Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Genet ; 11: 928, 2020.
Article in English | MEDLINE | ID: mdl-33061937

ABSTRACT

Alu elements are primate-specific repeats and represent the most abundant type of transposable elements (TE) in the human genome. Genome-wide analysis of the enrichment of histone post-translational modifications suggests that human Alu sequences could function as transcriptional enhancers; however, no functional experiments have evaluated the role of Alu sequences in the control of transcription in situ. The present study analyses the regulatory activity of a human Alu sequence from the AluSx family located in the second intron of the long intergenic non-coding RNA Linc00441, found in divergent orientation to the RB1 gene. We observed that the Alu sequence acts as an enhancer element based on reporter gene assays while CRISPR-Cas9 deletions of the Alu sequence in K562 cells resulted in a marked transcriptional upregulation of Linc00441 and a decrease in proliferation. Our results suggest that an intragenic Alu sequence with enhancer activity can act as a transcriptional attenuator of its host lincRNA.

2.
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
3.
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
4.
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
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...