Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1156077, 2023.
Article in English | MEDLINE | ID: mdl-37215121

ABSTRACT

Over the past decades, it has become increasingly clear that higher order chromatin folding and organization within the nucleus is involved in the regulation of genome activity and serves as an additional epigenetic mechanism that modulates cellular functions and gene expression programs in diverse biological processes. In particular, dynamic allelic interactions and nuclear locations can be of functional importance during the process of lymphoid differentiation and the regulation of immune responses. Analyses of the proximity between chromatin and/or nuclear regions can be performed on populations of cells with high-throughput sequencing approaches such as chromatin conformation capture ("3C"-based) or DNA adenine methyltransferase identification (DamID) methods, or, in individual cells, by the simultaneous visualization of genomic loci, their primary transcripts and nuclear compartments within the 3-dimensional nuclear space using Fluorescence In Situ Hybridization (FISH) and immunostaining. Here, we present a detailed protocol to simultaneously detect nascent RNA transcripts (3D RNA FISH), their genomic loci (3D DNA FISH) and/or their chromosome territories (CT paint DNA FISH) combined with the antibody-based detection of various nuclear factors (immunofluorescence). We delineate the application and effectiveness of this robust and reproducible protocol in several murine T lymphocyte subtypes (from differentiating thymic T cells, to activated splenic and peripheral T cells) as well as other murine cells, including embryonic stem cells, B cells, megakaryocytes and macrophages.


Subject(s)
Chromatin , T-Lymphocytes , Animals , Mice , In Situ Hybridization, Fluorescence/methods , T-Lymphocytes/metabolism , Chromatin/genetics , DNA/metabolism , Genomics
2.
PLoS One ; 14(11): e0223759, 2019.
Article in English | MEDLINE | ID: mdl-31682635

ABSTRACT

microRNAs are of vital importance for the regulation of the adaptive and innate immune responses, modulating gene expression at the post transcriptional level. Although there is cumulative information regarding the steady state mature microRNA levels and their respective targets, little is known about the effect of the three-dimensional chromatin architecture on the transcriptional regulation of microRNA gene loci. Here, we sought to investigate the effect of subnuclear localization on the transcriptional activation of eight murine microRNA loci in the immune system. Our results show that microRNA genes display a preferential monoallelic gene expression profile accompanied with perinuclear localization irrespectively of their transcription status or differentiation state. The expression profile and perinuclear localization are developmentally conserved while microRNA gene loci localization outside constitutive lamin associated domains is cross-species conserved. Our findings provide support for an active nuclear periphery and its role in chromatin organization of the non-coding genome.


Subject(s)
Cell Nucleus/genetics , MicroRNAs/genetics , Animals , Cell Differentiation/genetics , Chromatin/genetics , Gene Expression Regulation/genetics , Immune System/physiology , Lamins/genetics , Mice , Transcription, Genetic/genetics , Transcriptional Activation/genetics
3.
J Immunol ; 203(6): 1532-1547, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31405954

ABSTRACT

The infiltration and subsequent in situ subtype specification of monocytes to effector/inflammatory and repair macrophages is indispensable for tissue repair upon acute sterile injury. However, the chromatin-level mediators and regulatory events controlling this highly dynamic macrophage phenotype switch are not known. In this study, we used a murine acute muscle injury model to assess global chromatin accessibility and gene expression dynamics in infiltrating macrophages during sterile physiological inflammation and tissue regeneration. We identified a heme-binding transcriptional repressor, BACH1, as a novel regulator of this process. Bach1 knockout mice displayed impaired muscle regeneration, altered dynamics of the macrophage phenotype transition, and transcriptional deregulation of key inflammatory and repair-related genes. We also found that BACH1 directly binds to and regulates distal regulatory elements of these genes, suggesting a novel role for BACH1 in controlling a broad spectrum of the repair response genes in macrophages upon injury. Inactivation of heme oxygenase-1 (Hmox1), one of the most stringently deregulated genes in the Bach1 knockout in macrophages, impairs muscle regeneration by changing the dynamics of the macrophage phenotype switch. Collectively, our data suggest the existence of a heme-BACH1--HMOX1 regulatory axis, that controls the phenotype and function of the infiltrating myeloid cells upon tissue damage, shaping the overall tissue repair kinetics.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Regeneration/physiology , Animals , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Transcription, Genetic/physiology
4.
Proc Natl Acad Sci U S A ; 112(13): E1577-86, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25770217

ABSTRACT

Physiological processes rely on the regulation of total mRNA levels in a cell. In diploid organisms, the transcriptional activation of one or both alleles of a gene may involve trans-allelic interactions that provide a tight spatial and temporal level of gene expression regulation. The mechanisms underlying such interactions still remain poorly understood. Here, we demonstrate that lipopolysaccharide stimulation of murine macrophages rapidly resulted in the actin-mediated and transient homologous spatial proximity of Tnfα alleles, which was necessary for the mono- to biallelic switch in gene expression. We identified two new complementary long noncoding RNAs transcribed from the TNFα locus and showed that their knockdown had opposite effects in Tnfα spatial proximity and allelic expression. Moreover, the observed spatial proximity of Tnfα alleles depended on pyruvate kinase muscle isoform 2 (PKM2) and T-helper-inducing POZ-Krüppel-like factor (ThPOK). This study suggests a role for lncRNAs in the regulation of somatic homologous spatial proximity and allelic expression control necessary for fine-tuning mammalian immune responses.


Subject(s)
Lymphotoxin-alpha/genetics , Lymphotoxin-beta/genetics , RNA, Long Noncoding , Transcriptional Activation , Tumor Necrosis Factor-alpha/genetics , Alleles , Animals , Carrier Proteins/metabolism , Cell Line , Gene Expression Profiling , Gene Expression Regulation , In Situ Hybridization, Fluorescence , Lipopolysaccharides/chemistry , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thyroid Hormones/metabolism , Transcription Factors/metabolism , Thyroid Hormone-Binding Proteins
5.
Proc Natl Acad Sci U S A ; 112(13): E1587-93, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25770220

ABSTRACT

Naive CD4 T cells differentiate into several effector lineages, which generate a stronger and more rapid response to previously encountered immunological challenges. Although effector function is a key feature of adaptive immunity, the molecular basis of this process is poorly understood. Here, we investigated the spatiotemporal regulation of cytokine gene expression in resting and restimulated effector T helper 1 (Th1) cells. We found that the Lymphotoxin (LT)/TNF alleles, which encode TNF-α, were closely juxtaposed shortly after T-cell receptor (TCR) engagement, when transcription factors are limiting. Allelic pairing required a nuclear myosin, myosin VI, which is rapidly recruited to the LT/TNF locus upon restimulation. Furthermore, transcription was paused at the TNF locus and other related genes in resting Th1 cells and released in a myosin VI-dependent manner following activation. We propose that homologous pairing and myosin VI-mediated transcriptional pause release account for the rapid and efficient expression of genes induced by an external stimulus.


Subject(s)
Myosin Heavy Chains/physiology , Th1 Cells/metabolism , Transcription, Genetic , Alleles , Animals , Cell Nucleus/metabolism , Cytokines/metabolism , In Situ Hybridization, Fluorescence , Lymphotoxin-alpha/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Heavy Chains/genetics , RNA Polymerase II/metabolism , Receptors, Antigen, T-Cell/metabolism , Tumor Necrosis Factor-alpha/genetics
6.
Proc Natl Acad Sci U S A ; 110(17): 6955-60, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569250

ABSTRACT

The T helper type 2 (Th2) cytokine genes Il4, Il5, and Il13 are contained within a 140-kb region of mouse chromosome 11 and their expression is controlled by a locus control region (LCR) embedded within this locus. The LCR is composed of a number of DNase I-hypersensitive sites (HSs), which are believed to encompass the regulatory core of the LCR. To determine the function of these sites, mutant mice were generated in which combinations of these HSs had been deleted from the endogenous LCR, and the effect on Th2 cytokine expression was assessed through the use of in vivo and in vitro models. These experiments revealed that, although all of the hypersensitive sites analyzed are important for appropriate LCR function, some sites are more important than others in regulating cytokine expression. Interestingly, each LCR mutation showed contrasting effects on cytokine expression, in some cases with mutants displaying opposing phenotypes between in vitro cultures and in vivo immunizations. These studies indicated that Rad50 hypersensitive site 6 was the singularly most important HS for Th2 cytokine expression, displaying consistent reductions in cytokine levels in all models tested. Furthermore analysis of chromatin modifications revealed that deletion of Rad50 hypersensitive site 6 impacted epigenetic modifications at the promoters of the Il4, Il5, and Il13 genes as well as other regulatory sites within the Th2 locus.


Subject(s)
Cytokines/genetics , Deoxyribonuclease I/metabolism , Gene Expression Regulation/immunology , Locus Control Region/genetics , Th2 Cells/immunology , ATP-Binding Cassette Transporters/metabolism , Acid Anhydride Hydrolases , Analysis of Variance , Animals , Blotting, Western , Chromatin Immunoprecipitation , Cytokines/metabolism , DNA Primers/genetics , DNA-Binding Proteins , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Mutation/genetics , Ovalbumin/administration & dosage , Real-Time Polymerase Chain Reaction
7.
EMBO Rep ; 13(9): 819-26, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22836578

ABSTRACT

Current research on the cytokine-mediated signalling towards the polarization and differentiation of a T-helper cell lineage lacks mechanistic insights on the transcriptional regulation of cytokine receptor genes. Here, we propose a new mechanism for the transcriptional regulation of the interferon gamma receptor 1 gene via long-range intrachromosomal interactions with the Ifnγ locus mediated by the protein CTCF. These interactions sustain the monoallelic expression of the differentially methylated IfnγR1 gene and are persistent on blockade of active transcription. Our findings suggest that regulatory elements for a cytokine gene locus can also positively regulate the transcription of its receptor.


Subject(s)
Epigenesis, Genetic , Receptors, Interferon/genetics , Transcription, Genetic , Alleles , Animals , CCCTC-Binding Factor , CD4-Positive T-Lymphocytes/metabolism , DNA Methylation , Genome , Mice , Receptors, Interferon/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Interferon gamma Receptor
8.
J Immunol ; 184(10): 5686-95, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20404270

ABSTRACT

Nuclear architecture and chromatin reorganization have recently been shown to orchestrate gene expression and act as key players in developmental pathways. To investigate how regulatory elements in the mouse CD8 gene locus are arranged in space and in relation to each other, three-dimensional fluorescence in situ hybridization and chromosome conformation capture techniques were employed to monitor the repositioning of the locus in relation to its subchromosomal territory and to identify long-range interactions between the different elements during development. Our data demonstrate that CD8 gene expression in murine lymphocytes is accompanied by the relocation of the locus outside its subchromosomal territory. Similar observations in the CD4 locus point to a rather general phenomenon during T cell development. Furthermore, we show that this relocation of the CD8 gene locus is associated with a clustering of regulatory elements forming a tight active chromatin hub in CD8-expressing cells. In contrast, in nonexpressing cells, the gene remains close to the main body of its chromosomal domain and the regulatory elements appear not to interact with each other.


Subject(s)
CD8 Antigens/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Nucleus/genetics , Gene Expression Regulation, Developmental/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , CD4 Antigens/genetics , CD8 Antigens/biosynthesis , Chromosome Positioning/genetics , DNA Probes/genetics , Female , Imaging, Three-Dimensional , In Situ Hybridization, Fluorescence , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Protein Structure, Tertiary/genetics , Thymus Gland/cytology
9.
Trends Genet ; 26(4): 188-97, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20236724

ABSTRACT

The nucleus is an ordered three-dimensional entity, and organization of the genome within the nuclear space might have implications for orchestrating gene expression. Recent technological developments have revealed that chromatin is folded into loops bringing distal regulatory elements into intimate contact with the genes that they regulate. Such intrachromosomal contacts appear to be a general mechanism of enhancer-promoter communication in cis. Tantalizing evidence is emerging that regulatory elements might have the capacity to act in trans to regulate genes on other chromosomes. However, unequivocal data required to prove that interchromosomal gene regulation truly represents another level of control within the nucleus is lacking, and this concept remains highly contentious. Such controversy emphasizes that our current understanding of the mechanisms that govern gene expression are far from complete.


Subject(s)
Cell Nucleus/genetics , Chromosomes, Mammalian/metabolism , Gene Expression Regulation , Animals , Chromatin/genetics , Chromosomes, Mammalian/genetics , Enhancer Elements, Genetic , Humans , Promoter Regions, Genetic
10.
Curr Opin Immunol ; 21(2): 153-60, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19375293

ABSTRACT

Differentiation of T(H)1 and T(H)2 effector cells proceeds through several phases: First, naïve CD4(+) precursor cells are instructed to differentiate as appropriate to optimally fight the infectious threat encountered. This process is governed by the IL12 and IL4 cytokines, as well as by signaling through the Notch receptor. In response to these signals, transcription is initiated of lineage specific cytokine genes including the Ifngamma and Il4 genes as well as of genes encoding transcriptional regulators, such as T-bet and Gata3. The respective differentiation programs are reinforced by both positive and negative feedback mechanisms. Furthermore, epigenetic modifications of the lineage specific genes result in the emergence of regulatory elements, which control high level lineage restricted expression by both intrachromosomal and interchromosomal associations. Together, these mechanisms ensure stable inheritance of the differentiated fate in the numerous progeny of the original naïve CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Models, Biological , Th1 Cells/cytology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism
11.
J Physiol ; 586(21): 5161-79, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18787033

ABSTRACT

The rates of activation and unitary properties of Na+-activated K+ (K(Na)) currents have been found to vary substantially in different types of neurones. One class of K(Na) channels is encoded by the Slack gene. We have now determined that alternative RNA splicing gives rise to at least five different transcripts for Slack, which produce Slack channels that differ in their predicted cytoplasmic amino-termini and in their kinetic properties. Two of these, termed Slack-A channels, contain an amino-terminus domain closely resembling that of another class of K(Na) channels encoded by the Slick gene. Neuronal expression of Slack-A channels and of the previously described Slack isoform, now called Slack-B, are driven by independent promoters. Slack-A mRNAs were enriched in the brainstem and olfactory bulb and detected at significant levels in four different brain regions. When expressed in CHO cells, Slack-A channels activate rapidly upon depolarization and, in single channel recordings in Xenopus oocytes, are characterized by multiple subconductance states with only brief transient openings to the fully open state. In contrast, Slack-B channels activate slowly over hundreds of milliseconds, with openings to the fully open state that are approximately 6-fold longer than those for Slack-A channels. In numerical simulations, neurones in which outward currents are dominated by a Slack-A-like conductance adapt very rapidly to repeated or maintained stimulation over a wide range of stimulus strengths. In contrast, Slack-B currents promote rhythmic firing during maintained stimulation, and allow adaptation rate to vary with stimulus strength. Using an antibody that recognizes all amino-termini isoforms of Slack, Slack immunoreactivity is present at locations that have no Slack-B-specific staining, including olfactory bulb glomeruli and the dendrites of hippocampal neurones, suggesting that Slack channels with alternate amino-termini such as Slack-A channels are present at these locations. Our data suggest that alternative promoters of the Slack gene differentially modulate the properties of neurones.


Subject(s)
Action Potentials/physiology , Adaptation, Physiological/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Cloning, Molecular , Gene Expression Regulation/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Potassium Channels/genetics , Potassium Channels, Sodium-Activated , Promoter Regions, Genetic , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
13.
Immunity ; 24(4): 369-79, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16618596

ABSTRACT

Cytokine loci undergo changes in chromatin structure when naive CD4(+) T cells differentiate into Th1 or Th2 cells and have also been examined for regulatory sequences underlying such changes and their functional correlates. Studies have shown that distal regulatory elements control the Ifng and Th2 cytokine loci and are primary targets for tissue-specific transcription factors, serving as centers for epigenetic changes that mark heritable traits in effector cells. Reports of intra- and, remarkably, interchromosomal interactions between these regulatory elements shed light on the mechanisms by which they regulate gene expression, revealing an extraordinary new picture that conceptually extends our views on how genes are regulated from two to three dimensions. Here, we summarize these recent findings on the role of regulatory elements and their mechanisms of action, which are of broad significance for gene regulation, not only of the immune system but also of many, if not all, coregulated genes.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/immunology , Epigenesis, Genetic , Regulatory Elements, Transcriptional , Th2 Cells/cytology , Animals , Cytokines/genetics , Cytokines/immunology , Gene Expression/immunology , Glycosyltransferases/genetics , Glycosyltransferases/immunology , Humans , Th2 Cells/physiology
15.
Eur J Immunol ; 35(12): 3400-4, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16331704

ABSTRACT

The immune system is influenced by environmental factors such as hormones and nutrients. Previous studies have suggested that vitamins A and D influence the process of naive T helper (Th) cell differentiation into Th1 or Th2 cells. Vitamins A and D signal through the retinoid X receptor (RXR), which partners with either the retinoic acid receptor or the vitamin D receptor. Most previous studies into the role of RXR in Th differentiation have been performed in vitro and it was necessary for these to be verified in a physiological environment. However, in vivo study has been hindered since RXRalpha deficient mice are embryonic lethal. Du et al. in this issue of the European Journal of Immunology, overcome this obstacle using "pinkie" mice that harbor a hypomorphic mutation in the Rxralpha gene. The authors report that the mutant mice have an exaggerated Th1 immune response which is attributed to the aberrant antigen-presenting cell and CD4 T cell function. This study confirms previous studies indicating that RXR signaling plays an important role in Th cell differentiation and also provides a valuable tool with which to study the mechanisms and the in vivo functions of this signaling pathway.


Subject(s)
Retinoid X Receptors/genetics , Retinoid X Receptors/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Humans
16.
Nature ; 435(7042): 637-45, 2005 Jun 02.
Article in English | MEDLINE | ID: mdl-15880101

ABSTRACT

The T-helper-cell 1 and 2 (T(H)1 and T(H)2) pathways, defined by cytokines interferon-gamma (IFN-gamma) and interleukin-4 (IL-4), respectively, comprise two alternative CD4+ T-cell fates, with functional consequences for the host immune system. These cytokine genes are encoded on different chromosomes. The recently described T(H)2 locus control region (LCR) coordinately regulates the T(H)2 cytokine genes by participating in a complex between the LCR and promoters of the cytokine genes Il4, Il5 and Il13. Although they are spread over 120 kilobases, these elements are closely juxtaposed in the nucleus in a poised chromatin conformation. In addition to these intrachromosomal interactions, we now describe interchromosomal interactions between the promoter region of the IFN-gamma gene on chromosome 10 and the regulatory regions of the T(H)2 cytokine locus on chromosome 11. DNase I hypersensitive sites that comprise the T(H)2 LCR developmentally regulate these interchromosomal interactions. Furthermore, there seems to be a cell-type-specific dynamic interaction between interacting chromatin partners whereby interchromosomal interactions are apparently lost in favour of intrachromosomal ones upon gene activation. Thus, we provide an example of eukaryotic genes located on separate chromosomes associating physically in the nucleus via interactions that may have a function in coordinating gene expression.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Chromosome Positioning/genetics , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , Cytokines/genetics , Gene Expression Regulation , Alleles , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic/genetics , In Situ Hybridization, Fluorescence , Interferon-gamma/genetics , Interleukins/genetics , Locus Control Region/genetics , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Th1 Cells/cytology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism , Transcriptional Activation
17.
Nat Immunol ; 6(1): 42-8, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15608641

ABSTRACT

Several regulatory regions are important for the expression of genes encoding T helper type 2 (T(H)2) cytokines, including T(H)2-specific DNase I hypersensitivity sites in the T(H)2 cytokine locus control region. Among these sites, Rad50 hypersensitive site 7 (RHS7) shows rapid T(H)2-specific demethylation after antigenic stimulation. To investigate the function of RHS7 in T(H)2 cell differentiation, we have generated RHS7-deficient mice. CD4(+) T cells and mast cells showed a notable reduction in T(H)2 cytokine expression in vitro and T(H)2 responses in vivo were considerably impaired in RHS7-deficient mice. Deletion of RHS7 did not affect the expression of a linked Rad50 gene, but it did reduce long-range intrachromosomal interactions between the locus control region and promoters of the T(H)2 cytokine genes. Our findings show that RHS7 is essential for the proper regulation of T(H)2 cytokine gene expression.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Cytokines/immunology , Locus Control Region/genetics , Th2 Cells/immunology , ATP-Binding Cassette Transporters/genetics , Acid Anhydride Hydrolases , Animals , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins , Mice , Mice, Inbred C57BL
18.
Nat Immunol ; 5(10): 1017-27, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15378057

ABSTRACT

The T helper type 2 (T(H)2) locus control region is important in the regulation of the genes encoding the cytokines interleukins 4, 5 and 13. Using the chromosome conformation capture technique, we found that in T cells, natural killer cells, B cells and fibroblasts, the promoters for the genes encoding T(H)2 cytokines are located in close spatial proximity, forming an initial chromatin core configuration. In CD4(+) T cells and natural killer cells, but not B cells and fibroblasts, the T(H)2 locus control region participates in this configuration. The transcription factors GATA3 and STAT6 are essential for the establishment and/or maintenance of these interactions. Intrachromosomal interactions in the T(H)2 cytokine locus may form the basis for the coordinated transcriptional regulation of cytokine-encoding genes by the T(H)2 locus control region.


Subject(s)
Cytokines/genetics , Locus Control Region , Th2 Cells/immunology , Animals , Calcium/physiology , Chromatin/chemistry , Chromosome Mapping , DNA-Binding Proteins/physiology , GATA3 Transcription Factor , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Conformation , STAT6 Transcription Factor , Trans-Activators/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...