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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 151(5): 951-63, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23178118

RESUMEN

The inactive X chromosome's (Xi) physical territory is microscopically devoid of transcriptional hallmarks and enriched in silencing-associated modifications. How these microscopic signatures relate to specific Xi sequences is unknown. Therefore, we profiled Xi gene expression and chromatin states at high resolution via allele-specific sequencing in mouse trophoblast stem cells. Most notably, X-inactivated transcription start sites harbored distinct epigenetic signatures relative to surrounding Xi DNA. These sites displayed H3-lysine27-trimethylation enrichment and DNaseI hypersensitivity, similar to autosomal Polycomb targets, yet excluded Pol II and other transcriptional hallmarks, similar to nontranscribed genes. CTCF bound X-inactivated and escaping genes, irrespective of measured chromatin boundaries. Escape from X inactivation occurred within, and X inactivation was maintained exterior to, the area encompassed by Xist in cells subject to imprinted and random X inactivation. The data support a model whereby inactivation of specific regulatory elements, rather than a simple chromosome-wide separation from transcription machinery, governs gene silencing over the Xi.


Asunto(s)
Silenciador del Gen , Elementos Reguladores de la Transcripción , Inactivación del Cromosoma X , Animales , Factor de Unión a CCCTC , Cromatina/metabolismo , Desoxirribonucleasa I/metabolismo , Código de Histonas , Elementos de Nucleótido Esparcido Largo , Ratones , Proteínas del Grupo Polycomb/metabolismo , ARN Polimerasa II/metabolismo , Proteínas Represoras/metabolismo , Células Madre/citología , Células Madre/metabolismo , Trofoblastos/citología
2.
Mol Cell ; 75(3): 523-537.e10, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31256989

RESUMEN

Long noncoding RNAs (lncRNAs) cause Polycomb repressive complexes (PRCs) to spread over broad regions of the mammalian genome. We report that in mouse trophoblast stem cells, the Airn and Kcnq1ot1 lncRNAs induce PRC-dependent chromatin modifications over multi-megabase domains. Throughout the Airn-targeted domain, the extent of PRC-dependent modification correlated with intra-nuclear distance to the Airn locus, preexisting genome architecture, and the abundance of Airn itself. Specific CpG islands (CGIs) displayed characteristics indicating that they nucleate the spread of PRCs upon exposure to Airn. Chromatin environments surrounding Xist, Airn, and Kcnq1ot1 suggest common mechanisms of PRC engagement and spreading. Our data indicate that lncRNA potency can be tightly linked to lncRNA abundance and that within lncRNA-targeted domains, PRCs are recruited to CGIs via lncRNA-independent mechanisms. We propose that CGIs that autonomously recruit PRCs interact with lncRNAs and their associated proteins through three-dimensional space to nucleate the spread of PRCs in lncRNA-targeted domains.


Asunto(s)
ARN Largo no Codificante/genética , Animales , Cromatina/genética , Ensamble y Desensamble de Cromatina , Islas de CpG/genética , Genoma/genética , Impresión Genómica/genética , Humanos , Ratones , Complejo Represivo Polycomb 1/genética , Regiones Promotoras Genéticas , Células Madre/metabolismo , Trofoblastos/metabolismo
3.
Immunity ; 43(4): 703-14, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26431949

RESUMEN

Epigenetic changes, including histone methylation, control T cell differentiation and memory formation, though the enzymes that mediate these processes are not clear. We show that UTX, a histone H3 lysine 27 (H3K27) demethylase, supports T follicular helper (Tfh) cell responses that are essential for B cell antibody generation and the resolution of chronic viral infections. Mice with a T cell-specific UTX deletion had fewer Tfh cells, reduced germinal center responses, lacked virus-specific immunoglobulin G (IgG), and were unable to resolve chronic lymphocytic choriomeningitis virus infections. UTX-deficient T cells showed decreased expression of interleukin-6 receptor-α and other Tfh cell-related genes that were associated with increased H3K27 methylation. Additionally, Turner Syndrome subjects, who are predisposed to chronic ear infections, had reduced UTX expression in immune cells and decreased circulating CD4(+) CXCR5(+) T cell frequency. Thus, we identify a critical link between UTX in T cells and immunity to infection.


Asunto(s)
Histona Demetilasas/deficiencia , Histona Demetilasas/fisiología , Virus de la Coriomeningitis Linfocítica/inmunología , Proteínas Nucleares/deficiencia , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Viremia/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Diferenciación Celular , Femenino , Dosificación de Gen , Regulación de la Expresión Génica/inmunología , Predisposición Genética a la Enfermedad , Histonas/metabolismo , Humanos , Memoria Inmunológica , Subunidad alfa del Receptor de Interleucina-6/biosíntesis , Subunidad alfa del Receptor de Interleucina-6/genética , Cooperación Linfocítica , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Metilación , Ratones , Modelos Inmunológicos , Otitis Media/etiología , Procesamiento Proteico-Postraduccional , Receptores CXCR5/análisis , Especificidad de la Especie , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/virología , Linfocitos T Colaboradores-Inductores/enzimología , Linfocitos T Colaboradores-Inductores/virología , Transcripción Genética , Síndrome de Turner/complicaciones , Síndrome de Turner/enzimología , Virulencia , Inactivación del Cromosoma X
4.
Genes Dev ; 29(13): 1377-92, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26159997

RESUMEN

Histone H3.3 is a highly conserved histone H3 replacement variant in metazoans and has been implicated in many important biological processes, including cell differentiation and reprogramming. Germline and somatic mutations in H3.3 genomic incorporation pathway components or in H3.3 encoding genes have been associated with human congenital diseases and cancers, respectively. However, the role of H3.3 in mammalian development remains unclear. To address this question, we generated H3.3-null mouse models through classical genetic approaches. We found that H3.3 plays an essential role in mouse development. Complete depletion of H3.3 leads to developmental retardation and early embryonic lethality. At the cellular level, H3.3 loss triggers cell cycle suppression and cell death. Surprisingly, H3.3 depletion does not dramatically disrupt gene regulation in the developing embryo. Instead, H3.3 depletion causes dysfunction of heterochromatin structures at telomeres, centromeres, and pericentromeric regions of chromosomes, leading to mitotic defects. The resulting karyotypical abnormalities and DNA damage lead to p53 pathway activation. In summary, our results reveal that an important function of H3.3 is to support chromosomal heterochromatic structures, thus maintaining genome integrity during mammalian development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Inestabilidad Genómica/genética , Crecimiento y Desarrollo/genética , Histonas/metabolismo , Animales , Muerte Celular/genética , Línea Celular , Proliferación Celular/genética , Células Cultivadas , Fertilidad/genética , Genes Letales/genética , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/genética , Ratones , Mutación
5.
Genes Dev ; 28(18): 2056-69, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25228648

RESUMEN

Polycomb-repressive complex 2 (PRC2) catalyzes the methylation of histone H3 Lys27 (H3K27) and functions as a critical epigenetic regulator of both stem cell pluripotency and somatic differentiation, but its role in male germ cell development is unknown. Using conditional mutagenesis to remove the core PRC2 subunits EED and SUZ12 during male germ cell development, we identified a requirement for PRC2 in both mitotic and meiotic germ cells. We observed a paucity of mutant spermatogonial stem cells (SSCs), which appears independent of repression of the known cell cycle inhibitors Ink4a/Ink4b/Arf. Moreover, mutant spermatocytes exhibited ectopic expression of somatic lamins and an abnormal distribution of SUN1 proteins on the nuclear envelope. These defects were coincident with abnormal chromosome dynamics, affecting homologous chromosome pairing and synapsis. We observed acquisition of H3K27me3 on stage-specific genes during meiotic progression, indicating a requirement for PRC2 in regulating the meiotic transcriptional program. Together, these data demonstrate that transcriptional repression of soma-specific genes by PRC2 facilitates homeostasis and differentiation during mammalian spermatogenesis.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Complejo Represivo Polycomb 2/metabolismo , Espermatocitos/citología , Transcriptoma/genética , Animales , Cromosomas/genética , Cromosomas/metabolismo , Silenciador del Gen , Infertilidad Masculina/genética , Laminas/genética , Masculino , Meiosis/genética , Ratones , Complejo Represivo Polycomb 2/genética
6.
Kidney Int ; 98(3): 744-757, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32446935

RESUMEN

ANCA vasculitis is an autoimmune disease with increased expression of the autoantigen genes, myeloperoxidase (MPO) and proteinase 3 (PRTN3), but the origin and significance of expression is less distinct. To clarify this, we measured MPO and PRTN3 messenger RNA in monocytes, normal-density neutrophils, and in enriched leukocytes from peripheral blood mononuclear cells. Increased autoantigen gene expression was detected in normal-density neutrophils and enriched leukocytes from patients during active disease compared to healthy individuals, with the largest difference in enriched leukocytes. RNA-seq of enriched leukocytes comparing active-remission pairs identified a gene signature for low-density neutrophils. Cell sorting revealed low-density neutrophils contained mature and immature neutrophils depending on the presence or absence of CD10. Both populations contributed to autoantigen expression but the frequency of immature cells in low-density neutrophils did not correlate with low-density neutrophil MPO or PRTN3 expression. Low-density neutrophils were refractory to MPO-ANCA induced oxidative burst, suggesting an alternative role for low-density neutrophils in ANCA vasculitis pathogenesis. In contrast, normal-density neutrophils were activated by MPO-ANCA and monoclonal anti-PR3 antibody. Normal-density neutrophil activation correlated with MPO and PRTN3 mRNA. Increased autoantigen gene expression originating from the mature low-density and normal-density neutrophils suggests transcriptional dysregulation is a hallmark of ANCA vasculitis. Thus, the correlation between autoantigen gene expression and antibody-mediated normal-density neutrophil activation connects autoantigen gene expression with disease pathogenesis.


Asunto(s)
Anticuerpos Anticitoplasma de Neutrófilos , Neutrófilos , Autoantígenos/genética , Expresión Génica , Humanos , Leucocitos Mononucleares , Mieloblastina , Activación Neutrófila , Peroxidasa/genética
7.
Proc Natl Acad Sci U S A ; 114(43): E9046-E9055, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073101

RESUMEN

Kabuki syndrome, a congenital craniofacial disorder, manifests from mutations in an X-linked histone H3 lysine 27 demethylase (UTX/KDM6A) or a H3 lysine 4 methylase (KMT2D). However, the cellular and molecular etiology of histone-modifying enzymes in craniofacial disorders is unknown. We now establish Kabuki syndrome as a neurocristopathy, whereby the majority of clinical features are modeled in mice carrying neural crest (NC) deletion of UTX, including craniofacial dysmorphism, cardiac defects, and postnatal growth retardation. Female UTX NC knockout (FKO) demonstrates enhanced phenotypic severity over males (MKOs), due to partial redundancy with UTY, a Y-chromosome demethylase-dead homolog. Thus, NC cells may require demethylase-independent UTX activity. Consistently, Kabuki causative point mutations upstream of the JmjC domain do not disrupt UTX demethylation. We have isolated primary NC cells at a phenocritical postmigratory timepoint in both FKO and MKO mice, and genome-wide expression and histone profiling have revealed UTX molecular function in establishing appropriate chromatin structure to regulate crucial NC stem-cell signaling pathways. However, the majority of UTX regulated genes do not experience aberrations in H3K27me3 or H3K4me3, implicating alternative roles for UTX in transcriptional control. These findings are substantiated through demethylase-dead knockin mutation of UTX, which supports appropriate facial development.


Asunto(s)
Anomalías Múltiples/etiología , Cara/anomalías , Enfermedades Hematológicas/etiología , Histona Demetilasas/metabolismo , Cresta Neural/fisiopatología , Enfermedades Vestibulares/etiología , Animales , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Histona Demetilasas/genética , Humanos , Lisina/metabolismo , Masculino , Ratones Noqueados , Ratones Transgénicos , Mutación , Cresta Neural/metabolismo , Proteínas Nucleares/genética , Cráneo/embriología
8.
Gastroenterology ; 155(5): 1508-1523.e10, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30055169

RESUMEN

BACKGROUND & AIMS: The intestinal epithelium is maintained by intestinal stem cells (ISCs), which produce postmitotic absorptive and secretory epithelial cells. Initial fate specification toward enteroendocrine, goblet, and Paneth cell lineages requires the transcription factor Atoh1, which regulates differentiation of the secretory cell lineage. However, less is known about the origin of tuft cells, which participate in type II immune responses to parasite infections and appear to differentiate independently of Atoh1. We investigated the role of Sox4 in ISC differentiation. METHODS: We performed experiments in mice with intestinal epithelial-specific disruption of Sox4 (Sox4fl/fl:vilCre; SOX4 conditional knockout [cKO]) and mice without disruption of Sox4 (control mice). Crypt- and single-cell-derived organoids were used in assays to measure proliferation and ISC potency. Lineage allocation and gene expression changes were studied by immunofluorescence, real-time quantitative polymerase chain reaction, and RNA-seq analyses. Intestinal organoids were incubated with the type 2 cytokine interleukin 13 and gene expression was analyzed. Mice were infected with the helminth Nippostrongylus brasiliensis and intestinal tissues were collected 7 days later for analysis. Intestinal tissues collected from mice that express green fluorescent protein regulated by the Atoh1 promoter (Atoh1GFP mice) and single-cell RNA-seq analysis were used to identify cells that coexpress Sox4 and Atoh1. We generated SOX4-inducible intestinal organoids derived from Atoh1fl/fl:vilCreER (ATOH1 inducible knockout) mice and assessed differentiation. RESULTS: Sox4cKO mice had impaired ISC function and secretory differentiation, resulting in decreased numbers of tuft and enteroendocrine cells. In control mice, numbers of SOX4+ cells increased significantly after helminth infection, coincident with tuft cell hyperplasia. Sox4 was activated by interleukin 13 in control organoids; SOX4cKO mice had impaired tuft cell hyperplasia and parasite clearance after infection with helminths. In single-cell RNA-seq analysis, Sox4+/Atoh1- cells were enriched for ISC, progenitor, and tuft cell genes; 12.5% of Sox4-expressing cells coexpressed Atoh1 and were enriched for enteroendocrine genes. In organoids, overexpression of Sox4 was sufficient to induce differentiation of tuft and enteroendocrine cells-even in the absence of Atoh1. CONCLUSIONS: We found Sox4 promoted tuft and enteroendocrine cell lineage allocation independently of Atoh1. These results challenge the longstanding model in which Atoh1 is the sole regulator of secretory differentiation in the intestine and are relevant for understanding epithelial responses to parasitic infection.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células Enteroendocrinas/citología , Células Caliciformes/citología , Mucosa Intestinal/citología , Factores de Transcripción SOXC/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Receptores de Hialuranos/análisis , Ratones , Factores de Transcripción SOXC/análisis
9.
Nature ; 501(7465): 58-62, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23995680

RESUMEN

Topoisomerases are expressed throughout the developing and adult brain and are mutated in some individuals with autism spectrum disorder (ASD). However, how topoisomerases are mechanistically connected to ASD is unknown. Here we find that topotecan, a topoisomerase 1 (TOP1) inhibitor, dose-dependently reduces the expression of extremely long genes in mouse and human neurons, including nearly all genes that are longer than 200 kilobases. Expression of long genes is also reduced after knockdown of Top1 or Top2b in neurons, highlighting that both enzymes are required for full expression of long genes. By mapping RNA polymerase II density genome-wide in neurons, we found that this length-dependent effect on gene expression was due to impaired transcription elongation. Interestingly, many high-confidence ASD candidate genes are exceptionally long and were reduced in expression after TOP1 inhibition. Our findings suggest that chemicals and genetic mutations that impair topoisomerases could commonly contribute to ASD and other neurodevelopmental disorders.


Asunto(s)
Trastorno Autístico/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Elongación de la Transcripción Genética , Animales , ADN-Topoisomerasas de Tipo I/deficiencia , ADN-Topoisomerasas de Tipo II/deficiencia , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Impresión Genómica/genética , Humanos , Ratones , Mutación/genética , Proteínas de Unión a Poli-ADP-Ribosa , ARN Polimerasa II/metabolismo , Sinapsis/metabolismo , Inhibidores de Topoisomerasa/farmacología , Topotecan/farmacología , Elongación de la Transcripción Genética/efectos de los fármacos
10.
Dev Biol ; 424(2): 198-207, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28254491

RESUMEN

We previously reported the requirement of Polycomb Repressive Complex 2 (PRC2) for spermatogenesis through transcriptional repression of somatic genes and meiosis-specific genes. To characterize how PRC2's two methyltransferase subunits, EZH1 and EZH2, regulate histone H3 lysine 27 (H3K27) methylation during germ cell development, we generated mouse models with a germline ablation of EZH1 and/or EHZ2. Only the combined loss of EZH1 and EZH2 caused a depletion of global H3K27me3 marks and meiotic arrest in spermatocytes. Genome-wide analysis of H3K27me3 in spermatogenic cells revealed that a noncanonical EZH1-PRC2 could establish and maintain this histone mark on somatic genes and certain meiotic genes. Consistent with it having active enhancers in testis, Ezh1 was not only abundant in highly differentiated spermatocytes but also in actively proliferating progenitor and stem germ cells. Taken together, our findings suggest that the expression level of Ezh1 determines the restoration of H3K27 methylation in the absence of the canonical EZH2-PRC2.


Asunto(s)
Complejo Represivo Polycomb 2/metabolismo , Espermatogénesis , Espermatozoides/metabolismo , Animales , Secuencia de Bases , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Fertilidad , Eliminación de Gen , Genoma , Histonas/metabolismo , Lisina/metabolismo , Masculino , Metilación , Ratones Noqueados , Mitosis , Modelos Biológicos , Testículo/metabolismo
11.
J Am Soc Nephrol ; 28(4): 1175-1187, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27821628

RESUMEN

ANCA-associated vasculitis is an autoimmune condition characterized by vascular inflammation and organ damage. Pharmacologically induced remission of this condition is complicated by relapses. Potential triggers of relapse are immunologic challenges and environmental insults, both of which associate with changes in epigenetic silencing modifications. Altered histone modifications implicated in gene silencing associate with aberrant autoantigen expression. To establish a link between DNA methylation, a model epigenetic gene silencing modification, and autoantigen gene expression and disease status in ANCA-associated vasculitis, we measured gene-specific DNA methylation of the autoantigen genes myeloperoxidase (MPO) and proteinase 3 (PRTN3) in leukocytes of patients with ANCA-associated vasculitis observed longitudinally (n=82) and of healthy controls (n=32). Patients with active disease demonstrated hypomethylation of MPO and PRTN3 and increased expression of the autoantigens; in remission, DNA methylation generally increased. Longitudinal analysis revealed that patients with ANCA-associated vasculitis could be divided into two groups, on the basis of whether DNA methylation increased or decreased from active disease to remission. In patients with increased DNA methylation, MPO and PRTN3 expression correlated with DNA methylation. Kaplan-Meier estimate of relapse revealed patients with increased DNA methylation at the PRTN3 promoter had a significantly greater probability of a relapse-free period (P<0.001), independent of ANCA serotype. Patients with decreased DNA methylation at the PRTN3 promoter had a greater risk of relapse (hazard ratio, 4.55; 95% confidence interval, 2.09 to 9.91). Thus, changes in the DNA methylation status of the PRTN3 promoter may predict the likelihood of stable remission and explain autoantigen gene regulation.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/genética , Autoantígenos/genética , Metilación de ADN , Mieloblastina/genética , Peroxidasa/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Inducción de Remisión
12.
PLoS Genet ; 10(8): e1004507, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25101834

RESUMEN

The early mammalian embryo utilizes histone H3 lysine 27 trimethylation (H3K27me3) to maintain essential developmental genes in a repressive chromatin state. As differentiation progresses, H3K27me3 is removed in a distinct fashion to activate lineage specific patterns of developmental gene expression. These rapid changes in early embryonic chromatin environment are thought to be dependent on H3K27 demethylases. We have taken a mouse genetics approach to remove activity of both H3K27 demethylases of the Kdm6 gene family, Utx (Kdm6a, X-linked gene) and Jmjd3 (Kdm6b, autosomal gene). Male embryos null for active H3K27 demethylation by the Kdm6 gene family survive to term. At mid-gestation, embryos demonstrate proper patterning and activation of Hox genes. These male embryos retain the Y-chromosome UTX homolog, UTY, which cannot demethylate H3K27me3 due to mutations in catalytic site of the Jumonji-C domain. Embryonic stem (ES) cells lacking all enzymatic KDM6 demethylation exhibit a typical decrease in global H3K27me3 levels with differentiation. Retinoic acid differentiations of these ES cells demonstrate loss of H3K27me3 and gain of H3K4me3 to Hox promoters and other transcription factors, and induce expression similar to control cells. A small subset of genes exhibit decreased expression associated with reduction of promoter H3K4me3 and some low-level accumulation of H3K27me3. Finally, Utx and Jmjd3 mutant mouse embryonic fibroblasts (MEFs) demonstrate dramatic loss of H3K27me3 from promoters of several Hox genes and transcription factors. Our results indicate that early embryonic H3K27me3 repression can be alleviated in the absence of active demethylation by the Kdm6 gene family.


Asunto(s)
Diferenciación Celular/genética , Desarrollo Embrionario/genética , Histona Demetilasas/genética , Histona Demetilasas con Dominio de Jumonji/genética , Animales , Cromatina/genética , Embrión de Mamíferos , Células Madre Embrionarias , Femenino , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas/biosíntesis , Histona Demetilasas con Dominio de Jumonji/biosíntesis , Masculino , Ratones , Mutación , Embarazo
13.
BMC Bioinformatics ; 17: 144, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27009150

RESUMEN

BACKGROUND: Correctly identifying genomic regions enriched with histone modifications and transcription factors is key to understanding their regulatory and developmental roles. Conceptually, these regions are divided into two categories, narrow peaks and broad domains, and different algorithms are used to identify each one. Datasets that span these two categories are often analyzed with a single program for peak calling combined with an ad hoc method for domains. RESULTS: We developed hiddenDomains, which identifies both peaks and domains, and compare it to the leading algorithms using H3K27me3, H3K36me3, GABP, ESR1 and FOXA ChIP-seq datasets. The output from the programs was compared to qPCR-validated enriched and depleted sites, predicted transcription factor binding sites, and highly-transcribed gene bodies. With every method, hiddenDomains, performed as well as, if not better than algorithms dedicated to a specific type of analysis. CONCLUSIONS: hiddenDomains performs as well as the best domain and peak calling algorithms, making it ideal for analyzing ChIP-seq datasets, especially those that contain a mixture of peaks and domains.


Asunto(s)
Algoritmos , Inmunoprecipitación de Cromatina , Receptor alfa de Estrógeno/metabolismo , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Histonas/metabolismo , Humanos , Cadenas de Markov
14.
Nucleic Acids Res ; 42(8): e68, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24561615

RESUMEN

The ability to correlate chromosome conformation and gene expression gives a great deal of information regarding the strategies used by a cell to properly regulate gene activity. 4C-Seq is a relatively new and increasingly popular technology where the set of genomic interactions generated by a single point in the genome can be determined. 4C-Seq experiments generate large, complicated data sets and it is imperative that signal is properly distinguished from noise. Currently, there are a limited number of methods for analyzing 4C-Seq data. Here, we present a new method, fourSig, which in addition to being precise and simple to use also includes a new feature that prioritizes detected interactions. Our results demonstrate the efficacy of fourSig with previously published and novel 4C-Seq data sets and show that our significance prioritization correlates with the ability to reproducibly detect interactions among replicates.


Asunto(s)
Cromosomas/química , Programas Informáticos , Alelos , Animales , Interpretación Estadística de Datos , Expresión Génica , Sitios Genéticos , Genómica/métodos , Hibridación Fluorescente in Situ , Ratones , Conformación de Ácido Nucleico , Globinas beta/genética
15.
Nature ; 460(7255): 647-51, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19571810

RESUMEN

XX female mammals undergo transcriptional silencing of most genes on one of their two X chromosomes to equalize X-linked gene dosage with XY males in a process referred to as X-chromosome inactivation (XCI). XCI is an example of epigenetic regulation. Once enacted in individual cells of the early female embryo, XCI is stably transmitted such that most descendant cells maintain silencing of that X chromosome. In eutherian mammals, XCI is thought to be triggered by the expression of the non-coding Xist RNA from the future inactive X chromosome (Xi); Xist RNA in turn is proposed to recruit protein complexes that bring about heterochromatinization of the Xi. Here we test whether imprinted XCI, which results in preferential inactivation of the paternal X chromosome (Xp), occurs in mouse embryos inheriting an Xp lacking Xist. We find that silencing of Xp-linked genes can initiate in the absence of paternal Xist; Xist is, however, required to stabilize silencing along the Xp. Xp-linked gene silencing associated with mouse imprinted XCI, therefore, can initiate in the embryo independently of Xist RNA.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genes Ligados a X/genética , Impresión Genómica/genética , ARN/metabolismo , Inactivación del Cromosoma X/genética , Animales , Femenino , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
16.
PLoS Genet ; 8(1): e1002468, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22275877

RESUMEN

Changes in the spatial positioning of genes within the mammalian nucleus have been associated with transcriptional differences and thus have been hypothesized as a mode of regulation. In particular, the localization of genes to the nuclear and nucleolar peripheries is associated with transcriptional repression. However, the mechanistic basis, including the pertinent cis- elements, for such associations remains largely unknown. Here, we provide evidence that demonstrates a 119 bp 5S rDNA can influence nucleolar association in mammals. We found that integration of transgenes with 5S rDNA significantly increases the association of the host region with the nucleolus, and their degree of association correlates strongly with repression of a linked reporter gene. We further show that this mechanism may be functional in endogenous contexts: pseudogenes derived from 5S rDNA show biased conservation of their internal transcription factor binding sites and, in some cases, are frequently associated with the nucleolus. These results demonstrate that 5S rDNA sequence can significantly contribute to the positioning of a locus and suggest a novel, endogenous mechanism for nuclear organization in mammals.


Asunto(s)
Nucléolo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , ADN Ribosómico/genética , Heterocromatina/genética , ARN Ribosómico 5S/genética , ARN Ribosómico 5S/metabolismo , Transcripción Genética , Animales , Sitios de Unión , Línea Celular , Nucléolo Celular/metabolismo , Dactinomicina/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo , Ratones , Nucleosomas/genética , Nucleosomas/metabolismo , Seudogenes/genética , ARN Polimerasa I/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transgenes/genética
17.
Development ; 138(10): 2049-57, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21471155

RESUMEN

Repression of Xist RNA expression is considered a prerequisite to reversal of X-chromosome inactivation (XCI) in the mouse inner cell mass (ICM), and reactivation of X-linked genes is thought to follow loss of Xist RNA coating and heterochromatic markers of inactivation, such as methylation of histone H3. We analyzed X-chromosome activity in developing ICMs and show that reactivation of gene expression from the inactive-X initiates in the presence of Xist coating and H3K27me3. Furthermore, depletion of Xist RNA coating through forced upregulation of NANOG does not result in altered reactivation kinetics. Taken together, our observations suggest that in the ICM, X-linked gene transcription and Xist coating are uncoupled. These data fundamentally alter our perception of the reactivation process and support the existence of a mechanism to reactivate Xp-linked genes in the ICM that operates independently of loss of Xist RNA and H3K27me3 from the imprinted inactive-X.


Asunto(s)
Masa Celular Interna del Blastocisto/metabolismo , Histonas/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo , Animales , Masa Celular Interna del Blastocisto/citología , Epigénesis Genética , Femenino , Impresión Genómica , Hibridación Fluorescente in Situ , Masculino , Metilación , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo , ARN Largo no Codificante , Transcripción Genética , Inactivación del Cromosoma X
18.
Biol Reprod ; 89(1): 17, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23740947

RESUMEN

Recent advances in the cryopreservation of mouse sperm have resulted in dramatically improved in vitro fertilization (IVF) rates, but the biological mechanisms underlying the techniques remain unclear. Two different classes of compounds have been widely utilized to improve the IVF rates of cryopreserved mouse sperm: antioxidants and cyclodextrins. To determine how cryopreservation reduces mouse sperm IVF and how antioxidants and cyclodextrins mitigate this effect, we examined sperm function and oxidative damage after cryopreservation, with and without treatments, in mouse strains important for biomedical research. Our investigation revealed mouse strain-specific effects on IVF by modulation of oxidative stress and cholesterol efflux of cryopreserved sperm. Antioxidants improved IVF rates of C57Bl6/J cryopreserved mouse sperm by reducing hydrogen peroxide produced by sperm mitochondria and ameliorating peroxidative damage to the sperm acrosome. Enhancing cholesterol efflux with cyclodextrin restored capacitation-dependent sperm function and IVF after cryopreservation of C57Bl/6J, C57Bl/6N, and 129X1 mouse sperm. Our results highlight two accessible pathways for continued development of IVF techniques for mouse sperm and provide novel endpoints prognostic of IVF success. These insights may improve sperm cryopreservation methods of other mouse strains and species.


Asunto(s)
Colesterol/metabolismo , Criopreservación , Fertilización In Vitro , Estrés Oxidativo , Espermatozoides/fisiología , Animales , Peróxido de Hidrógeno/farmacología , Endogamia , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Estrés Oxidativo/fisiología
19.
Genet Epidemiol ; 34(1): 100-5, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19434714

RESUMEN

A major challenge in genome-wide association studies (GWASs) is to derive the multiple testing threshold when hypothesis tests are conducted using a large number of single nucleotide polymorphisms. Permutation tests are considered the gold standard in multiple testing adjustment in genetic association studies. However, it is computationally intensive, especially for GWASs, and can be impractical if a large number of random shuffles are used to ensure accuracy. Many researchers have developed approximation algorithms to relieve the computing burden imposed by permutation. One particularly attractive alternative to permutation is to calculate the effective number of independent tests, M(eff), which has been shown to be promising in genetic association studies. In this study, we compare recently developed M(eff) methods and validate them by the permutation test with 10,000 random shuffles using two real GWAS data sets: an Illumina 1M BeadChip and an Affymetrix GeneChip Human Mapping 500K Array Set. Our results show that the simpleM method produces the best approximation of the permutation threshold, and it does so in the shortest amount of time. We also show that M(eff) is indeed valid on a genome-wide scale in these data sets based on statistical theory and significance tests. The significance thresholds derived can provide practical guidelines for other studies using similar population samples and genotyping platforms.


Asunto(s)
Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Biometría/métodos , Interpretación Estadística de Datos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/estadística & datos numéricos , Polimorfismo de Nucleótido Simple
20.
Cell Rep ; 27(2): 514-524.e5, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30970254

RESUMEN

Obesity in humans is associated with poorer health outcomes after infections compared with non-obese individuals. Here, we examined the effects of white adipose tissue and obesity on T cell responses to viral infection in mice. We show that lymphocytic choriomeningitis virus (LCMV) grows to high titer in adipose tissue. Virus-specific T cells enter the adipose tissue to resolve infection but then remain as a memory population distinct from memory T cells in lymphoid tissues. Memory T cells in adipose tissue are abundant in lean mice, and diet-induced obesity further increases memory T cell number in adipose tissue and spleen. Upon re-challenge infection, memory T cells rapidly cause severe pathogenesis, leading to increases in lipase levels, calcification of adipose tissue, pancreatitis, and reduced survival in obese mice but not lean mice. Thus, obesity leads to a unique form of viral pathogenesis involving memory T cell-dependent adipocyte destruction and damage to other tissues.


Asunto(s)
Tejido Adiposo/fisiología , Obesidad/genética , Linfocitos T/metabolismo , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Obesidad/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA