Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Genet ; 15: 1384167, 2024.
Article in English | MEDLINE | ID: mdl-38706797

ABSTRACT

Background: Cis-regulatory elements (CREs) play crucial roles in regulating gene expression during erythroid cell differentiation. Genome-wide erythroid-specific CREs have not been characterized in chicken erythroid cells, which is an organism model used to study epigenetic regulation during erythropoiesis. Methods: Analysis of public genome-wide accessibility (ATAC-seq) maps, along with transcription factor (TF) motif analysis, CTCF, and RNA Pol II occupancy, as well as transcriptome analysis in fibroblasts and erythroid HD3 cells, were used to characterize erythroid-specific CREs. An α-globin CRE was identified, and its regulatory activity was validated in vitro and in vivo by luciferase activity and genome-editing assays in HD3 cells, respectively. Additionally, circular chromosome conformation capture (UMI-4C) assays were used to distinguish its role in structuring the α-globin domain in erythroid chicken cells. Results: Erythroid-specific CREs displayed occupancy by erythroid TF binding motifs, CTCF, and RNA Pol II, as well as an association with genes involved in hematopoiesis and cell differentiation. An α-globin CRE, referred to as CRE-2, was identified as exhibiting enhancer activity over αD and αA genes in vitro and in vivo. Induction of terminal erythroid differentiation showed that α-globin CRE-2 is required for the induction of αD and αA. Analysis of TF binding motifs at α-globin CRE-2 shows apparent regulation mediated by GATA-1, YY1, and CTCF binding. Conclusion: Our findings demonstrate that cell-specific CREs constitute a key mechanism that contributes to the fine-tuning gene regulation of erythroid cell differentiation and provide insights into the annotation and characterization of CREs in chicken cells.

2.
Nat Genet ; 54(2): 202-212, 2022 02.
Article in English | MEDLINE | ID: mdl-35145304

ABSTRACT

CCCTC-binding factor (CTCF) is critical to three-dimensional genome organization. Upon differentiation, CTCF insulates active and repressed genes within Hox gene clusters. We conducted a genome-wide CRISPR knockout (KO) screen to identify genes required for CTCF-boundary activity at the HoxA cluster, complemented by biochemical approaches. Among the candidates, we identified Myc-associated zinc-finger protein (MAZ) as a cofactor in CTCF insulation. MAZ colocalizes with CTCF at chromatin borders and, similar to CTCF, interacts with the cohesin subunit RAD21. MAZ KO disrupts gene expression and local contacts within topologically associating domains. Similar to CTCF motif deletions, MAZ motif deletions lead to derepression of posterior Hox genes immediately after CTCF boundaries upon differentiation, giving rise to homeotic transformations in mouse. Thus, MAZ is a factor contributing to appropriate insulation, gene expression and genomic architecture during development.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Genes, Homeobox , Homeodomain Proteins/genetics , Transcription Factors/metabolism , Animals , CCCTC-Binding Factor/chemistry , CCCTC-Binding Factor/genetics , CRISPR-Cas Systems , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Gene Editing , Gene Expression , Gene Expression Regulation, Developmental , Mice , Transcription Factors/chemistry , Transcription Factors/genetics
3.
Clin Cancer Res ; 27(20): 5669-5680, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34433651

ABSTRACT

PURPOSE: The epigenetic mechanisms involved in transcriptional regulation leading to malignant phenotype in gliomas remains poorly understood. Topoisomerase IIB (TOP2B), an enzyme that decoils and releases torsional forces in DNA, is overexpressed in a subset of gliomas. Therefore, we investigated its role in epigenetic regulation in these tumors. EXPERIMENTAL DESIGN: To investigate the role of TOP2B in epigenetic regulation in gliomas, we performed paired chromatin immunoprecipitation sequencing for TOP2B and RNA-sequencing analysis of glioma cell lines with and without TOP2B inhibition and in human glioma specimens. These experiments were complemented with assay for transposase-accessible chromatin using sequencing, gene silencing, and mouse xenograft experiments to investigate the function of TOP2B and its role in glioma phenotypes. RESULTS: We discovered that TOP2B modulates transcription of multiple oncogenes in human gliomas. TOP2B regulated transcription only at sites where it was enzymatically active, but not at all native binding sites. In particular, TOP2B activity localized in enhancers, promoters, and introns of PDGFRA and MYC, facilitating their expression. TOP2B levels and genomic localization was associated with PDGFRA and MYC expression across glioma specimens, which was not seen in nontumoral human brain tissue. In vivo, TOP2B knockdown of human glioma intracranial implants prolonged survival and downregulated PDGFRA. CONCLUSIONS: Our results indicate that TOP2B activity exerts a pleiotropic role in transcriptional regulation of oncogenes in a subset of gliomas promoting a proliferative phenotype.


Subject(s)
Brain Neoplasms/genetics , DNA Topoisomerases, Type II/physiology , Epigenesis, Genetic/physiology , Glioma/genetics , Introns/physiology , Oncogenes/physiology , Poly-ADP-Ribose Binding Proteins/physiology , Promoter Regions, Genetic/physiology , Animals , Brain Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Glioma/enzymology , Humans , Mice
4.
Oncogene ; 39(27): 5068-5081, 2020 07.
Article in English | MEDLINE | ID: mdl-32528131

ABSTRACT

Topoisomerase II poisons are one of the most common class of chemotherapeutics used in cancer. We and others had shown that a subset of glioblastomas, the most malignant of all primary brain tumors in adults, is responsive to TOP2 poisons. To identify genes that confer susceptibility to this drug in gliomas, we performed a genome-scale CRISPR knockout screen with etoposide. Genes involved in protein synthesis and DNA damage were implicated in etoposide susceptibility. To define potential biomarkers for TOP2 poisons, CRISPR hits were overlapped with genes whose expression correlates with susceptibility to this drug across glioma cell lines, revealing ribosomal protein subunit RPS11, 16, and 18 as putative biomarkers for response to TOP2 poisons. Loss of RPS11 led to resistance to etoposide and doxorubicin and impaired the induction of proapoptotic gene APAF1 following treatment. The expression of these ribosomal subunits was also associated with susceptibility to TOP2 poisons across cell lines from gliomas and multiple other cancers.


Subject(s)
Brain Neoplasms/drug therapy , Etoposide/pharmacology , Glioblastoma/drug therapy , Ribosomal Proteins/metabolism , Topoisomerase II Inhibitors/pharmacology , Apoptotic Protease-Activating Factor 1/metabolism , Brain Neoplasms/genetics , CRISPR-Cas Systems , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA Topoisomerases, Type II/metabolism , Doxorubicin/pharmacology , Gene Knockout Techniques , Glioblastoma/genetics , Humans
5.
Clin Cancer Res ; 26(16): 4390-4401, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32430477

ABSTRACT

PURPOSE: Cancer immunoediting shapes tumor progression by the selection of tumor cell variants that can evade immune recognition. Given the immune evasion and intratumor heterogeneity characteristic of gliomas, we hypothesized that CD8+ T cells mediate immunoediting in these tumors. EXPERIMENTAL DESIGN: We developed retrovirus-induced PDGF+ Pten -/- murine gliomas and evaluated glioma progression and tumor immunogenicity in the absence of CD8+ T cells by depleting this immune cell population. Furthermore, we characterized the genomic alterations present in gliomas that developed in the presence and absence of CD8+ T cells. RESULTS: Upon transplantation, gliomas that developed in the absence of CD8+ T cells engrafted poorly in recipients with intact immunity but engrafted well in those with CD8+ T-cell depletion. In contrast, gliomas that developed under pressure from CD8+ T cells were able to fully engraft in both CD8+ T-cell-depleted mice and immunocompetent mice. Remarkably, gliomas developed in the absence of CD8+ T cells exhibited increased aneuploidy, MAPK pathway signaling, gene fusions, and macrophage/microglial infiltration, and showed a proinflammatory phenotype. MAPK activation correlated with macrophage/microglia recruitment in this model and in the human disease. CONCLUSIONS: Our studies indicate that, in these tumor models, CD8+ T cells influence glioma oncogenic pathways, tumor genotype, and immunogenicity. This suggests immunoediting of immunogenic tumor clones through their negative selection by CD8+ T cells during glioma formation.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Immune Evasion/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Glioma/genetics , Glioma/pathology , Humans , Macrophages/immunology , Macrophages/pathology , Mice , Microglia/immunology , Microglia/pathology , T-Lymphocytes/pathology
6.
Int J Cancer ; 146(8): 2218-2228, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31443114

ABSTRACT

Glioblastoma (GBM) is one of the most aggressive primary brain tumors with frequent recurrences following the standard methods of treatment-temozolomide (TMZ), ionizing radiation and surgical resection. The objective of our study was to investigate GBM resistance mediated via MMP14 (matrix metalloproteinase 14). We used multiple PDX GBM models and established glioma cell lines to characterize expression and subcellular localization of MMP14 after TMZ treatment. We performed a Kiloplex ELISA-based array to evaluate changes in cellular proteins induced by MMP14 expression and translocation. Lastly, we conducted functional and mechanistic studies to elucidate the role of DLL4 (delta-like canonical notch ligand 4) in regulation of glioma stemness, particularly in the context of its relationship to MMP14. We detected that TMZ treatment promotes nuclear translocation of MMP14 followed by extracellular release of DLL4. DLL4 in turn stimulates cleavage of Notch3, its nuclear translocation and induction of sphering capacity and stemness.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Intracellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 14/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/drug effects , Receptor, Notch3/metabolism , Temozolomide/pharmacology , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Early Growth Response Protein 1/metabolism , Fibroblast Growth Factors/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Matrix Metalloproteinase 14/biosynthesis , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 26(2): 477-486, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31831565

ABSTRACT

PURPOSE: Paclitaxel shows little benefit in the treatment of glioma due to poor penetration across the blood-brain barrier (BBB). Low-intensity pulsed ultrasound (LIPU) with microbubble injection transiently disrupts the BBB allowing for improved drug delivery to the brain. We investigated the distribution, toxicity, and efficacy of LIPU delivery of two different formulations of paclitaxel, albumin-bound paclitaxel (ABX) and paclitaxel dissolved in cremophor (CrEL-PTX), in preclinical glioma models. EXPERIMENTAL DESIGN: The efficacy and biodistribution of ABX and CrEL-PTX were compared with and without LIPU delivery. Antiglioma activity was evaluated in nude mice bearing intracranial patient-derived glioma xenografts (PDX). Paclitaxel biodistribution was determined in sonicated and nonsonicated nude mice. Sonications were performed using a 1 MHz LIPU device (SonoCloud), and fluorescein was used to confirm and map BBB disruption. Toxicity of LIPU-delivered paclitaxel was assessed through clinical and histologic examination of treated mice. RESULTS: Despite similar antiglioma activity in vitro, ABX extended survival over CrEL-PTX and untreated control mice with orthotropic PDX. Ultrasound-mediated BBB disruption enhanced paclitaxel brain concentration by 3- to 5-fold for both formulations and further augmented the therapeutic benefit of ABX. Repeated courses of LIPU-delivered CrEL-PTX and CrEL alone were lethal in 42% and 37.5% of mice, respectively, whereas similar delivery of ABX at an equivalent dose was well tolerated. CONCLUSIONS: Ultrasound delivery of paclitaxel across the BBB is a feasible and effective treatment for glioma. ABX is the preferred formulation for further investigation in the clinical setting due to its superior brain penetration and tolerability compared with CrEL-PTX.


Subject(s)
Albumins/pharmacology , Albumins/pharmacokinetics , Drug Compounding/methods , Drug Delivery Systems/methods , Glioma/drug therapy , Paclitaxel/pharmacology , Paclitaxel/pharmacokinetics , Polyethylene Glycols/chemistry , Ultrasonography/methods , Animals , Blood-Brain Barrier/drug effects , Female , Glioma/pathology , Male , Mice , Mice, Nude , Microbubbles/therapeutic use , Nanoparticles/chemistry , Survival Rate , Tissue Distribution , Xenograft Model Antitumor Assays
8.
BMC Cancer ; 16: 226, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26983574

ABSTRACT

BACKGROUND: Post-transcriptional regulation by microRNAs is recognized as one of the major pathways for the control of cellular homeostasis. Less well understood is the transcriptional and epigenetic regulation of genes encoding microRNAs. In the present study we addressed the epigenetic regulation of the miR-181c in normal and malignant brain cells. METHODS: To explore the epigenetic regulation of the miR-181c we evaluated its expression using RT-qPCR and the in vivo binding of the CCCTC-binding factor (CTCF) to its regulatory region in different glioblastoma cell lines. DNA methylation survey, chromatin immunoprecipitation and RNA interference assays were used to assess the role of CTCF in the miR-181c epigenetic silencing. RESULTS: We found that miR-181c is downregulated in glioblastoma cell lines, as compared to normal brain tissues. Loss of expression correlated with a notorious gain of DNA methylation at the miR-181c promoter region and the dissociation of the multifunctional nuclear factor CTCF. Taking advantage of the genomic distribution of CTCF in different cell types we propose that CTCF has a local and cell type specific regulatory role over the miR-181c and not an architectural one through chromatin loop formation. This is supported by the depletion of CTCF in glioblastoma cells affecting the expression levels of NOTCH2 as a target of miR-181c. CONCLUSION: Together, our results point to the epigenetic role of CTCF in the regulation of microRNAs implicated in tumorigenesis.


Subject(s)
Biomarkers, Tumor/biosynthesis , Glioblastoma/genetics , MicroRNAs/biosynthesis , Receptor, Notch2/biosynthesis , Repressor Proteins/biosynthesis , Biomarkers, Tumor/genetics , CCCTC-Binding Factor , Cell Line, Tumor , DNA Methylation/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Glioblastoma/pathology , Humans , Receptor, Notch2/genetics , Repressor Proteins/genetics
9.
Biochim Biophys Acta ; 1849(8): 955-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26079690

ABSTRACT

The three-dimensional architecture of genomes provides new insights about genome organization and function, but many aspects remain unsolved at the local genomic scale. Here we investigate the regulation of two erythroid-specific loci, a folate receptor gene (FOLR1) and the ß-globin gene cluster, which are separated by 16kb of constitutive heterochromatin. We found that in early erythroid differentiation the FOLR1 gene presents a permissive chromatin configuration that allows its expression. Once the transition to the next differentiation state occurs, the heterochromatin spreads into the FOLR1 domain, concomitant with the dissociation of CTCF from a novel binding site, thereby resulting in irreversible silencing of the FOLR1 gene. We demonstrate that the sequences surrounding the CTCF-binding site possess classical insulator properties in vitro and in vivo. In contrast, the chicken cHS4 ß-globin insulator present on the other side of the heterochromatic segment is in a constitutive open chromatin configuration, with CTCF constantly bound from the early stages of erythroid differentiation. Therefore, this study demonstrates that the 16kb of constitutive heterochromatin contributes to silencing of the FOLR1 gene during erythroid differentiation.


Subject(s)
Folate Receptor 1/genetics , Genetic Loci , Insulator Elements/physiology , beta-Globins/genetics , Animals , Cell Differentiation/genetics , Cell Line, Transformed , Chick Embryo , Chickens , Chromatin/genetics , Chromatin/metabolism , Erythropoiesis/genetics , Folate Receptor 1/metabolism , Gene Expression Regulation , Heterochromatin/genetics , Heterochromatin/metabolism
10.
Methods Mol Biol ; 1288: 413-28, 2015.
Article in English | MEDLINE | ID: mdl-25827894

ABSTRACT

Comprehensive genomic and computational studies in the era of high-throughput sequencing revealed that the major proportion of the human genome is transcribed. This novel insight confronted the scientific community with new questions concerning the expanded role of RNA, especially noncoding RNA (ncRNA), in cellular pathways. In recent years, there has been mounting evidence that ncRNAs and RNA binding proteins (RBPs) are involved in a wide range of biological processes, such as developmental transitions, cell differentiation, stress response, genome organization, and regulation of gene expression. In particular, in the chromatin field long noncoding RNAs (lncRNAs) have drawn increasing attention to their function in epigenetic regulation due to the fact that they were found to interact with multiple chromatin regulators and modifiers. Recently, techniques to study the extent of RNA-protein interactions have been developed in many research laboratories. Here we describe protocols for RNA Immunoprecipitation-Sequencing (RIP-Seq) and Photoactivatable-Ribonucleoside-Enhanced Cross-linking and Immunoprecipitation combined with deep sequencing (PAR-CLIP-Seq) to identify RNA targets of RNA-binding proteins (RBPs) on a transcriptome-wide level, discussing advantages and drawbacks.


Subject(s)
High-Throughput Nucleotide Sequencing , Immunoprecipitation/methods , RNA-Binding Proteins/metabolism , RNA/genetics , RNA/metabolism , Transcriptome , Binding Sites , High-Throughput Nucleotide Sequencing/methods , Humans , Protein Binding , RNA, Long Noncoding/genetics
11.
Methods Mol Biol ; 1165: 53-69, 2014.
Article in English | MEDLINE | ID: mdl-24839018

ABSTRACT

Cellular homeostasis is the result of an intricate and coordinated combinatorial of biochemical and molecular processes. Among them is the control of gene expression in the context of the chromatin structure which is central for cell survival. Interdependent action of transcription factors, cofactors, chromatin remodeling activities, and three-dimensional organization of the genome are responsible to reach exquisite levels of gene expression. Among such transcription factors there is a subset of highly specialized nuclear factors with features resembling master regulators with a large variety of functions. This is turning to be the case of the multifunctional nuclear factor CCCTC-binding protein (CTCF) which is involved in gene regulation, chromatin organization, and three-dimensional conformation of the genome inside the cell nucleus. Technically its study has turned to be challenging, in particular its posttranscriptional interference by small interference RNAs. Here we describe three main strategies to downregulate the overall abundance of CTCF in culture cell lines.


Subject(s)
Genetic Techniques , Repressor Proteins/deficiency , Repressor Proteins/genetics , Anti-Bacterial Agents/pharmacology , CCCTC-Binding Factor , Cell Line , Down-Regulation , Drug Resistance, Bacterial , Flow Cytometry , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Lentivirus/genetics , Luminescent Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Transduction, Genetic
12.
Genes Dev ; 28(7): 723-34, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24696455

ABSTRACT

The multifunctional CCCTC-binding factor (CTCF) protein exhibits a broad range of functions, including that of insulator and higher-order chromatin organizer. We found that CTCF comprises a previously unrecognized region that is necessary and sufficient to bind RNA (RNA-binding region [RBR]) and is distinct from its DNA-binding domain. Depletion of cellular CTCF led to a decrease in not only levels of p53 mRNA, as expected, but also those of Wrap53 RNA, an antisense transcript originated from the p53 locus. PAR-CLIP-seq (photoactivatable ribonucleoside-enhanced cross-linking and immunoprecipitation [PAR-CLIP] combined with deep sequencing) analyses indicate that CTCF binds a multitude of transcripts genome-wide as well as to Wrap53 RNA. Apart from its established role at the p53 promoter, CTCF regulates p53 expression through its physical interaction with Wrap53 RNA. Cells harboring a CTCF mutant in its RBR exhibit a defective p53 response to DNA damage. Moreover, the RBR facilitates CTCF multimerization in an RNA-dependent manner, which may bear directly on its role in establishing higher-order chromatin structures in vivo.


Subject(s)
Gene Expression Regulation , Genes, p53/genetics , RNA/metabolism , Repressor Proteins/metabolism , Telomerase/metabolism , CCCTC-Binding Factor , Cell Line, Tumor , DNA Damage/genetics , Humans , Molecular Chaperones , Mutation , Protein Binding , Protein Multimerization , Repressor Proteins/genetics , Sequence Deletion/genetics
13.
BMC Cancer ; 11: 232, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663659

ABSTRACT

BACKGROUND: Long-term gene silencing throughout cell division is generally achieved by DNA methylation and other epigenetic processes. Aberrant DNA methylation is now widely recognized to be associated with cancer and other human diseases. Here we addressed the contribution of the multifunctional nuclear factor CTCF to the epigenetic regulation of the human retinoblastoma (Rb) gene promoter in different tumoral cell lines. METHODS: To assess the DNA methylation status of the Rb promoter, genomic DNA from stably transfected human erythroleukemic K562 cells expressing a GFP reporter transgene was transformed with sodium bisulfite, and then PCR-amplified with modified primers and sequenced. Single- and multi-copy integrants with the CTCF binding site mutated were isolated and characterized by Southern blotting. Silenced transgenes were reactivated using 5-aza-2'-deoxycytidine and Trichostatin-A, and their expression was monitored by fluorescent cytometry. Rb gene expression and protein abundance were assessed by RT-PCR and Western blotting in three different glioma cell lines, and DNA methylation of the promoter region was determined by sodium bisulfite sequencing, together with CTCF dissociation and methyl-CpG-binding protein incorporation by chromatin immunoprecipitation assays. RESULTS: We found that the inability of CTCF to bind to the Rb promoter causes a dramatic loss of gene expression and a progressive gain of DNA methylation. CONCLUSIONS: This study indicates that CTCF plays an important role in maintaining the Rb promoter in an optimal chromatin configuration. The absence of CTCF induces a rapid epigenetic silencing through a progressive gain of DNA methylation. Consequently, CTCF can now be seen as one of the epigenetic components that allows the proper configuration of tumor suppressor gene promoters. Its aberrant dissociation can then predispose key genes in cancer cells to acquire DNA methylation and epigenetic silencing.


Subject(s)
DNA Methylation/physiology , Genes, Retinoblastoma , Promoter Regions, Genetic/genetics , Repressor Proteins/physiology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , CCCTC-Binding Factor , Cell Line, Tumor , DNA Methylation/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Decitabine , Down-Regulation/genetics , Genes, Reporter , Glioma/pathology , HeLa Cells , Humans , Hydroxamic Acids/pharmacology , K562 Cells/chemistry , Mutation , Nucleic Acid Conformation , Repressor Proteins/deficiency , Repressor Proteins/genetics , Sequence Analysis, DNA , Transgenes
14.
Nucleic Acids Res ; 39(1): 89-103, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20813760

ABSTRACT

Genome organization into transcriptionally active domains denotes one of the first levels of gene expression regulation. Although the chromatin domain concept is generally accepted, only little is known on how domain organization impacts the regulation of differential gene expression. Insulators might hold answers to address this issue as they delimit and organize chromatin domains. We have previously identified a CTCF-dependent insulator with enhancer-blocking activity embedded in the 5' non-coding region of the chicken α-globin domain. Here, we demonstrate that this element, called the αEHS-1.4 insulator, protects a transgene against chromosomal position effects in stably transfected cell lines and transgenic mice. We found that this insulator can create a regulated chromatin environment that coincides with the onset of adult α-globin gene expression. Furthermore, such activity is in part dependent on the in vivo regulated occupancy of CTCF at the αEHS-1.4 element. Insulator function is also regulated by CTCF poly(ADP-ribosyl)ation. Our results suggest that the αEHS-1.4 insulator contributes in organizing the chromatin structure of the α-globin gene domain and prevents activation of adult α-globin gene expression at the erythroblast stage via CTCF.


Subject(s)
Chromatin/chemistry , Gene Expression Regulation , Insulator Elements , alpha-Globins/genetics , Animals , Binding Sites , CCCTC-Binding Factor , Cell Differentiation , Cell Line , Chickens/genetics , Chromatin/genetics , Chromatin/metabolism , Chromosomal Position Effects , Erythroid Cells/cytology , Erythroid Cells/metabolism , Genetic Loci , Locus Control Region , Mice , Mice, Transgenic , Poly(ADP-ribose) Polymerase Inhibitors , Repressor Proteins/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , alpha-Globins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...