Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 25(5): 611-23, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25862382

RESUMEN

Genomic imprinting is an epigenetic mechanism resulting in parental allele-specific gene expression. Defects in normal imprinting are found in cancer, assisted reproductive technologies, and several human syndromes. In mouse models, germline-derived DNA methylation is shown to regulate imprinting. Though imprinting is largely conserved between mammals, species- and tissue-specific domains of imprinted expression exist. Using the cynomolgus macaque (Macaca fascicularis) to assess primate-specific imprinting, we present a comprehensive view of tissue-specific imprinted expression and DNA methylation at established imprinted gene clusters. For example, like mouse and unlike human, macaque IGF2R is consistently imprinted, and the PLAGL1, INPP5F transcript variant 2, and PEG3 imprinting control regions are not methylated in the macaque germline but acquire this post-fertilization. Methylome data from human early embryos appear to support this finding. These suggest fundamental differences in imprinting control mechanisms between primate species and rodents at some imprinted domains, with implications for our understanding of the epigenetic programming process in humans and its influence on disease.


Asunto(s)
Metilación de ADN , Impresión Genómica , Oocitos/metabolismo , Animales , Secuencia de Bases , Proteínas de Unión al ADN/genética , Femenino , Humanos , Inositol Polifosfato 5-Fosfatasas , Factores de Transcripción de Tipo Kruppel/genética , Macaca fascicularis , Masculino , Ratones , Datos de Secuencia Molecular , Especificidad de Órganos , Monoéster Fosfórico Hidrolasas/genética , ARN Largo no Codificante/genética , Especificidad de la Especie
2.
Genome Res ; 24(7): 1064-74, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24709820

RESUMEN

Integrating the genotype with epigenetic marks holds the promise of better understanding the biology that underlies the complex interactions of inherited and environmental components that define the developmental origins of a range of disorders. The quality of the in utero environment significantly influences health over the lifecourse. Epigenetics, and in particular DNA methylation marks, have been postulated as a mechanism for the enduring effects of the prenatal environment. Accordingly, neonate methylomes contain molecular memory of the individual in utero experience. However, interindividual variation in methylation can also be a consequence of DNA sequence polymorphisms that result in methylation quantitative trait loci (methQTLs) and, potentially, the interaction between fixed genetic variation and environmental influences. We surveyed the genotypes and DNA methylomes of 237 neonates and found 1423 punctuate regions of the methylome that were highly variable across individuals, termed variably methylated regions (VMRs), against a backdrop of homogeneity. MethQTLs were readily detected in neonatal methylomes, and genotype alone best explained ∼25% of the VMRs. We found that the best explanation for 75% of VMRs was the interaction of genotype with different in utero environments, including maternal smoking, maternal depression, maternal BMI, infant birth weight, gestational age, and birth order. Our study sheds new light on the complex relationship between biological inheritance as represented by genotype and individual prenatal experience and suggests the importance of considering both fixed genetic variation and environmental factors in interpreting epigenetic variation.


Asunto(s)
Metilación de ADN , Ambiente , Epigénesis Genética , Interacción Gen-Ambiente , Heterogeneidad Genética , Genotipo , Transcriptoma , Biología Computacional/métodos , Islas de CpG , Epigenómica/métodos , Femenino , Humanos , Recién Nacido , Masculino , Polimorfismo de Nucleótido Simple , Embarazo , Sitios de Carácter Cuantitativo , Factores de Riesgo
3.
J Hum Genet ; 59(9): 504-11, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25102097

RESUMEN

CXCL14 is a chemokine that has previously been implicated in insulin resistance in mice. In humans, the role of CXCL14 in metabolic processes is not well established, and we sought to determine whether CXCL14 is a risk susceptibility gene important in fetal programming of metabolic disease. For this purpose, we investigated whether CXCL14 is differentially regulated in human umbilical cords of infants with varying birth weights. We found an elevated expression of CXCL14 in human low birth weight (LBW) cords, as well as in cords from nutritionally restricted Macaca fascicularis macaques. To further analyze the regulatory mechanisms underlying the expression of CXCL14, we examined CXCL14 in umbilical cord-derived mesenchymal stem cells (MSCs) that provide an in vitro cell-based system amenable to experimental manipulation. Using both whole frozen cords and MSCs, we determined that site-specific CpG methylation in the CXCL14 promoter is associated with altered expression, and that changes in methylation are evident in LBW infant-derived umbilical cords that may indicate future metabolic compromise through CXCL14.


Asunto(s)
Quimiocinas CXC/genética , Metilación de ADN , Perfilación de la Expresión Génica , Recién Nacido de Bajo Peso/metabolismo , Adulto , Animales , Restricción Calórica , Células Cultivadas , Islas de CpG/genética , Femenino , Humanos , Recién Nacido , Macaca fascicularis/genética , Masculino , Edad Materna , Células Madre Mesenquimatosas/metabolismo , Embarazo , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
4.
Am J Clin Nutr ; 114(3): 1219-1228, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33963740

RESUMEN

BACKGROUND: Previous studies focusing on the association between gestational diabetes and breastfeeding duration have been inconclusive. OBJECTIVES: We aimed to determine whether maternal gestational hyperglycemia is associated with the duration of breastfeeding and the concentrations of markers linked to breastmilk production. METHODS: Data from the prospective, multiethnic Growing Up in Singapore Towards Healthy Outcomes study were used to assess the association of fasting plasma glucose (FPG) and 2-h postglucose challenge (2hPG) measured at 26-28 wk of gestation with duration of breastfeeding and concentrations of protein, lactose, citrate, sodium, potassium, and zinc in breastmilk 3 wk postpartum. RESULTS: Of the 1035 participants, 5.2% and 9.5% had elevated FPG and 2hPG, respectively, consistent with a diagnosis of gestational diabetes mellitus based on International Association of Diabetes and Pregnancy Study Groups criteria. FPG ≥5.1 mmol/L was associated with a crude reduction in median breastfeeding duration of 2.3 mo. In a model adjusted for maternal prepregnancy BMI and intention to breastfeed, FPG ≥5.1 mmol/L predicted earlier termination of any breastfeeding (adjusted HR: 1.47; 95% CI: 1.04, 2.08) but not full breastfeeding (adjusted HR: 1.08; 0.76, 1.55). 2hPG ≥8.5 mmol/L was not significantly associated with the durations of any (adjusted HR: 0.86; 95% CI: 0.62, 1.19) or full (adjusted HR: 0.85; 95% CI: 0.62, 1.18) breastfeeding. Maternal FPG was significantly and positively associated with breastmilk sodium (adjusted coefficient: 1.28; 95% CI: 1.08, 1.51) and sodium-to-potassium ratio (adjusted coefficient: 1.29; 95% CI: 1.08, 1.54) but not with other measured breastmilk components. CONCLUSIONS: Women with FPG ≥5.1 mmol/L during pregnancy breastfeed for a shorter duration. Future work involving measurement of milk production is needed to determine whether low milk production predicts breastfeeding duration among women with elevated FPG. This trial was registered at www.clinicaltrials.gov as NCT01174875.


Asunto(s)
Glucemia/metabolismo , Lactancia Materna , Diabetes Gestacional , Hiperglucemia/complicaciones , Leche Humana/metabolismo , Adulto , Animales , Biomarcadores/metabolismo , Complicaciones de la Diabetes/sangre , Diabetes Gestacional/sangre , Ayuno , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/sangre , Hiperglucemia/epidemiología , Potasio/metabolismo , Embarazo , Estudios Prospectivos , Factores de Riesgo , Singapur , Sodio/metabolismo
5.
J Cell Biochem ; 110(2): 288-93, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20336663

RESUMEN

The dynamics of embryonic stem cell pluripotency is orchestrated by an interplay of transcriptional and epigenetic regulation in a systematic and modular manner. While the ES cell stage is marked by multiple loci with bivalent chromatin marks that prepare genes for imminent activation on differentiation, this open chromatin conformation is tempered by repressive machinery that prevent premature expression of key developmental genes. This review serves to highlight key ES transcription factors and their known links to the epigenetic machinery via known protein complexes.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Ciclo Celular , Epigénesis Genética , Humanos
6.
Epigenetics ; 13(9): 941-958, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30232931

RESUMEN

Accounting for cellular heterogeneity is essential in neonatal epigenome-wide association studies (EWAS) performed on heterogeneous tissues, such as umbilical cord tissue (CT) or cord blood (CB). Using a reference-panel-based statistical approach, the cell type composition of heterogeneous tissues can be estimated by comparison of whole tissue DNA methylation profiles with cell type-specific DNA methylation signatures. Currently, there is no adequate DNA methylation reference panel for CT, and existing CB panels have been generated on lower coverage Infinium HumanMethylation450 arrays. In this study, we generate a reference panel for CT and improve available CB panels by using the higher coverage Infinium MethylationEPIC arrays. We performed DNA methylation profiling of 9 cell types isolated from CT and CB samples from 14 neonates. In addition to these cell types, we profiled DNA methylation of unfractionated CT and CB. Cell type composition of these unfractionated tissue samples, as estimated by our reference panels, was in agreement with that obtained by flow cytometry. Expectedly, DNA methylation profiles from CT and CB were distinct, reflecting their mesenchymal and hematopoietic stem cell origins. Variable CpGs from both unfractionated CT and its isolated cell types were more likely to be located in open seas and intronic regions than those in CB. Cell type specific CpGs in CT were enriched in intercellular matrix pathways, while those from CB were enriched in immune-related pathways. This study provides an open source reference panel for estimation and adjustment of cellular heterogeneity in CT and CB, and broadens the scope of tissue utilization assessed in future neonatal EWAS studies.


Asunto(s)
Células Sanguíneas/metabolismo , Metilación de ADN , Epigenómica/normas , Sangre Fetal/metabolismo , Análisis de Secuencia de ADN/normas , Cordón Umbilical/metabolismo , Adulto , Islas de CpG , Femenino , Sangre Fetal/citología , Humanos , Recién Nacido , Especificidad de Órganos , Embarazo , Estándares de Referencia , Análisis de Secuencia de ADN/métodos , Cordón Umbilical/citología
7.
EBioMedicine ; 19: 60-72, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28473239

RESUMEN

Experimental studies show a substantial contribution of early life environment to obesity risk through epigenetic processes. We examined inter-individual DNA methylation differences in human birth tissues associated with child's adiposity. We identified a novel association between the level of CpG methylation at birth within the promoter of the long non-coding RNA ANRIL (encoded at CDKN2A) and childhood adiposity at age 6-years. An association between ANRIL methylation and adiposity was also observed in three additional populations; in birth tissues from ethnically diverse neonates, in peripheral blood from adolescents, and in adipose tissue from adults. Additionally, CpG methylation was associated with ANRIL expression in vivo, and CpG mutagenesis in vitro inhibited ANRIL promoter activity. Furthermore, CpG methylation enhanced binding to an Estrogen Response Element within the ANRIL promoter. Our findings demonstrate that perinatal methylation at loci relevant to gene function may be a robust marker of later adiposity, providing substantial support for epigenetic processes in mediating long-term consequences of early life environment on human health.


Asunto(s)
Adiposidad/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Adolescente , Adulto , Anciano , Biomarcadores , Línea Celular Tumoral , Niño , Islas de CpG , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Metilación de ADN , Epigénesis Genética , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Obesidad/genética , Adulto Joven
8.
Epigenetics ; 11(1): 36-48, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26786415

RESUMEN

Interindividual variability in the epigenome has gained tremendous attention for its potential in pathophysiological investigation, disease diagnosis, and evaluation of clinical intervention. DNA methylation is the most studied epigenetic mark in epigenome-wide association studies (EWAS) as it can be detected from limited starting material. Infinium 450K methylation array is the most popular platform for high-throughput profiling of this mark in clinical samples, as it is cost-effective and requires small amounts of DNA. However, this method suffers from low genome coverage and errors introduced by probe cross-hybridization. Whole-genome bisulfite sequencing can overcome these limitations but elevates the costs tremendously. Methyl-Capture Sequencing (MC Seq) is an attractive intermediate solution to increase the methylome coverage in large sample sets. Here we first demonstrate that MC Seq can be employed using DNA amounts comparable to the amounts used for Infinium 450K. Second, to provide guidance when choosing between the 2 platforms for EWAS, we evaluate and compare MC Seq and Infinium 450K in terms of coverage, technical variation, and concordance of methylation calls in clinical samples. Last, since the focus in EWAS is to study interindividual variation, we demonstrate the utility of MC Seq in studying interindividual variation in subjects from different ethnicities.


Asunto(s)
Metilación de ADN , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ADN/métodos , Epigénesis Genética , Etnicidad/genética , Femenino , Genoma Humano , Humanos , Masculino , Alineación de Secuencia
9.
G3 (Bethesda) ; 4(7): 1227-34, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24815017

RESUMEN

The Infinium Human Methylation450 BeadChip Array (Infinium 450K) is a robust and cost-efficient survey of genome-wide DNA methylation patterns. Macaca fascicularis (Cynomolgus macaque) is an important disease model; however, its genome sequence is only recently published, and few tools exist to interrogate the molecular state of Cynomolgus macaque tissues. Although the Infinium 450K is a hybridization array designed to the human genome, the relative conservation between the macaque and human genomes makes its use in macaques feasible. Here, we used the Infinium 450K array to assay DNA methylation in 11 macaque muscle biopsies. We showed that probe hybridization efficiency was related to the degree of sequence identity between the human probes and the macaque genome sequence. Approximately 61% of the Human Infinium 450K probes could be reliably mapped to the Cynomolgus macaque genome and contain a CpG site of interest. We also compared the Infinium 450K data to reduced representation bisulfite sequencing data generated on the same samples and found a high level of concordance between the two independent methodologies, which can be further improved by filtering for probe sequence identity and mismatch location. We conclude that the Infinium 450K array can be used to measure the DNA methylome of Cynomolgus macaque tissues using the provided filters. We also provide a pipeline for validation of the array in other species using a simple BLAST-based sequence identify filter.


Asunto(s)
Genoma , Macaca fascicularis/genética , Animales , Islas de CpG , ADN/genética , ADN/metabolismo , Metilación de ADN , Genoma Humano , Humanos , Músculo Esquelético/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ADN
10.
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
11.
PLoS One ; 7(7): e39744, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22808055

RESUMEN

BACKGROUND: Babies born at lower gestational ages or smaller birthweights have a greater risk of poorer health in later life. Both the causes of these sub-optimal birth outcomes and the mechanism by which the effects are transmitted over decades are the subject of extensive study. We investigated whether a transcriptomic signature of either birthweight or gestational age could be detected in umbilical cord RNA. METHODS: The gene expression patterns of 32 umbilical cords from Singaporean babies of Chinese ethnicity across a range of birthweights (1698-4151 g) and gestational ages (35-41 weeks) were determined. We confirmed the differential expression pattern by gestational age for 12 genes in a series of 127 umbilical cords of Chinese, Malay and Indian ethnicity. RESULTS: We found that the transcriptome is substantially influenced by gestational age; but less so by birthweight. We show that some of the expression changes dependent on gestational age are enriched in signal transduction pathways, such as Hedgehog and in genes with roles in cytokine signalling and angiogenesis. We show that some of the gene expression changes we report are reflected in the epigenome. CONCLUSIONS: We studied the umbilical cord which is peripheral to disease susceptible tissues. The results suggest that soma-wide transcriptome changes, preserved at the epigenetic level, may be a mechanism whereby birth outcomes are linked to the risk of adult metabolic and arthritic disease and suggest that greater attention be given to the association between premature birth and later disease risk.


Asunto(s)
Citocinas/genética , Proteínas Hedgehog/genética , Recién Nacido Pequeño para la Edad Gestacional , Nacimiento Prematuro/genética , Transcriptoma , Cordón Umbilical/química , Adulto , Peso al Nacer , Citocinas/metabolismo , Etnicidad , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Proteínas Hedgehog/metabolismo , Humanos , Recién Nacido , Masculino , Análisis por Micromatrices , Embarazo , Nacimiento Prematuro/etnología , Transducción de Señal , Singapur
12.
Stem Cells Int ; 2011: 560261, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21776282

RESUMEN

Much of embryonic stem cell biology has focused on transcriptional expression and regulation of genes that could mediate its unique potential in self-renewal or pluripotency. In alignment with our present understanding on the genetic, protein, and epigenetic factors that may direct cell fate, we present a short overview of the often overlooked contribution of alternative splice variants to regulatory diversity. Progressing beyond the limitations of a fixed genomic sequence, alternative splicing offers an additional layer of complexity to produce protein variants that may differ in function and localization that can direct embryonic stem cells to specific differentiation pathways. In light of the number of variants that can be produced at key ES cell genes alone, it is challenging to consider how much more multifaceted transcriptional regulation truly is, and if this can be captured more fully in future works.

13.
Stem Cell Res Ther ; 2(3): 26, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21569470

RESUMEN

INTRODUCTION: Octamer-binding transcription factor 4 (Oct4) is a master regulator of early mammalian development. Its expression begins from the oocyte stage, becomes restricted to the inner cell mass of the blastocyst and eventually remains only in primordial germ cells. Unearthing the interactions of Oct4 would provide insight into how this transcription factor is central to cell fate and stem cell pluripotency. METHODS: In the present study, affinity-tagged endogenous Oct4 cell lines were established via homologous recombination gene targeting in embryonic stem (ES) cells to express tagged Oct4. This allows tagged Oct4 to be expressed without altering the total Oct4 levels from their physiological levels. RESULTS: Modified ES cells remained pluripotent. However, when modified ES cells were tested for their functionality, cells with a large tag failed to produce viable homozygous mice. Use of a smaller tag resulted in mice with normal development, viability and fertility. This indicated that the choice of tags can affect the performance of Oct4. Also, different tags produce a different repertoire of Oct4 interactors. CONCLUSIONS: Using a total of four different tags, we found 33 potential Oct4 interactors, of which 30 are novel. In addition to transcriptional regulation, the molecular function associated with these Oct4-associated proteins includes various other catalytic activities, suggesting that, aside from chromosome remodeling and transcriptional regulation, Oct4 function extends more widely to other essential cellular mechanisms. Our findings show that multiple purification approaches are needed to uncover a comprehensive Oct4 protein interaction network.


Asunto(s)
Cromatografía de Afinidad/métodos , Factor 3 de Transcripción de Unión a Octámeros/aislamiento & purificación , Mapeo de Interacción de Proteínas , Marcadores de Afinidad , Secuencia de Aminoácidos , Animales , Western Blotting , Línea Celular/química , Células Madre Embrionarias/metabolismo , Genes Letales , Espectrometría de Masas , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/aislamiento & purificación , Factores de Transcripción/metabolismo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA