Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Nat Struct Mol Biol ; 31(3): 404-412, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38499830

RESUMEN

Cytosine DNA methylation is a highly conserved epigenetic mark in eukaryotes. Although the role of DNA methylation at gene promoters and repetitive elements has been extensively studied, the function of DNA methylation in other genomic contexts remains less clear. In the nucleus of mammalian cells, the genome is spatially organized at different levels, and strongly influences myriad genomic processes. There are a number of factors that regulate the three-dimensional (3D) organization of the genome, with the CTCF insulator protein being among the most well-characterized. Pertinently, CTCF binding has been reported as being DNA methylation-sensitive in certain contexts, perhaps most notably in the process of genomic imprinting. Therefore, it stands to reason that DNA methylation may play a broader role in the regulation of chromatin architecture. Here we summarize the current understanding that is relevant to both the mammalian DNA methylation and chromatin architecture fields and attempt to assess the extent to which DNA methylation impacts the folding of the genome. The focus is in early embryonic development and cellular transitions when the epigenome is in flux, but we also describe insights from pathological contexts, such as cancer, in which the epigenome and 3D genome organization are misregulated.


Asunto(s)
Metilación de ADN , Proteínas Represoras , Animales , Proteínas Represoras/metabolismo , Factor de Unión a CCCTC/metabolismo , Impresión Genómica , Cromatina , Mamíferos/genética
2.
Elife ; 122023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38149847

RESUMEN

The transmission of DNA through extracellular vesicles (EVs) represents a novel genetic material transfer mechanism that may impact genome evolution and tumorigenesis. We aimed to investigate the potential for vertical DNA transmission within maternal endometrial EVs to the pre-implantation embryo and describe any effect on embryo bioenergetics. We discovered that the human endometrium secretes all three general subtypes of EV - apoptotic bodies (ABs), microvesicles (MVs), and exosomes (EXOs) - into the human endometrial fluid (EF) within the uterine cavity. EVs become uniformly secreted into the EF during the menstrual cycle, with the proportion of different EV populations remaining constant; however, MVs contain significantly higher levels of mitochondrial (mt)DNA than ABs or EXOs. During the window of implantation, MVs contain an eleven-fold higher level of mtDNA when compared to cells-of-origin within the receptive endometrium, which possesses a lower mtDNA content and displays the upregulated expression of mitophagy-related genes. Furthermore, we demonstrate the internalization of EV-derived nuclear-encoded (n)DNA/mtDNA by trophoblast cells of murine embryos, which associates with a reduction in mitochondrial respiration and ATP production. These findings suggest that the maternal endometrium suffers a reduction in mtDNA content during the preconceptional period, that nDNA/mtDNA become packaged into secreted EVs that the embryo uptakes, and that the transfer of DNA to the embryo within EVs occurs alongside the modulation of bioenergetics during implantation.


Asunto(s)
Exosomas , Vesículas Extracelulares , Femenino , Humanos , Animales , Ratones , Vesículas Extracelulares/metabolismo , Implantación del Embrión , Exosomas/metabolismo , Embrión de Mamíferos/metabolismo , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo
3.
Genome Biol ; 24(1): 48, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36918927

RESUMEN

BACKGROUND: Genomic imprinting affects gene expression in a parent-of-origin manner and has a profound impact on complex traits including growth and behavior. While the rat is widely used to model human pathophysiology, few imprinted genes have been identified in this murid. To systematically identify imprinted genes and genomic imprints in the rat, we use low input methods for genome-wide analyses of gene expression and DNA methylation to profile embryonic and extraembryonic tissues at allele-specific resolution. RESULTS: We identify 14 and 26 imprinted genes in these tissues, respectively, with 10 of these genes imprinted in both tissues. Comparative analyses with mouse reveal that orthologous imprinted gene expression and associated canonical DNA methylation imprints are conserved in the embryo proper of the Muridae family. However, only 3 paternally expressed imprinted genes are conserved in the extraembryonic tissue of murids, all of which are associated with non-canonical H3K27me3 imprints. The discovery of 8 novel non-canonical imprinted genes unique to the rat is consistent with more rapid evolution of extraembryonic imprinting. Meta-analysis of novel imprinted genes reveals multiple mechanisms by which species-specific imprinted expression may be established, including H3K27me3 deposition in the oocyte, the appearance of ZFP57 binding motifs, and the insertion of endogenous retroviral promoters. CONCLUSIONS: In summary, we provide an expanded list of imprinted loci in the rat, reveal the extent of conservation of imprinted gene expression, and identify potential mechanisms responsible for the evolution of species-specific imprinting.


Asunto(s)
Histonas , Muridae , Ratones , Humanos , Ratas , Animales , Muridae/genética , Muridae/metabolismo , Histonas/metabolismo , Estudio de Asociación del Genoma Completo , Metilación de ADN , Impresión Genómica , Alelos
4.
Cell Rep ; 42(2): 112100, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36763500

RESUMEN

During pre-implantation stages of mammalian development, maternally stored material promotes both the erasure of the sperm and oocyte epigenetic profiles and is responsible for concomitant genome activation. Here, we have utilized single-cell methylome and transcriptome sequencing (scM&T-seq) to quantify both mRNA expression and DNA methylation in oocytes and a developmental series of human embryos at single-cell resolution. We fully characterize embryonic genome activation and maternal transcript degradation and map key epigenetic reprogramming events in developmentally high-quality embryos. By comparing these signatures with early embryos that have undergone spontaneous cleavage-stage arrest, as determined by time-lapse imaging, we identify embryos that fail to appropriately activate their genomes or undergo epigenetic reprogramming. Our results indicate that a failure to successfully accomplish these essential milestones impedes the developmental potential of pre-implantation embryos and is likely to have important implications, similar to aneuploidy, for the success of assisted reproductive cycles.


Asunto(s)
Multiómica , Semen , Animales , Humanos , Masculino , Desarrollo Embrionario/genética , Embrión de Mamíferos/metabolismo , Oocitos/metabolismo , Epigénesis Genética , Blastocisto/metabolismo , Mamíferos
5.
Clin Epigenetics ; 14(1): 71, 2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35643636

RESUMEN

BACKGROUND: Beckwith-Wiedemann syndrome (BWS) and Pseudohypoparathyroidism type 1B (PHP1B) are imprinting disorders (ID) caused by deregulation of the imprinted gene clusters located at 11p15.5 and 20q13.32, respectively. In both of these diseases a subset of the patients is affected by multi-locus imprinting disturbances (MLID). In several families, MLID is associated with damaging variants of maternal-effect genes encoding protein components of the subcortical maternal complex (SCMC). However, frequency, penetrance and recurrence risks of these variants are still undefined. In this study, we screened two cohorts of BWS patients and one cohort of PHP1B patients for the presence of MLID, and analysed the positive cases for the presence of maternal variants in the SCMC genes by whole exome-sequencing and in silico functional studies. RESULTS: We identified 10 new cases of MLID associated with the clinical features of either BWS or PHP1B, in which segregate 13 maternal putatively damaging missense variants of the SCMC genes. The affected genes also included KHDC3L that has not been associated with MLID to date. Moreover, we highlight the possible relevance of relatively common variants in the aetiology of MLID. CONCLUSION: Our data further add to the list of the SCMC components and maternal variants that are involved in MLID, as well as of the associated clinical phenotypes. Also, we propose that in addition to rare variants, common variants may play a role in the aetiology of MLID and imprinting disorders by exerting an additive effect in combination with rarer putatively damaging variants. These findings provide useful information for the molecular diagnosis and recurrence risk evaluation of MLID-associated IDs in genetic counselling.


Asunto(s)
Síndrome de Beckwith-Wiedemann , Seudohipoparatiroidismo , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Metilación de ADN , Impresión Genómica , Humanos , Proteínas/genética , Seudohipoparatiroidismo/genética , Seudohipoparatiroidismo
6.
J Med Genet ; 59(3): 253-261, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33579810

RESUMEN

INTRODUCTION: Kagami-Ogata syndrome (KOS14) and Temple syndrome (TS14) are two disorders associated with reciprocal alterations within the chr14q32 imprinted domain. Here, we present a work-up strategy for preimplantation genetic testing (PGT) to avoid the transmission of a causative micro-deletion. METHODS: We analysed DNA from the KOS14 index case and parents using methylation-sensitive ligation-mediated probe amplification and methylation pyrosequencing. The extent of the deletion was mapped using SNP arrays. PGT was performed in trophectoderm samples in order to identify unaffected embryos. Samples were amplified using multiple displacement amplification, followed by genome-wide SNP genotyping to determine the at-risk haplotype and next-generation sequencing to determine aneuploidies. RESULTS: A fully methylated pattern at the normally paternally methylated IG-DMR and MEG3 DMR in the KOS14 proband, accompanied by an unmethylated profile in the TS14 mother was consistent with maternal and paternal transmission of a deletion, respectively. Further analysis revealed a 108 kb deletion in both cases. The inheritance of the deletion on different parental alleles was consistent with the opposing phenotypes. In vitro fertilisation with intracytoplasmatic sperm injection and PGT were used to screen for deletion status and to transfer an unaffected embryo in this couple. A single euploid-unaffected embryo was identified resulting in a healthy baby born. DISCUSSION: We identify a microdeletion responsible for multigeneration KOS14 and TS14 within a single family where carriers have a 50% risk of transmitting the deletion to their offspring. We show that PGT can successfully be offered to couples with IDs caused by genetic anomalies.


Asunto(s)
Anomalías Múltiples , Diagnóstico Preimplantación , Anomalías Múltiples/genética , Aneuploidia , Cromosomas Humanos Par 14 , Femenino , Pruebas Genéticas/métodos , Humanos , Embarazo , Disomía Uniparental
7.
Nucleic Acids Res ; 48(20): 11394-11407, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33053156

RESUMEN

Genomic imprinting is an epigenetic process regulated by germline-derived DNA methylation that is resistant to embryonic reprogramming, resulting in parental origin-specific monoallelic gene expression. A subset of individuals affected by imprinting disorders (IDs) displays multi-locus imprinting disturbances (MLID), which may result from aberrant establishment of imprinted differentially methylated regions (DMRs) in gametes or their maintenance in early embryogenesis. Here we investigated the extent of MLID in a family harbouring a ZFP57 truncating variant and characterize the interactions between human ZFP57 and the KAP1 co-repressor complex. By ectopically targeting ZFP57 to reprogrammed loci in mouse embryos using a dCas9 approach, we confirm that ZFP57 recruitment is sufficient to protect oocyte-derived methylation from reprogramming. Expression profiling in human pre-implantation embryos and oocytes reveals that unlike in mice, ZFP57 is only expressed following embryonic-genome activation, implying that other KRAB-zinc finger proteins (KZNFs) recruit KAP1 prior to blastocyst formation. Furthermore, we uncover ZNF202 and ZNF445 as additional KZNFs likely to recruit KAP1 to imprinted loci during reprogramming in the absence of ZFP57. Together, these data confirm the perplexing link between KZFPs and imprint maintenance and highlight the differences between mouse and humans in this respect.


Asunto(s)
Metilación de ADN , Embrión de Mamíferos/metabolismo , Impresión Genómica , Células Germinativas/metabolismo , Oocitos/metabolismo , Proteínas Represoras/metabolismo , Síndrome de Beckwith-Wiedemann/metabolismo , Estudios de Cohortes , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Humanos , Análisis por Micromatrices , Mutación , Linaje , Seudohipoparatiroidismo/metabolismo , RNA-Seq , Proteínas Represoras/genética , Hermanos , Transcriptoma , Proteína 28 que Contiene Motivos Tripartito
8.
Clin Epigenetics ; 11(1): 35, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808399

RESUMEN

BACKGROUND: Genome-wide studies have begun to link subtle variations in both allelic DNA methylation and parent-of-origin genetic effects with early development. Numerous reports have highlighted that the placenta plays a critical role in coordinating fetal growth, with many key functions regulated by genomic imprinting. With the recent description of wide-spread polymorphic placenta-specific imprinting, the molecular mechanisms leading to this curious polymorphic epigenetic phenomenon is unknown, as is their involvement in pregnancies complications. RESULTS: Profiling of 35 ubiquitous and 112 placenta-specific imprinted differentially methylated regions (DMRs) using high-density methylation arrays and pyrosequencing revealed isolated aberrant methylation at ubiquitous DMRs as well as abundant hypomethylation at placenta-specific DMRs. Analysis of the underlying chromatin state revealed that the polymorphic nature is not only evident at the level of allelic methylation, but DMRs can also adopt an unusual epigenetic signature where the underlying histones are biallelically enrichment of H3K4 methylation, a modification normally mutually exclusive with DNA methylation. Quantitative expression analysis in placenta identified two genes, GPR1-AS1 and ZDBF2, that were differentially expressed between IUGRs and control samples after adjusting for clinical factors, revealing coordinated deregulation at the chromosome 2q33 imprinted locus. CONCLUSIONS: DNA methylation is less stable at placenta-specific imprinted DMRs compared to ubiquitous DMRs and contributes to privileged state of the placenta epigenome. IUGR-associated expression differences were identified for several imprinted transcripts independent of allelic methylation. Further work is required to determine if these differences are the cause IUGR or reflect unique adaption by the placenta to developmental stresses.


Asunto(s)
Metilación de ADN , Retardo del Crecimiento Fetal/genética , Perfilación de la Expresión Génica/métodos , Placenta/química , Cromosomas Humanos Par 2/genética , Islas de CpG , Proteínas de Unión al ADN/genética , Femenino , Retardo del Crecimiento Fetal/metabolismo , Regulación de la Expresión Génica , Impresión Genómica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Histonas/metabolismo , Humanos , Linaje , Embarazo
9.
Epigenomics ; 10(7): 941-954, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29962238

RESUMEN

AIM: This study aimed to establish a catalog of probes corresponding to imprinted differentially methylated regions (DMRs) on the Infinium HumanMethylationEPIC BeadChip. MATERIALS & METHODS: Reciprocal uniparental diploidies with low normal biparental mosaic contribution, together with normal diploid controls, were subjected to EPIC BeadChip hybridization. The methylation profiles were assessed for imprinted differential methylation. Top candidates were validated using locus-specific PCR-based assays. RESULTS: Seven hundred and eighty-nine CpG probes coincided with 50 known imprinted DMRs and 467 CpG probes corresponding to 124 novel imprinted DMR candidates were identified. Validation led to identification of several subtle DMRs within known imprinted domains as well as novel maternally methylated regions associated with PTCHD3 and JAKMIP1. CONCLUSION: Our comprehensive list of bona fide-imprinted DMR probes will simplify and facilitate methylation profiling of individuals with imprinting disorders and is applicable to other diseases in which aberrant imprinting has been implicated, such as cancer and fetal growth.


Asunto(s)
Metilación de ADN , Genoma Humano , Impresión Genómica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Islas de CpG , Sondas de ADN/genética , Estudio de Asociación del Genoma Completo , Voluntarios Sanos , Humanos
10.
Nat Med ; 24(5): 598-603, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29736028

RESUMEN

The chances to develop Alzheimer's disease (AD) result from a combination of genetic and non-genetic risk factors 1 , the latter likely being mediated by epigenetic mechanisms 2 . In the past, genome-wide association studies (GWAS) have identified an important number of risk loci associated with AD pathology 3 , but a causal relationship remains difficult to establish. In contrast, locus-specific or epigenome-wide association studies (EWAS) have revealed site-specific epigenetic alterations, which provide mechanistic insights for a particular risk gene but often lack the statistical power of GWAS 4 . Here, combining both approaches, we report a previously unidentified association of the peptidase M20-domain-containing protein 1 (PM20D1) with AD. We find that PM20D1 is a methylation and expression quantitative trait locus coupled to an AD-risk associated haplotype, which displays enhancer-like characteristics and contacts the PM20D1 promoter via a haplotype-dependent, CCCTC-binding-factor-mediated chromatin loop. Furthermore, PM20D1 is increased following AD-related neurotoxic insults at symptomatic stages in the APP/PS1 mouse model of AD and in human patients with AD who are carriers of the non-risk haplotype. In line, genetically increasing or decreasing the expression of PM20D1 reduces and aggravates AD-related pathologies, respectively. These findings suggest that in a particular genetic background, PM20D1 contributes to neuroprotection against AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Amidohidrolasas/genética , Sitios de Carácter Cuantitativo/genética , Anciano , Amidohidrolasas/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Femenino , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Estudio de Asociación del Genoma Completo , Humanos , Peróxido de Hidrógeno/metabolismo , Desequilibrio de Ligamiento/genética , Masculino , Ratones , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética
11.
Methods Mol Biol ; 1766: 109-121, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29605849

RESUMEN

Imprinting disorders are a group of congenital diseases that can result from multiple mechanisms affecting imprinted gene dosage including cytogenetic aberration and epigenetic anomalies. Quantification of CpG methylation and correct copy-number calling is required for molecular diagnosis. Methylation-sensitive multiplex ligation-dependent probe amplification (MS-MLPA) is a multiplex method that accurately measures both parameters in a single assay. This technique relies upon the ligation of MLPA probe oligonucleotides and digestion of the genomic DNA-probe hybrid complexes with the Hha1 methylation-sensitive restriction endonuclease prior to fluorescent PCR amplification with a single primer pair. Since each targeted probe contains stuffer sequence of varying length, each interrogated position is visualized as an amplicon of different size upon capillary electrophoresis.


Asunto(s)
Metilación de ADN , ADN/química , ADN/metabolismo , Impresión Genómica , Islas de CpG , ADN/efectos de los fármacos , ADN/genética , Cartilla de ADN/química , Cartilla de ADN/genética , Cartilla de ADN/metabolismo , Enzimas de Restricción del ADN/química , Enzimas de Restricción del ADN/farmacología , Electroforesis Capilar , Epigénesis Genética , Dosificación de Gen , Humanos , Reacción en Cadena de la Polimerasa Multiplex , Programas Informáticos
12.
Nat Commun ; 8(1): 467, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28883545

RESUMEN

It has been postulated that imprinting aberrations are common in tumors. To understand the role of imprinting in cancer, we have characterized copy-number and methylation in over 280 cancer cell lines and confirm our observations in primary tumors. Imprinted differentially methylated regions (DMRs) regulate parent-of-origin monoallelic expression of neighboring transcripts in cis. Unlike single-copy CpG islands that may be prone to hypermethylation, imprinted DMRs can either loose or gain methylation during tumorigenesis. Here, we show that methylation profiles at imprinted DMRs often not represent genuine epigenetic changes but simply the accumulation of underlying copy-number aberrations (CNAs), which is independent of the genome methylation state inferred from cancer susceptible loci. Our results reveal that CNAs also influence allelic expression as loci with copy-number neutral loss-of-heterozygosity or amplifications may be expressed from the appropriate parental chromosomes, which is indicative of maintained imprinting, although not observed as a single expression foci by RNA FISH.Altered genomic imprinting is frequently reported in cancer. Here, the authors analyze copy number and methylation in cancer cell lines and primary tumors to show that imprinted methylation profiles represent the accumulation of copy number alteration, rather than epigenetic alterations.


Asunto(s)
Epigénesis Genética , Dosificación de Gen , Impresión Genómica , Neoplasias/genética , Alelos , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Sitios Genéticos , Humanos
13.
Hum Mutat ; 38(6): 615-620, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28256047

RESUMEN

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare cause of pulmonary hypertension in newborns. Maternally inherited point mutations in Forkhead Box F1 gene (FOXF1), deletions of the gene, or its long-range enhancers on the maternal allele are responsible for this neonatal lethal disorder. Here, we describe monozygotic twins and one full-term newborn with ACD and gastrointestinal malformations caused by de novo mutations of FOXF1 on the maternal-inherited alleles. Since this parental transmission is consistent with genomic imprinting, the parent-of-origin specific monoallelic expression of genes, we have undertaken a detailed analysis of both allelic expression and DNA methylation. FOXF1 and its neighboring gene FENDRR were both biallelically expressed in a wide range of fetal tissues, including lung and intestine. Furthermore, detailed methylation screening within the 16q24.1 regions failed to identify regions of allelic methylation, suggesting that disrupted imprinting is not responsible for ACDMPV.


Asunto(s)
Factores de Transcripción Forkhead/genética , Impresión Genómica , Síndrome de Circulación Fetal Persistente/genética , Alveolos Pulmonares/anomalías , Hibridación Genómica Comparativa , Metilación de ADN/genética , Femenino , Humanos , Hipertensión Pulmonar , Recién Nacido , Herencia Materna/genética , Mutación , Síndrome de Circulación Fetal Persistente/complicaciones , Síndrome de Circulación Fetal Persistente/patología , Embarazo , Alveolos Pulmonares/patología , Gemelos Monocigóticos
14.
PLoS Genet ; 12(11): e1006427, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27835649

RESUMEN

Thousands of regions in gametes have opposing methylation profiles that are largely resolved during the post-fertilization epigenetic reprogramming. However some specific sequences associated with imprinted loci survive this demethylation process. Here we present the data describing the fate of germline-derived methylation in humans. With the exception of a few known paternally methylated germline differentially methylated regions (DMRs) associated with known imprinted domains, we demonstrate that sperm-derived methylation is reprogrammed by the blastocyst stage of development. In contrast a large number of oocyte-derived methylation differences survive to the blastocyst stage and uniquely persist as transiently methylated DMRs only in the placenta. Furthermore, we demonstrate that this phenomenon is exclusive to primates, since no placenta-specific maternal methylation was observed in mouse. Utilizing single cell RNA-seq datasets from human preimplantation embryos we show that following embryonic genome activation the maternally methylated transient DMRs can orchestrate imprinted expression. However despite showing widespread imprinted expression of genes in placenta, allele-specific transcriptional profiling revealed that not all placenta-specific DMRs coordinate imprinted expression and that this maternal methylation may be absent in a minority of samples, suggestive of polymorphic imprinted methylation.


Asunto(s)
Metilación de ADN/genética , Impresión Genómica/genética , Células Germinativas/metabolismo , Oocitos/metabolismo , Animales , Blastocisto/metabolismo , Islas de CpG/genética , Femenino , Humanos , Masculino , Ratones , Placenta/metabolismo , Embarazo , Primates/genética , Primates/crecimiento & desarrollo , Espermatozoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...