Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 154(6): 1232-45, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034247

ABSTRACT

The histone variant H2A.Z is a genome-wide signature of nucleosomes proximal to eukaryotic regulatory DNA. Whereas the multisubunit chromatin remodeler SWR1 is known to catalyze ATP-dependent deposition of H2A.Z, the mechanism of SWR1 recruitment to S. cerevisiae promoters has been unclear. A sensitive assay for competitive binding of dinucleosome substrates revealed that SWR1 preferentially binds long nucleosome-free DNA and the adjoining nucleosome core particle, allowing discrimination of gene promoters over gene bodies. Analysis of mutants indicates that the conserved Swc2/YL1 subunit and the adenosine triphosphatase domain of Swr1 are mainly responsible for binding to substrate. SWR1 binding is enhanced on nucleosomes acetylated by the NuA4 histone acetyltransferase, but recognition of nucleosome-free and nucleosomal DNA is dominant over interaction with acetylated histones. Such hierarchical cooperation between DNA and histone signals expands the dynamic range of genetic switches, unifying classical gene regulation by DNA-binding factors with ATP-dependent nucleosome remodeling and posttranslational histone modifications.


Subject(s)
Chromatin Assembly and Disassembly , Histones/metabolism , Multiprotein Complexes/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Acetylation , Adenosine Triphosphatases/metabolism , Base Sequence , Histone Acetyltransferases/metabolism , Molecular Sequence Data , Protein Processing, Post-Translational , Saccharomyces cerevisiae/genetics
2.
Proc Natl Acad Sci U S A ; 113(5): 1238-43, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787893

ABSTRACT

The architecture of higher-order chromatin in eukaryotic cell nuclei is largely unknown. Here, we use electron microscopy-assisted nucleosome interaction capture (EMANIC) cross-linking experiments in combination with mesoscale chromatin modeling of 96-nucleosome arrays to investigate the internal organization of condensed chromatin in interphase cell nuclei and metaphase chromosomes at nucleosomal resolution. The combined data suggest a novel hierarchical looping model for chromatin higher-order folding, similar to rope flaking used in mountain climbing and rappelling. Not only does such packing help to avoid tangling and self-crossing, it also facilitates rope unraveling. Hierarchical looping is characterized by an increased frequency of higher-order internucleosome contacts for metaphase chromosomes compared with chromatin fibers in vitro and interphase chromatin, with preservation of a dominant two-start zigzag organization associated with the 30-nm fiber. Moreover, the strong dependence of looping on linker histone concentration suggests a hierarchical self-association mechanism of relaxed nucleosome zigzag chains rather than longitudinal compaction as seen in 30-nm fibers. Specifically, concentrations lower than one linker histone per nucleosome promote self-associations and formation of these looped networks of zigzag fibers. The combined experimental and modeling evidence for condensed metaphase chromatin as hierarchical loops and bundles of relaxed zigzag nucleosomal chains rather than randomly coiled threads or straight and stiff helical fibers reconciles aspects of other models for higher-order chromatin structure; it constitutes not only an efficient storage form for the genomic material, consistent with other genome-wide chromosome conformation studies that emphasize looping, but also a convenient organization for local DNA unraveling and genome access.


Subject(s)
Chromosomes, Human , Metaphase , Nucleosomes/chemistry , Animals , Chickens , Chromatin/chemistry , Erythrocytes/ultrastructure , HeLa Cells , Humans , Microscopy, Electron
3.
Mol Cell ; 37(6): 865-78, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20347428

ABSTRACT

FANCM remodels branched DNA structures and plays essential roles in the cellular response to DNA replication stress. Here, we show that FANCM forms a conserved DNA-remodeling complex with a histone-fold heterodimer, MHF. We find that MHF stimulates DNA binding and replication fork remodeling by FANCM. In the cell, FANCM and MHF are rapidly recruited to forks stalled by DNA interstrand crosslinks, and both are required for cellular resistance to such lesions. In vertebrates, FANCM-MHF associates with the Fanconi anemia (FA) core complex, promotes FANCD2 monoubiquitination in response to DNA damage, and suppresses sister-chromatid exchanges. Yeast orthologs of these proteins function together to resist MMS-induced DNA damage and promote gene conversion at blocked replication forks. Thus, FANCM-MHF is an essential DNA-remodeling complex that protects replication forks from yeast to human.


Subject(s)
DNA Helicases/metabolism , DNA/metabolism , Genomic Instability , Histones/metabolism , Protein Folding , Protein Multimerization , Amino Acid Sequence , Animals , Cell Line , Chickens , DNA/genetics , DNA Damage , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Evolution, Molecular , Fanconi Anemia Complementation Group Proteins , Humans , Molecular Sequence Data , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Sequence Alignment , Sister Chromatid Exchange
4.
Mol Cell ; 35(6): 769-81, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19782027

ABSTRACT

Heterochromatin assembly in budding yeast requires the SIR complex, which contains the NAD-dependent deacetylase Sir2 and the Sir3 and Sir4 proteins. Sir3 binds to nucleosomes containing deacetylated histone H4 lysine 16 (H4K16) and, with Sir4, promotes spreading of Sir2 and deacetylation along the chromatin fiber. Combined action of histone modifying and binding activities is a conserved hallmark of heterochromatin, but the relative contribution of each activity to silencing has remained unclear. Here, we reconstitute SIR-chromatin complexes using purified components and show that the SIR complex efficiently deacetylates chromatin templates and promotes the assembly of altered structures that silence Gal4-VP16-activated transcription. Silencing requires all three Sir proteins, even with fully deacetylated chromatin, and involves the specific association of Sir3 with deacetylated H4K16. These results define a minimal set of components that mediate heterochromatic gene silencing and demonstrate distinct contributions for histone deacetylation and nucleosome binding in the silencing mechanism.


Subject(s)
Chromatin Assembly and Disassembly , Gene Expression Regulation, Fungal , Gene Silencing , Heterochromatin/metabolism , Histones/metabolism , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Acetylation , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Restriction Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Lysine , Multiprotein Complexes , Mutation , NAD/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosome Assembly Protein 1 , Protein Binding , RNA Polymerase II/metabolism , RNA Polymerase III/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuin 2 , Sirtuins/genetics , Sirtuins/metabolism
5.
Mol Cell ; 32(4): 503-18, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-19026781

ABSTRACT

Polycomb group proteins are critical to maintaining gene repression established during Drosophila development. Part of this group forms the PRC2 complex containing Ez that catalyzes di- and trimethylation of histone H3 lysine 27 (H3K37me2/3), marks repressive to transcription. We report that the mammalian homologs Ezh1 and Ezh2 form similar PRC2 complexes but exhibit contrasting repressive roles. While PRC2-Ezh2 catalyzes H3K27me2/3 and its knockdown affects global H3K27me2/3 levels, PRC2-Ezh1 performs this function weakly. In accordance, Ezh1 knockdown was ineffectual on global H3K27me2/3 levels. Instead, PRC2-Ezh1 directly and robustly represses transcription from chromatinized templates and compacts chromatin in the absence of the methyltransferase cofactor SAM, as evidenced by electron microscopy. Ezh1 targets a subset of Ezh2 genes, yet Ezh1 is more abundant in nonproliferative adult organs while Ezh2 expression is tightly associated with proliferation, as evidenced when analyzing aging mouse kidney. These results might reflect subfunctionalization of a PcG protein during evolution.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors/metabolism , Animals , Baculoviridae/genetics , Cell Line , Cell Line, Tumor , Chromatin/genetics , Chromatin/isolation & purification , Chromatin/ultrastructure , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Genes, Reporter , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Immunohistochemistry , Jurkat Cells , Kidney/cytology , Luciferases/metabolism , Mice , Mutation , NIH 3T3 Cells , Polycomb Repressive Complex 2 , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/genetics , Transfection
6.
J Biol Chem ; 289(49): 33827-37, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25294883

ABSTRACT

The structure of eukaryotic chromatin directly influences gene function, and is regulated by chemical modifications of the core histone proteins. Modification of the human histone H4 N-terminal tail region by the small ubiquitin-like modifier protein, SUMO-3, is associated with transcription repression. However, the direct effect of sumoylation on chromatin structure and function remains unknown. Therefore, we employed a disulfide-directed strategy to generate H4 homogenously and site-specifically sumoylated at Lys-12 (suH4ss). Chromatin compaction and oligomerization assays with nucleosomal arrays containing suH4ss established that SUMO-3 inhibits array folding and higher order oligomerization, which underlie chromatin fiber formation. Moreover, the effect of sumoylation differed from that of acetylation, and could be recapitulated with the structurally similar protein ubiquitin. Mechanistic studies at the level of single nucleosomes revealed that, unlike acetylation, the effect of SUMO-3 arises from the attenuation of long-range internucleosomal interactions more than from the destabilization of a compacted dinucleosome state. Altogether, our results present the first insight on the direct structural effects of histone H4 sumoylation and reveal a novel mechanism by which SUMO-3 inhibits chromatin compaction.


Subject(s)
Histones/chemistry , Lysine/chemistry , Nucleosomes/chemistry , Protein Processing, Post-Translational , Ubiquitin/chemistry , Acetylation , Chromatin Assembly and Disassembly , Disulfides/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Histones/genetics , Histones/metabolism , Humans , Kinetics , Lysine/metabolism , Models, Molecular , Nucleosomes/genetics , Nucleosomes/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Sumoylation , Transcription, Genetic , Ubiquitin/genetics , Ubiquitin/metabolism
7.
J Biol Chem ; 289(10): 6850-6861, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24415761

ABSTRACT

HP1(Hsα)-containing heterochromatin is located near centric regions of chromosomes and regulates DNA-mediated processes such as DNA repair and transcription. The higher-order structure of heterochromatin contributes to this regulation, yet the structure of heterochromatin is not well understood. We took a multidisciplinary approach to determine how HP1(Hsα)-nucleosome interactions contribute to the structure of heterochromatin. We show that HP1(Hsα) preferentially binds histone H3K9Me3-containing nucleosomal arrays in favor of non-methylated nucleosomal arrays and that nonspecific DNA interactions and pre-existing chromatin compaction promote binding. The chromo and chromo shadow domains of HP1(Hsα) play an essential role in HP1(Hsα)-nucleosome interactions, whereas the hinge region appears to have a less significant role. Electron microscopy of HP1(Hsα)-associated nucleosomal arrays showed that HP1(Hsα) caused nucleosome associations within an array, facilitating chromatin condensation. Differential sedimentation of HP1(Hsα)-associated nucleosomal arrays showed that HP1(Hsα) promotes interactions between arrays. These strand-to-strand interactions are supported by in vivo studies where tethering the Drosophila homologue HP1a to specific sites promotes interactions with distant chromosomal sites. Our findings demonstrate that HP1(Hsα)-nucleosome interactions cause chromatin condensation, a process that regulates many chromosome events.


Subject(s)
Chromatin/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Nucleosomes/chemistry , Animals , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Computer Simulation , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Histones/chemistry , Humans , Models, Chemical
8.
J Struct Biol ; 188(2): 142-55, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25264147

ABSTRACT

A consistent feature of sperm nuclei is its exceptionally compact state in comparison with somatic nuclei. Here, we have examined the structural organization of sperm chromatin from representatives of three vertebrate lineages, bony fish (Danio rerio), birds (Gallus gallus domesticus) and mammals (Mus musculus) using light and transmission electron microscopy (TEM). Although the three sperm nuclei are all highly compact, they differ in morphology and in the complement of compaction-inducing proteins. Whereas zebrafish sperm retain somatic histones and a nucleosomal organization, in the rooster and mouse, histones are largely replaced by small, arginine-rich protamines. In contrast to the mouse, the rooster protamine contains no cysteine residues and lacks the potential stabilizing effects of S-S bonds. Protamine driven chromatin compaction results in a stable, highly condensed chromatin, markedly different from the somatic nucleosome-based beads-on-a-string architecture, but its structure remains poorly understood. When prepared gently for whole mount TEM, the rooster and mouse sperm chromatin reveal striking rod-like units 40-50 nm in width. Also present in the mouse, which has very flattened sperm nuclei, but not rooster, where nuclei take the form of elongated cylinders, are toroidal shaped structures, with an external diameter of about 90 nm. In contrast, similarly prepared zebrafish sperm exhibit nucleosomal chromatin. We also examined the early stages in the binding of salmine (the salmon protamine) to defined sequence DNA. These images suggest an initial side-by-side binding of linear DNA-protamine complexes leading to the nucleation of thin, flexible rods with the potential to bend, allowing the ends to come into contact and fuse to form toroidal structures. We discuss the relationship between these in vitro observations and the rods and toroids seen in nuclei, and suggest an explanation for the apparent absence of these structures in TEM images of fully condensed sperm nuclei.


Subject(s)
Chromatin/metabolism , Spermatozoa/metabolism , Vertebrates/metabolism , Animals , Arginine/metabolism , Cell Nucleus/metabolism , Cysteine/metabolism , DNA/metabolism , Male , Protamines/metabolism , Salmine/metabolism
9.
Proc Natl Acad Sci U S A ; 108(40): 16588-93, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21949362

ABSTRACT

Centromere protein A (CENP-A) is a histone H3 variant that marks centromere location on the chromosome. To study the subunit structure and folding of human CENP-A-containing chromatin, we generated a set of nucleosomal arrays with canonical core histones and another set with CENP-A substituted for H3. At the level of quaternary structure and assembly, we find that CENP-A arrays are composed of octameric nucleosomes that assemble in a stepwise mechanism, recapitulating conventional array assembly with canonical histones. At intermediate structural resolution, we find that CENP-A-containing arrays are globally condensed relative to arrays with the canonical histones. At high structural resolution, using hydrogen-deuterium exchange coupled to mass spectrometry (H/DX-MS), we find that the DNA superhelical termini within each nucleosome are loosely connected to CENP-A, and we identify the key amino acid substitution that is largely responsible for this behavior. Also the C terminus of histone H2A undergoes rapid hydrogen exchange relative to canonical arrays and does so in a manner that is independent of nucleosomal array folding. These findings have implications for understanding CENP-A-containing nucleosome structure and higher-order chromatin folding at the centromere.


Subject(s)
Autoantigens/genetics , Centromere/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA, Superhelical/metabolism , Nucleic Acid Conformation , Nucleosomes/genetics , Protein Conformation , Centromere Protein A , Electrophoresis, Polyacrylamide Gel , Epigenomics , Histones/chemistry , Histones/genetics , Humans , Mass Spectrometry , Microscopy, Electron, Transmission , Nucleosomes/ultrastructure , Ultracentrifugation
10.
Exp Cell Res ; 318(12): 1448-55, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22394510

ABSTRACT

Despite over 30 years of work, the fundamental structure of eukaryotic chromatin remains controversial. Here, we review the roots of this controversy in disparities between results derived from studies of chromatin in nuclei, chromatin isolated from nuclei, and chromatin reconstituted from defined components. Thanks to recent advances in imaging, modeling, and other approaches, it is now possible to recognize some unifying principles driving chromatin architecture at the level of the ubiquitous '30 nm' chromatin fiber. These suggest that fiber architecture involves both zigzag and bent linker motifs, and that such heteromorphic structures facilitate the observed high packing ratios. Interactions between neighboring fibers in highly compact chromatin lead to extensive interdigitation of nucleosomes and the inability to resolve individual fibers in compact chromatin in situ.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/chemistry , DNA/chemistry , Nucleic Acid Conformation , Animals , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin/ultrastructure , DNA/metabolism , DNA Packaging/genetics , DNA Packaging/physiology , Humans , Models, Biological , Models, Molecular
11.
Proc Natl Acad Sci U S A ; 106(32): 13317-22, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19651606

ABSTRACT

The architecture of the chromatin fiber, which determines DNA accessibility for transcription and other template-directed biological processes, remains unknown. Here we investigate the internal organization of the 30-nm chromatin fiber, combining Monte Carlo simulations of nucleosome chain folding with EM-assisted nucleosome interaction capture (EMANIC). We show that at physiological concentrations of monovalent ions, linker histones lead to a tight 2-start zigzag dominated by interactions between alternate nucleosomes (i +/- 2) and sealed by histone N-tails. Divalent ions further compact the fiber by promoting bending in some linker DNAs and hence raising sequential nucleosome interactions (i +/- 1). Remarkably, both straight and bent linker DNA conformations are retained in the fully compact chromatin fiber as inferred from both EMANIC and modeling. This conformational variability is energetically favorable as it helps accommodate DNA crossings within the fiber axis. Our results thus show that the 2-start zigzag topology and the type of linker DNA bending that defines solenoid models may be simultaneously present in a structurally heteromorphic chromatin fiber with uniform 30 nm diameter. Our data also suggest that dynamic linker DNA bending by linker histones and divalent cations in vivo may mediate the transition between tight nucleosome packing within discrete 30-nm fibers and self-associated higher-order chromosomal forms.


Subject(s)
Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/metabolism , Animals , Cations , Chickens , Histones/metabolism , Models, Molecular , Nucleosomes/ultrastructure
12.
Biochemistry ; 49(20): 4395-410, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20405910

ABSTRACT

Methylated DNA binding protein 2 (MeCP2) is a methyl CpG binding protein whose key role is the recognition of epigenetic information encoded in DNA methylation patterns. Mutation or misregulation of MeCP2 function leads to Rett syndrome as well as a variety of other autism spectrum disorders. Here, we have analyzed in detail the properties of six individually expressed human MeCP2 domains spanning the entire protein with emphasis on their interactions with each other, with DNA, and with nucleosomal arrays. Each domain contributes uniquely to the structure and function of the full-length protein. MeCP2 is approximately 60% unstructured, with nine interspersed alpha-molecular recognition features (alpha-MoRFs), which are polypeptide segments predicted to acquire secondary structure upon forming complexes with binding partners. Large increases in secondary structure content are induced in some of the isolated MeCP2 domains and in the full-length protein by binding to DNA. Interactions between some MeCP2 domains in cis and trans seen in our assays likely contribute to the structure and function of the intact protein. We also show that MeCP2 has two functional halves. The N-terminal portion contains the methylated DNA binding domain (MBD) and two highly disordered flanking domains that modulate MBD-mediated DNA binding. One of these flanking domains is also capable of autonomous DNA binding. In contrast, the C-terminal portion of the protein that harbors at least two independent DNA binding domains and a chromatin-specific binding domain is largely responsible for mediating nucleosomal array compaction and oligomerization. These findings led to new mechanistic and biochemical insights regarding the conformational modulations of this intrinsically disordered protein, and its context-dependent in vivo roles.


Subject(s)
Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/metabolism , Protein Interaction Domains and Motifs/physiology , Binding Sites , Chromatin/metabolism , DNA/metabolism , Humans , Methyl-CpG-Binding Protein 2/physiology , Models, Molecular , Protein Binding/physiology , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary/physiology , Substrate Specificity , Temperature
13.
IUBMB Life ; 62(10): 732-8, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21031501

ABSTRACT

Methylated CpG Binding Protein 2 (MeCP2) is a nuclear protein named for its ability to selectively recognize methylated DNA. Much attention has been focused on understanding MeCP2 structure and function in the context of its role in Rett syndrome, a severe neurodevelopmental disorder that afflicts one in 10,000-15,000 girls. Early studies suggested a connection between DNA methylation, MeCP2, and establishment of a repressive chromatin structure at specific gene promoters. However, it is now recognized that MeCP2 can both activate and repress specific genes depending on the context. Likewise, in the cell, MeCP2 is bound to unmethylated DNA and chromatin in addition to methylated DNA. Thus, to understand the molecular basis of MeCP2 functionality, it is necessary to unravel the complex interrelationships between MeCP2 binding to unmethylated and methylated regions of the genome. MeCP2 is unusual and interesting in that it is an intrinsically disordered protein, that is, much of its primary sequence fails to fold into secondary structure and yet is functional. The unique structure of MeCP2 is the subject of the first section of this article. We then discuss recent investigations of the in vitro binding of MeCP2 to unmethylated and methylated DNA, and the potential ramifications of this work for in vivo function. We close by focusing on mechanistic studies indicating that the binding of MeCP2 to chromatin results in compaction into local (secondary) and global (tertiary) higher order structures. MeCP2 also competes with histone H1 for nucleosomal binding sites. The recent finding that MeCP2 is found at near stoichiometric levels with nucleosomes in neuronal cells underscores the multiple modes of engagement of MeCP2 with the genome, which include the cooperative tracking of methylation density.


Subject(s)
Chromatin/metabolism , DNA/metabolism , Histones/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Binding Sites , DNA/chemistry , DNA/genetics , DNA Methylation , Female , Genome, Human , Histones/genetics , Humans , Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Secondary , Rett Syndrome/genetics
14.
Mol Cell Biol ; 27(3): 864-77, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17101771

ABSTRACT

Mutations of the methylated DNA binding protein MeCP2, a multifunctional protein that is thought to transmit epigenetic information encoded as methylated CpG dinucleotides to the transcriptional machinery, give rise to the debilitating neurodevelopmental disease Rett syndrome (RTT). In this in vitro study, the methylation-dependent and -independent interactions of wild-type and mutant human MeCP2 with defined DNA and chromatin substrates were investigated. A combination of electrophoretic mobility shift assays and visualization by electron microscopy made it possible to understand the different conformational changes underlying the gel shifts. MeCP2 is shown to have, in addition to its well-established methylated DNA binding domain, a methylation-independent DNA binding site (or sites) in the first 294 residues, while the C-terminal portion of MeCP2 (residues 295 to 486) contains one or more essential chromatin interaction regions. All of the RTT-inducing mutants tested were quantitatively bound to chromatin under our conditions, but those that tend to be associated with the more severe RTT symptoms failed to induce the extensive compaction observed with wild-type MeCP2. Two modes of MeCP2-driven compaction were observed, one promoting nucleosome clustering and the other forming DNA-MeCP2-DNA complexes. MeCP2 binding to DNA and chromatin involves a number of different molecular interactions, some of which result in compaction and oligomerization. The multifunctional roles of MeCP2 may be reflected in these different interactions.


Subject(s)
Chromatin/metabolism , DNA Methylation , Methyl-CpG-Binding Protein 2/metabolism , Chromatin/ultrastructure , DNA/chemistry , DNA/ultrastructure , Electrophoretic Mobility Shift Assay , Humans , Methyl-CpG-Binding Protein 2/chemistry , Methyl-CpG-Binding Protein 2/ultrastructure , Models, Biological , Mutant Proteins/metabolism , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/ultrastructure , Protein Binding , Protein Conformation
15.
Biochemistry ; 48(2): 276-88, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19099415

ABSTRACT

Recent studies of the mechanisms involved in the regulation of gene expression in eukaryotic organisms depict a highly complex process requiring a coordinated rearrangement of numerous molecules to mediate DNA accessibility. Silencing in Saccharomyces cerevisiae involves the Sir family of proteins. Sir3p, originally described as repressing key areas of the yeast genome through interactions with the tails of histones H3 and H4, appears to have additional roles in that process, including involvement with a DNA binding component. Our in vitro studies focused on the characterization of Sir3p-nucleic acid interactions and their biological functions in Sir3p-mediated silencing using binding assays, EM imaging, and theoretical modeling. Our results suggest that the initial Sir3p recruitment is partially DNA-driven, highly cooperative, and dependent on nucleosomal features other than histone tails. The initial step appears to be rapidly followed by the spreading of silencing using linker DNA as a track.


Subject(s)
Chromatin/metabolism , DNA, Fungal/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Algorithms , Animals , Baculoviridae/genetics , Base Pairing , Base Sequence , Biological Assay , Chromatin/ultrastructure , DNA, Fungal/chemistry , DNA, Fungal/genetics , DNA, Fungal/ultrastructure , DNA, Ribosomal/isolation & purification , DNA, Single-Stranded/metabolism , Gene Silencing , Genome, Fungal , Histones/metabolism , Lytechinus/chemistry , Models, Molecular , Models, Theoretical , Molecular Sequence Data , Nucleic Acid Conformation , Nucleosomes/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/isolation & purification , Silent Information Regulator Proteins, Saccharomyces cerevisiae/ultrastructure , Spodoptera/cytology , Templates, Genetic
16.
J Cell Biol ; 166(2): 161-5, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15249582

ABSTRACT

The termini of eukaryotic chromosomes contain specialized protective structures, the telomeres, composed of TTAGGG repeats and associated proteins which, together with telomerase, control telomere length. Telomere shortening is associated with senescence and inappropriate telomerase activity may lead to cancer. Little is known about the chromatin context of telomeres, because, in most cells, telomere chromatin is tightly anchored within the nucleus. We now report the successful release of telomere chromatin from chicken erythrocyte and mouse lymphocyte nuclei, both of which have a reduced karyoskeleton. Electron microscopy reveals telomere chromatin fibers in the form of closed terminal loops, which correspond to the "t-loop" structures adopted by telomere DNA. The ability to recognize isolated telomeres in their native chromatin conformation opens the way for detailed structural and compositional studies.


Subject(s)
Chromatin/chemistry , Chromosomes/chemistry , Animals , Cell Nucleus , Chickens , Chromatin/isolation & purification , Erythrocytes/ultrastructure , Lymphocytes/ultrastructure , Mice , Microscopy, Electron , Nucleic Acid Conformation , Telomere/genetics
17.
Curr Opin Struct Biol ; 16(2): 213-20, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16540311

ABSTRACT

A complete understanding of the structure-function relationships of chromatin requires extending primarily one dimensional information, obtained from molecular genetic techniques and based on the underlying linear DNA sequence, to the three dimensional conformation. Recent progress in this endeavor has included the examination of fully defined nucleosomes and nucleosomal arrays assembled in vitro using X-ray diffraction, NMR spectroscopy, electron microscopy and atomic force microscopy. These studies have provided valuable insights into the structural roles of histone variants, the impact of histone mutations and the compaction of nucleosomal arrays. In addition, the diverse structural consequences of the binding of specific chromatin 'architectural' proteins are becoming apparent. These approaches provide an essential basis for understanding the conformation of the 'epigenome'.


Subject(s)
Chromatin/chemistry , Animals , Chromatin/physiology , Histones/chemistry , Histones/physiology , Humans , Microscopy, Atomic Force , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/physiology , Protein Binding , Protein Conformation
18.
Mol Cell Biol ; 23(13): 4559-72, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12808097

ABSTRACT

Most eukaryotic cells contain nearly equimolar amounts of nucleosomes and H1 linker histones. Despite their abundance and the potential functional specialization of H1 subtypes in multicellular organisms, gene inactivation studies have failed to reveal essential functions for linker histones in vivo. Moreover, in vitro studies suggest that H1 subtypes may not be absolutely required for assembly of chromosomes or nuclei. By sequentially inactivating the genes for three mouse H1 subtypes (H1c, H1d, and H1e), we showed that linker histones are essential for mammalian development. Embryos lacking the three H1 subtypes die by mid-gestation with a broad range of defects. Triple-H1-null embryos have about 50% of the normal ratio of H1 to nucleosomes. Mice null for five of these six H1 alleles are viable but are underrepresented in litters and are much smaller than their littermates. Marked reductions in H1 content were found in certain tissues of these mice and in another compound H1 mutant. These results demonstrate that the total amount of H1 is crucial for proper embryonic development. Extensive reduction of H1 in certain tissues did not lead to changes in nuclear size, but it did result in global shortening of the spacing between nucleosomes.


Subject(s)
Histones/physiology , Nucleosomes/physiology , Alleles , Animals , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatography, High Pressure Liquid , Chromosomes/metabolism , DNA/metabolism , Gene Expression Regulation, Developmental , Genotype , Heterozygote , Histones/chemistry , Histones/metabolism , Homozygote , Mice , Mice, Knockout , Micrococcal Nuclease/metabolism , Models, Biological , Mutation , Nucleosomes/metabolism , Phenotype , Polymerase Chain Reaction , Thymus Gland/metabolism , Time Factors
20.
Cold Spring Harb Perspect Biol ; 2(5): a000596, 2010 May.
Article in English | MEDLINE | ID: mdl-20452954

ABSTRACT

The primary role of the nucleus as an information storage, retrieval, and replication site requires the physical organization and compaction of meters of DNA. Although it has been clear for many years that nucleosomes constitute the first level of chromatin compaction, this contributes a relatively small fraction of the condensation needed to fit the typical genome into an interphase nucleus or set of metaphase chromosomes, indicating that there are additional "higher order" levels of chromatin condensation. Identifying these levels, their interrelationships, and the principles that govern their occurrence has been a challenging and much discussed problem. In this article, we focus on recent experimental advances and the emerging evidence indicating that structural plasticity and chromatin dynamics play dominant roles in genome organization. We also discuss novel approaches likely to yield important insights in the near future, and suggest research areas that merit further study.


Subject(s)
Chromatin/chemistry , Chromatin/physiology , Nucleic Acid Conformation , Protein Conformation , Cell Nucleus/chemistry , Computer Simulation
SELECTION OF CITATIONS
SEARCH DETAIL