Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-17381328

ABSTRACT

Assembly of heterochromatin at centromeric DNA regions in the fission yeast Schizosaccharomyces pombe involves an intimate interplay between chromatin modifying complexes and components of the RNAi pathway. The RNA-induced transcriptional silencing (RITS) complex, containing Chp1, Ago1, Tas3, and centromeric siRNAs, localizes to centromeric DNA repeats and is required for the assembly and maintenance of heterochromatin. RITS brings together two types of molecular recognition modules: a chromodomain protein, which binds to lysine 9 methylated histone H3 (H3K9), and Argonaute, which binds to specific sequences by siRNA-directed base-pairing interactions. The RNA-directed RNA polymerase complex (RDRC), composed of Rdp1, the Hrr1 helicase, and the Cid12 Poly(A) polymerase family member, synthesizes double-stranded RNA and creates the substrate for Dicer to generate siRNAs. RDRC physically associates with RITS, and both complexes localize to noncoding centromeric RNAs and centromeric DNA repeats, suggesting that recognition of nascent RNA transcripts may be involved in localization of these complexes to specific chromosome regions. In support of this possibility, tethering of the RITS complex to the transcript of the normally euchromatic ura4 (+) gene results in siRNA generation and RNAi- and heterochromatin-dependent silencing of the ura4 (+) gene. Finally, silencing of a subset of endogenous and transgene promoters within heterochromatic DNA domains occurs by RNAi-dependent degradation of nascent transcripts by a mechanism that we have termed co-transcriptional gene silencing (CTGS).


Subject(s)
Chromatin Assembly and Disassembly/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , RNA Interference , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Centromere/genetics , Centromere/metabolism , Chromosomes, Fungal/genetics , Chromosomes, Fungal/metabolism , Genes, Fungal , Models, Biological , Models, Genetic , Multiprotein Complexes , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism
2.
Mol Cell Biol ; 21(23): 8035-44, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11689694

ABSTRACT

The immunopurification of the endogenous cytoplasmic murine histone deacetylase 6 (mHDAC6), a member of the class II HDACs, from mouse testis cytosolic extracts allowed the identification of two associated proteins. Both were mammalian homologues of yeast proteins known to interact with each other and involved in the ubiquitin signaling pathway: p97/VCP/Cdc48p, a homologue of yeast Cdc48p, and phospholipase A2-activating protein, a homologue of yeast UFD3 (ubiquitin fusion degradation protein 3). Moreover, in the C-terminal region of mHDAC6, a conserved zinc finger-containing domain named ZnF-UBP, also present in several ubiquitin-specific proteases, was discovered and was shown to mediate the specific binding of ubiquitin by mHDAC6. By using a ubiquitin pull-down approach, nine major ubiquitin-binding proteins were identified in mouse testis cytosolic extracts, and mHDAC6 was found to be one of them. All of these findings strongly suggest that mHDAC6 could be involved in the control of protein ubiquitination. The investigation of biochemical properties of the mHDAC6 complex in vitro further supported this hypothesis and clearly established a link between protein acetylation and protein ubiquitination.


Subject(s)
Histone Deacetylases/metabolism , Signal Transduction/physiology , Ubiquitins/metabolism , Acetylation , Adenosine Triphosphatases , Animals , Cell Cycle Proteins/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Enzyme Activation/physiology , Histone Deacetylase 6 , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Macromolecular Substances , Male , Mice , Molecular Sequence Data , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Proteins/metabolism , Sequence Homology, Amino Acid , Spermatogenesis/physiology , Testis/chemistry , Testis/cytology , Testis/enzymology , Ubiquitin-Specific Proteases , Valosin Containing Protein , Zinc Fingers/physiology
3.
J Biol Chem ; 276(50): 47496-507, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11585834

ABSTRACT

Here we show that HDAC7, a member of the class II histone deacetylases, specifically targets several members of myocyte enhancer factors, MEF2A, -2C, and -2D, and inhibits their transcriptional activity. Furthermore, we demonstrate that DNA-bound MEF2C is capable of recruiting HDAC7, demonstrating that the HDAC7-dependent repression of transcription is not due to the inhibition of the MEF2 DNA binding activity. The data also suggest that the promoter bound MEF2 is potentially capable of remodeling adjacent nucleosomes via the recruitment of HDAC7. We have also observed a nucleocytoplasmic shuttling of HDAC7 and dissected the mechanism involved. In NIH3T3 cells, HDAC7 was primarily localized in the cytoplasm, essentially due to an active CRM1-dependent export of the protein from the nucleus. Interestingly, in HeLa cells, HDAC7 was predominantly nuclear. In these cells we could restore the cytoplasmic localization of HDAC7 by expressing CaMK I. This CaMK I-induced nuclear export of HDAC7 was abolished when three critical serines, Ser-178, Ser-344, and Ser-479, of HDAC7 were mutated. We show that these serines are involved in the direct interaction of HDAC7 with 14-3-3. Mutations of these serine residues weakened the association with 14-3-3 and dramatically enhanced the repression activity of HDAC7 in NIH3T3 cells, but not in HeLa cells. Data presented in this work clearly show that the signal dependent subcellular localization of HDAC7 is essential in controlling its activities. The data also show that the cellular concentration of factors such as 14-3-3, CaMK I, and other yet unknown molecules may determine the subcellular localization of an individual HDAC member in a cell type and HDAC-specific manner.


Subject(s)
Active Transport, Cell Nucleus , Histone Deacetylases/metabolism , Saccharomyces cerevisiae Proteins , 14-3-3 Proteins , 3T3 Cells , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Fungal Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins , Haplorhini , HeLa Cells , Humans , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Mutation , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Serine/chemistry , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism
4.
Curr Opin Genet Dev ; 11(2): 162-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250139

ABSTRACT

Nucleocytoplasmic shuttling of histone deacetylases is emerging as a major step in determining the composition, and hence the activity, of the corresponding nuclear regulatory complexes. This shuttling process is one of the distinctive characteristics of these enzymes, themselves belonging to structurally and functionally different classes. Considering the specific features of each class of deacetylases, it is possible to determine how each member can contribute to particular cellular functions.


Subject(s)
Cell Nucleus/enzymology , Cytoplasm/enzymology , Histone Deacetylases/metabolism , Histones/metabolism , Animals , Histone Deacetylases/chemistry , Histone Deacetylases/classification , Humans
5.
Mol Cell Biol ; 20(18): 6627-37, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10958660

ABSTRACT

The histone H1(0)-encoding gene is expressed in vertebrates in differentiating cells during the arrest of proliferation. In the H1(0) promoter, a specific regulatory element, which we named the H4 box, exhibits features which implicate a role in mediating H1(0) gene expression in response to both differentiation and cell cycle control signals. For instance, within the linker histone gene family, the H4 box is found only in the promoters of differentiation-associated subtypes, suggesting that it is specifically involved in differentiation-dependent expression of these genes. In addition, an element nearly identical to the H4 box is conserved in the promoters of histone H4-encoding genes and is known to be involved in their cell cycle-dependent expression. The transcription factors interacting with the H1(0) H4 box were therefore expected to link differentiation-dependent expression of H1(0) to the cell cycle control machinery. The aim of this work was to identify such transcription factors and to obtain information concerning the regulatory pathway involved. Interestingly, our cloning strategy led to the isolation of a retinoblastoma protein (RB) partner known as HBP1. HBP1, a high-mobility group box transcription factor, interacted specifically with the H1(0) H4 box and moreover was expressed in a differentiation-dependent manner. We also showed that the HBP1-encoding gene is able to produce different forms of HBP1. Finally, we demonstrated that both HBP1 and RB were involved in the activation of H1(0) gene expression. We therefore propose that HBP1 mediates a link between the cell cycle control machinery and cell differentiation signals. Through modulating the expression of specific chromatin-associated proteins such as histone H1(0), HBP1 plays a vital role in chromatin remodeling events during the arrest of cell proliferation in differentiating cells.


Subject(s)
Gene Expression Regulation , High Mobility Group Proteins/metabolism , Histones/genetics , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Adult , Animals , Base Sequence , Cell Differentiation , DNA, Complementary , HMGB1 Protein , High Mobility Group Proteins/genetics , Humans , Mice , Molecular Sequence Data , Rats , Regulatory Sequences, Nucleic Acid , Repressor Proteins/genetics , Tumor Cells, Cultured
6.
Curr Biol ; 10(12): 747-9, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10873806

ABSTRACT

The intracellular localization, and thereby the function, of a number of key regulator proteins tagged with a short leucine-rich motif (the nuclear export signal or NES) is controlled by CRM1/exportin1, which is involved in the export of these proteins from the nucleus [1]. A common characteristic of these regulators is their transient action in the nucleus during either a specific phase of the cell cycle or in response to specific signals [1]. Here, we show that a particular member of the class II histone-deacetylases mHDA2/mHDAC6 [2] belongs to this family of cellular regulators that are present predominantly in the cytoplasm, but are also capable of shuttling between the nucleus and the cytoplasm. A very potent NES present at the amino terminus of mHDAC6 was found to play an essential role in this shuttling process. The sub-cellular localization of mHDAC6 appeared to be controlled by specific signals, since the arrest of cell proliferation was found to be associated with the translocation of a fraction of the protein into the nucleus. Data presented here suggest that mHDAC6 might be the first member of a functionally distinct class of deacetylases, responsible for activities not shared by other known histone deacetylases.


Subject(s)
Cytoplasm/enzymology , Histone Deacetylases/metabolism , Amino Acid Sequence , HeLa Cells , Histone Deacetylases/chemistry , Humans , Molecular Sequence Data
7.
J Biol Chem ; 275(20): 15594-9, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10748098

ABSTRACT

Recently we identified a new family of histone deacetylases in higher eukaryotes related to yeast HDA1 and showed their differentiation-dependent expression. Data presented here indicate that HDAC5 (previously named mHDA1), one member of this family, might be a potent regulator of cell differentiation by interacting specifically with determinant transcription factors. We found that HDAC5 was able to interact in vivo and in vitro with MEF2A, a MADS box transcription factor, and to strongly inhibit its transcriptional activity. Surprisingly, this repression was independent of HDAC5 deacetylase domain. The N-terminal non-deacetylase domain of HDAC5 was able to ensure an efficient repression of MEF2A-dependent transcription. We then mapped protein domains involved in the HDAC5-MEF2A interaction and showed that MADS box/MEF2-domain region of MEF2A interacts specifically with a limited region in the N-terminal part of HDAC5 which also possesses a distinct repressor domain. These data show that two independent class II histone deacetylases HDAC4 and HDAC5 are able to interact with members of the MEF2 transcription factor family and regulate their transcriptional activity, thus suggesting a critical role for these deacetylases in the control of cell proliferation/differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Blotting, Western , DNA-Binding Proteins/isolation & purification , Genes, Reporter , Glutathione Transferase/genetics , HeLa Cells , Histone Deacetylases/chemistry , Histone Deacetylases/isolation & purification , Humans , Luciferases/genetics , MADS Domain Proteins , MEF2 Transcription Factors , Myogenic Regulatory Factors , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Transcription Factors/isolation & purification , Transfection
8.
J Biol Chem ; 274(4): 2440-5, 1999 Jan 22.
Article in English | MEDLINE | ID: mdl-9891014

ABSTRACT

The histone deacetylase domain of almost all members of higher eukaryotic histone deacetylases already identified (HDAC family) is highly homologous to that of yeast RPD3. In this paper we report the cloning of two cDNAs encoding members of a new family of histone deacetylase in mouse that show a better homology to yeast HDA1 histone deacetylase. These cDNAs encode relatively large proteins, presenting an in vitro trichostatin A-sensitive histone deacetylase activity. Interestingly, one, mHDA2, encodes a protein with two putative deacetylase domains, and the other, mHDA1, contains only one deacetylase homology domain, located at the C-terminal half of the protein. Our data showed that these newly identified genes could belong to a network of genes coordinately regulated and involved in the remodeling of chromatin during cell differentiation. Indeed, the expression of mHDA1 and mHDA2 is tightly linked to the state of cell differentiation, behaving therefore like the histone H1 degrees-encoding gene. Moreover, like histone H1(0) gene, mHDA1 and mHDA2 gene expression is induced upon deacetylase inhibitor treatment. We postulate the existence of a regulatory mechanism, commanding a coordinate expression of a group of genes involved in the remodeling of chromatin not only during cell differentiation but also after abnormal histone acetylation.


Subject(s)
Chromatin/metabolism , Histone Deacetylases/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Cloning, Molecular , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...