolism
4.
Int J Mol Sci ; 19(12)2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30513694

ABSTRACT

CCCTC-binding factor (CTCF) is a conserved transcription factor that performs diverse roles in transcriptional regulation and chromatin architecture. Cancer genome sequencing reveals diverse acquired mutations in CTCF, which we have shown functions as a tumour suppressor gene. While CTCF is essential for embryonic development, little is known of its absolute requirement in somatic cells and the consequences of CTCF haploinsufficiency. We examined the consequences of CTCF depletion in immortalised human and mouse cells using shRNA knockdown and CRISPR/Cas9 genome editing as well as examined the growth and development of heterozygous Ctcf (Ctcf+/-) mice. We also analysed the impact of CTCF haploinsufficiency by examining gene expression changes in CTCF-altered endometrial carcinoma. Knockdown and CRISPR/Cas9-mediated editing of CTCF reduced the cellular growth and colony-forming ability of K562 cells. CTCF knockdown also induced cell cycle arrest and a pro-survival response to apoptotic insult. However, in p53 shRNA-immortalised Ctcf+/- MEFs we observed the opposite: increased cellular proliferation, colony formation, cell cycle progression, and decreased survival after apoptotic insult compared to wild-type MEFs. CRISPR/Cas9-mediated targeting in Ctcf+/- MEFs revealed a predominance of in-frame microdeletions in Ctcf in surviving clones, however protein expression could not be ablated. Examination of CTCF mutations in endometrial cancers showed locus-specific alterations in gene expression due to CTCF haploinsufficiency, in concert with downregulation of tumour suppressor genes and upregulation of estrogen-responsive genes. Depletion of CTCF expression imparts a dramatic negative effect on normal cell function. However, CTCF haploinsufficiency can have growth-promoting effects consistent with known cancer hallmarks in the presence of additional genetic hits. Our results confirm the absolute requirement for CTCF expression in somatic cells and provide definitive evidence of CTCF's role as a haploinsufficient tumour suppressor gene. CTCF genetic alterations in endometrial cancer indicate that gene dysregulation is a likely consequence of CTCF loss, contributing to, but not solely driving cancer growth.


Subject(s)
CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Survival/physiology , Endometrial Neoplasms/genetics , Gene Editing , Animals , CRISPR-Cas Systems , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival/genetics , Female , Haploinsufficiency/genetics , Haploinsufficiency/physiology , Humans , K562 Cells , Mice , RNA, Small Interfering/genetics
5.
Nature ; 459(7243): 108-12, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19295514

ABSTRACT

The human body is composed of diverse cell types with distinct functions. Although it is known that lineage specification depends on cell-specific gene expression, which in turn is driven by promoters, enhancers, insulators and other cis-regulatory DNA sequences for each gene, the relative roles of these regulatory elements in this process are not clear. We have previously developed a chromatin-immunoprecipitation-based microarray method (ChIP-chip) to locate promoters, enhancers and insulators in the human genome. Here we use the same approach to identify these elements in multiple cell types and investigate their roles in cell-type-specific gene expression. We observed that the chromatin state at promoters and CTCF-binding at insulators is largely invariant across diverse cell types. In contrast, enhancers are marked with highly cell-type-specific histone modification patterns, strongly correlate to cell-type-specific gene expression programs on a global scale, and are functionally active in a cell-type-specific manner. Our results define over 55,000 potential transcriptional enhancers in the human genome, significantly expanding the current catalogue of human enhancers and highlighting the role of these elements in cell-type-specific gene expression.


Subject(s)
Cell Physiological Phenomena , Gene Expression Regulation , Histones/metabolism , Transcription Factors/genetics , Binding Sites , Cell Line , Chromatin/genetics , Genome, Human/genetics , HeLa Cells , Humans , K562 Cells , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism
6.
Genome Biol ; 25(1): 40, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38297316

ABSTRACT

BACKGROUND: Pervasive usage of alternative promoters leads to the deregulation of gene expression in carcinogenesis and may drive the emergence of new genes in spermatogenesis. However, little is known regarding the mechanisms underpinning the activation of alternative promoters. RESULTS: Here we describe how alternative cancer-testis-specific transcription is activated. We show that intergenic and intronic CTCF binding sites, which are transcriptionally inert in normal somatic cells, could be epigenetically reprogrammed into active de novo promoters in germ and cancer cells. BORIS/CTCFL, the testis-specific paralog of the ubiquitously expressed CTCF, triggers the epigenetic reprogramming of CTCF sites into units of active transcription. BORIS binding initiates the recruitment of the chromatin remodeling factor, SRCAP, followed by the replacement of H2A histone with H2A.Z, resulting in a more relaxed chromatin state in the nucleosomes flanking the CTCF binding sites. The relaxation of chromatin around CTCF binding sites facilitates the recruitment of multiple additional transcription factors, thereby activating transcription from a given binding site. We demonstrate that the epigenetically reprogrammed CTCF binding sites can drive the expression of cancer-testis genes, long noncoding RNAs, retro-pseudogenes, and dormant transposable elements. CONCLUSIONS: Thus, BORIS functions as a transcription factor that epigenetically reprograms clustered CTCF binding sites into transcriptional start sites, promoting transcription from alternative promoters in both germ cells and cancer cells.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Male , Humans , DNA-Binding Proteins/metabolism , CCCTC-Binding Factor/metabolism , Transcription Factors/metabolism , Histones/metabolism , Chromatin , Binding Sites
7.
Front Mol Neurosci ; 16: 1185796, 2023.
Article in English | MEDLINE | ID: mdl-37324587

ABSTRACT

Introduction: CTCF-related disorder (CRD) is a neurodevelopmental disorder (NDD) caused by monoallelic pathogenic variants in CTCF. The first CTCF variants in CRD cases were documented in 2013. To date, 76 CTCF variants have been further described in the literature. In recent years, due to the increased application of next-generation sequencing (NGS), growing numbers of CTCF variants are being identified, and multiple genotype-phenotype databases cataloging such variants are emerging. Methods: In this study, we aimed to expand the genotypic spectrum of CRD, by cataloging NDD phenotypes associated with reported CTCF variants. Here, we systematically reviewed all known CTCF variants reported in case studies and large-scale exome sequencing cohorts. We also conducted a meta-analysis using public variant data from genotype-phenotype databases to identify additional CTCF variants, which we then curated and annotated. Results: From this combined approach, we report an additional 86 CTCF variants associated with NDD phenotypes that have not yet been described in the literature. Furthermore, we describe and explain inconsistencies in the quality of reported variants, which impairs the reuse of data for research of NDDs and other pathologies. Discussion: From this integrated analysis, we provide a comprehensive and annotated catalog of all currently known CTCF mutations associated with NDD phenotypes, to aid diagnostic applications, as well as translational and basic research.

8.
Nat Commun ; 12(1): 3846, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34158481

ABSTRACT

CTCF is a key organizer of the 3D genome. Its specialized paralog, BORIS, heterodimerizes with CTCF but is expressed only in male germ cells and in cancer states. Unexpectedly, BORIS-null mice have only minimal germ cell defects. To understand the CTCF-BORIS relationship, mouse models with varied CTCF and BORIS levels were generated. Whereas Ctcf+/+Boris+/+, Ctcf+/-Boris+/+, and Ctcf+/+Boris-/- males are fertile, Ctcf+/-Boris-/- (Compound Mutant; CM) males are sterile. Testes with combined depletion of both CTCF and BORIS show reduced size, defective meiotic recombination, increased apoptosis, and malformed spermatozoa. Although CM germ cells exhibit only 25% of CTCF WT expression, chromatin binding of CTCF is preferentially lost from CTCF-BORIS heterodimeric sites. Furthermore, CM testes lose the expression of a large number of spermatogenesis genes and gain the expression of developmentally inappropriate genes that are "toxic" to fertility. Thus, a combined action of CTCF and BORIS is required to both repress pre-meiotic genes and activate post-meiotic genes for a complete spermatogenesis program.


Subject(s)
CCCTC-Binding Factor/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Spermatogenesis/genetics , Testis/metabolism , Animals , CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Humans , Infertility, Male/genetics , Male , Meiosis/genetics , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding , RNA-Seq/methods , Recombination, Genetic , Spermatozoa/metabolism
9.
Mol Cell Biol ; 27(7): 2636-47, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17242189

ABSTRACT

Paternal deletion of the imprinting control region (ICR) KvDMR1 results in loss of expression of the Kcnq1ot1 noncoding RNA and derepression of flanking paternally silenced genes. Truncation of Kcnq1ot1 also results in the loss of imprinted expression of these genes in most cases, demonstrating a role for the RNA or its transcription in gene silencing. However, enhancer-blocking studies indicate that KvDMR1 also contains chromatin insulator or silencer activity. In this report we demonstrate by electrophoretic mobility shift assays and chromatin immunoprecipitation the existence of two CTCF binding sites within KvDMR1 that are occupied in vivo only on the unmethylated paternally derived allele. Methylation interference and mutagenesis allowed the precise mapping of protein-DNA contact sites for CTCF within KvDMR1. Using a luciferase reporter assay, we mapped the putative transcriptional promoter for Kcnq1ot1 upstream and to a site functionally separable from enhancer-blocking activity and CTCF binding sites. Luciferase reporter assays also suggest the presence of an additional cis-acting element in KvDMR1 upstream of the putative promoter that can function as an enhancer. These results suggest that the KvDMR1 ICR consists of multiple, independent cis-acting modules. Dissection of KvDMR1 into its functional components should help elucidate the mechanism of its function in vivo.


Subject(s)
DNA-Binding Proteins/metabolism , Genomic Imprinting , Repressor Proteins/metabolism , Alleles , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Chromatin Immunoprecipitation , CpG Islands , DNA Methylation , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , Female , Humans , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Repressor Proteins/genetics , Transcription Initiation Site
10.
Mol Cancer Res ; 17(10): 2051-2062, 2019 10.
Article in English | MEDLINE | ID: mdl-31292201

ABSTRACT

High-grade serous carcinoma (HGSC) is the most aggressive and predominant form of epithelial ovarian cancer and the leading cause of gynecologic cancer-related death. We have previously shown that CTCFL (also known as BORIS, Brother of the Regulator of Imprinted Sites) is expressed in most ovarian cancers, and is associated with global and promoter-specific DNA hypomethylation, advanced tumor stage, and poor prognosis. To explore its role in HGSC, we expressed BORIS in human fallopian tube secretory epithelial cells (FTSEC), the presumptive cells of origin for HGSC. BORIS-expressing cells exhibited increased motility and invasion, and BORIS expression was associated with alterations in several cancer-associated gene expression networks, including fatty acid metabolism, TNF signaling, cell migration, and ECM-receptor interactions. Importantly, GALNT14, a glycosyltransferase gene implicated in cancer cell migration and invasion, was highly induced by BORIS, and GALNT14 knockdown significantly abrogated BORIS-induced cell motility and invasion. In addition, in silico analyses provided evidence for BORIS and GALNT14 coexpression in several cancers. Finally, ChIP-seq demonstrated that expression of BORIS was associated with de novo and enhanced binding of CTCF at hundreds of loci, many of which correlated with activation of transcription at target genes, including GALNT14. Taken together, our data indicate that BORIS may promote cell motility and invasion in HGSC via upregulation of GALNT14, and suggests BORIS as a potential therapeutic target in this malignancy. IMPLICATIONS: These studies provide evidence that aberrant expression of BORIS may play a role in the progression to HGSC by enhancing the migratory and invasive properties of FTSEC.


Subject(s)
CCCTC-Binding Factor/genetics , DNA-Binding Proteins/genetics , N-Acetylgalactosaminyltransferases/genetics , Ovarian Neoplasms/genetics , CCCTC-Binding Factor/metabolism , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Fallopian Tubes/metabolism , Fallopian Tubes/pathology , Female , Humans , N-Acetylgalactosaminyltransferases/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , Transfection
11.
Mol Cell Biol ; 25(4): 1511-25, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15684400

ABSTRACT

The murine Igh locus has a 3' regulatory region (3' RR) containing four enhancers (hs3A, hs1,2, hs3B, and hs4) at DNase I-hypersensitive sites. The 3' RR exerts long-range effects on class switch recombination (CSR) to several isotypes through its control of germ line transcription. By measuring levels of acetylated histones H3 and H4 and of dimethylated H3 (K4) with chromatin immunoprecipitation assays, we found that early in B-cell development, chromatin encompassing the enhancers of the 3' RR began to attain stepwise modifications typical of an open conformation. The hs4 enhancer was associated with active chromatin initially in pro- and pre-B cells and then together with hs3A, hs1,2, and hs3B in B and plasma cells. Histone modifications were similar in resting splenic B cells and in splenic B cells induced by lipopolysaccharide to undergo CSR. From the pro-B-cell stage onward, the approximately 11-kb region immediately downstream of hs4 displayed H3 and H4 modifications indicative of open chromatin. This region contained newly identified DNase I-hypersensitive sites and several CTCF target sites, some of which were occupied in vivo in a developmentally regulated manner. The open chromatin environment of the extended 3' RR in mature B cells was flanked by regions associated with dimethylated K9 of histone H3. Together, these data suggest that 3' RR elements are located within a specific chromatin subdomain that contains CTCF binding sites and developmentally regulated modules.


Subject(s)
3' Flanking Region/genetics , B-Lymphocytes/metabolism , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Histones/metabolism , Repressor Proteins/metabolism , Acetylation/drug effects , Animals , B-Lymphocytes/immunology , Bone Marrow/immunology , Bone Marrow/metabolism , CCCTC-Binding Factor , Chromatin/genetics , Chromatin/immunology , DNA Primers/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic/genetics , Histones/genetics , Histones/immunology , Immunoglobulin Class Switching/drug effects , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Lipopolysaccharides/pharmacology , Locus Control Region/genetics , Mice , Repressor Proteins/genetics , Repressor Proteins/immunology , Spleen/immunology , Spleen/metabolism , Transcription, Genetic/genetics
12.
Mol Cell Biol ; 25(24): 11184-90, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314537

ABSTRACT

Imprinted methylation of the paternal Rasgrf1 allele in mice occurs at a differentially methylated domain (DMD) 30 kbp 5' of the promoter. A repeated sequence 3' of the DMD regulates imprinted methylation, which is required for imprinted expression. Here we identify the mechanism by which methylation controls imprinting. The DMD is an enhancer blocker that binds CTCF in a methylation-sensitive manner. CTCF bound to the unmethylated maternal allele silences expression. CTCF binding to the paternal allele is prevented by repeat-mediated methylation, allowing expression. Optimal in vitro enhancer-blocking activity requires CTCF binding sites. The enhancer blocker can be bypassed in vivo and imprinting abolished by placing an extra enhancer proximal to the promoter. Together, the repeats and the DMD constitute a binary switch that regulates Rasgrf1 imprinting.


Subject(s)
Carrier Proteins/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Genomic Imprinting , Repressor Proteins/metabolism , Animals , CCCTC-Binding Factor , GTPase-Activating Proteins , Mice , Models, Genetic
13.
Nucleus ; 9(1): 33-41, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29077515

ABSTRACT

CCCTC-binding factor (CTCF) is a conserved, essential regulator of chromatin architecture containing a unique array of 11 zinc fingers (ZFs). Gene duplication and sequence divergence during early amniote evolution generated the CTCF paralog Brother Of the Regulator of Imprinted Sites (BORIS), which has a DNA binding specificity identical to that of CTCF but divergent N- and C-termini. While healthy somatic tissues express only CTCF, CTCF and BORIS are normally co-expressed in meiotic and post-meiotic germ cells, and aberrant activation of BORIS occurs in tumors and some cancer cell lines. This has led to a model in which CTCF and BORIS compete for binding to some but not all genomic target sites; however, regulation of CTCF and BORIS genomic co-occupancy is not well understood. We recently addressed this issue, finding evidence for two major classes of CTCF target sequences, some of which contain single CTCF target sites (1xCTSes) and others containing two adjacent CTCF motifs (2xCTSes). The functional and chromatin structural features of 2xCTSes are distinct from those of 1xCTS-containing regions bound by a CTCF monomer. We suggest that these previously overlooked classes of CTCF binding regions may have different roles in regulating diverse chromatin-based phenomena, and may impact our understanding of heritable epigenetic regulation in cancer cells and normal germ cells.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Chromatin/chemistry , Chromatin/metabolism , Humans , Promoter Regions, Genetic/genetics
14.
Cancer Res ; 65(17): 7751-62, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140943

ABSTRACT

Brother of the Regulator of Imprinted Sites (BORIS) is a mammalian CTCF paralog with the same central 11Zn fingers (11ZF) that mediate specific interactions with varying approximately 50-bp target sites. Regulated in vivo occupancy of such sites may yield structurally and functionally distinct CTCF/DNA complexes involved in various aspects of gene regulation, including epigenetic control of gene imprinting and X chromosome inactivation. The latter functions are mediated by meCpG-sensitive 11ZF binding. Because CTCF is normally present in all somatic cells, whereas BORIS is active only in CTCF- and 5-methylcytosine-deficient adult male germ cells, switching DNA occupancy from CTCF to BORIS was suggested to regulate site specificity and timing of epigenetic reprogramming. In addition to 11ZF-binding paternal imprinting control regions, cancer-testis gene promoters also undergo remethylation during CTCF/BORIS switching in germ cells. Only promoters of cancer testis genes are normally silenced in all somatic cells but activated during spermatogenesis when demethylated in BORIS-positive germ cells and are found aberrantly derepressed in various tumors. We show here that BORIS is also expressed in multiple cancers and is thus itself a cancer-testis gene and that conditional expression of BORIS in normal fibroblasts activates cancer-testis genes selectively. We tested if replacement of CTCF by BORIS on regulatory DNA occurs in vivo on activation of a prototype cancer-testis gene, MAGE-A1. Transition from a hypermethylated/silenced to a hypomethylated/activated status induced in normal cells by 5-aza-2'-deoxycytidine (5-azadC) was mimicked by conditional input of BORIS and is associated with complete switching from CTCF to BORIS occupancy at a single 11ZF target. This site manifested a novel type of CTCF/BORIS 11ZF binding insensitive to CpG methylation. Whereas 5-azadC induction of BORIS takes only few hours, derepression of MAGE-A1 occurred 1 to 2 days later, suggesting that BORIS mediates cancer-testis gene activation by 5-azadC. Indeed, infection of normal fibroblasts with anti-BORIS short hairpin RNA retroviruses before treatment with 5-azadC blocked reactivation of MAGE-A1. We suggest that BORIS is likely tethering epigenetic machinery to a novel class of CTCF/BORIS 11ZF target sequences that mediate induction of cancer-testis genes.


Subject(s)
DNA Methylation , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Proteins/genetics , Animals , Antigens, Neoplasm , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Line, Tumor , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Decitabine , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Genetic Vectors/genetics , Humans , Immunohistochemistry , Melanoma-Specific Antigens , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Nucleic Acid Conformation , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , Retroviridae/genetics , Transcriptional Activation , Transfection
15.
Cancer Res ; 65(17): 7763-74, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140944

ABSTRACT

Regulatory sequences recognized by the unique pair of paralogous factors, CTCF and BORIS, have been implicated in epigenetic regulation of imprinting and X chromosome inactivation. Lung cancers exhibit genome-wide demethylation associated with derepression of a specific class of genes encoding cancer-testis (CT) antigens such as NY-ESO-1. CT genes are normally expressed in BORIS-positive male germ cells deficient in CTCF and meCpG contents, but are strictly silenced in somatic cells. The present study was undertaken to ascertain if aberrant activation of BORIS contributes to derepression of NY-ESO-1 during pulmonary carcinogenesis. Preliminary experiments indicated that NY-ESO-1 expression coincided with derepression of BORIS in cultured lung cancer cells. Quantitative reverse transcription-PCR analysis revealed robust, coincident induction of BORIS and NY-ESO-1 expression in lung cancer cells, but not normal human bronchial epithelial cells following 5-aza-2'-deoxycytidine (5-azadC), Depsipeptide FK228 (DP), or sequential 5-azadC/DP exposure under clinically relevant conditions. Bisulfite sequencing, methylation-specific PCR, and chromatin immunoprecipitation (ChIP) experiments showed that induction of BORIS coincided with direct modulation of chromatin structure within a CpG island in the 5'-flanking noncoding region of this gene. Cotransfection experiments using promoter-reporter constructs confirmed that BORIS modulates NY-ESO-1 expression in lung cancer cells. Gel shift and ChIP experiments revealed a novel CTCF/BORIS-binding site in the NY-ESO-1 promoter, which unlike such sites in the H19-imprinting control region and X chromosome, is insensitive to CpG methylation in vitro. In vivo occupancy of this site by CTCF was associated with silencing of the NY-ESO-1 promoter, whereas switching from CTCF to BORIS occupancy coincided with derepression of NY-ESO-1. Collectively, these data indicate that reciprocal binding of CTCF and BORIS to the NY-ESO-1 promoter mediates epigenetic regulation of this CT gene in lung cancer cells, and suggest that induction of BORIS may be a novel strategy to augment immunogenicity of pulmonary carcinomas.


Subject(s)
Antigens, Neoplasm/genetics , DNA-Binding Proteins/metabolism , Lung Neoplasms/genetics , Membrane Proteins/genetics , Repressor Proteins/metabolism , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/metabolism , Base Sequence , CCCTC-Binding Factor , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Histones/metabolism , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sulfites/pharmacology
17.
Sci Rep ; 7: 41279, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28145452

ABSTRACT

Despite sharing the same sequence specificity in vitro and in vivo, CCCTC-binding factor (CTCF) and its paralog brother of the regulator of imprinted sites (BORIS) are simultaneously expressed in germ cells. Recently, ChIP-seq analysis revealed two classes of CTCF/BORIS-bound regions: single CTCF target sites (1xCTSes) that are bound by CTCF alone (CTCF-only) or double CTCF target sites (2xCTSes) simultaneously bound by CTCF and BORIS (CTCF&BORIS) or BORIS alone (BORIS-only) in germ cells and in BORIS-positive somatic cancer cells. BORIS-bound regions (CTCF&BORIS and BORIS-only sites) are, on average, enriched for RNA polymerase II (RNAPII) binding and histone retention in mature spermatozoa relative to CTCF-only sites, but little else is known about them. We show that subsets of CTCF&BORIS and BORIS-only sites are occupied by several testis-specific transcriptional regulators (TSTRs) and associated with highly expressed germ cell-specific genes and histone retention in mature spermatozoa. We also demonstrate a physical interaction between BORIS and one of the analyzed TSTRs, TATA-binding protein (TBP)-associated factor 7-like (TAF7L). Our data suggest that CTCF and BORIS cooperate with additional TSTRs to regulate gene expression in developing male gametes and histone retention in mature spermatozoa, potentially priming certain regions of the genome for rapid activation following fertilization.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Spermatozoa/metabolism , Testis/metabolism , Transcription, Genetic , Animals , Gene Expression Regulation , HEK293 Cells , Histones/metabolism , Humans , Male , Mice , Nuclear Proteins/metabolism , Organ Specificity , Protein Binding , Regulatory Factor X Transcription Factors/metabolism , Spermatids/metabolism , Spermatozoa/cytology , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcription Factors/metabolism
18.
Gene ; 375: 26-36, 2006 Jun 21.
Article in English | MEDLINE | ID: mdl-16647825

ABSTRACT

CTCF is a nuclear phosphoprotein capable of using different subsets of its 11 Zn fingers (ZF) for sequence-specific binding to many dissimilar DNA CTCF-target sites. Such sites were identified in the genomic DNA of various multicellular organisms, in which the CTCF gene was cloned, including insects, birds, rodents, and primates. CTCF/DNA-complexes formed in vivo with different 50-bp-long sequences mediate diverse functions such as positive and negative regulation of promoters, and organization of all known enhancer-blocking elements ("chromatin insulators") including constitutive and epigenetically regulated elements. Abnormal functions of certain CTCF sites are implicated in cancer and in epigenetic syndromes such as BWS and skewed X-inactivation. We describe here the cloning and characterization of the CTCF cDNA and promoter region from zebrafish, a valuable vertebrate model organism. The full-length zebrafish CTCF cDNA clone is 4244 bp in length with an open reading frame (ORF) of 2391 bp that encodes 797 amino acids. The zebrafish CTCF amino acid sequence shows high identity (up to 98% in the zinc finger region) with human CTCF, and perfect conservation of exon-intron organization. Southern blot analyses indicated that the zebrafish genome contains a single copy of the CTCF gene. In situ hybridization revealed the presence of zebrafish CTCF transcripts in all early stages of embryogenesis. Transfection assays with luciferase reporter-constructs identified a core promoter region within 146 bp immediately upstream of the transcriptional start site of zebrafish CTCF that is located at a highly conserved YY1/Initiator element.


Subject(s)
DNA-Binding Proteins/genetics , Evolution, Molecular , Promoter Regions, Genetic , Repressor Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , CCCTC-Binding Factor , Cloning, Molecular , DNA Primers , Humans , In Situ Hybridization , Molecular Sequence Data , Plasmids , Sequence Homology, Amino Acid , Transcription, Genetic
19.
Cancer Res ; 62(1): 48-52, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11782357

ABSTRACT

CTCF is a widely expressed 11-zinc finger (ZF) transcription factor that is involved in different aspects of gene regulation including promoter activation or repression, hormone-responsive gene silencing, methylation-dependent chromatin insulation, and genomic imprinting. Because CTCF targets include oncogenes and tumor suppressor genes, we screened over 100 human tumor samples for mutations that might disrupt CTCF activity. We did not observe any CTCF mutations leading to truncations/premature stops. Rather, in breast, prostate, and Wilms' tumors, we observed four different CTCF somatic missense mutations involving amino acids within the ZF domain. Each ZF mutation abrogated CTCF binding to a subset of target sites within the promoters/insulators of certain genes involved in regulating cell proliferation but did not alter binding to the regulatory sequences of other genes. These observations suggest that CTCF may represent a novel tumor suppressor gene that displays tumor-specific "change of function" rather than complete "loss of function."


Subject(s)
DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mutation, Missense , Repressor Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers/genetics , Amino Acid Sequence , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Female , Genes, Tumor Suppressor , Globins/genetics , Humans , Male , Molecular Sequence Data , Muramidase/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Conformation , Substrate Specificity , Wilms Tumor/genetics , Wilms Tumor/metabolism
20.
Genome Biol ; 16: 161, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26268681

ABSTRACT

BACKGROUND: CTCF and BORIS (CTCFL), two paralogous mammalian proteins sharing nearly identical DNA binding domains, are thought to function in a mutually exclusive manner in DNA binding and transcriptional regulation. RESULTS: Here we show that these two proteins co-occupy a specific subset of regulatory elements consisting of clustered CTCF binding motifs (termed 2xCTSes). BORIS occupancy at 2xCTSes is largely invariant in BORIS-positive cancer cells, with the genomic pattern recapitulating the germline-specific BORIS binding to chromatin. In contrast to the single-motif CTCF target sites (1xCTSes), the 2xCTS elements are preferentially found at active promoters and enhancers, both in cancer and germ cells. 2xCTSes are also enriched in genomic regions that escape histone to protamine replacement in human and mouse sperm. Depletion of the BORIS gene leads to altered transcription of a large number of genes and the differentiation of K562 cells, while the ectopic expression of this CTCF paralog leads to specific changes in transcription in MCF7 cells. CONCLUSIONS: We discover two functionally and structurally different classes of CTCF binding regions, 2xCTSes and 1xCTSes, revealed by their predisposition to bind BORIS. We propose that 2xCTSes play key roles in the transcriptional program of cancer and germ cells.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Promoter Regions, Genetic , Repressor Proteins/metabolism , Animals , Binding Sites , CCCTC-Binding Factor , Cell Line , Chromatin/chemistry , DNA/chemistry , DNA/metabolism , Genome , Humans , K562 Cells , Male , Mice , Neoplasms/genetics , Nucleotide Motifs , Protein Binding , Spermatids/metabolism , Spermatozoa/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL