Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 51(20): 10992-11009, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37791849

ABSTRACT

A wide range of nuclear proteins are involved in the spatio-temporal organization of the genome through diverse biological processes such as gene transcription and DNA replication. Upon stimulation by testosterone and translocation to the nucleus, multiple androgen receptors (ARs) accumulate in microscopically discernable foci which are irregularly distributed in the nucleus. Here, we investigated the formation and physical nature of these foci, by combining novel fluorescent labeling techniques to visualize a defined chromatin locus of AR-regulated genes-PTPRN2 or BANP-simultaneously with either AR foci or individual AR molecules. Quantitative colocalization analysis showed evidence of AR foci formation induced by R1881 at both PTPRN2 and BANP loci. Furthermore, single-particle tracking (SPT) revealed three distinct subdiffusive fractional Brownian motion (fBm) states: immobilized ARs were observed near the labeled genes likely as a consequence of DNA-binding, while the intermediate confined state showed a similar spatial behavior but with larger displacements, suggesting compartmentalization by liquid-liquid phase separation (LLPS), while freely mobile ARs were diffusing in the nuclear environment. All together, we show for the first time in living cells the presence of AR-regulated genes in AR foci.


Subject(s)
Cell Nucleus , Receptors, Androgen , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Receptors, Androgen/metabolism , Humans , Mice , Cell Line, Tumor
2.
Development ; 147(10)2020 05 26.
Article in English | MEDLINE | ID: mdl-32253238

ABSTRACT

The transcription factor Zeb2 controls fate specification and subsequent differentiation and maturation of multiple cell types in various embryonic tissues. It binds many protein partners, including activated Smad proteins and the NuRD co-repressor complex. How Zeb2 subdomains support cell differentiation in various contexts has remained elusive. Here, we studied the role of Zeb2 and its domains in neurogenesis and neural differentiation in the young postnatal ventricular-subventricular zone (V-SVZ), in which neural stem cells generate olfactory bulb-destined interneurons. Conditional Zeb2 knockouts and separate acute loss- and gain-of-function approaches indicated that Zeb2 is essential for controlling apoptosis and neuronal differentiation of V-SVZ progenitors before and after birth, and we identified Sox6 as a potential downstream target gene of Zeb2. Zeb2 genetic inactivation impaired the differentiation potential of the V-SVZ niche in a cell-autonomous fashion. We also provide evidence that its normal function in the V-SVZ also involves non-autonomous mechanisms. Additionally, we demonstrate distinct roles for Zeb2 protein-binding domains, suggesting that Zeb2 partners co-determine neuronal output from the mouse V-SVZ in both quantitative and qualitative ways in early postnatal life.


Subject(s)
Lateral Ventricles/embryology , Lateral Ventricles/growth & development , Neurogenesis/genetics , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Apoptosis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Gene Knockout Techniques , Interneurons/metabolism , Lateral Ventricles/metabolism , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Olfactory Bulb/metabolism , SOXD Transcription Factors/metabolism , Signal Transduction/immunology , Zinc Finger E-box Binding Homeobox 2/genetics
3.
Cell ; 133(2): 265-79, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423198

ABSTRACT

The immunoglobulin heavy-chain (Igh) locus is organized into distinct regions that contain multiple variable (V(H)), diversity (D(H)), joining (J(H)) and constant (C(H)) coding elements. How the Igh locus is structured in 3D space is unknown. To probe the topography of the Igh locus, spatial distance distributions were determined between 12 genomic markers that span the entire Igh locus. Comparison of the distance distributions to computer simulations of alternative chromatin arrangements predicted that the Igh locus is organized into compartments containing clusters of loops separated by linkers. Trilateration and triple-point angle measurements indicated the mean relative 3D positions of the V(H), D(H), J(H), and C(H) elements, showed compartmentalization and striking conformational changes involving V(H) and D(H)-J(H) elements during early B cell development. In pro-B cells, the entire repertoire of V(H) regions (2 Mbp) appeared to have merged and juxtaposed to the D(H) elements, mechanistically permitting long-range genomic interactions to occur with relatively high frequency.


Subject(s)
Genes, Immunoglobulin Heavy Chain , Animals , B-Lymphocytes/chemistry , B-Lymphocytes/metabolism , Cell Lineage , Cells, Cultured , Mice , Mice, Inbred C57BL , Models, Molecular , Nucleic Acid Conformation , VDJ Exons
4.
Hum Mol Genet ; 29(15): 2535-2550, 2020 08 29.
Article in English | MEDLINE | ID: mdl-32628253

ABSTRACT

The transcription factor zinc finger E-box binding protein 2 (ZEB2) controls embryonic and adult cell fate decisions and cellular maturation in many stem/progenitor cell types. Defects in these processes in specific cell types underlie several aspects of Mowat-Wilson syndrome (MOWS), which is caused by ZEB2 haplo-insufficiency. Human ZEB2, like mouse Zeb2, is located on chromosome 2 downstream of a ±3.5 Mb-long gene-desert, lacking any protein-coding gene. Using temporal targeted chromatin capture (T2C), we show major chromatin structural changes based on mapping in-cis proximities between the ZEB2 promoter and this gene desert during neural differentiation of human-induced pluripotent stem cells, including at early neuroprogenitor cell (NPC)/rosette state, where ZEB2 mRNA levels increase significantly. Combining T2C with histone-3 acetylation mapping, we identified three novel candidate enhancers about 500 kb upstream of the ZEB2 transcription start site. Functional luciferase-based assays in heterologous cells and NPCs reveal co-operation between these three enhancers. This study is the first to document in-cis Regulatory Elements located in ZEB2's gene desert. The results further show the usability of T2C for future studies of ZEB2 REs in differentiation and maturation of multiple cell types and the molecular characterization of newly identified MOWS patients that lack mutations in ZEB2 protein-coding exons.


Subject(s)
Chromatin/ultrastructure , Enhancer Elements, Genetic/genetics , Hirschsprung Disease/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Zinc Finger E-box Binding Homeobox 2/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Facies , Gene Expression Regulation/genetics , Hirschsprung Disease/pathology , Homeodomain Proteins/genetics , Humans , Intellectual Disability/pathology , Mice , Microcephaly/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/ultrastructure , Regulatory Sequences, Nucleic Acid
5.
Stem Cells ; 38(2): 202-217, 2020 02.
Article in English | MEDLINE | ID: mdl-31675135

ABSTRACT

Cooperative actions of extrinsic signals and cell-intrinsic transcription factors alter gene regulatory networks enabling cells to respond appropriately to environmental cues. Signaling by transforming growth factor type ß (TGFß) family ligands (eg, bone morphogenetic proteins [BMPs] and Activin/Nodal) exerts cell-type specific and context-dependent transcriptional changes, thereby steering cellular transitions throughout embryogenesis. Little is known about coordinated regulation and transcriptional interplay of the TGFß system. To understand intrafamily transcriptional regulation as part of this system's actions during development, we selected 95 of its components and investigated their mRNA-expression dynamics, gene-gene interactions, and single-cell expression heterogeneity in mouse embryonic stem cells transiting to neural progenitors. Interrogation at 24 hour intervals identified four types of temporal gene transcription profiles that capture all stages, that is, pluripotency, epiblast formation, and neural commitment. Then, between each stage we performed esiRNA-based perturbation of each individual component and documented the effect on steady-state mRNA levels of the remaining 94 components. This exposed an intricate system of multilevel regulation whereby the majority of gene-gene interactions display a marked cell-stage specific behavior. Furthermore, single-cell RNA-profiling at individual stages demonstrated the presence of detailed co-expression modules and subpopulations showing stable co-expression modules such as that of the core pluripotency genes at all stages. Our combinatorial experimental approach demonstrates how intrinsically complex transcriptional regulation within a given pathway is during cell fate/state transitions.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Embryonic Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Cell Differentiation , Humans
6.
Hum Genomics ; 14(1): 39, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33066815

ABSTRACT

The expression of the human ß-like globin genes follows a well-orchestrated developmental pattern, undergoing two essential switches, the first one during the first weeks of gestation (ε to γ), and the second one during the perinatal period (γ to ß). The γ- to ß-globin gene switching mechanism includes suppression of fetal (γ-globin, HbF) and activation of adult (ß-globin, HbA) globin gene transcription. In hereditary persistence of fetal hemoglobin (HPFH), the γ-globin suppression mechanism is impaired leaving these individuals with unusual elevated levels of fetal hemoglobin (HbF) in adulthood. Recently, the transcription factors KLF1 and BCL11A have been established as master regulators of the γ- to ß-globin switch. Previously, a genomic variant in the KLF1 gene, identified by linkage analysis performed on twenty-seven members of a Maltese family, was found to be associated with HPFH. However, variation in the levels of HbF among family members, and those from other reported families carrying genetic variants in KLF1, suggests additional contributors to globin switching. ASF1B was downregulated in the family members with HPFH. Here, we investigate the role of ASF1B in γ- to ß-globin switching and erythropoiesis in vivo. Mouse-human interspecies ASF1B protein identity is 91.6%. By means of knockdown functional assays in human primary erythroid cultures and analysis of the erythroid lineage in Asf1b knockout mice, we provide evidence that ASF1B is a novel contributor to steady-state erythroid differentiation, and while its loss affects the balance of globin expression, it has no major role in hemoglobin switching.


Subject(s)
Cell Cycle Proteins/genetics , Erythropoiesis/genetics , Histone Chaperones/genetics , beta-Globins/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Line , Gene Expression Regulation , HEK293 Cells , Histone Chaperones/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice, Knockout , Polymorphism, Single Nucleotide , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , gamma-Globins/genetics
7.
Nature ; 523(7558): 53-8, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26106861

ABSTRACT

In response to DNA damage, tissue homoeostasis is ensured by protein networks promoting DNA repair, cell cycle arrest or apoptosis. DNA damage response signalling pathways coordinate these processes, partly by propagating gene-expression-modulating signals. DNA damage influences not only the abundance of messenger RNAs, but also their coding information through alternative splicing. Here we show that transcription-blocking DNA lesions promote chromatin displacement of late-stage spliceosomes and initiate a positive feedback loop centred on the signalling kinase ATM. We propose that initial spliceosome displacement and subsequent R-loop formation is triggered by pausing of RNA polymerase at DNA lesions. In turn, R-loops activate ATM, which signals to impede spliceosome organization further and augment ultraviolet-irradiation-triggered alternative splicing at the genome-wide level. Our findings define R-loop-dependent ATM activation by transcription-blocking lesions as an important event in the DNA damage response of non-replicating cells, and highlight a key role for spliceosome displacement in this process.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage/physiology , Signal Transduction , Spliceosomes/metabolism , Alternative Splicing/physiology , Cell Line , Chromatin/metabolism , DNA-Directed RNA Polymerases/metabolism , Enzyme Activation , Humans , Ultraviolet Rays
8.
Genes Dev ; 27(7): 767-77, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23592796

ABSTRACT

Transcription steps are marked by different modifications of the C-terminal domain of RNA polymerase II (RNAPII). Phosphorylation of Ser5 and Ser7 by cyclin-dependent kinase 7 (CDK7) as part of TFIIH marks initiation, whereas phosphorylation of Ser2 by CDK9 marks elongation. These processes are thought to take place in localized transcription foci in the nucleus, known as "transcription factories," but it has been argued that the observed clusters/foci are mere fixation or labeling artifacts. We show that transcription factories exist in living cells as distinct foci by live-imaging fluorescently labeled CDK9, a kinase known to associate with active RNAPII. These foci were observed in different cell types derived from CDK9-mCherry knock-in mice. We show that these foci are very stable while highly dynamic in exchanging CDK9. Chromatin immunoprecipitation (ChIP) coupled with deep sequencing (ChIP-seq) data show that the genome-wide binding sites of CDK9 and initiating RNAPII overlap on transcribed genes. Immunostaining shows that CDK9-mCherry foci colocalize with RNAPII-Ser5P, much less with RNAPII-Ser2P, and not with CDK12 (a kinase reported to be involved in the Ser2 phosphorylation) or with splicing factor SC35. In conclusion, transcription factories exist in living cells, and initiation and elongation of transcripts takes place in different nuclear compartments.


Subject(s)
RNA Polymerase II/metabolism , Time-Lapse Imaging , Transcription, Genetic , Animals , Cells, Cultured , Cyclin-Dependent Kinase 9/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Protein Structure, Tertiary , Protein Transport , RNA Polymerase II/chemistry , Red Fluorescent Protein
9.
Haematologica ; 105(7): 1802-1812, 2020 07.
Article in English | MEDLINE | ID: mdl-31582556

ABSTRACT

GATA1 is an essential transcriptional regulator of myeloid hematopoietic differentiation towards red blood cells. During erythroid differentiation, GATA1 forms different complexes with other transcription factors such as LDB1, TAL1, E2A and LMO2 ("the LDB1 complex") or with FOG1. The functions of GATA1 complexes have been studied extensively in definitive erythroid differentiation; however, the temporal and spatial formation of these complexes during erythroid development is unknown. We applied proximity ligation assay (PLA) to detect, localize and quantify individual interactions during embryonic stem cell differentiation and in mouse fetal liver (FL) tissue. We show that GATA1/LDB1 interactions appear before the proerythroblast stage and increase in a subset of the CD71+/TER119- cells to activate the terminal erythroid differentiation program in 12.5 day FL. Using Ldb1 and Gata1 knockdown FL cells, we studied the functional contribution of the GATA1/LDB1 complex during differentiation. This shows that the active LDB1 complex appears quite late at the proerythroblast stage of differentiation and confirms the power of PLA in studying the dynamic interaction of proteins in cell differentiation at the single cell level. We provide dynamic insight into the temporal and spatial formation of the GATA1 and LDB1 transcription factor complexes during hematopoietic development and differentiation.


Subject(s)
Embryonic Stem Cells/cytology , GATA1 Transcription Factor , LIM Domain Proteins , Animals , Cell Differentiation , DNA-Binding Proteins , GATA1 Transcription Factor/genetics , Liver , Mice , Transcription Factors
10.
Genome Res ; 26(11): 1478-1489, 2016 11.
Article in English | MEDLINE | ID: mdl-27633323

ABSTRACT

Mammalian cells have developed intricate mechanisms to interpret, integrate, and respond to extracellular stimuli. For example, tumor necrosis factor (TNF) rapidly activates proinflammatory genes, but our understanding of how this occurs against the ongoing transcriptional program of the cell is far from complete. Here, we monitor the early phase of this cascade at high spatiotemporal resolution in TNF-stimulated human endothelial cells. NF-κB, the transcription factor complex driving the response, interferes with the regulatory machinery by binding active enhancers already in interaction with gene promoters. Notably, >50% of these enhancers do not encode canonical NF-κB binding motifs. Using a combination of genomics tools, we find that binding site selection plays a key role in NF-κΒ-mediated transcriptional activation and repression. We demonstrate the latter by describing the synergy between NF-κΒ and the corepressor JDP2. Finally, detailed analysis of a 2.8-Mbp locus using sub-kbp-resolution targeted chromatin conformation capture and genome editing uncovers how NF-κΒ that has just entered the nucleus exploits pre-existing chromatin looping to exert its multimodal role. This work highlights the involvement of topology in cis-regulatory element function during acute transcriptional responses, where primary DNA sequence and its higher-order structure constitute a regulatory context leading to either gene activation or repression.


Subject(s)
Consensus Sequence , NF-kappa B/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Cells, Cultured , Chromatin/metabolism , Gene Editing , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , NF-kappa B/genetics , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
Mol Syst Biol ; 14(6): e8214, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29858282

ABSTRACT

The last decade has radically renewed our understanding of higher order chromatin folding in the eukaryotic nucleus. As a result, most current models are in support of a mostly hierarchical and relatively stable folding of chromosomes dividing chromosomal territories into A- (active) and B- (inactive) compartments, which are then further partitioned into topologically associating domains (TADs), each of which is made up from multiple loops stabilized mainly by the CTCF and cohesin chromatin-binding complexes. Nonetheless, the structure-to-function relationship of eukaryotic genomes is still not well understood. Here, we focus on recent work highlighting the biophysical and regulatory forces that contribute to the spatial organization of genomes, and we propose that the various conformations that chromatin assumes are not so much the result of a linear hierarchy, but rather of both converging and conflicting dynamic forces that act on it.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/chemistry , Eukaryota/genetics , Genome , Animals , Humans , Nucleic Acid Conformation , Protein Folding , Transcription, Genetic
12.
Stem Cells ; 35(3): 611-625, 2017 03.
Article in English | MEDLINE | ID: mdl-27739137

ABSTRACT

In human embryonic stem cells (ESCs) the transcription factor Zeb2 regulates neuroectoderm versus mesendoderm formation, but it is unclear how Zeb2 affects the global transcriptional regulatory network in these cell-fate decisions. We generated Zeb2 knockout (KO) mouse ESCs, subjected them as embryoid bodies (EBs) to neural and general differentiation and carried out temporal RNA-sequencing (RNA-seq) and reduced representation bisulfite sequencing (RRBS) analysis in neural differentiation. This shows that Zeb2 acts preferentially as a transcriptional repressor associated with developmental progression and that Zeb2 KO ESCs can exit from their naïve state. However, most cells in these EBs stall in an early epiblast-like state and are impaired in both neural and mesendodermal differentiation. Genes involved in pluripotency, epithelial-to-mesenchymal transition (EMT), and DNA-(de)methylation, including Tet1, are deregulated in the absence of Zeb2. The observed elevated Tet1 levels in the mutant cells and the knowledge of previously mapped Tet1-binding sites correlate with loss-of-methylation in neural-stimulating conditions, however, after the cells initially acquired the correct DNA-methyl marks. Interestingly, cells from such Zeb2 KO EBs maintain the ability to re-adapt to 2i + LIF conditions even after prolonged differentiation, while knockdown of Tet1 partially rescues their impaired differentiation. Hence, in addition to its role in EMT, Zeb2 is critical in ESCs for exit from the epiblast state, and links the pluripotency network and DNA-methylation with irreversible commitment to differentiation. Stem Cells 2017;35:611-625.


Subject(s)
Cell Lineage , Germ Layers/cytology , Germ Layers/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Cell Differentiation , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Mice , Mice, Knockout , Neurons/cytology , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Principal Component Analysis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Sequence Analysis, RNA , Transcription, Genetic
13.
Mol Syst Biol ; 12(12): 891, 2016 Dec 09.
Article in English | MEDLINE | ID: mdl-27940490

ABSTRACT

Mammalian interphase chromosomes fold into a multitude of loops to fit the confines of cell nuclei, and looping is tightly linked to regulated function. Chromosome conformation capture (3C) technology has significantly advanced our understanding of this structure-to-function relationship. However, all 3C-based methods rely on chemical cross-linking to stabilize spatial interactions. This step remains a "black box" as regards the biases it may introduce, and some discrepancies between microscopy and 3C studies have now been reported. To address these concerns, we developed "i3C", a novel approach for capturing spatial interactions without a need for cross-linking. We apply i3C to intact nuclei of living cells and exploit native forces that stabilize chromatin folding. Using different cell types and loci, computational modeling, and a methylation-based orthogonal validation method, "TALE-iD", we show that native interactions resemble cross-linked ones, but display improved signal-to-noise ratios and are more focal on regulatory elements and CTCF sites, while strictly abiding to topologically associating domain restrictions.


Subject(s)
Cell Nucleus/genetics , Chromosomes, Human/chemistry , Chromosomes, Human/genetics , Animals , High-Throughput Nucleotide Sequencing/methods , Human Umbilical Vein Endothelial Cells , Humans , Interphase , K562 Cells , Mammals/genetics , Models, Genetic , Nucleic Acid Conformation , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA/methods
14.
Proc Natl Acad Sci U S A ; 111(3): 996-1001, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24335803

ABSTRACT

Recent studies of genome-wide chromatin interactions have revealed that the human genome is partitioned into many self-associating topological domains. The boundary sequences between domains are enriched for binding sites of CTCC-binding factor (CTCF) and the cohesin complex, implicating these two factors in the establishment or maintenance of topological domains. To determine the role of cohesin and CTCF in higher-order chromatin architecture in human cells, we depleted the cohesin complex or CTCF and examined the consequences of loss of these factors on higher-order chromatin organization, as well as the transcriptome. We observed a general loss of local chromatin interactions upon disruption of cohesin, but the topological domains remain intact. However, we found that depletion of CTCF not only reduced intradomain interactions but also increased interdomain interactions. Furthermore, distinct groups of genes become misregulated upon depletion of cohesin and CTCF. Taken together, these observations suggest that CTCF and cohesin contribute differentially to chromatin organization and gene regulation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , Repressor Proteins/metabolism , Binding Sites , CCCTC-Binding Factor , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA-Binding Proteins , Gene Expression Profiling , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Mitosis , Multigene Family , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , Transcriptome , Cohesins
15.
Nucleic Acids Res ; 42(21)2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25260593

ABSTRACT

The characterization of transcription factor complexes and their binding sites in the genome by affinity purification has yielded tremendous new insights into how genes are regulated. The affinity purification requires either the use of antibodies raised against the factor of interest itself or by high-affinity binding of a C- or N-terminally added tag sequence to the factor. Unfortunately, fusing extra amino acids to the termini of a factor can interfere with its biological function or the tag may be inaccessible inside the protein. Here, we describe an effective solution to that problem by integrating the 'tag' close to the nuclear localization sequence domain of the factor. We demonstrate the effectiveness of this approach with the transcription factors Fli-1 and Irf2bp2, which cannot be tagged at their extremities without loss of function. This resulted in the identification of novel proteins partners and a new hypothesis on the contribution of Fli-1 to hematopoiesis.


Subject(s)
Nuclear Localization Signals , Nuclear Proteins/analysis , Transcription Factors/analysis , Animals , Cell Differentiation , Cell Line, Tumor , Nuclear Proteins/chemistry , Proto-Oncogene Protein c-fli-1/analysis , Proto-Oncogene Protein c-fli-1/chemistry , Proto-Oncogene Protein c-fli-1/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
16.
Am J Hum Genet ; 91(3): 533-40, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22939636

ABSTRACT

Polymicrogyria is a malformation of the developing cerebral cortex caused by abnormal organization and characterized by many small gyri and fusion of the outer molecular layer. We have identified autosomal-recessive mutations in RTTN, encoding Rotatin, in individuals with bilateral diffuse polymicrogyria from two separate families. Rotatin determines early embryonic axial rotation, as well as anteroposterior and dorsoventral patterning in the mouse. Human Rotatin has recently been identified as a centrosome-associated protein. The Drosophila melanogaster homolog of Rotatin, Ana3, is needed for structural integrity of centrioles and basal bodies and maintenance of sensory neurons. We show that Rotatin colocalizes with the basal bodies at the primary cilium. Cultured fibroblasts from affected individuals have structural abnormalities of the cilia and exhibit downregulation of BMP4, WNT5A, and WNT2B, which are key regulators of cortical patterning and are expressed at the cortical hem, the cortex-organizing center that gives rise to Cajal-Retzius (CR) neurons. Interestingly, we have shown that in mouse embryos, Rotatin colocalizes with CR neurons at the subpial marginal zone. Knockdown experiments in human fibroblasts and neural stem cells confirm a role for RTTN in cilia structure and function. RTTN mutations therefore link aberrant ciliary function to abnormal development and organization of the cortex in human individuals.


Subject(s)
Carrier Proteins/genetics , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Cilia/physiology , Malformations of Cortical Development/genetics , Adolescent , Cell Cycle Proteins , Cell Line , Child , Female , Gene Knockout Techniques , Genes, Recessive , Humans , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/diagnosis , Mutation
17.
Nucleic Acids Res ; 41(13): e132, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23671339

ABSTRACT

The coupling of chromosome conformation capture (3C) with next-generation sequencing technologies enables the high-throughput detection of long-range genomic interactions, via the generation of ligation products between DNA sequences, which are closely juxtaposed in vivo. These interactions involve promoter regions, enhancers and other regulatory and structural elements of chromosomes and can reveal key details of the regulation of gene expression. 3C-seq is a variant of the method for the detection of interactions between one chosen genomic element (viewpoint) and the rest of the genome. We present r3Cseq, an R/Bioconductor package designed to perform 3C-seq data analysis in a number of different experimental designs. The package reads a common aligned read input format, provides data normalization, allows the visualization of candidate interaction regions and detects statistically significant chromatin interactions, thus greatly facilitating hypothesis generation and the interpretation of experimental results. We further demonstrate its use on a series of real-world applications.


Subject(s)
Chromosomes/chemistry , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Software , Animals , Chromatin Immunoprecipitation , Genes, myb , Genomics/methods , Mice , beta-Globins/genetics
18.
Am J Respir Cell Mol Biol ; 51(2): 311-22, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24669837

ABSTRACT

Lung development is determined by the coordinated expression of several key genes. Previously, we and others have shown the importance of the sex determining region Y-box 2 (Sox2) gene in lung development. Transgenic expression of Sox2 during lung development resulted in cystic airways, and here we show that modulating the timing of ectopic Sox2 expression in the branching regions of the developing lung results in variable cystic lesions resembling the spectrum of the human congenital disorder congenital cystic adenomatoid malformation (CCAM). Sox2 dominantly differentiated naive epithelial cells into the proximal lineage irrespective of the presence of Fgf10. Sox2 directly induced the expression of Trp63, the master switch toward the basal cell lineage and induced the expression of Gata6, a factor involved in the emergence of bronchoalveolar stem cells. We showed that SOX2 and TRP63 are coexpressed in the lungs of human patients with type II CCAM. The combination of premature differentiation toward the proximal cell lineage and the induction of proliferation resulted in the cyst-like structures. Thus, we show that Sox2 is directly responsible for the emergence of two lung progenitor cells: basal cells by regulating the master gene Trp63 and bronchoalveolar stem cells by regulating Gata6.


Subject(s)
Cystic Adenomatoid Malformation of Lung, Congenital/metabolism , Epithelial Cells/metabolism , Lung/metabolism , Phosphoproteins/metabolism , SOXB1 Transcription Factors/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Cell Proliferation , Cystic Adenomatoid Malformation of Lung, Congenital/genetics , Cystic Adenomatoid Malformation of Lung, Congenital/pathology , Epithelial Cells/pathology , Fibroblast Growth Factor 10/metabolism , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Genotype , Gestational Age , HEK293 Cells , Humans , Lung/pathology , Mice , Mice, Transgenic , Phenotype , Phosphoproteins/genetics , SOXB1 Transcription Factors/genetics , Stem Cells/pathology , Tissue Culture Techniques , Trans-Activators/genetics , Transcription Factors/genetics , Transfection , Tumor Suppressor Proteins/genetics , Up-Regulation
19.
J Cell Sci ; 125(Pt 19): 4630-9, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22767512

ABSTRACT

The chromatin architecture is constantly changing because of cellular processes such as proliferation, differentiation and changes in the expression profile during gene activation or silencing. Unravelling the changes that occur in the chromatin structure during these processes has been a topic of interest for many years. It is known that gene activation of large gene loci is thought to occur by means of an active looping mechanism. It was also shown for the ß-globin locus that the gene promoter interacts with an active chromatin hub by means of an active looping mechanism. This means that the locus changes in three-dimensional (3D) nuclear volume and chromatin shape. As a means of visualizing and measuring these dynamic changes in chromatin structure of the ß-globin locus, we used a 3D DNA-FISH method in combination with 3D image acquisition to volume render fluorescent signals into 3D objects. These 3D chromatin structures were geometrically analysed, and results prior to and after gene activation were quantitatively compared. Confocal and super-resolution imaging revealed that the inactive locus occurs in several different conformations. These conformations change in shape and surface structure upon cell differentiation into a more folded and rounded structure that has a substantially smaller size and volume. These physical measurements represent the first non-biochemical evidence that, upon gene activation, an actively transcribing chromatin hub is formed by means of additional chromatin looping.


Subject(s)
Genetic Loci/genetics , Imaging, Three-Dimensional/methods , Nucleic Acid Conformation , Transcriptional Activation , beta-Globins/chemistry , beta-Globins/genetics , Animals , Cell Line , Chromatin/chemistry , Chromatin/genetics , DNA/chemistry , In Situ Hybridization, Fluorescence , Mice , Microscopy, Confocal , Microspheres , Models, Biological
20.
Nat Cell Biol ; 9(2): 201-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17187062

ABSTRACT

The transcription factor Gata-3 is a defining marker of the 'luminal' subtypes of breast cancer. To gain insight into the role of Gata-3 in breast epithelial development and oncogenesis, we have explored its normal function within the mammary gland by conditionally deleting Gata-3 at different stages of development. We report that Gata-3 has essential roles in the morphogenesis of the mammary gland in both the embryo and adult. Through the discovery of a novel marker (beta3-integrin) of luminal progenitor cells and their purification, we demonstrate that Gata-3 deficiency leads to an expansion of luminal progenitors and a concomitant block in differentiation. Remarkably, introduction of Gata-3 into a stem cell-enriched population induced maturation along the alveolar luminal lineage. These studies provide evidence for the existence of an epithelial hierarchy within the mammary gland and establish Gata-3 as a critical regulator of luminal differentiation.


Subject(s)
GATA3 Transcription Factor/metabolism , Mammary Glands, Animal/metabolism , Morphogenesis/drug effects , Animals , Cell Differentiation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , GATA3 Transcription Factor/deficiency , GATA3 Transcription Factor/pharmacology , Integrin beta3/biosynthesis , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/growth & development , Mice , Pregnancy , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL