Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nature ; 475(7356): 381-5, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21776083

RESUMEN

The gene for the atypical NOTCH ligand delta-like homologue 1 (Dlk1) encodes membrane-bound and secreted isoforms that function in several developmental processes in vitro and in vivo. Dlk1, a member of a cluster of imprinted genes, is expressed from the paternally inherited chromosome. Here we show that mice that are deficient in Dlk1 have defects in postnatal neurogenesis in the subventricular zone: a developmental continuum that results in depletion of mature neurons in the olfactory bulb. We show that DLK1 is secreted by niche astrocytes, whereas its membrane-bound isoform is present in neural stem cells (NSCs) and is required for the inductive effect of secreted DLK1 on self-renewal. Notably, we find that there is a requirement for Dlk1 to be expressed from both maternally and paternally inherited chromosomes. Selective absence of Dlk1 imprinting in both NSCs and niche astrocytes is associated with postnatal acquisition of DNA methylation at the germ-line-derived imprinting control region. The results emphasize molecular relationships between NSCs and the niche astrocyte cells of the microenvironment, identifying a signalling system encoded by a single gene that functions coordinately in both cell types. The modulation of genomic imprinting in a stem-cell environment adds a new level of epigenetic regulation to the establishment and maintenance of the niche, raising wider questions about the adaptability, function and evolution of imprinting in specific developmental contexts.


Asunto(s)
Animales Recién Nacidos/metabolismo , Astrocitos/metabolismo , Impresión Genómica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Nicho de Células Madre/citología , Envejecimiento/genética , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Membrana Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Genotipo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Bulbo Olfatorio/citología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Nicho de Células Madre/metabolismo
2.
PLoS Genet ; 8(4): e1002605, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22511876

RESUMEN

Environmental factors during early life are critical for the later metabolic health of the individual and of future progeny. In our obesogenic environment, it is of great socioeconomic importance to investigate the mechanisms that contribute to the risk of metabolic ill health. Imprinted genes, a class of functionally mono-allelic genes critical for early growth and metabolic axis development, have been proposed to be uniquely susceptible to environmental change. Furthermore, it has also been suggested that perturbation of the epigenetic reprogramming of imprinting control regions (ICRs) may play a role in phenotypic heritability following early life insults. Alternatively, the presence of multiple layers of epigenetic regulation may in fact protect imprinted genes from such perturbation. Unbiased investigation of these alternative hypotheses requires assessment of imprinted gene expression in the context of the response of the whole transcriptome to environmental assault. We therefore analyse the role of imprinted genes in multiple tissues in two affected generations of an established murine model of the developmental origins of health and disease using microarrays and quantitative RT-PCR. We demonstrate that, despite the functional mono-allelicism of imprinted genes and their unique mechanisms of epigenetic dosage control, imprinted genes as a class are neither more susceptible nor protected from expression perturbation induced by maternal undernutrition in either the F1 or the F2 generation compared to other genes. Nor do we find any evidence that the epigenetic reprogramming of ICRs in the germline is susceptible to nutritional restriction. However, we propose that those imprinted genes that are affected may play important roles in the foetal response to undernutrition and potentially its long-term sequelae. We suggest that recently described instances of dosage regulation by relaxation of imprinting are rare and likely to be highly regulated.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Interacción Gen-Ambiente , Impresión Genómica , Desnutrición , Animales , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Desnutrición/genética , Desnutrición/metabolismo , Ratones , Placenta/metabolismo , Placentación , Embarazo
3.
Biochem Soc Trans ; 41(3): 711-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23697930

RESUMEN

Cultured pluripotent stem cells hold great promise for regenerative medicine. Considerable efforts have been invested into the refinement and definition of improved culture systems that sustain self-renewal and avoid differentiation of pluripotent cells in vitro. Recent studies have, however, found that the choice of culture condition has a significant impact on epigenetic profiles of cultured pluripotent cells. Mouse and human ESCs (embryonic stem cells) show substantial epigenetic differences that are dependent on the culture condition, including global changes to DNA methylation and histone modifications and, in female human ESCs, to the epigenetic process of X chromosome inactivation. Epigenetic perturbations have also been detected during culture of pre-implantation embryos; limited research undertaken in mouse suggests a direct effect of the in vitro environment on epigenetic processes in this system. Widespread epigenetic changes induced by the culture condition in stem cells thus emphasize the necessity for extensive research into both immediate and long-term epigenetic effects of embryo culture during assisted reproductive technologies.


Asunto(s)
Blastocisto/citología , Técnicas de Cultivo de Célula/métodos , Epigénesis Genética/fisiología , Células Madre Pluripotentes/citología , Animales , Blastocisto/metabolismo , Blastocisto/fisiología , Células Cultivadas , Técnicas de Cultivo de Embriones , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Ratones , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología , Técnicas Reproductivas Asistidas
4.
Nat Commun ; 10(1): 3840, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477698

RESUMEN

Resistant tumours are thought to arise from the action of Darwinian selection on genetically heterogenous cancer cell populations. However, simple clonal selection is inadequate to describe the late relapses often characterising luminal breast cancers treated with endocrine therapy (ET), suggesting a more complex interplay between genetic and non-genetic factors. Here, we dissect the contributions of clonal genetic diversity and transcriptional plasticity during the early and late phases of ET at single-cell resolution. Using single-cell RNA-sequencing and imaging we disentangle the transcriptional variability of plastic cells and define a rare subpopulation of pre-adapted (PA) cells which undergoes further transcriptomic reprogramming and copy number changes to acquire full resistance. We find evidence for sub-clonal expression of a PA signature in primary tumours and for dominant expression in clustered circulating tumour cells. We propose a multi-step model for ET resistance development and advocate the use of stage-specific biomarkers.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Antineoplásicos Hormonales/uso terapéutico , Mama/citología , Mama/patología , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Microscopía Intravital , Células MCF-7 , Aprendizaje Automático , Mutación , Células Neoplásicas Circulantes/efectos de los fármacos , RNA-Seq , Análisis de la Célula Individual , Esferoides Celulares
5.
Genome Biol ; 16: 112, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-26025256

RESUMEN

BACKGROUND: Selective maintenance of genomic epigenetic imprints during pre-implantation development is required for parental origin-specific expression of imprinted genes. The Kruppel-like zinc finger protein ZFP57 acts as a factor necessary for maintaining the DNA methylation memory at multiple imprinting control regions in early mouse embryos and embryonic stem (ES) cells. Maternal-zygotic deletion of ZFP57 in mice presents a highly penetrant phenotype with no animals surviving to birth. Additionally, several cases of human transient neonatal diabetes are associated with somatic mutations in the ZFP57 coding sequence. RESULTS: Here, we comprehensively map sequence-specific ZFP57 binding sites in an allele-specific manner using hybrid ES cell lines from reciprocal crosses between C57BL/6J and Cast/EiJ mice, assigning allele specificity to approximately two-thirds of all binding sites. While half of these are biallelic and include endogenous retrovirus (ERV) targets, the rest show monoallelic binding based either on parental origin or on genetic background of the allele. Parental-origin allele-specific binding is methylation-dependent and maps only to imprinting control differentially methylated regions (DMRs) established in the germline. We identify a novel imprinted gene, Fkbp6, which has a critical function in mouse male germ cell development. Genetic background-specific sequence differences also influence ZFP57 binding, as genetic variation that disrupts the consensus binding motif and its methylation is often associated with monoallelic expression of neighboring genes. CONCLUSIONS: The work described here uncovers further roles for ZFP57-mediated regulation of genomic imprinting and identifies a novel mechanism for genetically determined monoallelic gene expression.


Asunto(s)
Alelos , Proteínas de Unión al ADN/metabolismo , Impresión Genómica , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Represoras/metabolismo , Animales , Sitios de Unión , Línea Celular , Metilación de ADN , Variación Genética , Código de Histonas , Ratones , Ratones Endogámicos C57BL
6.
Nat Struct Mol Biol ; 20(3): 311-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23416945

RESUMEN

Naive pluripotent embryonic stem cells (ESCs) and embryonic germ cells (EGCs) are derived from the preimplantation epiblast and primordial germ cells (PGCs), respectively. We investigated whether differences exist between ESCs and EGCs, in view of their distinct developmental origins. PGCs are programmed to undergo global DNA demethylation; however, we find that EGCs and ESCs exhibit equivalent global DNA methylation levels. Inhibition of MEK and Gsk3b by 2i conditions leads to pronounced reduction in DNA methylation in both cell types. This is driven by Prdm14 and is associated with downregulation of Dnmt3a and Dnmt3b. However, genomic imprints are maintained in 2i, and we report derivation of EGCs with intact genomic imprints. Collectively, our findings establish that culture in 2i instills a naive pluripotent state with a distinctive epigenetic configuration that parallels molecular features observed in both the preimplantation epiblast and nascent PGCs.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/fisiología , Células Germinativas/citología , Células Madre Pluripotentes/fisiología , Animales , Benzamidas/farmacología , Diferenciación Celular , Células Cultivadas/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Proteínas de Unión al ADN , Difenilamina/análogos & derivados , Difenilamina/farmacología , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Perfilación de la Expresión Génica , Impresión Genómica , Células Germinativas/fisiología , Estratos Germinativos/citología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Pluripotentes/citología , Piridinas/farmacología , Pirimidinas/farmacología , Proteínas de Unión al ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma , ADN Metiltransferasa 3B
7.
J Clin Invest ; 123(5): 2169-82, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23543057

RESUMEN

Large regions of recurrent genomic loss are common in cancers; however, with a few well-characterized exceptions, how they contribute to tumor pathogenesis remains largely obscure. Here we identified primate-restricted imprinting of a gene cluster on chromosome 20 in the region commonly deleted in chronic myeloid malignancies. We showed that a single heterozygous 20q deletion consistently resulted in the complete loss of expression of the imprinted genes L3MBTL1 and SGK2, indicative of a pathogenetic role for loss of the active paternally inherited locus. Concomitant loss of both L3MBTL1 and SGK2 dysregulated erythropoiesis and megakaryopoiesis, 2 lineages commonly affected in chronic myeloid malignancies, with distinct consequences in each lineage. We demonstrated that L3MBTL1 and SGK2 collaborated in the transcriptional regulation of MYC by influencing different aspects of chromatin structure. L3MBTL1 is known to regulate nucleosomal compaction, and we here showed that SGK2 inactivated BRG1, a key ATP-dependent helicase within the SWI/SNF complex that regulates nucleosomal positioning. These results demonstrate a link between an imprinted gene cluster and malignancy, reveal a new pathogenetic mechanism associated with acquired regions of genomic loss, and underline the complex molecular and cellular consequences of "simple" cancer-associated chromosome deletions.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 20 , Regulación de la Expresión Génica , Impresión Genómica , Alelos , Animales , Linaje de la Célula , Proteínas Cromosómicas no Histona/genética , Femenino , Silenciador del Gen , Heterocigoto , Humanos , Proteínas Inmediatas-Precoces/genética , Macaca , Macropodidae , Masculino , Modelos Genéticos , Familia de Multigenes , Trastornos Mieloproliferativos/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras , Transcripción Genética , Proteínas Supresoras de Tumor
8.
Epigenetics Chromatin ; 3(1): 2, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20180964

RESUMEN

BACKGROUND: The field of epigenetics is developing rapidly, however we are only beginning to comprehend the complexity of its influence on gene regulation. Using genomic imprinting as a model we examine epigenetic profiles associated with different forms of gene regulation. Imprinting refers to the expression of a gene from only one of the chromosome homologues in a parental-origin-specific manner. This is dependent on heritable germline epigenetic control at a cis-acting imprinting control region that influences local epigenetic states. Epigenetic modifications associated with imprinting regulation can be compared to those associated with the more canonical developmental regulation, important for processes such as differentiation and tissue specificity. Here we test the hypothesis that these two mechanisms are associated with different histone modification enrichment patterns. RESULTS: Using high-throughput data extraction with subsequent analysis, we have found that particular histone modifications are more likely to be associated with either imprinting repression or developmental repression of imprinted genes. H3K9me3 and H4K20me3 are together enriched at imprinted genes with differentially methylated promoters and do not show a correlation with developmental regulation. H3K27me3 and H3K4me3, however, are more often associated with developmental regulation. We find that imprinted genes are subject to developmental regulation through bivalency with H3K4me3 and H3K27me3 enrichment on the same allele. Furthermore, a specific tri-mark signature comprising H3K4me3, H3K9me3 and H4K20me3 has been identified at all imprinting control regions. CONCLUSION: A large amount of data is produced from whole-genome expression and epigenetic profiling studies of cellular material. We have shown that such publicly available data can be mined and analysed in order to generate novel findings for categories of genes or regulatory elements. Comparing two types of gene regulation, imprinting and developmental, our results suggest that different histone modifications associate with these distinct processes. This form of analysis is therefore a useful tool to elucidate the complex epigenetic code associated with genome function and to determine the underlying features conferring epigenetic states.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA