Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1242481, 2023.
Article in English | MEDLINE | ID: mdl-37635874

ABSTRACT

Intra-thymic T cell development is coordinated by the regulatory actions of SATB1 genome organizer. In this report, we show that SATB1 is involved in the regulation of transcription and splicing, both of which displayed deregulation in Satb1 knockout murine thymocytes. More importantly, we characterized a novel SATB1 protein isoform and described its distinct biophysical behavior, implicating potential functional differences compared to the commonly studied isoform. SATB1 utilized its prion-like domains to transition through liquid-like states to aggregated structures. This behavior was dependent on protein concentration as well as phosphorylation and interaction with nuclear RNA. Notably, the long SATB1 isoform was more prone to aggregate following phase separation. Thus, the tight regulation of SATB1 isoforms expression levels alongside with protein post-translational modifications, are imperative for SATB1's mode of action in T cell development. Our data indicate that deregulation of these processes may also be linked to disorders such as cancer.

2.
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
3.
Nat Commun ; 13(1): 6954, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36376298

ABSTRACT

Mechanisms of tissue-specific gene expression regulation via 3D genome organization are poorly understood. Here we uncover the regulatory chromatin network of developing T cells and identify SATB1, a tissue-specific genome organizer, enriched at the anchors of promoter-enhancer loops. We have generated a T-cell specific Satb1 conditional knockout mouse which allows us to infer the molecular mechanisms responsible for the deregulation of its immune system. H3K27ac HiChIP and Hi-C experiments indicate that SATB1-dependent promoter-enhancer loops regulate expression of master regulator genes (such as Bcl6), the T cell receptor locus and adhesion molecule genes, collectively being critical for cell lineage specification and immune system homeostasis. SATB1-dependent regulatory chromatin loops represent a more refined layer of genome organization built upon a high-order scaffold provided by CTCF and other factors. Overall, our findings unravel the function of a tissue-specific factor that controls transcription programs, via spatial chromatin arrangements complementary to the chromatin structure imposed by ubiquitously expressed genome organizers.


Subject(s)
Matrix Attachment Region Binding Proteins , Mice , Animals , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes/metabolism , Chromatin/genetics , Genome , Promoter Regions, Genetic/genetics
4.
Front Immunol ; 13: 921375, 2022.
Article in English | MEDLINE | ID: mdl-35812421

ABSTRACT

In the heart of innate and adaptive immunity lies the proper spatiotemporal development of several immune cell lineages. Multiple studies have highlighted the necessity of epigenetic and transcriptional regulation in cell lineage specification. This mode of regulation is mediated by transcription factors and chromatin remodelers, controlling developmentally essential gene sets. The core of transcription and epigenetic regulation is formulated by different epigenetic modifications determining gene expression. Apart from "classic" epigenetic modifications, 3D chromatin architecture is also purported to exert fundamental roles in gene regulation. Chromatin conformation both facilitates cell-specific factor binding at specified regions and is in turn modified as such, acting synergistically. The interplay between global and tissue-specific protein factors dictates the epigenetic landscape of T and innate lymphoid cell (ILC) lineages. The expression of global genome organizers such as CTCF, YY1, and the cohesin complexes, closely cooperate with tissue-specific factors to exert cell type-specific gene regulation. Special AT-rich binding protein 1 (SATB1) is an important tissue-specific genome organizer and regulator controlling both long- and short-range chromatin interactions. Recent indications point to SATB1's cooperation with the aforementioned factors, linking global to tissue-specific gene regulation. Changes in 3D genome organization are of vital importance for proper cell development and function, while disruption of this mechanism can lead to severe immuno-developmental defects. Newly emerging data have inextricably linked chromatin architecture deregulation to tissue-specific pathophysiological phenotypes. The combination of these findings may shed light on the mechanisms behind pathological conditions.


Subject(s)
Matrix Attachment Region Binding Proteins , Chromatin/genetics , Epigenesis, Genetic , Immunity, Innate , Lymphocytes/metabolism , Matrix Attachment Region Binding Proteins/genetics , T-Lymphocytes/metabolism
5.
Methods Protoc ; 4(3)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34287381

ABSTRACT

The functional implications of the three-dimensional genome organization are becoming increasingly recognized. The Hi-C and HiChIP research approaches belong among the most popular choices for probing long-range chromatin interactions. A few methodical protocols have been published so far, yet their reproducibility and efficiency may vary. Most importantly, the high frequency of the dangling ends may dramatically affect the number of usable reads mapped to valid interaction pairs. Additionally, more obstacles arise from the chromatin compactness of certain investigated cell types, such as primary T cells, which due to their small and compact nuclei, impede limitations for their use in various genomic approaches. Here we systematically optimized all the major steps of the HiChIP protocol in T cells. As a result, we reduced the number of dangling ends to nearly zero and increased the proportion of long-range interaction pairs. Moreover, using three different mouse genotypes and multiple biological replicates, we demonstrated the high reproducibility of the optimized protocol. Although our primary goal was to optimize HiChIP, we also successfully applied the optimized steps to Hi-C, given their significant protocol overlap. Overall, we describe the rationale behind every optimization step, followed by a detailed protocol for both HiChIP and Hi-C experiments.

6.
Nucleus ; 11(1): 117-131, 2020 12.
Article in English | MEDLINE | ID: mdl-33356851

ABSTRACT

The regulatory circuits that define developmental decisions of thymocytes are still incompletely resolved. SATB1 protein is predominantly expressed at the CD4+CD8+cell stage exerting its broad transcription regulation potential with both activatory and repressive roles. A series of post-translational modifications and the presence of potential SATB1 protein isoforms indicate the complexity of its regulatory potential. The most apparent mechanism of its involvement in gene expression regulation is via the orchestration of long-range chromatin loops between genes and their regulatory elements. Multiple SATB1 perturbations in mice uncovered a link to autoimmune diseases while clinical investigations on cancer research uncovered that SATB1 has a promoting role in several types of cancer and can be used as a prognostic biomarker. SATB1 is a multivalent tissue-specific factor with a broad and yet undetermined regulatory potential. Future investigations on this protein could further uncover T cell-specific regulatory pathways and link them to (patho)physiology.


Subject(s)
Autoimmune Diseases/metabolism , Biomarkers, Tumor/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chromatin/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Biomarkers, Tumor/genetics , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Chromatin/genetics , Chromatin/pathology , Humans , Matrix Attachment Region Binding Proteins/genetics , Mice , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology
7.
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
8.
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
9.
PLoS One ; 12(9): e0184788, 2017.
Article in English | MEDLINE | ID: mdl-28910376

ABSTRACT

It is becoming increasingly appreciated that the non-coding genome may have a great impact on the regulation of chromatin structure and gene expression. The innate immune response can be mediated upon lipopolysaccharide stimulation of macrophages which leads to immediate transcriptional activation of early responsive genes including tumor necrosis factor alpha (Tnfα). The functional role of non-coding RNAs, such as lncRNAs and microRNAs, on the transcriptional activation of proinflammatory genes and the subsequent regulation of the innate immune response is still lacking mechanistic insights. In this study we wanted to unravel the functional role of the lncRNA SeT, which is encoded from the murine Tnfα gene locus, and miR-155 on the transcriptional regulation of the Tnfα gene. We utilized genetically modified mice harboring either a deletion of the SeT promoter elements or the mature miR-155 and studied the response of macrophages to lipopolysaccharide (LPS) stimulation. We found that decreased expression of the lncRNA SeT in murine primary macrophages resulted in increased mortality of mice challenged with LPS, which was corroborated by increased Tnfα steady state mRNA levels and a higher frequency of biallelically expressing macrophages. On the contrary, miR-155 deletion resulted in reduced Tnfα mRNA levels supported by a lower frequency of biallelically expressing macrophages upon stimulation with LPS. In both cases, in the absence of either lncRNA SeT or miR-155 we observed a deregulation of the Tnfα allele homologous pairing, previously shown to regulate the switch from mono- to bi-allelic gene expression. Although lncRNA SeT was not found to be a direct target of miR-155 its stability was increased upon miR-155 deletion. This study suggests a role of the non-coding genome in mediating Tnfα mRNA dosage control based on the regulation of homologous pairing of gene alleles and their subsequent biallelic expression.


Subject(s)
Gene Expression Profiling/methods , Macrophages/cytology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Tumor Necrosis Factor-alpha/genetics , Alleles , Animals , Cells, Cultured , Gene Expression Regulation , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic
10.
J Immunol ; 195(12): 5750-61, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26538391

ABSTRACT

Endotoxin tolerance occurs to protect the organism from hyperactivation of innate immune responses, primarily mediated by macrophages. Regulation of endotoxin tolerance occurs at multiple levels of cell responses and requires significant changes in gene expression. In the process of macrophage activation, induced expression of microRNA (miR)-155 and miR-146a contributes to the regulation of the inflammatory response and endotoxin tolerance. In this article, we demonstrate that expression of both miRNAs is coordinately regulated during endotoxin tolerance by a complex mechanism that involves monoallelic interchromosomal association, alterations in histone methyl marks, and transcription factor binding. Upon activation of naive macrophages, Histone3 was trimethylated at lysine4 and NFκBp65 was bound on both miR-155 and miR-146a gene loci. However, at the stage of endotoxin tolerance, both miR gene loci were occupied by C/EBPß, NFκBp50, and the repressive Histone3 marks trimethylation of K9 of H3. DNA fluorescence in situ hybridization experiments revealed monoallelic interchromosomal colocalization of miR-155 and miR-146a gene loci at the stage of endotoxin tolerance, whereas RNA-DNA-fluorescence in situ hybridization experiments showed that the colocalized alleles were silenced, suggesting a common repression mechanism. Genetic ablation of Akt1, which is known to abrogate endotoxin tolerance, abolished induction of loci colocalization and C/EBPß binding, further supporting that this mechanism occurs specifically in endotoxin tolerance. Overall, this study demonstrates that two miRNAs are coordinately regulated via gene colocalization at the three-dimensional chromatin space, same transcriptional machinery, and similar Histone3 methylation profile, contributing to the development of endotoxin tolerance.


Subject(s)
Macrophages, Peritoneal/immunology , MicroRNAs/metabolism , Oncogene Protein v-akt/metabolism , Shock, Septic/immunology , Alleles , Animals , Cell Line , Cells, Cultured , Chromatin Assembly and Disassembly , Endotoxins/immunology , Epigenetic Repression , Immune Tolerance , Immunity, Innate , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Oncogene Protein v-akt/genetics , RNA Interference , RNA, Small Interfering/genetics
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
Proc Natl Acad Sci U S A ; 105(8): 2853-8, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18287022

ABSTRACT

Cilia are present on nearly all cell types in mammals and perform remarkably diverse functions. However, the mechanisms underlying ciliogenesis are unclear. Here, we cloned a previously uncharacterized highly conserved gene, stumpy, located on mouse chromosome 7. Stumpy was ubiquitously expressed, and conditional loss in mouse resulted in complete penetrance of perinatal hydrocephalus (HC) and severe polycystic kidney disease (PKD). We found that cilia in stumpy mutant brain and kidney cells were absent or markedly deformed, resulting in defective flow of cerebrospinal fluid. Stumpy colocalized with ciliary basal bodies, physically interacted with gamma-tubulin, and was present along ciliary axonemes, suggesting that stumpy plays a role in ciliary axoneme extension. Therefore, stumpy is essential for ciliogenesis and may be involved in the pathogenesis of human congenital malformations such as HC and PKD.


Subject(s)
Cilia/physiology , Genetic Predisposition to Disease , Hydrocephalus/genetics , Polycystic Kidney Diseases/genetics , Animals , Base Sequence , Blotting, Northern , Brain/pathology , Cloning, Molecular , Computational Biology , Gene Expression Profiling , Histocytochemistry , Hydrocephalus/metabolism , In Situ Hybridization , Kidney/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polycystic Kidney Diseases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...