Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Eur J Cell Biol ; 103(2): 151417, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729084

ABSTRACT

Nowadays, it is an established concept that the capability to reach a specialised cell identity via differentiation, as in the case of multi- and pluripotent stem cells, is not only determined by biochemical factors, but that also physical aspects of the microenvironment play a key role; interpreted by the cell through a force-based signalling pathway called mechanotransduction. However, the intricate ties between the elements involved in mechanotransduction, such as the extracellular matrix, the glycocalyx, the cell membrane, Integrin adhesion complexes, Cadherin-mediated cell/cell adhesion, the cytoskeleton, and the nucleus, are still far from being understood in detail. Here we report what is currently known about these elements in general and their specific interplay in the context of multi- and pluripotent stem cells. We furthermore merge this overview to a more comprehensive picture, that aims to cover the whole mechanotransductive pathway from the cell/microenvironment interface to the regulation of the chromatin structure in the nucleus. Ultimately, with this review we outline the current picture of the interplay between mechanotransductive cues and epigenetic regulation and how these processes might contribute to stem cell dynamics and fate.


Subject(s)
Mechanotransduction, Cellular , Humans , Animals , Stem Cells/metabolism , Stem Cells/cytology , Cell Differentiation
2.
Metabolites ; 12(7)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35888776

ABSTRACT

Activating KRAS mutations occur in about 30% of pulmonary adenocarcinoma (AC) cases and the discovery of specific inhibitors of G12C-mutated KRAS has considerably improved the prognosis for a subgroup of about 14% of non-small cell lung cancer (NSCLC) patients. However, even in patients with a KRAS G12C mutation, the overall response rate only reaches about 40% and mutations other than G12C still cannot be targeted. Despite the fact that one-carbon metabolism (1CM) and epigenetic regulation are known to be dysregulated by aberrant KRAS activity, we still lack evidence that co-treatment with drugs that regulate these factors might ameliorate response rates and patient prognosis. In this study, we show a direct dependency of Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) and Enhancer of Zeste Homolog 2 (EZH2) expression on mutationally activated KRAS and their prognostic relevance in KRAS-mutated AC. We show that aberrant KRAS activity generates a vulnerability of AC cancer cell lines to both MTHFD2 and EZH2 inhibitors. Importantly, co-inhibition of both factors was synergistically effective and comparable to KRASG12C inhibition alone, paving the way for their use in a therapeutic approach for NSCLC cancer patients.

3.
Genome Biol ; 18(1): 209, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089033

ABSTRACT

BACKGROUND: Herpesviruses can infect a wide range of animal species. Herpes simplex virus 1 (HSV-1) is one of the eight herpesviruses that can infect humans and is prevalent worldwide. Herpesviruses have evolved multiple ways to adapt the infected cells to their needs, but knowledge about these transcriptional and post-transcriptional modifications is sparse. RESULTS: Here, we show that HSV-1 induces the expression of about 1000 antisense transcripts from the human host cell genome. A subset of these is also activated by the closely related varicella zoster virus. Antisense transcripts originate either at gene promoters or within the gene body, and they show different susceptibility to the inhibition of early and immediate early viral gene expression. Overexpression of the major viral transcription factor ICP4 is sufficient to turn on a subset of antisense transcripts. Histone marks around transcription start sites of HSV-1-induced and constitutively transcribed antisense transcripts are highly similar, indicating that the genetic loci are already poised to transcribe these novel RNAs. Furthermore, an antisense transcript overlapping with the BBC3 gene (also known as PUMA) transcriptionally silences this potent inducer of apoptosis in cis. CONCLUSIONS: We show for the first time that a virus induces widespread antisense transcription of the host cell genome. We provide evidence that HSV-1 uses this to downregulate a strong inducer of apoptosis. Our findings open new perspectives on global and specific alterations of host cell transcription by viruses.


Subject(s)
Genome, Human , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions/genetics , RNA, Antisense/genetics , Transcription, Genetic , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Gene Expression Regulation, Viral/drug effects , HeLa Cells , Histone Code , Host-Pathogen Interactions/drug effects , Humans , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Transcription, Genetic/drug effects , Viral Proteins/metabolism
4.
Mol Syst Biol ; 13(10): 946, 2017 10 16.
Article in English | MEDLINE | ID: mdl-29038337

ABSTRACT

Polycomb repression in mouse embryonic stem cells (ESCs) is tightly associated with promoter co-occupancy of RNA polymerase II (RNAPII) which is thought to prime genes for activation during early development. However, it is unknown whether RNAPII poising is a general feature of Polycomb repression, or is lost during differentiation. Here, we map the genome-wide occupancy of RNAPII and Polycomb from pluripotent ESCs to non-dividing functional dopaminergic neurons. We find that poised RNAPII complexes are ubiquitously present at Polycomb-repressed genes at all stages of neuronal differentiation. We observe both loss and acquisition of RNAPII and Polycomb at specific groups of genes reflecting their silencing or activation. Strikingly, RNAPII remains poised at transcription factor genes which are silenced in neurons through Polycomb repression, and have major roles in specifying other, non-neuronal lineages. We conclude that RNAPII poising is intrinsically associated with Polycomb repression throughout differentiation. Our work suggests that the tight interplay between RNAPII poising and Polycomb repression not only instructs promoter state transitions, but also may enable promoter plasticity in differentiated cells.


Subject(s)
Dopaminergic Neurons/cytology , Genes, Developmental , Mouse Embryonic Stem Cells/cytology , Polycomb-Group Proteins/metabolism , RNA Polymerase II/metabolism , Animals , Cell Differentiation , Dopaminergic Neurons/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Promoter Regions, Genetic , Sequence Analysis, RNA , Transcription Factors/genetics
5.
Mol Syst Biol ; 11(12): 852, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26700852

ABSTRACT

Mammalian chromosomes fold into arrays of megabase-sized topologically associating domains (TADs), which are arranged into compartments spanning multiple megabases of genomic DNA. TADs have internal substructures that are often cell type specific, but their higher-order organization remains elusive. Here, we investigate TAD higher-order interactions with Hi-C through neuronal differentiation and show that they form a hierarchy of domains-within-domains (metaTADs) extending across genomic scales up to the range of entire chromosomes. We find that TAD interactions are well captured by tree-like, hierarchical structures irrespective of cell type. metaTAD tree structures correlate with genetic, epigenomic and expression features, and structural tree rearrangements during differentiation are linked to transcriptional state changes. Using polymer modelling, we demonstrate that hierarchical folding promotes efficient chromatin packaging without the loss of contact specificity, highlighting a role far beyond the simple need for packing efficiency.


Subject(s)
Chromatin/chemistry , Chromosomes/chemistry , Mouse Embryonic Stem Cells/cytology , Neurons/cytology , Transcription, Genetic , Animals , Cell Differentiation , Cells, Cultured , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Regulation , Mice
6.
Elife ; 42015 Dec 19.
Article in English | MEDLINE | ID: mdl-26687004

ABSTRACT

Dynamic post-translational modification of RNA polymerase II (RNAPII) coordinates the co-transcriptional recruitment of enzymatic complexes that regulate chromatin states and processing of nascent RNA. Extensive phosphorylation of serine residues at the largest RNAPII subunit occurs at its structurally-disordered C-terminal domain (CTD), which is composed of multiple heptapeptide repeats with consensus sequence Y1-S2-P3-T4-S5-P6-S7. Serine-5 and Serine-7 phosphorylation mark transcription initiation, whereas Serine-2 phosphorylation coincides with productive elongation. In vertebrates, the CTD has eight non-canonical substitutions of Serine-7 into Lysine-7, which can be acetylated (K7ac). Here, we describe mono- and di-methylation of CTD Lysine-7 residues (K7me1 and K7me2). K7me1 and K7me2 are observed during the earliest transcription stages and precede or accompany Serine-5 and Serine-7 phosphorylation. In contrast, K7ac is associated with RNAPII elongation, Serine-2 phosphorylation and mRNA expression. We identify an unexpected balance between RNAPII K7 methylation and acetylation at gene promoters, which fine-tunes gene expression levels.


Subject(s)
Gene Expression Regulation , Lysine/metabolism , Protein Processing, Post-Translational , RNA Polymerase II/metabolism , Transcription, Genetic , Animals , Consensus , Methylation , Mice , NIH 3T3 Cells , Phosphorylation , Serine/metabolism
7.
Science ; 347(6225): 1010-4, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25678556

ABSTRACT

Although it is generally accepted that cellular differentiation requires changes to transcriptional networks, dynamic regulation of promoters and enhancers at specific sets of genes has not been previously studied en masse. Exploiting the fact that active promoters and enhancers are transcribed, we simultaneously measured their activity in 19 human and 14 mouse time courses covering a wide range of cell types and biological stimuli. Enhancer RNAs, then messenger RNAs encoding transcription factors, dominated the earliest responses. Binding sites for key lineage transcription factors were simultaneously overrepresented in enhancers and promoters active in each cellular system. Our data support a highly generalizable model in which enhancer transcription is the earliest event in successive waves of transcriptional change during cellular differentiation or activation.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Stem Cells/cytology , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Cattle , Dogs , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Stem Cells/metabolism
8.
Cold Spring Harb Perspect Biol ; 2(6): a000588, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484389

ABSTRACT

Eukaryotic gene expression is an intricate multistep process, regulated within the cell nucleus through the activation or repression of RNA synthesis, processing, cytoplasmic export, and translation into protein. The major regulators of gene expression are chromatin remodeling and transcription machineries that are locally recruited to genes. However, enzymatic activities that act on genes are not ubiquitously distributed throughout the nucleoplasm, but limited to specific and spatially defined foci that promote preferred higher-order chromatin arrangements. The positioning of genes within the nuclear landscape relative to specific functional landmarks plays an important role in gene regulation and disease.


Subject(s)
Gene Expression Regulation/physiology , Gene Order/genetics , Gene Order/physiology , Chromatin/genetics , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease
9.
PLoS Biol ; 8(1): e1000270, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20052287

ABSTRACT

The position of genes in the interphase nucleus and their association with functional landmarks correlate with active and/or silent states of expression. Gene activation can induce chromatin looping from chromosome territories (CTs) and is thought to require de novo association with transcription factories. We identify two types of factory: "poised transcription factories," containing RNA polymerase II phosphorylated on Ser5, but not Ser2, residues, which differ from "active factories" associated with phosphorylation on both residues. Using the urokinase-type plasminogen activator (uPA) gene as a model system, we find that this inducible gene is predominantly associated with poised (S5p(+)S2p(-)) factories prior to activation and localized at the CT interior. Shortly after induction, the uPA locus is found associated with active (S5p(+)S2p(+)) factories and loops out from its CT. However, the levels of gene association with poised or active transcription factories, before and after activation, are independent of locus positioning relative to its CT. RNA-FISH analyses show that, after activation, the uPA gene is transcribed with the same frequency at each CT position. Unexpectedly, prior to activation, the uPA loci internal to the CT are seldom transcriptionally active, while the smaller number of uPA loci found outside their CT are transcribed as frequently as after induction. The association of inducible genes with poised transcription factories prior to activation is likely to contribute to the rapid and robust induction of gene expression in response to external stimuli, whereas gene positioning at the CT interior may be important to reinforce silencing mechanisms prior to induction.


Subject(s)
Gene Silencing/physiology , Transcriptional Activation/physiology , Urokinase-Type Plasminogen Activator/genetics , Antibodies/immunology , Chromatin Assembly and Disassembly/physiology , Chromosomal Position Effects/genetics , Chromosomal Position Effects/physiology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , Genes/genetics , Genetic Loci/genetics , Genetic Loci/physiology , Hep G2 Cells , Humans , Metalloendopeptidases/physiology , RNA Polymerase II/physiology , Transcriptional Activation/genetics , Urokinase-Type Plasminogen Activator/immunology
10.
Nucleic Acids Res ; 38(11): 3595-604, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20110257

ABSTRACT

PREP1 (PKNOX1) maps in the Down syndrome (DS) critical region of chromosome 21, is overexpressed in some DS tissues and might be involved in the DS phenotype. By using fibroblasts from DS patients and by overexpressing Prep1 in F9 teratocarcinoma and Prep1(i/i) MEF to single out the role of the protein, we report that excess Prep1 increases the sensitivity of cells to genotoxic stress and the extent of the apoptosis directly correlates with the level of Prep1. The apoptotic response of Prep1-overexpressing cells is mediated by the pro-apoptotic p53 protein that we show is a direct target of Prep1, as its depletion reverts the apoptotic phenotype. The induction of p53 overcomes the anti-apoptotic role of Bcl-X(L), previously shown to be also a Prep1 target, the levels of which are increased in Prep1-overexpressing cells as well. Our results provide a rationale for the involvement of PREP1 in the apoptotic phenotype of DS tissues and indicate that differences in Prep1 level can have drastic effects.


Subject(s)
Apoptosis , Down Syndrome/metabolism , Homeodomain Proteins/metabolism , Animals , Cells, Cultured , Down Syndrome/pathology , Embryonal Carcinoma Stem Cells , Etoposide/toxicity , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genetic Vectors , Homeodomain Proteins/genetics , Humans , Mice , Phenotype , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , bcl-X Protein/genetics , bcl-X Protein/metabolism
11.
J Neurosci ; 29(19): 6296-307, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19439607

ABSTRACT

Expression of neurosecretion by nerve cells requires the levels of the transcription repressor element-1 silencing transcription factor (REST) to be very low. However, when high-REST clones of PC12 cells, defective of neurosecretion, were fused to other high-REST, non-neurosecretory cells, some neurosecretion was recovered. To clarify the mechanism of this recovery, we fused defective PC12 cells with human lymphocytes. A cytogenetic analysis revealed all hybrid clones that recovered neurosecretion to contain a fragment of chromosome 11 including the gene encoding BHC80, a protein of one of the complexes that mediate REST repression. In these clones, REST levels were as high as in defective PC12, whereas BHC80, localized in the nucleus, was 4- to 5-fold higher. Transient transfection of defective PC12 with various amounts of BHC80 cDNA induced (1) in defective PC12, the reexpression of only neurosecretion mRNAs; (2) in defective PC12 cotransfected with the REST negative construct DNA-binding domain (to attenuate gene repression), the recovery of a weak, but complete neurosecretory phenotype, including dense-core granules and their regulated exocytosis. Chromatin immunoprecipitation and immunodepletion analyses revealed the extensive BHC80 association with REST at the genes of two neurosecretion proteins, chromograninB and SNAP25, however only in the low-REST PC12, whereas in high-REST defective PC12 no association was appreciable. In defective PC12 transfected with BHC80 some association was reestablished. Therefore, the recovery of neurosecretion observed after fusion/transfection of defective PC12 depends on the reciprocal level of BHC80 and REST, with BHC80 working as a negative modulator of REST repression. This role appears of possible cell physiological and pathological importance.


Subject(s)
Histone Deacetylases/metabolism , Neurosecretion/physiology , Repressor Proteins/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Chromogranin B/metabolism , DNA, Complementary/metabolism , Histone Deacetylases/genetics , Humans , Lymphocytes , PC12 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Synaptosomal-Associated Protein 25/metabolism , Transfection
12.
Mol Biol Cell ; 20(15): 3543-51, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19477923

ABSTRACT

We have analyzed the role of actin polymerization in retinoic acid (RA)-induced HoxB transcription, which is mediated by the HoxB regulator Prep1. RA induction of the HoxB genes can be prevented by the inhibition of actin polymerization. Importantly, inhibition of actin polymerization specifically affects the transcription of inducible Hox genes, but not that of their transcriptional regulators, the RARs, nor of constitutively expressed, nor of actively transcribed Hox genes. RA treatment induces the recruitment to the HoxB2 gene enhancer of a complex composed of "elongating" RNAPII, Prep1, beta-actin, and N-WASP as well as the accessory splicing components p54Nrb and PSF. We show that inhibition of actin polymerization prevents such recruitment. We conclude that inducible Hox genes are selectively sensitive to the inhibition of actin polymerization and that actin polymerization is required for the assembly of a transcription complex on the regulatory region of the Hox genes.


Subject(s)
Actins/metabolism , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Actins/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytochalasin D/pharmacology , DNA-Binding Proteins , Down-Regulation , Homeodomain Proteins/metabolism , Humans , Immunoblotting , Mutation , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , PTB-Associated Splicing Factor , Polymers/metabolism , RNA Interference , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism
13.
Dev Cell ; 16(1): 9-11, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19154714

ABSTRACT

Understanding the mechanisms that regulate gene expression during development is a major challenge in science. In this issue of Developmental Cell, Amano and colleagues report that expression of Sonic hedgehog (Shh) protein in the posterior mesenchyme of the mouse limb bud correlates with a long-range chromatin interaction with enhancer MFCS1 and looping of the Shh locus from its chromosome territory (CT).


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins , Limb Buds/physiology , Animals , Enhancer Elements, Genetic , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Limb Buds/anatomy & histology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Morphogenesis
14.
Mol Cell Biol ; 29(5): 1143-51, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19103748

ABSTRACT

The Prep1 homeodomain transcription factor is essential in embryonic development. Prep1 hypomorphic mutant mouse (Prep1(i/i)) embryos (embryonic day 9.5) display an increased terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling reaction compared to wild-type (WT) littermates. Prep1(i/i) mouse embryo fibroblasts (MEFs) show an increased basal level of annexin V binding activity, reduction of the mitochondrial-membrane potential, and increased caspase 9 and 3 activation, indicating increased apoptosis. Prep1(i/i) MEFs also respond faster than WT MEFs to genotoxic stress, indicating increased activation of the intrinsic apoptotic pathways. We did not observe an increase in p53 or an abnormal p53 response to apoptotic stimuli. However, hypomorphic MEFs have decreased endogenous levels of antiapoptotic Bcl-X(L) mRNA and protein, and Bcl-x overexpression rescues the defect of Prep1(i/i) MEFs. Using transient transfections and chromatin immunoprecipitation, we identified the Bcl-x promoter as a novel target of Prep1. Thus, Prep1 directly controls mitochondrial homeostasis (and the apoptotic potential) by modulating Bcl-x gene expression.


Subject(s)
Apoptosis , Gene Expression Regulation , Homeodomain Proteins/physiology , bcl-X Protein/genetics , Animals , Cells, Cultured , Embryo, Mammalian , Fibroblasts/cytology , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Promoter Regions, Genetic , Signal Transduction
15.
J Neurosci ; 28(42): 10674-86, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18923043

ABSTRACT

The Arx transcription factor is expressed in the developing ventral telencephalon and subsets of its derivatives. Mutation of human ARX ortholog causes neurological disorders including epilepsy, lissencephaly, and mental retardation. We have isolated the mouse Arx endogenous enhancer modules that control its tightly compartmentalized forebrain expression. Interestingly, they are scattered downstream of its coding region and partially included within the introns of the downstream PolA1 gene. These enhancers are ultraconserved noncoding sequences that are highly conserved throughout the vertebrate phylum. Functional characterization of the Arx GABAergic enhancer element revealed its strict dependence on the activity of Dlx transcription factors. Dlx overexpression induces ectopic expression of endogenous Arx and its isolated enhancer, whereas loss of Dlx expression results in reduced Arx expression, suggesting that Arx is a key mediator of Dlx function. To further elucidate the mechanisms involved, a combination of gain-of-function studies in mutant Arx or Dlx tissues was pursued. This analysis provided evidence that, although Arx is necessary for the Dlx-dependent promotion of interneuron migration, it is not required for the GABAergic cell fate commitment mediated by Dlx factors. Although Arx has additional functions independent of the Dlx pathway, we have established a direct genetic relationship that controls critical steps in the development of telencephalic GABAergic neurons. These findings contribute elucidating the genetic hierarchy that likely underlies the etiology of a variety of human neurodevelopmental disorders.


Subject(s)
Cell Movement/physiology , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Interneurons/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , gamma-Aminobutyric Acid/physiology , Animals , Base Sequence , Cells, Cultured , Gene Targeting/methods , Hippocampus/cytology , Hippocampus/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Interneurons/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Transcription Factors/biosynthesis , Transcription Factors/genetics
16.
Mol Cell Biol ; 27(22): 7981-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17875935

ABSTRACT

Prep1 is known to interact in vivo with Pbx1 to regulate development and organogenesis. We have identified a novel Prep1-interacting protein, p160 c-Myb binding protein (p160). p160 and Pbx1 compete for Prep1 in vitro, and p160 inhibits Prep1-dependent HoxB2 expression in retinoic acid-treated NT2-D1 cells. The N-terminal physiologically truncated form of p160, p67, binds the sequence 63LFPLL67 in the HR1 domain of Prep1. Mutation of both L63 and L66 impairs the binding of Prep1 to both p160/p67 and Pbx1. The sequences required to bind Prep1 are mainly located in residues 51 to 151. Immunofluorescence colocalization and coimmunoprecipitation of endogenous p160 and Prep1 are induced by ActD, which translocates p160 from the nucleolus to the nucleoplasm. These data therefore show that p160 is a novel regulator of Prep1-Pbx1 transcriptional activity.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins , Homeodomain Proteins/genetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , Nuclear Proteins/genetics , Pre-B-Cell Leukemia Transcription Factor 1 , RNA-Binding Proteins , Sequence Alignment , Transcription Factors/genetics , Transcription Factors/metabolism
17.
J Biol Chem ; 282(17): 12537-46, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17331942

ABSTRACT

We show the interaction between the enhancer and the minimal promoter of urokinase-type plasminogen activator gene during active transcription by coupling micrococcal nuclease digestion of cross-linked, sonicated chromatin, and chromatin immunoprecipitation. This approach allowed the precise identification of the interacting genomic fragments, one of which is resistant to micrococcal nuclease cleavage. The interacting fragments form a single transcriptional control unit, as indicated by their common protein content. Furthermore, we show that the enhancer-MP interaction persists during the early stages of transcription and is lost upon alpha-amanitin treatment, indicating the requirement for active transcription. Our results support a looping model of interaction between the enhancer and the MP of the urokinase-type plasminogen activator gene.


Subject(s)
Enhancer Elements, Genetic , Models, Genetic , Promoter Regions, Genetic , Transcription, Genetic , Urokinase-Type Plasminogen Activator/biosynthesis , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , Micrococcal Nuclease/chemistry , Rats , Urokinase-Type Plasminogen Activator/genetics
18.
Mol Cell ; 23(5): 749-55, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16949370

ABSTRACT

Myosin VI is the only myosin that moves toward the minus end of actin filaments, suggesting a unique biological function. Here, we show that myosin VI is present in the nucleus of mammalian cells where it colocalizes with newly transcribed mRNA and with RNA polymerase II (RNAPII) and is detected in the RNAPII complex. The colocalization and interaction of myosin VI with RNAPII require transcriptional activity. Chromatin immunoprecipitation (ChIP) demonstrates that myosin VI is recruited to the promoter and intragenic regions of active genes, encoding urokinase plasminogen activator (uPA), eukaryotic initiation factor 6 (p27/eIF6), and low-density lipoprotein receptor (LDLR), but not to noncoding, nonregulatory intergenic regions. Downregulation of myosin VI reduces steady-state mRNA levels of these genes in vivo, and antibodies to myosin VI reduce transcription in vitro. We suggest that myosin VI modulates RNAPII-dependent transcription of active genes, implicating the possibility of an actin-myosin based mechanism of transcription.


Subject(s)
Cell Nucleus/metabolism , Myosin Heavy Chains/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , Cells, Cultured , HT29 Cells , HeLa Cells , Humans , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Blood ; 100(9): 3325-32, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12384434

ABSTRACT

Activated transcription of the urokinase-type plasminogen activator (uPA) gene depends on the enhancer, located approximately 2 kb from the start of transcription. The proximal promoter, driving basal transcription, contains a GC-/GA-rich sequence immediately upstream of the TATA box. We have investigated the role played by this element in the transcription of the uPA gene in HeLa and PC3 cells, which do not express or constitutively express the gene, respectively. This region binds either Sp1 or Sp3, as monomers or multimers, but not a combination of the 2 proteins. The more efficient binding of Sp1 to the proximal promoter in PC3 cells is correlated to its phosphorylation state. Polymerase chain reaction (PCR)-coupled, chromatin immunoprecipitation experiments with anti-Sp1 antibodies indeed show an enrichment of proximal promoter sequences in PC3 cells and support the observed difference in transcription levels from proximal promoter constructs in HeLa versus PC3 cells. Furthermore, overexpression of Sp1 increases transcription from the reporter construct in HeLa cells, whereas in PC3 cells, overexpression of Sp3 does not reduce transcription from the same construct, indicating that the Sp1/Sp3 balance cannot be shifted. We conclude that the GC-/GA-rich element of the uPA regulatory region is an independent functional element, regulated by Sp family proteins. Phosphorylation of Sp1 determines the presence in vivo and the functionality of this element in PC3 cells. Thus, the cellular context determines the relevance of the GC-/GA-rich region in uPA gene transcription, which contributes to constitutive gene expression, related, in turn, to the invasive phenotype.


Subject(s)
Adenocarcinoma/pathology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/pathology , Sp1 Transcription Factor/metabolism , Urokinase-Type Plasminogen Activator/genetics , Adenocarcinoma/genetics , Base Sequence , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/pharmacology , Genes, Reporter , HeLa Cells/metabolism , Humans , Luciferases/biosynthesis , Luciferases/genetics , Male , Molecular Sequence Data , Multigene Family , Neoplasm Proteins/biosynthesis , Phosphorylation , Polymerase Chain Reaction , Prostatic Neoplasms/genetics , Protein Binding , Protein Processing, Post-Translational , Recombinant Fusion Proteins/biosynthesis , Sp3 Transcription Factor , Transcription Factors/metabolism , Tumor Cells, Cultured/pathology , Urokinase-Type Plasminogen Activator/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...