Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 113(5): 1238-43, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26787893

RESUMEN

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.


Asunto(s)
Cromosomas Humanos , Metafase , Nucleosomas/química , Animales , Pollos , Cromatina/química , Eritrocitos/ultraestructura , Células HeLa , Humanos , Microscopía Electrónica
2.
J Biol Chem ; 289(49): 33827-37, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25294883

RESUMEN

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.


Asunto(s)
Histonas/química , Lisina/química , Nucleosomas/química , Procesamiento Proteico-Postraduccional , Ubiquitina/química , Acetilación , Ensamble y Desensamble de Cromatina , Disulfuros/química , Escherichia coli/genética , Escherichia coli/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Cinética , Lisina/metabolismo , Modelos Moleculares , Nucleosomas/genética , Nucleosomas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Sumoilación , Transcripción Genética , Ubiquitina/genética , Ubiquitina/metabolismo
3.
J Struct Biol ; 188(2): 142-55, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25264147

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , Espermatozoides/metabolismo , Vertebrados/metabolismo , Animales , Arginina/metabolismo , Núcleo Celular/metabolismo , Cisteína/metabolismo , ADN/metabolismo , Masculino , Protaminas/metabolismo , Salmina/metabolismo
4.
J Biol Chem ; 289(10): 6850-6861, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24415761

RESUMEN

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.


Asunto(s)
Cromatina/química , Proteínas Cromosómicas no Histona/química , Nucleosomas/química , Animales , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Simulación por Computador , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Histonas/química , Humanos , Modelos Químicos
5.
Cell ; 154(6): 1232-45, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-24034247

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina , Histonas/metabolismo , Complejos Multiproteicos/metabolismo , Nucleosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Acetilación , Adenosina Trifosfatasas/metabolismo , Secuencia de Bases , Histona Acetiltransferasas/metabolismo , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Saccharomyces cerevisiae/genética
6.
Exp Cell Res ; 318(12): 1448-55, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22394510

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Cromatina/química , ADN/química , Conformación de Ácido Nucleico , Animales , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cromatina/ultraestructura , ADN/metabolismo , Empaquetamiento del ADN/genética , Empaquetamiento del ADN/fisiología , Humanos , Modelos Biológicos , Modelos Moleculares
7.
Proc Natl Acad Sci U S A ; 108(40): 16588-93, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21949362

RESUMEN

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.


Asunto(s)
Autoantígenos/genética , Centrómero/genética , Proteínas Cromosómicas no Histona/genética , ADN Superhelicoidal/metabolismo , Conformación de Ácido Nucleico , Nucleosomas/genética , Conformación Proteica , Proteína A Centromérica , Electroforesis en Gel de Poliacrilamida , Epigenómica , Histonas/química , Histonas/genética , Humanos , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Nucleosomas/ultraestructura , Ultracentrifugación
8.
IUBMB Life ; 62(10): 732-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21031501

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , ADN/metabolismo , Histonas/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Sitios de Unión , ADN/química , ADN/genética , Metilación de ADN , Femenino , Genoma Humano , Histonas/genética , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Estructura Secundaria de Proteína , Síndrome de Rett/genética
9.
Mol Cell Biol ; 30(19): 4656-70, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20679481

RESUMEN

Sporadic mutations in the hMeCP2 gene, coding for a protein that preferentially binds symmetrically methylated CpGs, result in the severe neurological disorder Rett syndrome (RTT). In the present work, employing a wide range of experimental approaches, we shed new light on the many levels of MeCP2 interaction with DNA and chromatin. We show that strong methylation-independent as well as methylation-dependent binding by MeCP2 is influenced by DNA length. Although MeCP2 is strictly monomeric in solution, its binding to DNA is cooperative, with dimeric binding strongly correlated with methylation density, and strengthened by nearby A/T repeats. Dimeric binding is abolished in the F155S and R294X severe RTT mutants. MeCP2 also binds chromatin in vitro, resulting in compaction-related changes in nucleosome architecture that resemble the classical zigzag motif induced by histone H1 and considered important for 30-nm-fiber formation. In vivo chromatin binding kinetics and in vitro steady-state nucleosome binding of both MeCP2 and H1 provide strong evidence for competition between MeCP2 and H1 for common binding sites. This suggests that chromatin binding by MeCP2 and H1 in vivo should be viewed in the context of competitive multifactorial regulation.


Asunto(s)
Cromatina/metabolismo , ADN/metabolismo , Histonas/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Secuencia Rica en At/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células 3T3 BALB , Sitios de Unión , Unión Competitiva , Cromatina/genética , ADN/genética , Metilación de ADN/efectos de los fármacos , Decitabina , Inhibidores Enzimáticos/farmacología , Polarización de Fluorescencia , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/genética , Humanos , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Microscopía de Fuerza Atómica , Microscopía Electrónica , Microscopía Fluorescente , Mutación , Nucleosomas/genética , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Unión Proteica , Multimerización de Proteína
10.
Cold Spring Harb Perspect Biol ; 2(5): a000596, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20452954

RESUMEN

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.


Asunto(s)
Cromatina/química , Cromatina/fisiología , Conformación de Ácido Nucleico , Conformación Proteica , Núcleo Celular/química , Simulación por Computador
11.
Biochemistry ; 49(20): 4395-410, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20405910

RESUMEN

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.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Dominios y Motivos de Interacción de Proteínas/fisiología , Sitios de Unión , Cromatina/metabolismo , ADN/metabolismo , Humanos , Proteína 2 de Unión a Metil-CpG/fisiología , Modelos Moleculares , Unión Proteica/fisiología , Estabilidad Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína/fisiología , Especificidad por Sustrato , Temperatura
12.
Mol Cell ; 37(6): 865-78, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20347428

RESUMEN

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.


Asunto(s)
ADN Helicasas/metabolismo , ADN/metabolismo , Inestabilidad Genómica , Histonas/metabolismo , Pliegue de Proteína , Multimerización de Proteína , Secuencia de Aminoácidos , Animales , Línea Celular , Pollos , ADN/genética , Daño del ADN , ADN Helicasas/química , ADN Helicasas/genética , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Evolución Molecular , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Humanos , Datos de Secuencia Molecular , Unión Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Alineación de Secuencia , Intercambio de Cromátides Hermanas
13.
Mol Cell ; 35(6): 769-81, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19782027

RESUMEN

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.


Asunto(s)
Ensamble y Desensamble de Cromatina , Regulación Fúngica de la Expresión Génica , Silenciador del Gen , Heterocromatina/metabolismo , Histonas/metabolismo , Saccharomyces cerevisiae/genética , Transcripción Genética , Acetilación , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Enzimas de Restricción del ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/genética , Lisina , Complejos Multiproteicos , Mutación , NAD/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína 1 de Ensamblaje de Nucleosomas , Unión Proteica , ARN Polimerasa II/metabolismo , ARN Polimerasa III/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/metabolismo , Sirtuina 2 , Sirtuinas/genética , Sirtuinas/metabolismo
14.
Proc Natl Acad Sci U S A ; 106(32): 13317-22, 2009 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-19651606

RESUMEN

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.


Asunto(s)
Conformación de Ácido Nucleico , Nucleosomas/química , Nucleosomas/metabolismo , Animales , Cationes , Pollos , Histonas/metabolismo , Modelos Moleculares , Nucleosomas/ultraestructura
15.
Biochemistry ; 48(2): 276-88, 2009 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-19099415

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , ADN de Hongos/metabolismo , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/metabolismo , Algoritmos , Animales , Baculoviridae/genética , Emparejamiento Base , Secuencia de Bases , Bioensayo , Cromatina/ultraestructura , ADN de Hongos/química , ADN de Hongos/genética , ADN de Hongos/ultraestructura , ADN Ribosómico/aislamiento & purificación , ADN de Cadena Simple/metabolismo , Silenciador del Gen , Genoma Fúngico , Histonas/metabolismo , Lytechinus/química , Modelos Moleculares , Modelos Teóricos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Nucleosomas/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/aislamiento & purificación , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/ultraestructura , Spodoptera/citología , Moldes Genéticos
16.
Mol Cell ; 32(4): 503-18, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-19026781

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Factores de Transcripción/metabolismo , Animales , Baculoviridae/genética , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Cromatina/aislamiento & purificación , Cromatina/ultraestructura , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2 , Genes Reporteros , Células HeLa , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Inmunohistoquímica , Células Jurkat , Riñón/citología , Luciferasas/metabolismo , Ratones , Mutación , Células 3T3 NIH , Complejo Represivo Polycomb 2 , Regiones Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Transfección
17.
Mol Cell Biol ; 28(19): 6010-21, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18644858

RESUMEN

The SWI/SNF complex disrupts and mobilizes chromatin in an ATP-dependent manner. SWI/SNF interactions with nucleosomes were mapped by DNA footprinting and site-directed DNA and protein cross-linking when SWI/SNF was recruited by a transcription activator. SWI/SNF was found by DNA footprinting to contact tightly around one gyre of DNA spanning approximately 50 bp from the nucleosomal entry site to near the dyad axis. The DNA footprint is consistent with nucleosomes binding to an asymmetric trough of SWI/SNF that was revealed by the improved imaging of free SWI/SNF. The DNA site-directed cross-linking revealed that the catalytic subunit Swi2/Snf2 is associated with nucleosomes two helical turns from the dyad axis and that the Snf6 subunit is proximal to the transcription factor recruiting SWI/SNF. The highly conserved Snf5 subunit associates with the histone octamer and not with nucleosomal DNA. The model of the binding trough of SWI/SNF illustrates how nucleosomal DNA can be mobilized while SWI/SNF remains bound.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Adenosina Trifosfatasas , Animales , Proteínas Cromosómicas no Histona/metabolismo , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Histonas/genética , Histonas/metabolismo , Modelos Moleculares , Mutación , Nucleosomas/metabolismo , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo , Xenopus laevis
18.
J Biol Chem ; 283(29): 20523-34, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18499664

RESUMEN

Most cases of Rett syndrome (RTT) are caused by mutations in the methylated DNA-binding protein, MeCP2. Here, we have shown that frequent RTT-causing missense mutations (R106W, R133C, F155S, T158M) located in the methylated DNA-binding domain (MBD) of MeCP2 have profound and diverse effects on its structure, stability, and DNA-binding properties. Fluorescence spectroscopy, which reports on the single tryptophan in the MBD, indicated that this residue is strongly protected from the aqueous environment in the wild type but is more exposed in the R133C and F155S mutations. In the mutant proteins R133C, F155S, and T158M, the thermal stability of the domain was strongly reduced. Thermal stability of the wild-type protein was increased in the presence of unmethylated DNA and was further enhanced by DNA methylation. DNA-induced thermal stability was also seen, but to a lesser extent, in each of the mutant proteins. Circular dichroism (CD) of the MBD revealed differences in the secondary structure of the four mutants. Upon binding to methylated DNA, the wild type showed a subtle but reproducible increase in alpha-helical structure, whereas the F155S and R106W did not acquire secondary structure with DNA. Each of the mutant proteins studied is unique in terms of the properties of the MBD and the structural changes induced by DNA binding. For each mutation, we examined the extent to which the magnitude of these differences correlated with the severity of RTT patient symptoms.


Asunto(s)
ADN/metabolismo , Proteína 2 de Unión a Metil-CpG/química , Proteína 2 de Unión a Metil-CpG/metabolismo , Pliegue de Proteína , Síndrome de Rett/metabolismo , Dicroismo Circular , ADN/genética , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Modelos Moleculares , Mutación/genética , Desnaturalización de Ácido Nucleico , Unión Proteica , Estructura Terciaria de Proteína , Síndrome de Rett/genética , Temperatura
19.
Mol Cell Biol ; 28(11): 3563-72, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18362167

RESUMEN

The telomeres and mating-type loci of budding yeast adopt a condensed, heterochromatin-like state through recruitment of the silent information regulator (SIR) proteins SIR2p, SIR3p, and SIR4p. In this study we characterize the chromatin binding determinants of recombinant SIR3p and identify how SIR3p mediates chromatin fiber condensation in vitro. Purified full-length SIR3p was incubated with naked DNA, nucleosome core particles, or defined nucleosomal arrays, and the resulting complexes were analyzed by electrophoretic shift assays, sedimentation velocity, and electron microscopy. SIR3p bound avidly to all three types of templates. SIR3p loading onto its nucleosomal sites in chromatin produced thickened condensed fibers that retained a beaded morphology. At higher SIR3p concentrations, individual nucleosomal arrays formed oligomeric suprastructures bridged by SIR3p oligomers. When condensed SIR3p-bound chromatin fibers were incubated in Mg(2+), they folded and oligomerized even further to produce hypercondensed higher-order chromatin structures. Collectively, these results define how SIR3p may function as a chromatin architectural protein and provide new insight into the interplay between endogenous and protein-mediated chromatin fiber condensation pathways.


Asunto(s)
Cromatina/química , Cromatina/ultraestructura , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/química , ADN/química , ADN/ultraestructura , Nucleosomas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Sales (Química)/química , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/genética
20.
J Biol Chem ; 282(38): 28237-45, 2007 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17660293

RESUMEN

hMeCP2 (human methylated DNA-binding protein 2), mutations of which cause most cases of Rett syndrome (RTT), is involved in the transmission of repressive epigenetic signals encoded by DNA methylation. The present work focuses on the modifications of chromatin architecture induced by MeCP2 and the effects of RTT-causing mutants. hMeCP2 binds to nucleosomes close to the linker DNA entry-exit site and protects approximately 11 bp of linker DNA from micrococcal nuclease. MeCP2 mutants differ in this property; the R106W mutant gives very little extra protection beyond the approximately 146-bp nucleosome core, whereas the large C-terminal truncation R294X reveals wild type behavior. Gel mobility assays show that linker DNA is essential for proper MeCP2 binding to nucleosomes, and electron microscopy visualization shows that the protein induces distinct conformational changes in the linker DNA. When bound to nucleosomes, MeCP2 is in close proximity to histone H3, which exits the nucleosome core close to the proposed MeCP2-binding site. These findings firmly establish nucleosomal linker DNA as a crucial binding partner of MeCP2 and show that different RTT-causing mutations of MeCP2 are correspondingly defective in different aspects of the interactions that alter chromatin architecture.


Asunto(s)
Cromatina/metabolismo , Proteína 2 de Unión a Metil-CpG/fisiología , Síndrome de Rett/genética , Sitios de Unión , Unión Competitiva , Proteínas Cromosómicas no Histona/química , Cromosomas/metabolismo , ADN/química , Histonas/química , Humanos , Proteína 2 de Unión a Metil-CpG/química , Microscopía Electrónica , Modelos Genéticos , Mutación , Nucleosomas/metabolismo , Síndrome de Rett/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...