Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Clin Epigenetics ; 15(1): 196, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-38124114

RESUMEN

BACKGROUND: Phaeochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumours. Pathogenic variants have been identified in more than 15 susceptibility genes; associated tumours are grouped into three Clusters, reinforced by their transcriptional profiles. Cluster 1A PPGLs have pathogenic variants affecting enzymes of the tricarboxylic acid cycle, including succinate dehydrogenase. Within inherited PPGLs, these are the most common. PPGL tumours are known to undergo epigenetic reprograming, and here, we report on global histone post-translational modifications and DNA methylation levels, alongside clinical phenotypes. RESULTS: Out of the 25 histone post-translational modifications examined, Cluster 1A PPGLs were distinguished from other tumours by a decrease in hyper-acetylated peptides and an increase in H3K4me2. DNA methylation was compared between tumours from individuals who developed metastatic disease versus those that did not. The majority of differentially methylated sites identified tended to be completely methylated or unmethylated in non-metastatic tumours, with low inter-sample variance. Metastatic tumours by contrast consistently had an intermediate DNA methylation state, including the ephrin receptor EPHA4 and its ligand EFNA3. Gene expression analyses performed to identify genes involved in metastatic tumour behaviour pin-pointed a number of genes previously described as mis-regulated in Cluster 1A tumours, as well as highlighting the tumour suppressor RGS22 and the pituitary tumour-transforming gene PTTG1. CONCLUSIONS: Combined transcriptomic and DNA methylation analyses revealed aberrant pathways, including ones that could be implicated in metastatic phenotypes and, for the first time, we report a decrease in hyper-acetylated histone marks in Cluster 1 PPGLs.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Paraganglioma , Feocromocitoma , Humanos , Feocromocitoma/genética , Feocromocitoma/metabolismo , Feocromocitoma/patología , Histonas/genética , Histonas/metabolismo , Metilación de ADN , Paraganglioma/genética , Paraganglioma/patología , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de las Glándulas Suprarrenales/patología , Perfilación de la Expresión Génica
2.
J Clin Endocrinol Metab ; 108(12): 3302-3310, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37285480

RESUMEN

CONTEXT: Somatic EPAS1 variants account for 5% to 8% of all pheochromocytoma and paragangliomas (PPGL) but are detected in over 90% of PPGL in patients with congenital cyanotic heart disease, where hypoxemia may select for EPAS1 gain-of-function variants. Sickle cell disease (SCD) is an inherited hemoglobinopathy associated with chronic hypoxia and there are isolated reports of PPGL in patients with SCD, but a genetic link between the conditions has yet to be established. OBJECTIVE: To determine the phenotype and EPAS1 variant status of patients with PPGL and SCD. METHODS: Records of 128 patients with PPGL under follow-up at our center from January 2017 to December 2022 were screened for SCD diagnosis. For identified patients, clinical data and biological specimens were obtained, including tumor, adjacent non-tumor tissue and peripheral blood. Sanger sequencing of exons 9 and 12 of EPAS1, followed by amplicon next-generation sequencing of identified variants was performed on all samples. RESULTS: Four patients with both PPGL and SCD were identified. Median age at PPGL diagnosis was 28 years. Three tumors were abdominal paragangliomas and 1 was a pheochromocytoma. No germline pathogenic variants in PPGL-susceptibility genes were identified in the cohort. Genetic testing of tumor tissue detected unique EPAS1 variants in all 4 patients. Variants were not detected in the germline, and 1 variant was detected in lymph node tissue of a patient with metastatic disease. CONCLUSION: We propose that somatic EPAS1 variants may be acquired through exposure to chronic hypoxia in SCD and drive PPGL development. Future work is needed to further characterize this association.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Anemia de Células Falciformes , Paraganglioma , Feocromocitoma , Adulto , Humanos , Neoplasias de las Glándulas Suprarrenales/patología , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Mutación de Línea Germinal , Hipoxia , Paraganglioma/patología , Feocromocitoma/patología
3.
Front Cell Dev Biol ; 10: 832348, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35223855

RESUMEN

The mammalian genome is depleted in CG dinucleotides, except at protected regions where they cluster as CpG islands (CGIs). CGIs are gene regulatory hubs and serve as transcription initiation sites and are as expected, associated with gene promoters. Advances in genomic annotations demonstrate that a quarter of CGIs are found within genes. Such intragenic regions are repressive environments, so it is surprising that CGIs reside here and even more surprising that some resist repression and are transcriptionally active within a gene. Hence, intragenic CGI positioning within genes is not arbitrary and is instead, selected for. As a wealth of recent studies demonstrate, intragenic CGIs are embedded within genes and consequently, influence 'host' gene mRNA isoform length and expand transcriptome diversity.

4.
PLoS One ; 16(11): e0254971, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34793449

RESUMEN

Tree ring features are affected by environmental factors and therefore are the basis for dendrochronological studies to reconstruct past environmental conditions. Oak wood often provides the data for these studies because of the durability of oak heartwood and hence the availability of samples spanning long time periods of the distant past. Wood formation is regulated in part by epigenetic mechanisms such as DNA methylation. Studies of the methylation state of DNA preserved in oak heartwood thus could identify epigenetic tree ring features informing on past environmental conditions. In this study, we aimed to establish protocols for the extraction of DNA, the high-throughput sequencing of whole-genome DNA libraries (WGS) and the profiling of DNA methylation by whole-genome bisulfite sequencing (WGBS) for oak (Quercus robur) heartwood drill cores taken from the trunks of living standing trees spanning the AD 1776-2014 time period. Heartwood contains little DNA, and large amounts of phenolic compounds known to hinder the preparation of high-throughput sequencing libraries. Whole-genome and DNA methylome library preparation and sequencing consistently failed for oak heartwood samples more than 100 and 50 years of age, respectively. DNA fragmentation increased with sample age and was exacerbated by the additional bisulfite treatment step during methylome library preparation. Relative coverage of the non-repetitive portion of the oak genome was sparse. These results suggest that quantitative methylome studies of oak hardwood will likely be limited to relatively recent samples and will require a high sequencing depth to achieve sufficient genome coverage.


Asunto(s)
Metilación de ADN , ADN de Plantas , Quercus/genética , Islas de CpG , Epigenoma , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Secuenciación Completa del Genoma
5.
Front Cell Dev Biol ; 9: 676543, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34239874

RESUMEN

Dopa decarboxylase (DDC) synthesizes serotonin in the developing mouse heart where it is encoded by Ddc_exon1a, a tissue-specific paternally expressed imprinted gene. Ddc_exon1a shares an imprinting control region (ICR) with the imprinted, maternally expressed (outside of the central nervous system) Grb10 gene on mouse chromosome 11, but little else is known about the tissue-specific imprinted expression of Ddc_exon1a. Fluorescent immunostaining localizes DDC to the developing myocardium in the pre-natal mouse heart, in a region susceptible to abnormal development and implicated in congenital heart defects in human. Ddc_exon1a and Grb10 are not co-expressed in heart nor in brain where Grb10 is also paternally expressed, despite sharing an ICR, indicating they are mechanistically linked by their shared ICR but not by Grb10 gene expression. Evidence from a Ddc_exon1a gene knockout mouse model suggests that it mediates the growth of the developing myocardium and a thinning of the myocardium is observed in a small number of mutant mice examined, with changes in gene expression detected by microarray analysis. Comparative studies in the human developing heart reveal a paternal expression bias with polymorphic imprinting patterns between individual human hearts at DDC_EXON1a, a finding consistent with other imprinted genes in human.

6.
Genome Res ; 31(8): 1381-1394, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34244229

RESUMEN

Hydroxycarbamide (HC, hydroxyurea) is a cytoreductive drug inducing cell cycle blockade. However, emerging evidence suggests that HC plays a role in the modulation of transcription through the activity of transcription factors and DNA methylation. Examining the global mechanism of action of HC in the context of myeloproliferative neoplasms (MPNs), for which HC is the first-line treatment, will provide a better understanding of its molecular effects. To explore the effects of HC genome-wide, transcriptomic analyses were performed on two clinically relevant cell types at different stages of differentiation treated with HC in a murine MPN model. This study was replicated in MPN patients by profiling genome-wide gene expression and DNA methylation using patient blood samples collected longitudinally, before and following HC exposure. The effects of HC on the transcriptome were not only associated with cell cycle interruption but also with hematopoietic functions. Moreover, a group of genes were restored to normal expression levels in murine hematopoietic stem cells (HSCs) following drug treatment, including the master regulator of hematopoiesis, RUNX1 In humans, HC significantly modifies DNA methylation levels in HSCs at several distal regulatory regions, which we show to be associated with SPI1 binding sites and at the SPI1 locus itself. We have identified novel targets of HC that include pivotal transcription factors involved in hematopoiesis, and for the first time we report abnormal methylation patterns in MPN patients at the master regulator gene SPI1 and its distal binding sites, which HC is able to restore to normal levels.


Asunto(s)
Metilación de ADN , Neoplasias , Animales , Hematopoyesis/genética , Humanos , Hidroxiurea/farmacología , Ratones , Neoplasias/genética , Transcriptoma
7.
Cancer Genet ; 256-257: 110-114, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34107390

RESUMEN

We report a novel case of multiple paragangliomas in a patient who was identified with pathogenic variants in both NF1 and SDHD genes. The proband is a man with known familial NF1 disease, diagnosed clinically in childhood. Multiple head and neck paragangliomas (HNPGL) were found during investigations for acute left sided neurological symptoms, in the region of his known plexiform neurofibroma. He was referred for genetic counselling. He underwent surgery to remove a left carotid body tumor (CBT). A pheochromocytoma and paraganglioma gene panel was tested. Blood and HNPGL tumor DNA were analyzed by whole exome sequencing. In addition to the NF1 truncating variant c.5107delA, p.(Ser1703AlafsTer7), the SDHD truncating pathogenic variant c.3G > A, p.(Met1?) was found. Tumor sequencing showed no LOH of SDHD or NF1, but monoallelic loss of 11p15 and 11q12.2-q12.3 was observed. Co-occurrence of pathogenic variants in multiple cancer susceptibility genes is rare but possible, identified by the increased use of panel testing. This is the first description of a patient presenting with NF1 and SDHD dual pathology, with HNPGL development due to SDHD. This case illustrates the central role of genetic sequencing in PPGLs and the strong genotype-phenotype correlations of different genes.


Asunto(s)
Mutación de Línea Germinal/genética , Neoplasias de Cabeza y Cuello/genética , Neurofibromina 1/genética , Paraganglioma/genética , Succinato Deshidrogenasa/genética , Adulto , Secuencia de Bases , Femenino , Neoplasias de Cabeza y Cuello/diagnóstico por imagen , Humanos , Masculino , Paraganglioma/diagnóstico por imagen , Paraganglioma/patología , Linaje
8.
Nat Commun ; 11(1): 5469, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33122634

RESUMEN

Zbtb11 is a conserved transcription factor mutated in families with hereditary intellectual disability. Its precise molecular and cellular functions are currently unknown, precluding our understanding of the aetiology of this disease. Using a combination of functional genomics, genetic and biochemical approaches, here we show that Zbtb11 plays essential roles in maintaining the homeostasis of mitochondrial function. Mechanistically, we find Zbtb11 facilitates the recruitment of nuclear respiratory factor 2 (NRF-2) to its target promoters, activating a subset of nuclear genes with roles in the biogenesis of respiratory complex I and the mitoribosome. Genetic inactivation of Zbtb11 resulted in a severe complex I assembly defect, impaired mitochondrial respiration, mitochondrial depolarisation, and ultimately proliferation arrest and cell death. Experimental modelling of the pathogenic human mutations showed these have a destabilising effect on the protein, resulting in reduced Zbtb11 dosage, downregulation of its target genes, and impaired complex I biogenesis. Our study establishes Zbtb11 as an essential mitochondrial regulator, improves our understanding of the transcriptional mechanisms of nuclear control over mitochondria, and may help to understand the aetiology of Zbtb11-associated intellectual disability.


Asunto(s)
Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Discapacidad Intelectual/genética , Mitocondrias/metabolismo , Dedos de Zinc/genética , Animales , Línea Celular , ADN Mitocondrial , Complejo I de Transporte de Electrón/biosíntesis , Complejo I de Transporte de Electrón/metabolismo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Discapacidad Intelectual/etiología , Ratones , Mutación/genética , Regiones Promotoras Genéticas , Proteínas Represoras/genética
10.
Nucleic Acids Res ; 48(15): 8349-8359, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32621610

RESUMEN

Alternative splicing (AS) and alternative polyadenylation (APA) generate diverse transcripts in mammalian genomes during development and differentiation. Epigenetic marks such as trimethylation of histone H3 lysine 36 (H3K36me3) and DNA methylation play a role in generating transcriptome diversity. Intragenic CpG islands (iCGIs) and their corresponding host genes exhibit dynamic epigenetic and gene expression patterns during development and between different tissues. We hypothesise that iCGI-associated H3K36me3, DNA methylation and transcription can influence host gene AS and/or APA. We investigate H3K36me3 and find that this histone mark is not a major regulator of AS or APA in our model system. Genomewide, we identify over 4000 host genes that harbour an iCGI in the mammalian genome, including both previously annotated and novel iCGI/host gene pairs. The transcriptional activity of these iCGIs is tissue- and developmental stage-specific and, for the first time, we demonstrate that the premature termination of host gene transcripts upstream of iCGIs is closely correlated with the level of iCGI transcription in a DNA-methylation independent manner. These studies suggest that iCGI transcription, rather than H3K36me3 or DNA methylation, interfere with host gene transcription and pre-mRNA processing genomewide and contributes to the spatiotemporal diversification of both the transcriptome and proteome.


Asunto(s)
Epigénesis Genética , Procesamiento Proteico-Postraduccional/genética , Precursores del ARN/genética , Transcripción Genética , Animales , Diferenciación Celular/genética , Cromatina/genética , Islas de CpG/genética , Metilación de ADN/genética , Genoma/genética , Código de Histonas/genética , Humanos , Regiones Promotoras Genéticas , Seudogenes/genética , Precursores del ARN/metabolismo
11.
PLoS Genet ; 16(4): e1008599, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32271759

RESUMEN

In 1993, Denise Barlow proposed that genomic imprinting might have arisen from a host defense mechanism designed to inactivate retrotransposons. Although there were few examples at hand, she suggested that there should be maternal-specific and paternal-specific factors involved, with cognate imprinting boxes that they recognized; furthermore, the system should build on conserved biochemical factors, including DNA methylation, and maternal control should predominate for imprints. Here, we revisit this hypothesis in the light of recent advances in our understanding of host defense and DNA methylation and in particular, the link with Krüppel-associated box-zinc finger (KRAB-ZF) proteins.


Asunto(s)
Silenciador del Gen , Impresión Genómica , Modelos Genéticos , Retroelementos/genética , Animales , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo
12.
Development ; 146(9)2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-31023876

RESUMEN

The endocardium interacts with the myocardium to promote proliferation and morphogenesis during the later stages of heart development. However, the role of the endocardium in early cardiac ontogeny remains under-explored. Given the shared origin, subsequent juxtaposition, and essential cell-cell interactions of endocardial and myocardial cells throughout heart development, we hypothesized that paracrine signaling from the endocardium to the myocardium is crucial for initiating early differentiation of myocardial cells. To test this, we generated an in vitro, endocardial-specific ablation model using the diphtheria toxin receptor under the regulatory elements of the Nfatc1 genomic locus (NFATc1-DTR). Early treatment of NFATc1-DTR mouse embryoid bodies with diphtheria toxin efficiently ablated endocardial cells, which significantly attenuated the percentage of beating EBs in culture and expression of early and late myocardial differentiation markers. The addition of Bmp2 during endocardial ablation partially rescued myocyte differentiation, maturation and function. Therefore, we conclude that early stages of myocardial differentiation rely on endocardial paracrine signaling mediated in part by Bmp2. Our findings provide novel insight into early endocardial-myocardial interactions that can be explored to promote early myocardial development and growth.


Asunto(s)
Diferenciación Celular/fisiología , Endocardio/citología , Endocardio/metabolismo , Miocardio/citología , Miocardio/metabolismo , Animales , Diferenciación Celular/genética , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Organogénesis/genética , Organogénesis/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
13.
Proc Biol Sci ; 283(1824)2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26842569

RESUMEN

Genomic imprinting is essential for development and growth and plays diverse roles in physiology and behaviour. Imprinted genes have traditionally been studied in isolation or in clusters with respect to cis-acting modes of gene regulation, both from a mechanistic and evolutionary point of view. Recent studies in mammals, however, reveal that imprinted genes are often co-regulated and are part of a gene network involved in the control of cellular proliferation and differentiation. Moreover, a subset of imprinted genes acts in trans on the expression of other imprinted genes. Numerous studies have modulated levels of imprinted gene expression to explore phenotypic and gene regulatory consequences. Increasingly, the applied genome-wide approaches highlight how perturbation of one imprinted gene may affect other maternally or paternally expressed genes. Here, we discuss these novel findings and consider evolutionary theories that offer a rationale for such intricate interactions among imprinted genes. An evolutionary view of these trans-regulatory effects provides a novel interpretation of the logic of gene networks within species and has implications for the origin of reproductive isolation between species.


Asunto(s)
Evolución Biológica , Redes Reguladoras de Genes , Impresión Genómica , Mamíferos/genética , Animales
14.
Hum Mutat ; 36(12): 1135-44, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26394720

RESUMEN

Genetic heterogeneity presents a significant challenge for the identification of monogenic disease genes. Whole-exome sequencing generates a large number of candidate disease-causing variants and typical analyses rely on deleterious variants being observed in the same gene across several unrelated affected individuals. This is less likely to occur for genetically heterogeneous diseases, making more advanced analysis methods necessary. To address this need, we present HetRank, a flexible gene-ranking method that incorporates interaction network data. We first show that different genes underlying the same monogenic disease are frequently connected in protein interaction networks. This motivates the central premise of HetRank: those genes carrying potentially pathogenic variants and whose network neighbors do so in other affected individuals are strong candidates for follow-up study. By simulating 1,000 exome sequencing studies (20,000 exomes in total), we model varying degrees of genetic heterogeneity and show that HetRank consistently prioritizes more disease-causing genes than existing analysis methods. We also demonstrate a proof-of-principle application of the method to prioritize genes causing Adams-Oliver syndrome, a genetically heterogeneous rare disease. An implementation of HetRank in R is available via the Website http://sourceforge.net/p/hetrank/.


Asunto(s)
Biología Computacional/métodos , Exoma , Estudios de Asociación Genética/métodos , Heterogeneidad Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Programas Informáticos , Simulación por Computador , Epistasis Genética , Redes Reguladoras de Genes , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Mapeo de Interacción de Proteínas/métodos , Navegador Web
15.
Biol Reprod ; 91(5): 125, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25297545

RESUMEN

CCCTC-binding factor (CTCF) is the major protein involved in insulator activity in vertebrates, with widespread DNA binding sites in the genome. CTCF participates in many processes related to global chromatin organization and remodeling, contributing to the repression or activation of gene transcription. It is also involved in epigenetic reprogramming and is essential during gametogenesis and embryo development. Abnormal DNA methylation patterns at CTCF motifs may impair CTCF binding to DNA, and are related to fertility disorders in mammals. Therefore, CTCF and its binding sites are important candidate regions to be investigated as molecular markers for gamete and embryo quality. This article reviews the role of CTCF in genomic imprinting, gametogenesis, and early embryo development and, moreover, highlights potential opportunities for environmental influences associated with assisted reproductive techniques (ARTs) to affect CTCF-mediated processes. We discuss the potential use of CTCF as a molecular marker for assessing gamete and embryo quality in the context of improving the efficiency and safety of ARTs.


Asunto(s)
Impresión Genómica/genética , Crecimiento y Desarrollo/genética , Proteínas Represoras/fisiología , Reproducción/genética , Animales , Factor de Unión a CCCTC , Metilación de ADN , Desarrollo Embrionario/genética , Fertilidad/genética , Gametogénesis/genética , Humanos , Proteínas Represoras/química , Técnicas Reproductivas Asistidas
16.
PLoS Biol ; 12(2): e1001799, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586114

RESUMEN

Developmental programming links growth in early life with health status in adulthood. Although environmental factors such as maternal diet can influence the growth and adult health status of offspring, the genetic influences on this process are poorly understood. Using the mouse as a model, we identify the imprinted gene Grb10 as a mediator of nutrient supply and demand in the postnatal period. The combined actions of Grb10 expressed in the mother, controlling supply, and Grb10 expressed in the offspring, controlling demand, jointly regulate offspring growth. Furthermore, Grb10 determines the proportions of lean and fat tissue during development, thereby influencing energy homeostasis in the adult. Most strikingly, we show that the development of normal lean/fat proportions depends on the combined effects of Grb10 expressed in the mother, which has the greater effect on offspring adiposity, and Grb10 expressed in the offspring, which influences lean mass. These distinct functions of Grb10 in mother and pup act complementarily, which is consistent with a coadaptation model of imprinting evolution, a model predicted but for which there is limited experimental evidence. In addition, our findings identify Grb10 as a key genetic component of developmental programming, and highlight the need for a better understanding of mother-offspring interactions at the genetic level in predicting adult disease risk.


Asunto(s)
Tamaño Corporal/genética , Proteína Adaptadora GRB10/genética , Animales , Femenino , Proteína Adaptadora GRB10/metabolismo , Regulación del Desarrollo de la Expresión Génica , Impresión Genómica , Carioferinas/fisiología , Lactancia/genética , Ratones , Ratones Noqueados , Receptores Citoplasmáticos y Nucleares/fisiología , Factor de Transcripción STAT5/fisiología , Proteína Exportina 1
17.
Genome Res ; 23(10): 1624-35, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23804403

RESUMEN

DNA binding factors are essential for regulating gene expression. CTCF and cohesin are DNA binding factors with central roles in chromatin organization and gene expression. We determined the sites of CTCF and cohesin binding to DNA in mouse brain, genome wide and in an allele-specific manner with high read-depth ChIP-seq. By comparing our results with existing data for mouse liver and embryonic stem (ES) cells, we investigated the tissue specificity of CTCF binding sites. ES cells have fewer unique CTCF binding sites occupied than liver and brain, consistent with a ground-state pattern of CTCF binding that is elaborated during differentiation. CTCF binding sites without the canonical consensus motif were highly tissue specific. In brain, a third of CTCF and cohesin binding sites coincide, consistent with the potential for many interactions between cohesin and CTCF but also many instances of independent action. In the context of genomic imprinting, CTCF and/or cohesin bind to a majority but not all differentially methylated regions, with preferential binding to the unmethylated parental allele. Whether the parental allele-specific methylation was established in the parental germlines or post-fertilization in the embryo is not a determinant in CTCF or cohesin binding. These findings link CTCF and cohesin with the control regions of a subset of imprinted genes, supporting the notion that imprinting control is mechanistically diverse.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Metilación de ADN , ADN/metabolismo , Impresión Genómica , Proteínas Represoras/metabolismo , Alelos , Animales , Sitios de Unión , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cromatina/genética , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Cromosomas de los Mamíferos , Biología Computacional , Regulación de la Expresión Génica , Sitios Genéticos , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Especificidad de Órganos , Unión Proteica , Proteínas Represoras/química , Proteínas Represoras/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Cohesinas
18.
EMBO J ; 32(13): 1941-52, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23727884

RESUMEN

Germ cells and adult stem cells maintain tissue homeostasis through a finely tuned program of responses to both physiological and stress-related signals. PLZF (Promyelocytic Leukemia Zinc Finger protein), a member of the POK family of transcription factors, acts as an epigenetic regulator of stem cell maintenance in germ cells and haematopoietic stem cells. We identified L1 retrotransposons as the primary targets of PLZF. PLZF-mediated DNA methylation induces silencing of the full-length L1 gene and inhibits L1 retrotransposition. Furthermore, PLZF causes the formation of barrier-type boundaries by acting on inserted truncated L1 sequences in protein coding genes. Cell stress releases PLZF-mediated repression, resulting in L1 activation/retrotransposition and impaired spermatogenesis and myelopoiesis. These results reveal a novel mechanism of action by which, PLZF represses retrotransposons, safeguarding normal progenitor homeostasis.


Asunto(s)
Epigenómica , Regulación de la Expresión Génica , Células Germinativas/metabolismo , Factores de Transcripción de Tipo Kruppel/fisiología , Elementos de Nucleótido Esparcido Largo/genética , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Regiones no Traducidas 5'/genética , Animales , Diferenciación Celular , Inmunoprecipitación de Cromatina , Metilación de ADN , Células Germinativas/citología , Ratones , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Células Madre/citología , Transcripción Genética
19.
PLoS Genet ; 9(1): e1003234, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23358118

RESUMEN

What good are transposable elements (TEs)? Although their activity can be harmful to host genomes and can cause disease, they nevertheless represent an important source of genetic variation that has helped shape genomes. In this review, we examine the impact of TEs, collectively referred to as the mobilome, on the transcriptome. We explore how TEs-particularly retrotransposons-contribute to transcript diversity and consider their potential significance as a source of small RNAs that regulate host gene transcription. We also discuss a critical role for the mobilome in engineering transcriptional networks, permitting coordinated gene expression, and facilitating the evolution of novel physiological processes.


Asunto(s)
Elementos Transponibles de ADN/genética , ARN Mensajero/genética , Transcripción Genética , Transcriptoma/genética , Evolución Molecular , Regulación de la Expresión Génica , Variación Genética , Humanos
20.
Mol Cell ; 47(6): 909-20, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22902559

RESUMEN

Identifying loci with parental differences in DNA methylation is key to unraveling parent-of-origin phenotypes. By conducting a MeDIP-Seq screen in maternal-methylation free postimplantation mouse embryos (Dnmt3L-/+), we demonstrate that maternal-specific methylation exists very scarcely at midgestation. We reveal two forms of oocyte-specific methylation inheritance: limited to preimplantation, or with longer duration, i.e. maternally imprinted loci. Transient and imprinted maternal germline DMRs (gDMRs) are indistinguishable in gametes and preimplantation embryos, however, de novo methylation of paternal alleles at implantation delineates their fates and acts as a major leveling factor of parent-inherited differences. We characterize two new imprinted gDMRs, at the Cdh15 and AK008011 loci, with tissue-specific imprinting loss, again by paternal methylation gain. Protection against demethylation after fertilization has been emphasized as instrumental in maintaining parent-of-origin methylation inherited from the gametes. Here we provide evidence that protection against de novo methylation acts as an equal major pivot, at implantation and throughout life.


Asunto(s)
Cadherinas/genética , Metilación de ADN , Embrión de Mamíferos/metabolismo , Impresión Genómica , Células Germinativas/metabolismo , Oocitos/metabolismo , Animales , Blastocisto/metabolismo , Embrión de Mamíferos/citología , Fertilización , Pruebas Genéticas , Ratones , Seudogenes , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA