Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Annu Rev Biochem ; 89: 255-282, 2020 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-32259458

RESUMEN

Facultative heterochromatin (fHC) concerns the developmentally regulated heterochromatinization of different regions of the genome and, in the case of the mammalian X chromosome and imprinted loci, of only one allele of a homologous pair. The formation of fHC participates in the timely repression of genes, by resisting strong trans activators. In this review, we discuss the molecular mechanisms underlying the establishment and maintenance of fHC in mammals using a mouse model. We focus on X-chromosome inactivation (XCI) as a paradigm for fHC but also relate it to genomic imprinting and homeobox (Hox) gene cluster repression. A vital role for noncoding transcription and/or transcripts emerges as the general principle of triggering XCI and canonical imprinting. However, other types of fHC are established through an unknown mechanism, independent of noncoding transcription (Hox clusters and noncanonical imprinting). We also extensively discuss polycomb-group repressive complexes (PRCs), which frequently play a vital role in fHC maintenance.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Heterocromatina/metabolismo , Proteínas del Grupo Polycomb/genética , Inactivación del Cromosoma X , Cromosoma X/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Silenciador del Gen , Heterocromatina/química , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Oocitos/citología , Oocitos/crecimiento & desarrollo , Oocitos/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Espermatozoides/citología , Espermatozoides/crecimiento & desarrollo , Espermatozoides/metabolismo , Cromosoma X/química
2.
Cell ; 171(1): 85-102.e23, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28867287

RESUMEN

Chromatin modification and higher-order chromosome structure play key roles in gene regulation, but their functional interplay in controlling gene expression is elusive. We have discovered the machinery and mechanism underlying the dynamic enrichment of histone modification H4K20me1 on hermaphrodite X chromosomes during C. elegans dosage compensation and demonstrated H4K20me1's pivotal role in regulating higher-order chromosome structure and X-chromosome-wide gene expression. The structure and the activity of the dosage compensation complex (DCC) subunit DPY-21 define a Jumonji demethylase subfamily that converts H4K20me2 to H4K20me1 in worms and mammals. Selective inactivation of demethylase activity eliminates H4K20me1 enrichment in somatic cells, elevates X-linked gene expression, reduces X chromosome compaction, and disrupts X chromosome conformation by diminishing the formation of topologically associating domains (TADs). Unexpectedly, DPY-21 also associates with autosomes of germ cells in a DCC-independent manner to enrich H4K20me1 and trigger chromosome compaction. Our findings demonstrate the direct link between chromatin modification and higher-order chromosome structure in long-range regulation of gene expression.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Cromosoma X/química , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas Portadoras/genética , Compensación de Dosificación (Genética) , Embrión no Mamífero/metabolismo , Histona Demetilasas con Dominio de Jumonji/química , Histona Demetilasas con Dominio de Jumonji/metabolismo , Modelos Moleculares , Mutación , Piperidinas/metabolismo , Alineación de Secuencia , Tiofenos/metabolismo
3.
Nat Rev Genet ; 18(6): 377-389, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28479596

RESUMEN

Extensive 3D folding is required to package a genome into the tiny nuclear space, and this packaging must be compatible with proper gene expression. Thus, in the well-hierarchized nucleus, chromosomes occupy discrete territories and adopt specific 3D organizational structures that facilitate interactions between regulatory elements for gene expression. The mammalian X chromosome exemplifies this structure-function relationship. Recent studies have shown that, upon X-chromosome inactivation, active and inactive X chromosomes localize to different subnuclear positions and adopt distinct chromosomal architectures that reflect their activity states. Here, we review the roles of long non-coding RNAs, chromosomal organizational structures and the subnuclear localization of chromosomes as they relate to X-linked gene expression.


Asunto(s)
Cromosoma X/química , Animales , Expresión Génica , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Inactivación del Cromosoma X
4.
Mol Cell ; 60(3): 487-99, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26545078

RESUMEN

The MLE helicase remodels the roX lncRNAs, enabling the lncRNA-mediated assembly of the Drosophila dosage compensation complex. We identified a stable MLE core comprising the DExH helicase module and two auxiliary domains: a dsRBD and an OB-like fold. MLEcore is an unusual DExH helicase that can unwind blunt-ended RNA duplexes and has specificity for uridine nucleotides. We determined the 2.1 Å resolution structure of MLEcore bound to a U10 RNA and ADP-AlF4. The OB-like and dsRBD folds bind the DExH module and contribute to form the entrance of the helicase channel. Four uridine nucleotides engage in base-specific interactions, rationalizing the conservation of uridine-rich sequences in critical roX substrates. roX2 binding is orchestrated by MLE's auxiliary domains, which is prerequisite for MLE localization to the male X chromosome. The structure visualizes a transition-state mimic of the reaction and suggests how eukaryotic DEAH/RHA helicases couple ATP hydrolysis to RNA translocation.


Asunto(s)
Adenosina Trifosfato/química , Proteínas Cromosómicas no Histona/química , ADN Helicasas/química , Proteínas de Drosophila/química , ARN Helicasas/química , ARN/química , Factores de Transcripción/química , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Masculino , Estructura Terciaria de Proteína , ARN/genética , ARN/metabolismo , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromosoma X/química , Cromosoma X/genética , Cromosoma X/metabolismo
5.
Nature ; 535(7613): 575-9, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27437574

RESUMEN

X-chromosome inactivation (XCI) involves major reorganization of the X chromosome as it becomes silent and heterochromatic. During female mammalian development, XCI is triggered by upregulation of the non-coding Xist RNA from one of the two X chromosomes. Xist coats the chromosome in cis and induces silencing of almost all genes via its A-repeat region, although some genes (constitutive escapees) avoid silencing in most cell types, and others (facultative escapees) escape XCI only in specific contexts. A role for Xist in organizing the inactive X (Xi) chromosome has been proposed. Recent chromosome conformation capture approaches have revealed global loss of local structure on the Xi chromosome and formation of large mega-domains, separated by a region containing the DXZ4 macrosatellite. However, the molecular architecture of the Xi chromosome, in both the silent and expressed regions,remains unclear. Here we investigate the structure, chromatin accessibility and expression status of the mouse Xi chromosome in highly polymorphic clonal neural progenitors (NPCs) and embryonic stem cells. We demonstrate a crucial role for Xist and the DXZ4-containing boundary in shaping Xi chromosome structure using allele-specific genome-wide chromosome conformation capture (Hi-C) analysis, an assay for transposase-accessible chromatin with high throughput sequencing (ATAC-seq) and RNA sequencing. Deletion of the boundary disrupts mega-domain formation, and induction of Xist RNA initiates formation of the boundary and the loss of DNA accessibility. We also show that in NPCs, the Xi chromosome lacks active/inactive compartments and topologically associating domains (TADs), except around genes that escape XCI. Escapee gene clusters display TAD-like structures and retain DNA accessibility at promoter-proximal and CTCF-binding sites. Furthermore, altered patterns of facultative escape genes indifferent neural progenitor clones are associated with the presence of different TAD-like structures after XCI. These findings suggest a key role for transcription and CTCF in the formation of TADs in the context of the Xi chromosome in neural progenitors.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Inactivación del Cromosoma X , Cromosoma X/metabolismo , Alelos , Animales , Sitios de Unión , Factor de Unión a CCCTC , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Cromosomas de los Mamíferos/química , Cromosomas de los Mamíferos/genética , Células Madre Embrionarias/metabolismo , Femenino , Silenciador del Gen , Masculino , Ratones , Células-Madre Neurales/metabolismo , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Represoras/metabolismo , Análisis de Secuencia , Transcripción Genética , Cromosoma X/química , Cromosoma X/genética , Inactivación del Cromosoma X/genética
6.
Genes Dev ; 28(23): 2591-6, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452271

RESUMEN

The adjustment of X-linked gene expression to the X chromosome copy number (dosage compensation [DC]) has been widely studied as a model of chromosome-wide gene regulation. In Caenorhabditis elegans, DC is achieved by twofold down-regulation of gene expression from both Xs in hermaphrodites. We show that in males, the single X chromosome interacts with nuclear pore proteins, while in hermaphrodites, the DC complex (DCC) impairs this interaction and alters X localization. Our results put forward a structural model of DC in which X-specific sequences locate the X chromosome in transcriptionally active domains in males, while the DCC prevents this in hermaphrodites.


Asunto(s)
Caenorhabditis elegans/genética , Compensación de Dosificación (Genética)/genética , Cromosoma X/química , Cromosoma X/genética , Animales , Regulación de la Expresión Génica , Organismos Hermafroditas/genética , Masculino , Modelos Genéticos
7.
Genes Dev ; 28(23): 2652-62, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452275

RESUMEN

The male-specific lethal dosage compensation complex (MSL-DCC) selectively assembles on the X chromosome in Drosophila males and activates gene transcription by twofold through histone acetylation. An MSL recognition element (MRE) sequence motif nucleates the initial MSL association, but how it is recognized remains unknown. Here, we identified the CXC domain of MSL2 specifically recognizing the MRE motif and determined its crystal structure bound to specific and nonspecific DNAs. The CXC domain primarily contacts one strand of DNA duplex and employs a single arginine to directly read out dinucleotide sequences from the minor groove. The arginine is flexible when bound to nonspecific sequences. The core region of the MRE motif harbors two binding sites on opposite strands that can cooperatively recruit a CXC dimer. Specific DNA-binding mutants of MSL2 are impaired in MRE binding and X chromosome localization in vivo. Our results reveal multiple dynamic DNA-binding modes of the CXC domain that target the MSL-DCC to X chromosomes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Compensación de Dosificación (Genética) , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Modelos Moleculares , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Cromosoma X/química , Cromosoma X/metabolismo , Secuencias de Aminoácidos , Animales , Drosophila melanogaster/química , Unión Proteica , Estructura Terciaria de Proteína
8.
Stem Cells ; 37(7): 876-887, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30895693

RESUMEN

In spite of the progress in gene editing achieved in recent years, a subset of genetic diseases involving structural chromosome abnormalities, including aneuploidies, large deletions and complex rearrangements, cannot be treated with conventional gene therapy approaches. We have previously devised a strategy, dubbed chromosome transplantation (CT), to replace an endogenous mutated chromosome with an exogenous normal one. To establish a proof of principle for our approach, we chose as disease model the chronic granulomatous disease (CGD), an X-linked severe immunodeficiency due to abnormalities in CYBB (GP91) gene, including large genomic deletions. We corrected the gene defect by CT in induced pluripotent stem cells (iPSCs) from a CGD male mouse model. The Hprt gene of the endogenous X chromosome was inactivated by CRISPR/Cas9 technology thus allowing the exploitation of the hypoxanthine-aminopterin-thymidine selection system to introduce a normal donor X chromosome by microcell-mediated chromosome transfer. X-transplanted clones were obtained, and diploid XY clones which spontaneously lost the endogenous X chromosome were isolated. These cells were differentiated toward the myeloid lineage, and functional granulocytes producing GP91 protein were obtained. We propose the CT approach to correct iPSCs from patients affected by other X-linked diseases with large deletions, whose treatment is still unsatisfactory. Stem Cells 2019;37:876-887.


Asunto(s)
Cromosomas de los Mamíferos , Terapia Genética/métodos , Granulocitos/metabolismo , Enfermedad Granulomatosa Crónica/terapia , Hipoxantina Fosforribosiltransferasa/genética , Células Madre Pluripotentes Inducidas/metabolismo , NADPH Oxidasa 2/genética , Aminopterina/metabolismo , Aminopterina/farmacología , Animales , Secuencia de Bases , Sistemas CRISPR-Cas , Diferenciación Celular , Células Clonales , Medios de Cultivo/química , Modelos Animales de Enfermedad , Edición Génica/métodos , Granulocitos/citología , Granulocitos/efectos de los fármacos , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/metabolismo , Enfermedad Granulomatosa Crónica/patología , Humanos , Hipoxantina/metabolismo , Hipoxantina/farmacología , Hipoxantina Fosforribosiltransferasa/deficiencia , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , NADPH Oxidasa 2/deficiencia , Prueba de Estudio Conceptual , Eliminación de Secuencia , Tioguanina/metabolismo , Tioguanina/farmacología , Timidina/metabolismo , Timidina/farmacología , Cromosoma X/química , Cromosoma X/metabolismo
9.
Nature ; 502(7469): 59-64, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24067610

RESUMEN

Large-scale chromosome structure and spatial nuclear arrangement have been linked to control of gene expression and DNA replication and repair. Genomic techniques based on chromosome conformation capture (3C) assess contacts for millions of loci simultaneously, but do so by averaging chromosome conformations from millions of nuclei. Here we introduce single-cell Hi-C, combined with genome-wide statistical analysis and structural modelling of single-copy X chromosomes, to show that individual chromosomes maintain domain organization at the megabase scale, but show variable cell-to-cell chromosome structures at larger scales. Despite this structural stochasticity, localization of active gene domains to boundaries of chromosome territories is a hallmark of chromosomal conformation. Single-cell Hi-C data bridge current gaps between genomics and microscopy studies of chromosomes, demonstrating how modular organization underlies dynamic chromosome structure, and how this structure is probabilistically linked with genome activity patterns.


Asunto(s)
Cromosomas/química , Técnicas Genéticas , Modelos Moleculares , Animales , Núcleo Celular/genética , Cromatina/química , Cromosomas/genética , Masculino , Ratones , Conformación Molecular , Análisis de la Célula Individual , Cromosoma X/química , Cromosoma X/genética
10.
Genes Dev ; 25(13): 1371-83, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21690198

RESUMEN

Three-dimensional topology of DNA in the cell nucleus provides a level of transcription regulation beyond the sequence of the linear DNA. To study the relationship between the transcriptional activity and the spatial environment of a gene, we used allele-specific chromosome conformation capture-on-chip (4C) technology to produce high-resolution topology maps of the active and inactive X chromosomes in female cells. We found that loci on the active X form multiple long-range interactions, with spatial segregation of active and inactive chromatin. On the inactive X, silenced loci lack preferred interactions, suggesting a unique random organization inside the inactive territory. However, escapees, among which is Xist, are engaged in long-range contacts with each other, enabling identification of novel escapees. Deletion of Xist results in partial refolding of the inactive X into a conformation resembling the active X without affecting gene silencing or DNA methylation. Our data point to a role for Xist RNA in shaping the conformation of the inactive X chromosome at least partially independent of transcription.


Asunto(s)
Estructuras Cromosómicas , ARN no Traducido/genética , Cromosoma X/química , Animales , Femenino , Genes Ligados a X/genética , Ratones , ARN Largo no Codificante , ARN no Traducido/metabolismo
11.
Nature ; 485(7398): 381-5, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22495304

RESUMEN

In eukaryotes transcriptional regulation often involves multiple long-range elements and is influenced by the genomic environment. A prime example of this concerns the mouse X-inactivation centre (Xic), which orchestrates the initiation of X-chromosome inactivation (XCI) by controlling the expression of the non-protein-coding Xist transcript. The extent of Xic sequences required for the proper regulation of Xist remains unknown. Here we use chromosome conformation capture carbon-copy (5C) and super-resolution microscopy to analyse the spatial organization of a 4.5-megabases (Mb) region including Xist. We discover a series of discrete 200-kilobase to 1 Mb topologically associating domains (TADs), present both before and after cell differentiation and on the active and inactive X. TADs align with, but do not rely on, several domain-wide features of the epigenome, such as H3K27me3 or H3K9me2 blocks and lamina-associated domains. TADs also align with coordinately regulated gene clusters. Disruption of a TAD boundary causes ectopic chromosomal contacts and long-range transcriptional misregulation. The Xist/Tsix sense/antisense unit illustrates how TADs enable the spatial segregation of oppositely regulated chromosomal neighbourhoods, with the respective promoters of Xist and Tsix lying in adjacent TADs, each containing their known positive regulators. We identify a novel distal regulatory region of Tsix within its TAD, which produces a long intervening RNA, Linx. In addition to uncovering a new principle of cis-regulatory architecture of mammalian chromosomes, our study sets the stage for the full genetic dissection of the X-inactivation centre.


Asunto(s)
ARN no Traducido/genética , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Animales , Diferenciación Celular , ADN Intergénico/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Epigenómica , Femenino , Fibroblastos , Regulación de la Expresión Génica , Histonas/metabolismo , Hibridación Fluorescente in Situ , Masculino , Metilación , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante , Transcriptoma , Cromosoma X/química
12.
Genes Dev ; 23(21): 2490-5, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19884256

RESUMEN

The dosage compensation complex (DCC) in Drosophila globally increases transcription from the X chromosome in males to compensate for its monosomy. We discovered a male-specific conformation of the X chromosome that depends on the associations of high-affinity binding sites (HAS) of the DCC. The core DCC subunits MSL1-MSL2 are responsible for this male-specific organization. Contrary to emerging concepts, we found that neither DCC assembly nor the conformation of the male X chromosome are influenced by nuclear pore components. We propose that nuclear organization of HAS is central to the faithful distribution of the DCC along the X chromosome.


Asunto(s)
Compensación de Dosificación (Genética) , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Cromosoma X/química , Cromosoma X/genética , Animales , Núcleo Celular/metabolismo , Desarrollo Embrionario , Interfase/genética , Masculino , Unión Proteica
13.
Reproduction ; 145(1): R15-30, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23148085

RESUMEN

Pre-selection of spermatozoa based on the relative DNA difference between X- and Y-chromosome bearing populations by flow cytometry is an established method that has been introduced into commercial cattle production. Although several important improvements have increased the sort efficiency, the fertilising ability of sexed spermatozoa based on offspring per insemination is still behind farmers' expectations. The main stress factors, especially on mitochondria, that reduce the lifespan of spermatozoa are described, and new technical as well as biological solutions to maintain the natural sperm integrity and to increase the sorting efficiency are discussed. Among these methods are the identification of Y-chromosome bearing spermatozoa by bi-functionalised gold nanoparticles and triplex hybridisation in vivo as well as new laser-controlled deflection system that replaces the deflection of spermatozoa in the electrostatic field. Additionally, as well as a new nonsurgical transfer system of spermatozoa into the oviduct of cows has been developed and allows a significant reduction of spermatozoa per transfer. Altogether, the improvements made in the recent years will allow a broader use of sex-sorted spermatozoa even in those species that require more cells than cows and sheep.


Asunto(s)
Animales Domésticos , Cruzamiento/métodos , Inseminación Artificial/métodos , Preselección del Sexo/métodos , Espermatozoides/citología , Animales , Bovinos , ADN/análisis , Femenino , Citometría de Flujo/métodos , Masculino , Nanopartículas del Metal , Oveja Doméstica , Cromosoma X/química , Cromosoma Y/química
14.
Nat Genet ; 20(2): 207-11, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9771718

RESUMEN

Gene dosage variations occur in many diseases. In cancer, deletions and copy number increases contribute to alterations in the expression of tumour-suppressor genes and oncogenes, respectively. Developmental abnormalities, such as Down, Prader Willi, Angelman and Cri du Chat syndromes, result from gain or loss of one copy of a chromosome or chromosomal region. Thus, detection and mapping of copy number abnormalities provide an approach for associating aberrations with disease phenotype and for localizing critical genes. Comparative genomic hybridization (CGH) was developed for genome-wide analysis of DNA sequence copy number in a single experiment. In CGH, differentially labelled total genomic DNA from a 'test' and a 'reference' cell population are cohybridized to normal metaphase chromosomes, using blocking DNA to suppress signals from repetitive sequences. The resulting ratio of the fluorescence intensities at a location on the 'cytogenetic map', provided by the chromosomes, is approximately proportional to the ratio of the copy numbers of the corresponding DNA sequences in the test and reference genomes. CGH has been broadly applied to human and mouse malignancies. The use of metaphase chromosomes, however, limits detection of events involving small regions (of less than 20 Mb) of the genome, resolution of closely spaced aberrations and linking ratio changes to genomic/genetic markers. Therefore, more laborious locus-by-locus techniques have been required for higher resolution studies. Hybridization to an array of mapped sequences instead of metaphase chromosomes could overcome the limitations of conventional CGH (ref. 6) if adequate performance could be achieved. Copy number would be related to the test/reference fluorescence ratio on the array targets, and genomic resolution could be determined by the map distance between the targets, or by the length of the cloned DNA segments. We describe here our implementation of array CGH. We demonstrate its ability to measure copy number with high precision in the human genome, and to analyse clinical specimens by obtaining new information on chromosome 20 aberrations in breast cancer.


Asunto(s)
ADN/química , Dosificación de Gen , Hibridación de Ácido Nucleico/métodos , Animales , Neoplasias de la Mama/genética , Aberraciones Cromosómicas , Femenino , Marcadores Genéticos , Humanos , Hibridación Fluorescente in Situ , Ratones , Microquímica , Células Tumorales Cultivadas , Cromosoma X/química
15.
Chromosoma ; 120(4): 335-51, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21384262

RESUMEN

Drosophila males undergo meiosis without recombination or chiasmata but homologous chromosomes pair and disjoin regularly. The X-Y pair utilizes a specific repeated sequence within the heterochromatic ribosomal DNA blocks as a pairing site. No pairing sites have yet been identified for the autosomes. To search for such sites, we utilized probes targeting specific heterochromatic regions to assay heterochromatin pairing sequences and behavior in meiosis by fluorescence in situ hybridization (FISH). We found that the small fourth chromosome pairs at heterochromatic region 61 and associates with the X chromosome throughout prophase I. Homolog pairing of the fourth chromosome is disrupted when the homolog conjunction complex is perturbed by mutations in SNM or MNM. On the other hand, six tested heterochromatic regions of the major autosomes proved to be largely unpaired after early prophase I, suggesting that stable homolog pairing sites do not exist in heterochromatin of the major autosomes. Furthermore, FISH analysis revealed two distinct patterns of sister chromatid cohesion in heterochromatin: regions with stable cohesion and regions lacking cohesion. This suggests that meiotic sister chromatid cohesion is incomplete within heterochromatin and may occur at specific preferential sites.


Asunto(s)
Cromátides , Drosophila melanogaster/genética , Heterocromatina/química , Meiosis , Espermatocitos/metabolismo , Cromosoma X/química , Cromosoma Y/química , Animales , Centrómero/química , Centrómero/genética , Cromátides/química , Cromátides/genética , Cromátides/metabolismo , Emparejamiento Cromosómico , Segregación Cromosómica , Fluorescencia , Heterocromatina/genética , Hibridación Fluorescente in Situ , Masculino , Sondas Moleculares/análisis , Mutación , Espermatocitos/citología , Cromosoma X/genética , Cromosoma Y/genética
16.
Chromosoma ; 120(4): 387-97, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21533987

RESUMEN

Sciara coprophila (Diptera, Nematocera) constitutes a classic model to analyze unusual chromosome behavior such as the somatic elimination of paternal X chromosomes, the elimination of the whole paternal, plus non-disjunction of the maternal X chromosome at male meiosis. The molecular organization of the heterochromatin in S. coprophila is mostly unknown except for the ribosomal DNA located in the X chromosome pericentromeric heterochromatin. The characterization of the centromeric regions, thus, is an essential and required step for the establishment of S. coprophila as a model system to study fundamental mechanisms of chromosome segregation. To accomplish such a study, heterochromatic sections of the X chromosome centromeric region from salivary glands polytene chromosomes were microdissected and microcloned. Here, we report the identification and characterization of two tandem repeated DNA sequences from the pericentromeric region of the X chromosome, a pericentromeric RTE element and an AT-rich centromeric satellite. These sequences will be important tools for the cloning of S. coprophila centromeric heterochromatin using libraries of large genomic clones.


Asunto(s)
Centrómero/química , ADN/química , Dípteros/genética , Heterocromatina/química , Larva/genética , Cromosomas Politénicos/química , Secuencias Repetidas en Tándem/genética , Cromosoma X/química , Animales , Centrómero/genética , Mapeo Cromosómico , ADN/genética , Heterocromatina/genética , Hibridación Fluorescente in Situ , Masculino , Meiosis/genética , Datos de Secuencia Molecular , Filogenia , Cromosomas Politénicos/genética , Glándulas Salivales/química , Glándulas Salivales/citología , Fijación del Tejido , Cromosoma X/genética
17.
Cytogenet Genome Res ; 135(1): 79-82, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21811057

RESUMEN

Human and canine lymphoid neoplasms are characterized by non-random cytogenetic abnormalities. However, due to the low mitotic activity of the B cells, cytogenetic analyses of B-cell lymphoid proliferations are difficult to perform. In the present study we stimulated canine B-cell lymphoma cells with the immunostimulatory CpG-oligonucleotide DSP30 in combination with interleukin-2 (IL-2) and obtained an adequate number of metaphases. Cytogenetic analyses revealed the loss of one X chromosome as the sole cytogenetic aberration. Chromosome analysis of the corresponding blood showed a normal female karyotype. Monosomy X as the sole clonal chromosomal abnormality is found in human hematopoietic malignancies as well, thus the dog may serve as a promising animal model.


Asunto(s)
Linfocitos B/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos , Citogenética/métodos , Enfermedades de los Perros , Ganglios Linfáticos/patología , Linfoma de Células B , Monosomía , Cromosoma X/química , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Células Cultivadas , Enfermedades de los Perros/genética , Enfermedades de los Perros/inmunología , Perros , Femenino , Humanos , Interleucina-2/farmacología , Cariotipificación , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Linfoma de Células B/genética , Linfoma de Células B/inmunología , Metafase , Oligonucleótidos/farmacología , Cromosoma X/genética
18.
Genome Biol ; 22(1): 279, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34579774

RESUMEN

BACKGROUND: Mammalian development is associated with extensive changes in gene expression, chromatin accessibility, and nuclear structure. Here, we follow such changes associated with mouse embryonic stem cell differentiation and X inactivation by integrating, for the first time, allele-specific data from these three modalities obtained by high-throughput single-cell RNA-seq, ATAC-seq, and Hi-C. RESULTS: Allele-specific contact decay profiles obtained by single-cell Hi-C clearly show that the inactive X chromosome has a unique profile in differentiated cells that have undergone X inactivation. Loss of this inactive X-specific structure at mitosis is followed by its reappearance during the cell cycle, suggesting a "bookmark" mechanism. Differentiation of embryonic stem cells to follow the onset of X inactivation is associated with changes in contact decay profiles that occur in parallel on both the X chromosomes and autosomes. Single-cell RNA-seq and ATAC-seq show evidence of a delay in female versus male cells, due to the presence of two active X chromosomes at early stages of differentiation. The onset of the inactive X-specific structure in single cells occurs later than gene silencing, consistent with the idea that chromatin compaction is a late event of X inactivation. Single-cell Hi-C highlights evidence of discrete changes in nuclear structure characterized by the acquisition of very long-range contacts throughout the nucleus. Novel computational approaches allow for the effective alignment of single-cell gene expression, chromatin accessibility, and 3D chromosome structure. CONCLUSIONS: Based on trajectory analyses, three distinct nuclear structure states are detected reflecting discrete and profound simultaneous changes not only to the structure of the X chromosomes, but also to that of autosomes during differentiation. Our study reveals that long-range structural changes to chromosomes appear as discrete events, unlike progressive changes in gene expression and chromatin accessibility.


Asunto(s)
Diferenciación Celular/genética , Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Inactivación del Cromosoma X , Alelos , Animales , Ciclo Celular , Línea Celular , Núcleo Celular/genética , Femenino , Genoma , Masculino , Ratones , RNA-Seq , Análisis de la Célula Individual , Cromosoma X/química
19.
Methods Mol Biol ; 2329: 237-247, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34085227

RESUMEN

Posttranslational histone modifications are critical for the regulation of genome function. The levels of histone modifications oscillate during the cell cycle. Most modifications are diluted after DNA replication and then their levels are restored during the rest of the cell cycle with different kinetics depending on the modification. Some modifications, like histone H4 Lys20 monomethylation (H4K20me1), exhibit cell cycle-dependent dynamic changes. To track histone modifications in living cells, we have developed genetically encoded probes termed modification specific intracellular antibodies, or "mintbodies." As mintbodies shuttle between the cytoplasm and nucleus by diffusion, their nuclear concentration depends on the target modification level. By measuring the nuclear to cytoplasmic intensity ratio of H4K20me1-specific mintbody, we have monitored the increase of H4K20me1 in the G2 phase. Here we describe how the mintbody-based methods can be applied to track a specific chromosome, such as the inactive X chromosome (Xi), on which genes are repressed through histone H3 Lys27 trimethylation (H3K27me3). When H3K27me3-specific mintbodies are expressed in cells that harbor Xi, the mintbodies are concentrated on Xi and the dynamic behavior of Xi can be tracked using a confocal microscope. After acquiring 3D time-lapse images, an image analysis allows measuring the volume, shape and H3K27me3 level of Xi during the cell cycle.


Asunto(s)
Colorantes Fluorescentes/química , Histonas/metabolismo , Inactivación del Cromosoma X , Cromosoma X/genética , Animales , Ciclo Celular , Línea Celular Tumoral , Células HeLa , Código de Histonas , Histonas/química , Humanos , Metilación , Ratones , Microscopía Confocal , Imagen de Lapso de Tiempo , Cromosoma X/química
20.
Nucleic Acids Res ; 36(3): 950-62, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18086708

RESUMEN

Loss of function of the RNA helicase maleless (MLE) in Drosophila melanogaster leads to male-specific lethality due to a failure of X chromosome dosage compensation. MLE is presumably involved in incorporating the non-coding roX RNA into the dosage compensation complex (DCC), which is an essential but poorly understood requirement for faithful targeting of the complex to the X chromosome. Sequence comparison predicts several RNA-binding domains in MLE but their properties have not been experimentally verified. We evaluated the RNA-binding characteristics of these conserved motifs and their contributions to RNA-stimulated ATPase activity, to helicase activity, as well as to the targeting of MLE to the nucleus and to the X chromosome territory. We find that RB2 is the dominant, conditional RNA-binding module, which is indispensable for ATPase and helicase activity whereas the N-terminal RB1 motif does not bind RNA, but is involved in targeting MLE to the X chromosome. The C-terminal domain containing a glycine-rich heptad repeat adds potential dimerization and RNA-binding surfaces which are not required for helicase activity.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/química , ADN Helicasas/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , ARN Helicasas/química , ARN Helicasas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Nucleótidos de Adenina/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Sitios de Unión , Línea Celular , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Mutación , Estructura Terciaria de Proteína , ARN/química , ARN/metabolismo , ARN Helicasas/genética , Relación Estructura-Actividad , Factores de Transcripción/genética , Cromosoma X/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA