Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Neurosci ; 36(42): 10893-10907, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27798143

ABSTRACT

Face perception is subserved by a series of face-selective regions in the human ventral stream, which undergo prolonged development from childhood to adulthood. However, it is unknown how neural development of these regions relates to the development of face-perception abilities. Here, we used functional magnetic resonance imaging (fMRI) to measure brain responses of ventral occipitotemporal regions in children (ages, 5-12 years) and adults (ages, 19-34 years) when they viewed faces that parametrically varied in dissimilarity. Since similar faces generate lower responses than dissimilar faces due to fMRI adaptation, this design objectively evaluates neural sensitivity to face identity across development. Additionally, a subset of subjects participated in a behavioral experiment to assess perceptual discriminability of face identity. Our data reveal three main findings: (1) neural sensitivity to face identity increases with age in face-selective but not object-selective regions; (2) the amplitude of responses to faces increases with age in both face-selective and object-selective regions; and (3) perceptual discriminability of face identity is correlated with the neural sensitivity to face identity of face-selective regions. In contrast, perceptual discriminability is not correlated with the amplitude of response in face-selective regions or of responses of object-selective regions. These data suggest that developmental increases in neural sensitivity to face identity in face-selective regions improve perceptual discriminability of faces. Our findings significantly advance the understanding of the neural mechanisms of development of face perception and open new avenues for using fMRI adaptation to study the neural development of high-level visual and cognitive functions more broadly. SIGNIFICANCE STATEMENT: Face perception, which is critical for daily social interactions, develops from childhood to adulthood. However, it is unknown what developmental changes in the brain lead to improved performance. Using fMRI in children and adults, we find that from childhood to adulthood, neural sensitivity to changes in face identity increases in face-selective regions. Critically, subjects' perceptual discriminability among faces is linked to neural sensitivity: participants with higher neural sensitivity in face-selective regions demonstrate higher perceptual discriminability. Thus, our results suggest that developmental increases in face-selective regions' sensitivity to face identity improve perceptual discrimination of faces. These findings significantly advance understanding of the neural mechanisms underlying the development of face perception and have important implications for assessing both typical and atypical development.


Subject(s)
Discrimination, Psychological/physiology , Face , Recognition, Psychology/physiology , Adult , Aging/physiology , Brain Mapping , Child , Child, Preschool , Female , Functional Laterality/physiology , Humans , Linear Models , Locus Control Region/physiology , Magnetic Resonance Imaging , Male , Occipital Lobe/growth & development , Occipital Lobe/physiology , Photic Stimulation , Psychomotor Performance , Temporal Lobe/growth & development , Temporal Lobe/physiology , Visual Cortex/growth & development , Visual Cortex/physiology , Young Adult
2.
PLoS One ; 11(2): e0148576, 2016.
Article in English | MEDLINE | ID: mdl-26840450

ABSTRACT

The T helper type 2 (Th2) locus control region (LCR) regulates Th2 cell differentiation. Several transcription factors bind to the LCR to modulate the expression of Th2 cytokine genes, but the molecular mechanisms behind Th2 cytokine gene regulation are incompletely understood. Here, we used database analysis and an oligonucleotide competition/electrophoretic mobility shift assays to search for transcription factors binding to RHS5, a DNase I hypersensitive site (DHS) within the Th2 LCR. Consequently, we demonstrated that GATA-binding protein-3 (GATA-3), E26 transformation-specific protein 1 (Ets-1), octamer transcription factor-1 (Oct-1), and Oct-2 selectively associate with RHS5. Furthermore, chromatin immunoprecipitation and luciferase reporter assays showed that Oct-1 and Oct-2 bound within the Il4 promoter region and the Th2 LCR, and that Oct-1 and GATA-3 or Oct-2 synergistically triggered the transactivational activity of the Il4 promoter through RHS5. These results suggest that Oct-1 and GATA-3/Oct-2 direct Th2 cytokine gene expression in a cooperative manner.


Subject(s)
GATA3 Transcription Factor/metabolism , Gene Expression Regulation/physiology , Interleukin-4/biosynthesis , Locus Control Region/physiology , Octamer Transcription Factor-1/metabolism , Th2 Cells/metabolism , Animals , Cell Line , GATA3 Transcription Factor/genetics , Interleukin-4/genetics , Mice , Octamer Transcription Factor-1/genetics , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2 , Th2 Cells/cytology
3.
J Biol Chem ; 287(30): 25454-65, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22669946

ABSTRACT

Locus control regions (LCRs) comprise sets of DNA elements capable of establishing autonomous chromatin domains that support robust and physiologically appropriate expression of target genes, often working over extensive distances. Human growth hormone (hGH-N) expression in the pituitary is under the regulation of a well characterized LCR containing four DNase I hypersensitive sites (HSs). The two pituitary-specific HS, HSI and HSII, are located 14.5 and 15.5 kb 5' to the hGH-N promoter. HSI is essential for activation of hGH-N during pituitary development and for sustaining robust activity in the adult. To determine whether the closely linked HSII has a role in hGH-N expression, it was deleted from a previously validated hGH/P1 transgene. Analysis of three independent hGH/P1(ΔHSII) transgenic mouse lines revealed that this deletion had no adverse effect on the formation of HSI, yet resulted in a substantial loss (70%) in hGH-N mRNA expression. This loss of expression was accompanied by a corresponding reduction in recruitment of the pituitary-specific transcription factor Pit-1 to the hGH-N promoter and a selective decrease in promoter occupancy of the elongation-linked isoform of RNA polymerase II. Sufficiency of HSI and HSII in LCR activity was explored by establishing two additional sets of mouse transgenic lines in which DNA segments containing these HS were positioned within the λ phage genome. In this "neutral" DNA context, HSII was required for the recruitment of HAT activity. These data establish HSII as a nonredundant component of the hGH LCR essential for establishment of robust levels of hGH-N gene expression.


Subject(s)
DNA, Intergenic/metabolism , Gene Expression Regulation/physiology , Genetic Loci/physiology , Human Growth Hormone/biosynthesis , Locus Control Region/physiology , Pituitary Gland/metabolism , Animals , Base Sequence , DNA, Intergenic/genetics , Deoxyribonuclease I/chemistry , Human Growth Hormone/genetics , Humans , Mice , Mice, Transgenic , Pituitary Gland/growth & development , Sequence Deletion
4.
J Biol Chem ; 285(21): 15894-905, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20236933

ABSTRACT

The human beta-globin gene is expressed at high levels in erythroid cells and regulated by proximal and distal cis-acting DNA elements, including promoter, enhancer, and a locus control region (LCR). Transcription complexes are recruited not only to the globin gene promoters but also to the LCR. Previous studies have implicated the ubiquitously expressed transcription factor USF and the tissue-restricted activator NF-E2 in the recruitment of transcription complexes to the beta-globin gene locus. Here we demonstrate that although USF is required for the efficient association of RNA polymerase II (Pol II) with immobilized LCR templates, USF and NF-E2 together regulate the association of Pol II with the adult beta-globin gene promoter. Recruitment of Pol II to the LCR occurs in undifferentiated murine erythroleukemia cells, but phosphorylation of LCR-associated Pol II at serine 5 of the C-terminal domain is mediated by erythroid differentiation and requires the activity of NF-E2. Furthermore, we provide evidence showing that USF interacts with NF-E2 in erythroid cells. The data provide mechanistic insight into how ubiquitous and tissue-restricted transcription factors cooperate to regulate the recruitment and activity of transcription complexes in a tissue-specific chromatin domain.


Subject(s)
Genetic Loci/physiology , Locus Control Region/physiology , NF-E2 Transcription Factor, p45 Subunit/metabolism , RNA Polymerase II/metabolism , beta-Globins/biosynthesis , Animals , Cell Line, Tumor , Humans , Mice , NF-E2 Transcription Factor, p45 Subunit/genetics , Organ Specificity/physiology , Phosphorylation/physiology , Protein Structure, Tertiary , RNA Polymerase II/genetics , Transcription, Genetic/physiology , beta-Globins/genetics
5.
FASEB J ; 23(12): 4335-43, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19690216

ABSTRACT

In primitive erythroid cells of human beta-globin locus transgenic mice (TgM), the locus control region (LCR)-proximal epsilon- and gamma-globin genes are transcribed, whereas the distal delta- and beta-globin genes are silent. It is generally accepted that the beta-globin gene is competitively suppressed by gamma-globin gene expression at this developmental stage. Previously, however, we observed that epsilon-globin gene expression was severely attenuated when its distance from the LCR was extended, implying that beta-globin gene might also be silenced because of its great distance from the LCR. Here, to clarify the beta-globin gene silencing mechanism, we established TgM lines carrying either gamma- or epsilon- plus gamma-globin promoter deletions, without significantly altering the distance between the beta-globin gene and the LCR. Precocious expression of delta- and beta-globin genes was observed in primitive erythroid cells of mutant, but not wild-type TgM, which was most evident when both the epsilon and gamma promoters were deleted. Thus, we clearly demonstrated that the repression of the delta- and beta-globin genes in primitive erythroid cells is dominated by competitive silencing by the epsilon- and gamma-globin gene promoters, and that epsilon- and the other beta-like globin genes might be activated by two distinct mechanisms by the LCR.


Subject(s)
Chromosomes, Artificial, Yeast/genetics , Erythroid Cells/metabolism , Locus Control Region/physiology , beta-Globins/genetics , Animals , Gene Expression Regulation/physiology , Humans , Mice , Mice, Transgenic , Saccharomyces cerevisiae
6.
IUBMB Life ; 60(12): 824-33, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18767169

ABSTRACT

The mammalian beta-globin locus is a multigene locus containing several globin genes and a number of regulatory elements. During development, the expression of the genes changes in a process called "switching." The most important regulatory element in the locus is the locus control region (LCR) upstream of the globin genes that is essential for high-level expression of these genes. The discovery of the LCR initially raised the question how this element could exert its effect on the downstream globin genes. The question was solved by the finding that the LCR and activate globin genes are in physical contact, forming a chromatin structure named the active chromatin hub (ACH). Here we discuss the significance of ACH formation, provide an overview of the proteins implicated in chromatin looping at the beta-globin locus, and evaluate the relationship between nuclear organization and beta-globin gene expression.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation , Locus Control Region/physiology , beta-Globins/genetics , Animals , Cell Differentiation , Genes, Switch , Humans , Transcription, Genetic , beta-Globins/metabolism
7.
Int J Biochem Cell Biol ; 40(8): 1481-93, 2008.
Article in English | MEDLINE | ID: mdl-18308612

ABSTRACT

Evidences indicate that locus control region (LCR) of beta-globin spatially closes to the downstream active gene promoter to mediate the transcriptional activation by looping. DNA binding proteins may play an important role in the looping formation. NF-E2 is one of the key transcription factors in beta-globin gene transcriptional activation. To shed light on whether NF-E2 is involved in this process, DS19MafKsiRNA cell pools were established by specifically knocked down the expression of MafK/NF-E2 p18, one subunit of NF-E2 heterodimer. In the above cell pools, it was observed that the occupancy efficiency of NF-E2 on beta-globin gene locus and the expression level of beta-globin genes were decreased. H3 acetylation, H3-K4 methylation and the deposition of RNA polymerase II, but not the recruitment of GATA-1, were also found reduced at the beta-globin gene cluster. Chromosome Conformation Capture (3C) assay showed that the cross-linking frequency between the main NF-E2 binding site HS2 and downstream structural genes was reduced compared to the normal cell. This result demonstrated that MafK/NF-E2 p18 recruitment was involved in the physical proximity of LCR and active beta-globin genes upon beta-globin gene transcriptional activation.


Subject(s)
Gene Expression Regulation/physiology , Globins/genetics , Locus Control Region/physiology , MafK Transcription Factor/physiology , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Polymerase II/physiology , GATA1 Transcription Factor/physiology , Gene Silencing , Globins/biosynthesis , Histones/metabolism , Mice , RNA Interference
8.
Adv Genet ; 61: 107-42, 2008.
Article in English | MEDLINE | ID: mdl-18282504

ABSTRACT

Transcriptional activation in higher eukaryotes frequently involves the long-range action of a number of regulatory DNA elements. One of the main questions in transcriptional regulation is how cis-regulatory elements communicate with the promoter of a gene over large distances. There has been a lively debate in recent years whether this communication takes place via a noncontact mechanism (linking, tracking) or via a contact mechanism (looping). The demonstration that the major regulatory element of the beta-globin locus, the locus control region (LCR), is in close proximity to the active beta-globin genes validates the contact model for long-range activation. Here, we will review the beta-globin locus as a model system to study long-range activation, briefly describe the different models for long-range activation, and summarize the recent findings that the LCR of the beta-globin locus is in close proximity to the active promoters. Although it is now firmly established that looping takes place within the beta-globin locus (and other loci), it is not clear how these long-range contacts are established and what the precise role is of the LCR. We will argue that the main action of the LCR takes place at the promoter and open reading frame of the gene itself and we will discuss key rate-limiting steps in transcriptional activation and the possible mechanisms by which they are influenced by the LCR.


Subject(s)
Epistasis, Genetic , Gene Expression Regulation/physiology , Globins/genetics , Animals , Chromatin/chemistry , Chromatin/physiology , Gene Order/physiology , Humans , Locus Control Region/physiology , Models, Biological , Promoter Regions, Genetic/physiology , Transcription, Genetic/physiology
9.
Adv Genet ; 61: 339-88, 2008.
Article in English | MEDLINE | ID: mdl-18282513

ABSTRACT

The past two decades have seen great progress in the elucidation of the genetic basis of human genetic disease. Many clinical phenotypes have been linked with mutations or deletions in specific causative genes. However, it is often less recognized that in addition to the integrity of the protein-coding sequences, human health critically also depends on the spatially, temporally, and quantitatively correct expression of those genes. Genetic disease can therefore equally be caused by disruption of the regulatory mechanisms that ensure proper gene expression. The term "position effect" is used in those situations where the expression level of a gene is deleteriously affected by an alteration in its chromosomal environment, while maintaining an intact transcription unit. Here, we review recent advances in our understanding of the possible mechanisms of a number of "position effect" disease cases and discuss the findings with respect to current models for genome organization and long-range control of gene expression.


Subject(s)
Gene Expression Regulation/physiology , Genetic Diseases, Inborn/etiology , Animals , Bystander Effect/genetics , Forkhead Transcription Factors/physiology , Gene Dosage/physiology , Gene Order/physiology , High Mobility Group Proteins/physiology , Humans , Locus Control Region/genetics , Locus Control Region/physiology , RNA, Small Interfering/physiology , Regulatory Sequences, Nucleic Acid/physiology , SOX9 Transcription Factor , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/physiology
11.
Blood ; 110(12): 4101-7, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17881636

ABSTRACT

To delineate the relationship between epigenetic modifications and hemoglobin switching, we compared the pattern of histone acetylation and pol II binding across the beta-globin locus at fetal and adult stages of human development. To make this comparison possible, we introduced an external control into experimental samples in chromatin immunoprecipitation (ChIP) assays. Using this common standard, we found that the locus control region (LCR) was acetylated to the same level at all stages, whereas acetylation levels at the individual gene regions correlated with the state of transcription. In the active genes, the promoters were less acetylated compared with the coding regions. Furthermore, all globin promoters were acetylated to a similar level irrespective of the state of transcription. However, after correction for the loss of nucleosomes, the level of acetylation per histone at the active gamma and beta promoters was 5- to 7-fold greater than that at the inactive epsilon promoter. Although the histone acetylation level within the LCR was developmentally stable, pol II binding in fetal erythroblasts was 2- to 3-fold greater than that in adult erythroblasts. These results demonstrate that dynamic changes in histone acetylation and pol II take place as the human beta-globin gene region undergoes its developmental switches.


Subject(s)
Aging/physiology , Erythroblasts/metabolism , Globins/biosynthesis , Histones/metabolism , Locus Control Region/physiology , Quantitative Trait Loci/physiology , Acetylation , Adult , Chromatin Immunoprecipitation , DNA Polymerase II/metabolism , Erythroblasts/cytology , Fetus/cytology , Fetus/metabolism , Globins/genetics , Humans , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/physiology , Transcription, Genetic/physiology
12.
J Biol Chem ; 282(22): 16544-52, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17428799

ABSTRACT

Expression of the beta-globin genes proceeds from basal to exceptionally high levels during erythroid differentiation in vivo. High expression is dependent on the locus control region (LCR) and coincides with more frequent LCR-gene contacts. These contacts are established in the context of an active chromatin hub (ACH), a spatial chromatin configuration in which the LCR, together with other regulatory sequences, loops toward the active beta-globin-like genes. Here, we used recently established I/11 cells as a model system that faithfully recapitulates the in vivo erythroid differentiation program to study the molecular events that accompany and underlie ACH formation. Upon I/11 cell induction, histone modifications changed, the ACH was formed, and the beta-globin-like genes were transcribed at rates similar to those observed in vivo. The establishment of frequent LCR-gene contacts coincided with a more efficient loading of polymerase onto the beta-globin promoter. Binding of the transcription factors GATA-1 and EKLF to the locus, although previously shown to be required, was not sufficient for ACH formation. Moreover, we used knock-out mice to show that the erythroid transcription factor p45 NF-E2, which has been implicated in beta-globin gene regulation, is dispensable for beta-globin ACH formation.


Subject(s)
Cell Differentiation/physiology , Chromatin Assembly and Disassembly/physiology , Erythroid Cells/metabolism , Globins/biosynthesis , Locus Control Region/physiology , NF-E2 Transcription Factor, p45 Subunit/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Erythroid Cells/cytology , GATA1 Transcription Factor/metabolism , Globins/genetics , Histones/metabolism , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Knockout , Models, Biological , NF-E2 Transcription Factor, p45 Subunit/deficiency , Promoter Regions, Genetic/physiology , Quantitative Trait Loci/physiology
13.
Microbiology (Reading) ; 152(Pt 11): 3367-3382, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17074906

ABSTRACT

Porphyromonas gingivalis, an oral bacterium associated with periodontal disease, requires haemin for growth. Although several multigenic clusters encoding haemin-uptake systems are present on the genome of P. gingivalis, little is known regarding their transcriptional organization and expression. This study identified a 23 kDa iron-regulated haemin-binding protein encoded by a larger than previously reported variant of hmuY. It was shown that the hmu locus is larger than previously reported and is composed of six genes, hmuYRSTUV, encoding a novel hybrid haemin-uptake system. The locus has an operonic organization and the transcriptional start site is located 292 bp upstream of hmuY. The data indicate that the regulation of the operon is iron-dependent. Interestingly, differential regulation within the operon was demonstrated, resulting in excess of the hmuYR message encoding the outer-membrane proteins when compared to the full-length transcript. In addition, the hmuY transcript is more prevalent than the hmuR transcript. Secondary structure analysis of the hmuYRSTUV mRNA predicted the formation of several potential stem-loops in the 5' ends of hmuR- and hmuS-specific mRNAs, consistent with the differential regulation observed. Finally, it was demonstrated that haemin binding and uptake are elevated in iron-depleted conditions and are reduced 45 % and 70 %, respectively, in an hmu-deficient strain when compared to the parental strain, indicating that the hmu locus plays a major role in haemin acquisition in P. gingivalis. Since homologues of the hmu locus were also found in Bacteroides fragilis, Bacteroides thetaiotaomicron and Prevotella intermedia, these findings may have implications for a better understanding of haemin acquisition in those organisms as well.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Genome, Bacterial , Hemin/metabolism , Iron/metabolism , Locus Control Region/physiology , Operon , Porphyromonas gingivalis/genetics , Transcription, Genetic , Amino Acid Sequence , Bacterial Outer Membrane Proteins/metabolism , Bacteroidaceae Infections/microbiology , Locus Control Region/genetics , Molecular Sequence Data , Molecular Weight , Porphyromonas gingivalis/metabolism , Promoter Regions, Genetic
14.
PLoS Biol ; 4(11): e355, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17048991

ABSTRACT

Expression of imprinted genes is restricted to a single parental allele as a result of epigenetic regulation-DNA methylation and histone modifications. Igf2/H19 is a reciprocally imprinted locus exhibiting paternal Igf2 and maternal H19 expression. Their expression is regulated by a paternally methylated imprinting control region (ICR) located between the two genes. Although the de novo DNA methyltransferases have been shown to be necessary for the establishment of ICR methylation, the mechanism by which they are targeted to the region remains unknown. We demonstrate that CTCFL/BORIS, a paralog of CTCF, is an ICR-binding protein expressed during embryonic male germ cell development, coinciding with the timing of ICR methylation. PRMT7, a protein arginine methyltransferase with which CTCFL interacts, is also expressed during embryonic testis development. Symmetrical dimethyl arginine 3 of histone H4, a modification catalyzed by PRMT7, accumulates in germ cells during this developmental period. This modified histone is also found enriched in both H19 ICR and Gtl2 differentially methylated region (DMR) chromatin of testis by chromatin immunoprecipitation (ChIP) analysis. In vitro studies demonstrate that CTCFL stimulates the histone-methyltransferase activity of PRMT7 via interactions with both histones and PRMT7. Finally, H19 ICR methylation is demonstrated by nuclear co-injection of expression vectors encoding CTCFL, PRMT7, and the de novo DNA methyltransferases, Dnmt3a, -b and -L, in Xenopus oocytes. These results suggest that CTCFL and PRMT7 may play a role in male germline imprinted gene methylation.


Subject(s)
DNA Methylation , DNA-Binding Proteins/physiology , Genomic Imprinting/physiology , Methyltransferases/physiology , RNA, Untranslated/metabolism , Testis/cytology , Animals , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Embryo, Nonmammalian , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Germ Cells/physiology , Histones/chemistry , Histones/metabolism , Locus Control Region/physiology , Male , Methyltransferases/metabolism , Mice , Models, Biological , Molecular Sequence Data , Oocytes/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Methyltransferases/physiology , Protein-Arginine N-Methyltransferases , Proteins/genetics , RNA, Long Noncoding , RNA, Untranslated/genetics , Testis/embryology , Xenopus
16.
Mol Cell ; 23(3): 365-75, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16885026

ABSTRACT

Activation of eukaryotic genes often relies on remote chromatin determinants. How these determinants function remains poorly understood. The hGH gene is activated by a 5'-remote locus control region (LCR). Pituitary-specific DNase I hypersensitive site I (HSI), the dominant hGH LCR element, is separated from the hGH-N promoter by a 14.5 kb span that encompasses the B-lymphocyte-specific CD79b gene. Here, we describe a domain of noncoding Pol II transcription in pituitary somatotropes that includes the hGH LCR and adjacent CD79b locus. This entire "LCR domain of transcription" is HSI [corrected] dependent and terminates 3' to CD79b, leaving a gap in transcription between this domain and the target hGH-N promoter. Insertion of a Pol II terminator within the LCR blocks CD79b transcription and represses hGH-N expression. These data document an essential role for LCR transcription in long-range control, link "bystander"CD79b transcription to this process, and support a unique model for locus activation.


Subject(s)
Gene Expression Regulation , Human Growth Hormone/genetics , Locus Control Region/physiology , Transcription, Genetic/genetics , Acetylation , Animals , CD79 Antigens/genetics , Chromatin Immunoprecipitation , Gene Deletion , Gene Library , Growth Hormone-Releasing Hormone/genetics , Histones/metabolism , Humans , Liver/metabolism , Locus Control Region/genetics , Mice , Mice, Transgenic , Models, Genetic , Pituitary Gland/metabolism , RNA Polymerase II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Terminator Regions, Genetic/genetics , Transcriptional Activation
17.
Blood Cells Mol Dis ; 35(3): 339-44, 2005.
Article in English | MEDLINE | ID: mdl-16185901

ABSTRACT

Chromatin insulators are regulatory elements that determine domains of genetic functions. We have previously described the characterization of a 265 bp insulator element, termed sns, localized at the 3' end of the early histone H2A gene of the sea urchin Paracentrotus lividus. This sequence contains three cis-acting elements (Box A, Box B, and Box C + T) all needed for the enhancer-blocking activity in both sea urchin and human cells. The goal of this study was to further characterize the sea urchin sns insulator in the erythroid environment. We employed colony assays in human (K562) and mouse (MEL) erythroid cell lines. We tested the capability of sns to interfere with the communication between the 5'HS2 enhancer of the human beta-globin LCR and the gamma-globin promoter. We found that the sns sequence displays directional enhancer-blocking activity. By the use of antibodies against known DNA binding proteins, in electrophoretic mobility shift assays, we demonstrated the binding of the erythroid-specific GATA-1 and the ubiquitous Oct-1 and Sp1 transcription factors. These factors bind to Box A, Box B, and Box C + T, respectively, in both K562 and MEL nuclear extracts. These results may have significant implications for the conservation of insulator function in evolutionary distant organisms and may prove to be of practical benefit in gene transfer applications for erythroid disorders such as hemoglobinopathies and thalassemias.


Subject(s)
Chromatin/physiology , Erythroid Cells/metabolism , Insulator Elements/physiology , Sea Urchins/genetics , Animals , Cell Line , Chromatin/genetics , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic/genetics , GATA1 Transcription Factor/physiology , Gene Transfer Techniques , Globins/genetics , Humans , Insulator Elements/genetics , Locus Control Region/physiology , Mice , Octamer Transcription Factor-1/physiology , Paracentrotus/genetics , Sp1 Transcription Factor/physiology
18.
Mol Cell Biol ; 25(8): 3220-31, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15798207

ABSTRACT

The mouse immunoglobulin kappa (Igkappa) gene contains an intronic enhancer and two enhancers downstream of its transcription unit. Using chromosome conformation capture technology, we demonstrate that rearranged and actively transcribed Igkappa alleles in MPC-11 plasmacytoma cells exhibit mutual interactions over 22 kb between these three enhancers and Vkappa gene promoters. In addition, the 5' region of the active transcription unit exhibits a continuum of interactions with downstream chromatin segments. We also observe interactions between Ei and E3' with 3' boundary sequences 24 kb downstream of Ed, adjacent to a neighboring housekeeping gene. Very similar interactions between the enhancers are also exhibited by normal B cells isolated from mouse splenic tissue but not by germ line transcriptionally inactive alleles of T cells or P815 mastocytoma cells, which exhibit a seemingly linear chromatin organization. These results fit a looping mechanism for enhancer function like in the beta-globin locus and suggest a dynamic modulation of the spatial organization of the active Igkappa locus. Chromatin immunoprecipitation experiments reveal that the interacting Igkappa gene cis-acting sequences are associated with AP-4, E47, and p65NF-kappaB, potential protein candidates that may be responsible for initiating and/or maintaining the formation of these higher-order complexes. However, S107 plasmacytoma cells that lack NF-kappaB still exhibit mutual interactions between the Igkappa gene enhancers.


Subject(s)
3' Flanking Region , Enhancer Elements, Genetic/physiology , Immunoglobulin Variable Region/genetics , Immunoglobulin kappa-Chains/genetics , Transcription Factors/metabolism , Transcription, Genetic/physiology , Animals , B-Lymphocytes/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Globins/genetics , Locus Control Region/physiology , Mice , NF-kappa B/metabolism , Nucleic Acid Conformation , Plasmacytoma , Promoter Regions, Genetic/genetics , T-Lymphocytes/metabolism , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transcription Factor RelA
19.
J Immunol ; 173(10): 6200-10, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15528357

ABSTRACT

MHC class II (MHC-II) genes are regulated by an enhanceosome complex containing two gene-specific transcription factors, regulatory factor X complex (RFX) and CIITA. These factors assemble on a strictly conserved regulatory module (S-X-X2-Y) found immediately upstream of the promoters of all classical and nonclassical MHC-II genes as well as the invariant chain (Ii) gene. To identify new targets of RFX and CIITA, we developed a computational approach based on the unique and highly constrained architecture of the composite S-Y motif. We identified six novel S'-Y' modules situated far away from the promoters of known human RFX- and CIITA-controlled genes. Four are situated at strategic positions within the MHC-II locus, and two are found within the Ii gene. These S'-Y' modules function as transcriptional enhancers, are bona fide targets of RFX and CIITA in B cells and IFN-gamma-induced cells, and induce broad domains of histone hyperacetylation. These results reveal a hitherto unexpected level of complexity involving long distance control of MHC-II expression by multiple distal regulatory elements.


Subject(s)
DNA-Binding Proteins/physiology , Enhancer Elements, Genetic/physiology , Gene Expression Profiling , Genes, MHC Class II , Locus Control Region/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Acetylation , Acetyltransferases/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Base Sequence , Cell Line, Tumor , Computational Biology/methods , Conserved Sequence , DNA-Binding Proteins/metabolism , Gene Expression Profiling/methods , Genetic Markers/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histone Acetyltransferases , Histones/metabolism , Humans , Interferon-gamma/physiology , Molecular Sequence Data , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding/genetics , Protein Binding/immunology , Regulatory Factor X Transcription Factors , Trans-Activators/metabolism , Transcription Factors/metabolism
20.
Curr Biol ; 14(11): 1007-12, 2004 Jun 08.
Article in English | MEDLINE | ID: mdl-15182675

ABSTRACT

The H19 imprinted gene locus is regulated by an upstream 2 kb imprinting control region (ICR) that influences allele-specific expression, DNA methylation, and replication timing. This ICR becomes de novo methylated during late spermatogenesis in the male but emerges from oogenesis in an unmethylated form, and this allele-specific pattern is then maintained throughout early development and in all tissues of the mouse. We have used a genetic approach involving transfection into embryonic stem (ES) cells in order to decipher how the maternal allele is protected from de novo methylation at the time of implantation. Our studies show that CCCTC binding factor (CTCF) boundary elements within the ICR have the ability to prevent de novo methylation on the maternal allele. Since CTCF does not recognize its binding sequence when methylated, this reaction does not occur on the paternal allele, thus preserving the gamete-derived, allele-specific pattern. These results suggest that CTCF may play a general role in the maintenance of differential methylation patterns in vivo.


Subject(s)
Alleles , DNA Methylation , DNA-Binding Proteins/metabolism , Genomic Imprinting/physiology , Locus Control Region/physiology , Repressor Proteins/metabolism , Animals , Base Sequence , Blotting, Southern , CCCTC-Binding Factor , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Female , Male , Mice , Mice, Transgenic , Plasmids/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , Sex Factors , Stem Cells/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...