Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(5): 1309-1324.e18, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30078704

RESUMEN

We applied a combinatorial indexing assay, sci-ATAC-seq, to profile genome-wide chromatin accessibility in ∼100,000 single cells from 13 adult mouse tissues. We identify 85 distinct patterns of chromatin accessibility, most of which can be assigned to cell types, and ∼400,000 differentially accessible elements. We use these data to link regulatory elements to their target genes, to define the transcription factor grammar specifying each cell type, and to discover in vivo correlates of heterogeneity in accessibility within cell types. We develop a technique for mapping single cell gene expression data to single-cell chromatin accessibility data, facilitating the comparison of atlases. By intersecting mouse chromatin accessibility with human genome-wide association summary statistics, we identify cell-type-specific enrichments of the heritability signal for hundreds of complex traits. These data define the in vivo landscape of the regulatory genome for common mammalian cell types at single-cell resolution.


Asunto(s)
Cromatina/química , Análisis de la Célula Individual/métodos , Animales , Análisis por Conglomerados , Epigénesis Genética , Epigenómica , Regulación de la Expresión Génica , Genoma Humano , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Mamíferos , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción
2.
Mol Cell ; 83(15): 2624-2640, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37419111

RESUMEN

The four-dimensional nucleome (4DN) consortium studies the architecture of the genome and the nucleus in space and time. We summarize progress by the consortium and highlight the development of technologies for (1) mapping genome folding and identifying roles of nuclear components and bodies, proteins, and RNA, (2) characterizing nuclear organization with time or single-cell resolution, and (3) imaging of nuclear organization. With these tools, the consortium has provided over 2,000 public datasets. Integrative computational models based on these data are starting to reveal connections between genome structure and function. We then present a forward-looking perspective and outline current aims to (1) delineate dynamics of nuclear architecture at different timescales, from minutes to weeks as cells differentiate, in populations and in single cells, (2) characterize cis-determinants and trans-modulators of genome organization, (3) test functional consequences of changes in cis- and trans-regulators, and (4) develop predictive models of genome structure and function.


Asunto(s)
Núcleo Celular , Genoma , Genoma/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/metabolismo
3.
Nature ; 626(8001): 1084-1093, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38355799

RESUMEN

The house mouse (Mus musculus) is an exceptional model system, combining genetic tractability with close evolutionary affinity to humans1,2. Mouse gestation lasts only 3 weeks, during which the genome orchestrates the astonishing transformation of a single-cell zygote into a free-living pup composed of more than 500 million cells. Here, to establish a global framework for exploring mammalian development, we applied optimized single-cell combinatorial indexing3 to profile the transcriptional states of 12.4 million nuclei from 83 embryos, precisely staged at 2- to 6-hour intervals spanning late gastrulation (embryonic day 8) to birth (postnatal day 0). From these data, we annotate hundreds of cell types and explore the ontogenesis of the posterior embryo during somitogenesis and of kidney, mesenchyme, retina and early neurons. We leverage the temporal resolution and sampling depth of these whole-embryo snapshots, together with published data4-8 from earlier timepoints, to construct a rooted tree of cell-type relationships that spans the entirety of prenatal development, from zygote to birth. Throughout this tree, we systematically nominate genes encoding transcription factors and other proteins as candidate drivers of the in vivo differentiation of hundreds of cell types. Remarkably, the most marked temporal shifts in cell states are observed within one hour of birth and presumably underlie the massive physiological adaptations that must accompany the successful transition of a mammalian fetus to life outside the womb.


Asunto(s)
Animales Recién Nacidos , Embrión de Mamíferos , Desarrollo Embrionario , Gástrula , Análisis de la Célula Individual , Imagen de Lapso de Tiempo , Animales , Femenino , Ratones , Embarazo , Animales Recién Nacidos/embriología , Animales Recién Nacidos/genética , Diferenciación Celular/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Desarrollo Embrionario/genética , Gástrula/citología , Gástrula/embriología , Gastrulación/genética , Riñón/citología , Riñón/embriología , Mesodermo/citología , Mesodermo/enzimología , Neuronas/citología , Neuronas/metabolismo , Retina/citología , Retina/embriología , Somitos/citología , Somitos/embriología , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética , Especificidad de Órganos/genética
4.
Mol Cell ; 76(4): 676-690.e10, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31495564

RESUMEN

Conventional methods for single-cell genome sequencing are limited with respect to uniformity and throughput. Here, we describe sci-L3, a single-cell sequencing method that combines combinatorial indexing (sci-) and linear (L) amplification. The sci-L3 method adopts a 3-level (3) indexing scheme that minimizes amplification biases while enabling exponential gains in throughput. We demonstrate the generalizability of sci-L3 with proof-of-concept demonstrations of single-cell whole-genome sequencing (sci-L3-WGS), targeted sequencing (sci-L3-target-seq), and a co-assay of the genome and transcriptome (sci-L3-RNA/DNA). We apply sci-L3-WGS to profile the genomes of >10,000 sperm and sperm precursors from F1 hybrid mice, mapping 86,786 crossovers and characterizing rare chromosome mis-segregation events in meiosis, including instances of whole-genome equational chromosome segregation. We anticipate that sci-L3 assays can be applied to fully characterize recombination landscapes, to couple CRISPR perturbations and measurements of genome stability, and to other goals requiring high-throughput, high-coverage single-cell sequencing.


Asunto(s)
Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Técnicas de Amplificación de Ácido Nucleico , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos , Secuenciación Completa del Genoma , Animales , Segregación Cromosómica , Masculino , Meiosis/genética , Ratones , Prueba de Estudio Conceptual , Espermatozoides/fisiología , Transcriptoma , Flujo de Trabajo
5.
Proc Natl Acad Sci U S A ; 120(21): e2218478120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37192167

RESUMEN

Aneuploidy syndromes impact multiple organ systems but understanding of tissue-specific aneuploidy effects remains limited-especially for the comparison between peripheral tissues and relatively inaccessible tissues like brain. Here, we address this gap in knowledge by studying the transcriptomic effects of chromosome X, Y, and 21 aneuploidies in lymphoblastoid cell lines, fibroblasts and iPSC-derived neuronal cells (LCLs, FCL, and iNs, respectively). We root our analyses in sex chromosome aneuploidies, which offer a uniquely wide karyotype range for dosage effect analysis. We first harness a large LCL RNA-seq dataset from 197 individuals with one of 6 sex chromosome dosages (SCDs: XX, XXX, XY, XXY, XYY, and XXYY) to i) validate theoretical models of SCD sensitivity and ii) define an expanded set of 41 genes that show obligate dosage sensitivity to SCD and are all in cis (i.e., reside on the X or Y chromosome). We then use multiple complementary analyses to show that cis effects of SCD in LCLs are preserved in both FCLs (n = 32) and iNs (n = 24), whereas trans effects (i.e., those on autosomal gene expression) are mostly not preserved. Analysis of additional datasets confirms that the greater cross-cell type reproducibility of cis vs. trans effects is also seen in trisomy 21 cell lines. These findings i) expand our understanding of X, Y, and 21 chromosome dosage effects on human gene expression and ii) suggest that LCLs may provide a good model system for understanding cis effects of aneuploidy in harder-to-access cell types.


Asunto(s)
Aneuploidia , Síndrome de Down , Humanos , Reproducibilidad de los Resultados , Síndrome de Down/genética , Cromosomas Sexuales , Expresión Génica
6.
Hum Mol Genet ; 30(R2): R285-R295, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34387327

RESUMEN

The gene content of the X and Y chromosomes has dramatically diverged during evolution. The ensuing dosage imbalance within the genome of males and females has led to unique chromosome-wide regulatory mechanisms with significant and sex-specific impacts on X-linked gene expression. X inactivation or silencing of most genes on one X chromosome chosen at random in females profoundly affects the manifestation of X-linked diseases, as males inherit a single maternal allele, while females express maternal and paternal alleles in a mosaic manner. An additional complication is the existence of genes that escape X inactivation and thus are ubiquitously expressed from both alleles in females. The mosaic nature of X-linked gene expression and the potential for escape can vary between individuals, tissues, cell types and stages of life. Our understanding of the specialized nature of X-linked genes and of the multilayer epigenetic regulation that influence their expression throughout the organism has been helped by molecular studies conducted by tissue-specific and single-cell-specific approaches. In turn, the definition of molecular events that control X silencing has helped develop new approaches for the treatment of some X-linked disorders. This review focuses on the peculiarities of the X chromosome genetic content and epigenetic regulation in shaping the manifestation of congenital and acquired X-linked disorders in a sex-specific manner.


Asunto(s)
Genes Ligados a X , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Inactivación del Cromosoma X , Alelos , Aneuploidia , Cromosomas Humanos X , Femenino , Dosificación de Gen , Regulación de la Expresión Génica , Humanos , Masculino , Especificidad de Órganos/genética
7.
Methods ; 170: 61-68, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31536770

RESUMEN

The highly dynamic nature of chromosome conformation and three-dimensional (3D) genome organization leads to cell-to-cell variability in chromatin interactions within a cell population, even if the cells of the population appear to be functionally homogeneous. Hence, although Hi-C is a powerful tool for mapping 3D genome organization, this heterogeneity of chromosome higher order structure among individual cells limits the interpretive power of population based bulk Hi-C assays. Moreover, single-cell studies have the potential to enable the identification and characterization of rare cell populations or cell subtypes in a heterogeneous population. However, it may require surveying relatively large numbers of single cells to achieve statistically meaningful observations in single-cell studies. By applying combinatorial cellular indexing to chromosome conformation capture, we developed single-cell combinatorial indexed Hi-C (sci-Hi-C), a high throughput method that enables mapping chromatin interactomes in large number of single cells. We demonstrated the use of sci-Hi-C data to separate cells by karytoypic and cell-cycle state differences and to identify cellular variability in mammalian chromosomal conformation. Here, we provide a detailed description of method design and step-by-step working protocols for sci-Hi-C.


Asunto(s)
Mapeo Cromosómico/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de la Célula Individual/métodos , Animales , Línea Celular , Núcleo Celular/genética , Separación Celular/métodos , Cromatina/genética , Cromatina/aislamiento & purificación , Cromatina/metabolismo , Simulación por Computador , Biblioteca de Genes , Humanos , Ratones , Conformación de Ácido Nucleico
8.
Annu Rev Genet ; 46: 537-60, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22974302

RESUMEN

Differentiated sex chromosomes evolved because of suppressed recombination once sex became genetically controlled. In XX/XY and ZZ/ZW systems, the heterogametic sex became partially aneuploid after degeneration of the Y or W. Often, aneuploidy causes abnormal levels of gene expression throughout the entire genome. Dosage compensation mechanisms evolved to restore balanced expression of the genome. These mechanisms include upregulation of the heterogametic chromosome as well as repression in the homogametic sex. Remarkably, strategies for dosage compensation differ between species. In organisms where more is known about molecular mechanisms of dosage compensation, specific protein complexes containing noncoding RNAs are targeted to the X chromosome. In addition, the dosage-regulated chromosome often occupies a specific nuclear compartment. Some genes escape dosage compensation, potentially resulting in sex-specific differences in gene expression. This review focuses on dosage compensation in mammals, with comparisons to fruit flies, nematodes, and birds.


Asunto(s)
Compensación de Dosificación (Genética) , Cromosoma X/genética , Cromosoma Y/genética , Alelos , Aneuploidia , Animales , Aves/genética , Drosophila/genética , Evolución Molecular , Femenino , Variación Genética , Masculino , Mamíferos/genética , Nematodos/genética , Poliploidía , ARN no Traducido/genética , Factores Sexuales , Cromosoma X/metabolismo , Cromosoma Y/metabolismo
9.
Nat Methods ; 14(3): 263-266, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135255

RESUMEN

We present single-cell combinatorial indexed Hi-C (sciHi-C), a method that applies combinatorial cellular indexing to chromosome conformation capture. In this proof of concept, we generate and sequence six sciHi-C libraries comprising a total of 10,696 single cells. We use sciHi-C data to separate cells by karyotypic and cell-cycle state differences and identify cell-to-cell heterogeneity in mammalian chromosomal conformation. Our results demonstrate that combinatorial indexing is a generalizable strategy for single-cell genomics.


Asunto(s)
Cromosomas/genética , ADN/genética , Genoma Humano/genética , Genómica/métodos , Conformación Molecular , Análisis de la Célula Individual/métodos , Ciclo Celular/genética , Línea Celular Tumoral , ADN/análisis , Biblioteca de Genes , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Análisis de Secuencia de ADN/métodos
10.
Nat Rev Genet ; 15(6): 367-78, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24733023

RESUMEN

Genes on the mammalian X chromosome are present in one copy in males and two copies in females. The complex mechanisms that regulate the X chromosome lead to evolutionary and physiological variability in gene expression between species, the sexes, individuals, developmental stages, tissues and cell types. In early development, delayed and incomplete X chromosome inactivation (XCI) in some species causes variability in gene expression. Additional diversity stems from escape from XCI and from mosaicism or XCI skewing in females. This causes sex-specific differences that manifest as differential gene expression and associated phenotypes. Furthermore, the complexity and diversity of X dosage regulation affect the severity of diseases caused by X-linked mutations.


Asunto(s)
Trastornos de los Cromosomas , Cromosomas Humanos X , Regulación de la Expresión Génica , Enfermedades Genéticas Ligadas al Cromosoma X , Caracteres Sexuales , Inactivación del Cromosoma X , Animales , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/metabolismo , Cromosomas Humanos X/genética , Cromosomas Humanos X/metabolismo , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Humanos , Masculino , Mosaicismo
11.
Proc Natl Acad Sci U S A ; 114(14): E2882-E2890, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28320934

RESUMEN

DNA methylation at gene promoters in a CG context is associated with transcriptional repression, including at genes silenced on the inactive X chromosome in females. Non-CG methylation (mCH) is a distinct feature of the neuronal epigenome that is differentially distributed between males and females on the X chromosome. However, little is known about differences in mCH on the active (Xa) and inactive (Xi) X chromosomes because stochastic X-chromosome inactivation (XCI) confounds allele-specific epigenomic profiling. We used whole-genome bisulfite sequencing in a mouse model with nonrandom XCI to examine allele-specific DNA methylation in frontal cortex. Xi was largely devoid of mCH, whereas Xa contained abundant mCH similar to the male X chromosome and the autosomes. In contrast to the repressive association of DNA methylation at CG dinucleotides (mCG), mCH accumulates on Xi in domains with transcriptional activity, including the bodies of most genes that escape XCI and at the X-inactivation center, validating this epigenetic mark as a signature of transcriptional activity. Escape genes showing CH hypermethylation were the only genes with CG-hypomethylated promoters on Xi, a well-known mark of active transcription. Finally, we found extensive allele-specific mCH and mCG at autosomal imprinted regions, some with a negative correlation between methylation in the two contexts, further supporting their distinct functions. Our findings show that neuronal mCH functions independently of mCG and is a highly dynamic epigenomic correlate of allele-specific gene regulation.


Asunto(s)
Encéfalo/fisiología , Cromatina/metabolismo , Metilación de ADN , Alelos , Animales , Epigénesis Genética , Femenino , Impresión Genómica , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Polimorfismo de Nucleótido Simple , ARN Largo no Codificante/genética , Cromosoma X , Inactivación del Cromosoma X
12.
Am J Med Genet A ; 179(9): 1783-1790, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31294511

RESUMEN

Rare individuals with 20p11.2 proximal deletions have been previously reported, with a variable phenotype that includes heterotaxy, biliary atresia, midline brain defects associated with panhypopituitarism, intellectual disability, scoliosis, and seizures. Deletions have ranged in size from 277 kb to 11.96 Mb. We describe a newborn with a de novo 2.7 Mb deletion of 20p11.22p11.21 that partially overlaps previously reported deletions and encompasses FOXA2. Her clinical findings further expand the 20p11.2 deletion phenotype to include severe midline cranial and intracranial defects such as aqueductal stenosis with hydrocephalus, mesencephalosynapsis with diencephalic-mesencephalic junction dysplasia, and pyriform aperture stenosis. We also report one individual with a missense variant in FOXA2 who had abnormal glucose homeostasis, panhypopituitarism, and endodermal organ dysfunction. Together, these findings support the critical role of FOXA2 in panhypopituitarism and midline defects.


Asunto(s)
Encéfalo/anomalías , Constricción Patológica/genética , Factor Nuclear 3-beta del Hepatocito/genética , Hipopituitarismo/genética , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Deleción Cromosómica , Cromosomas Humanos Par 20/genética , Constricción Patológica/diagnóstico por imagen , Constricción Patológica/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Hidrocefalia/diagnóstico por imagen , Hidrocefalia/genética , Hidrocefalia/fisiopatología , Hipopituitarismo/diagnóstico por imagen , Hipopituitarismo/fisiopatología , Recién Nacido , Mutación Missense/genética , Fenotipo , Corteza Piriforme/diagnóstico por imagen , Corteza Piriforme/fisiopatología
13.
Methods ; 142: 59-73, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29382556

RESUMEN

The folding and three-dimensional (3D) organization of chromatin in the nucleus critically impacts genome function. The past decade has witnessed rapid advances in genomic tools for delineating 3D genome architecture. Among them, chromosome conformation capture (3C)-based methods such as Hi-C are the most widely used techniques for mapping chromatin interactions. However, traditional Hi-C protocols rely on restriction enzymes (REs) to fragment chromatin and are therefore limited in resolution. We recently developed DNase Hi-C for mapping 3D genome organization, which uses DNase I for chromatin fragmentation. DNase Hi-C overcomes RE-related limitations associated with traditional Hi-C methods, leading to improved methodological resolution. Furthermore, combining this method with DNA capture technology provides a high-throughput approach (targeted DNase Hi-C) that allows for mapping fine-scale chromatin architecture at exceptionally high resolution. Hence, targeted DNase Hi-C will be valuable for delineating the physical landscapes of cis-regulatory networks that control gene expression and for characterizing phenotype-associated chromatin 3D signatures. Here, we provide a detailed description of method design and step-by-step working protocols for these two methods.


Asunto(s)
Mapeo Cromosómico/métodos , Desoxirribonucleasa I/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Imagenología Tridimensional/métodos , Imagen Molecular/métodos , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/química , Cromatina/genética , Mapeo Cromosómico/instrumentación , Reactivos de Enlaces Cruzados/química , Enzimas de Restricción del ADN/química , Enzimas de Restricción del ADN/metabolismo , Desoxirribonucleasa I/química , Formaldehído/química , Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/instrumentación , Imagenología Tridimensional/instrumentación , Imagen Molecular/instrumentación , Técnicas de Cultivo de Tejidos/instrumentación , Técnicas de Cultivo de Tejidos/métodos , Secuenciación Completa del Genoma/instrumentación , Secuenciación Completa del Genoma/métodos
14.
Semin Cell Dev Biol ; 56: 9-18, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27112542

RESUMEN

Males are XY and females are XX in most mammalian species. Other species such as birds have a different sex chromosome make-up: ZZ in males and ZW in females. In both types of organisms one of the sex chromosomes, Y or W, has degenerated due to lack of recombination with its respective homolog X or Z. Since autosomes are present in two copies in diploid organisms the heterogametic sex has become a natural "aneuploid" with haploinsufficiency for X- or Z-linked genes. Specific mechanisms have evolved to restore a balance between critical gene products throughout the genome and between males and females. Some of these mechanisms were co-opted from and/or added to compensatory processes that alleviate autosomal aneuploidy. Surprisingly, several modes of dosage compensation have evolved. In this review we will consider the evidence for dosage compensation and the molecular mechanisms implicated.


Asunto(s)
Compensación de Dosificación (Genética) , Cromosomas Sexuales/genética , Alelos , Aneuploidia , Animales , Evolución Molecular , Humanos , Regulación hacia Arriba/genética
15.
Nat Methods ; 12(1): 71-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25437436

RESUMEN

High-throughput methods based on chromosome conformation capture have greatly advanced our understanding of the three-dimensional (3D) organization of genomes but are limited in resolution by their reliance on restriction enzymes. Here we describe a method called DNase Hi-C for comprehensively mapping global chromatin contacts. DNase Hi-C uses DNase I for chromatin fragmentation, leading to greatly improved efficiency and resolution over that of Hi-C. Coupling this method with DNA-capture technology provides a high-throughput approach for targeted mapping of fine-scale chromatin architecture. We applied targeted DNase Hi-C to characterize the 3D organization of 998 large intergenic noncoding RNA (lincRNA) promoters in two human cell lines. Our results revealed that expression of lincRNAs is tightly controlled by complex mechanisms involving both super-enhancers and the Polycomb repressive complex. Our results provide the first glimpse of the cell type-specific 3D organization of lincRNA genes.


Asunto(s)
Cromatina/fisiología , ARN no Traducido/genética , Cromatina/química , Cromatina/ultraestructura , Mapeo Cromosómico , Desoxirribonucleasa I/metabolismo , Genoma , Humanos , Células K562 , Conformación Proteica , Elementos Reguladores de la Transcripción/genética
16.
PLoS Genet ; 11(3): e1005079, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25785854

RESUMEN

X chromosome inactivation (XCI) silences most genes on one X chromosome in female mammals, but some genes escape XCI. To identify escape genes in vivo and to explore molecular mechanisms that regulate this process we analyzed the allele-specific expression and chromatin structure of X-linked genes in mouse tissues and cells with skewed XCI and distinguishable alleles based on single nucleotide polymorphisms. Using a binomial model to assess allelic expression, we demonstrate a continuum between complete silencing and expression from the inactive X (Xi). The validity of the RNA-seq approach was verified using RT-PCR with species-specific primers or Sanger sequencing. Both common escape genes and genes with significant differences in XCI status between tissues were identified. Such genes may be candidates for tissue-specific sex differences. Overall, few genes (3-7%) escape XCI in any of the mouse tissues examined, suggesting stringent silencing and escape controls. In contrast, an in vitro system represented by the embryonic-kidney-derived Patski cell line showed a higher density of escape genes (21%), representing both kidney-specific escape genes and cell-line specific escape genes. Allele-specific RNA polymerase II occupancy and DNase I hypersensitivity at the promoter of genes on the Xi correlated well with levels of escape, consistent with an open chromatin structure at escape genes. Allele-specific CTCF binding on the Xi clustered at escape genes and was denser in brain compared to the Patski cell line, possibly contributing to a more compartmentalized structure of the Xi and fewer escape genes in brain compared to the cell line where larger domains of escape were observed.


Asunto(s)
Inactivación del Cromosoma X , Animales , Factor de Unión a CCCTC , Desoxirribonucleasa I/metabolismo , Femenino , Ratones , Especificidad de Órganos , Polimorfismo de Nucleótido Simple , ARN Polimerasa II/metabolismo , Proteínas Represoras/metabolismo , Análisis de Secuencia de ARN
17.
PLoS Genet ; 9(5): e1003489, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23658530

RESUMEN

The Rhox cluster on the mouse X chromosome contains reproduction-related homeobox genes expressed in a sexually dimorphic manner. We report that two members of the Rhox cluster, Rhox6 and 9, are regulated by de-methylation of histone H3 at lysine 27 by KDM6A, a histone demethylase with female-biased expression. Consistent with other homeobox genes, Rhox6 and 9 are in bivalent domains prior to embryonic stem cell differentiation and thus poised for activation. In female mouse ES cells, KDM6A is specifically recruited to Rhox6 and 9 for gene activation, a process inhibited by Kdm6a knockdown in a dose-dependent manner. In contrast, KDM6A occupancy at Rhox6 and 9 is low in male ES cells and knockdown has no effect on expression. In mouse ovary where Rhox6 and 9 remain highly expressed, KDM6A occupancy strongly correlates with expression. Our study implicates Kdm6a, a gene that escapes X inactivation, in the regulation of genes important in reproduction, suggesting that KDM6A may play a role in the etiology of developmental and reproduction-related effects of X chromosome anomalies.


Asunto(s)
Células Madre Embrionarias/metabolismo , Histona Demetilasas/genética , Proteínas de Homeodominio/genética , Reproducción/genética , Animales , Metilación de ADN , Células Madre Embrionarias/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas/metabolismo , Proteínas de Homeodominio/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Reproducción/fisiología , Caracteres Sexuales , Inactivación del Cromosoma X/genética
18.
Nat Genet ; 38(1): 47-53, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16341221

RESUMEN

Monosomy of the X chromosome owing to divergence between the sex chromosomes leads to dosage compensation mechanisms to restore balanced expression between the X and the autosomes. In Drosophila melanogaster, upregulation of the male X leads to dosage compensation. It has been hypothesized that mammals likewise upregulate their active X chromosome. Together with X inactivation, this mechanism would maintain balanced expression between the X chromosome and autosomes and between the sexes. Here, we show that doubling of the global expression level of the X chromosome leads to dosage compensation in somatic tissues from several mammalian species. X-linked genes are highly expressed in brain tissues, consistent with a role in cognitive functions. Furthermore, the X chromosome is expressed but not upregulated in spermatids and secondary oocytes, preserving balanced expression of the genome in these haploid cells. Upon fertilization, upregulation of the active X must occur to achieve the observed dosage compensation in early embryos.


Asunto(s)
Compensación de Dosificación (Genética) , Mamíferos/genética , Cromosoma X/genética , Animales , Encéfalo/fisiología , Drosophila/genética , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Oocitos/fisiología , Espermátides/fisiología , Regulación hacia Arriba , Inactivación del Cromosoma X
19.
Genome Res ; 21(3): 402-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21282478

RESUMEN

In Mus spretus, the chloride channel 4 gene Clcn4-2 is X-linked and dosage compensated by X up-regulation and X inactivation, while in the closely related mouse species Mus musculus, Clcn4-2 has been translocated to chromosome 7. We sequenced Clcn4-2 in M. spretus and identified the breakpoints of the evolutionary translocation in the Mus lineage. Genetic and epigenetic differences were observed between the 5'ends of the autosomal and X-linked loci. Remarkably, Clcn4-2 introns have been truncated on chromosome 7 in M. musculus as compared with the X-linked loci from seven other eutherian mammals. Intron sequences specifically preserved in the X-linked loci were significantly enriched in AT-rich oligomers. Genome-wide analyses showed an overall enrichment in AT motifs unique to the eutherian X (except for genes that escape X inactivation), suggesting a role for these motifs in regulation of the X chromosome.


Asunto(s)
Canales de Cloruro/genética , Región de Flanqueo 5'/genética , Secuencia Rica en At , Animales , Secuencia de Bases , Canales de Cloruro/metabolismo , Rotura Cromosómica , Mapeo Cromosómico , Compensación de Dosificación (Genética) , Epigenómica , Evolución Molecular , Femenino , Dosificación de Gen , Genes , Genoma , Humanos , Intrones , Masculino , Ratones , Datos de Secuencia Molecular , Muridae , Estructura Terciaria de Proteína/genética , Ratas , Análisis de Secuencia de ADN , Especificidad de la Especie , Cromosoma X/genética
20.
Genome Res ; 20(5): 614-22, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20363980

RESUMEN

X inactivation equalizes the dosage of gene expression between the sexes, but some genes escape silencing and are thus expressed from both alleles in females. To survey X inactivation and escape in mouse, we performed RNA sequencing in Mus musculus x Mus spretus cells with complete skewing of X inactivation, relying on expression of single nucleotide polymorphisms to discriminate allelic origin. Thirteen of 393 (3.3%) mouse genes had significant expression from the inactive X, including eight novel escape genes. We estimate that mice have significantly fewer escape genes compared with humans. Furthermore, escape genes did not cluster in mouse, unlike the large escape domains in human, suggesting that expression is controlled at the level of individual genes. Our findings are consistent with the striking differences in phenotypes between female mice and women with a single X chromosome--a near normal phenotype in mice versus Turner syndrome and multiple abnormalities in humans. We found that escape genes are marked by the absence of trimethylation at lysine 27 of histone H3, a chromatin modification associated with genes subject to X inactivation. Furthermore, this epigenetic mark is developmentally regulated for some mouse genes.


Asunto(s)
Expresión Génica , Análisis de Secuencia de ARN , Inactivación del Cromosoma X/genética , Cromosoma X/genética , Alelos , Animales , Línea Celular , Femenino , Histonas/genética , Histonas/metabolismo , Humanos , Células Híbridas , Riñón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple/genética , Síndrome de Turner/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA