Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 83(10): 1542-1544, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37207620

RESUMEN

Three papers, Kawasaki et al.1 in this issue and Chen et al.2 and Li et al.3 in the previous issue of Molecular Cell, reveal regulatory roles for multiway chromatin interactions mediated by structural elements in Drosophila and human genomes.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Animales , Humanos , Regiones Promotoras Genéticas , Cromatina/genética , Drosophila/genética
2.
Artículo en Inglés | MEDLINE | ID: mdl-34400552

RESUMEN

Dramatic nuclear reorganization occurs during early development to convert terminally differentiated gametes to a totipotent zygote, which then gives rise to an embryo. Aberrant epigenome resetting severely impairs embryo development and even leads to lethality. How the epigenomes are inherited, reprogrammed, and reestablished in this critical developmental period has gradually been unveiled through the rapid development of technologies including ultrasensitive chromatin analysis methods. In this review, we summarize the latest findings on epigenetic reprogramming in gametogenesis and embryogenesis, and how it contributes to gamete maturation and parental-to-zygotic transition. Finally, we highlight the key questions that remain to be answered to fully understand chromatin regulation and nuclear reprogramming in early development.


Asunto(s)
Epigenómica , Cigoto , Animales , Reprogramación Celular , Cromatina , Desarrollo Embrionario/genética , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Cigoto/fisiología
3.
Genome Res ; 31(7): 1121-1135, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34140314

RESUMEN

Heterochromatin remodeling is critical for various cell processes. In particular, the "loss of heterochromatin" phenotype in cellular senescence is associated with the process of aging and age-related disorders. Although biological processes of senescent cells, including senescence-associated heterochromatin foci (SAHF) formation, chromosome compaction, and redistribution of key proteins, have been closely associated with high-order chromatin structure, the relationship between the high-order chromatin reorganization and the loss of heterochromatin phenotype during senescence has not been fully understood. By using senescent and deep senescent fibroblasts induced by DNA damage harboring the "loss of heterochromatin" phenotype, we observed progressive 3D reorganization of heterochromatin during senescence. Facultative and constitutive heterochromatin marked by H3K27me3 and H3K9me3, respectively, show different alterations. Facultative heterochromatin tends to switch from the repressive B-compartment to the active A-compartment, whereas constitutive heterochromatin shows no significant changes at the compartment level but enhanced interactions between themselves. Both types of heterochromatin show increased chromatin accessibility and gene expression leakage during senescence. Furthermore, increased chromatin accessibility in potential CTCF binding sites accompanies the establishment of novel loops in constitutive heterochromatin. Finally, we also observed aberrant expression of repetitive elements, including LTR (long terminal repeat) and satellite classes. Overall, facultative and constitutive heterochromatin show both similar and distinct multiscale alterations in the 3D map, chromatin accessibility, and gene expression leakage. This study provides an epigenomic map of heterochromatin reorganization during senescence.

4.
Mol Cell ; 79(2): 234-250.e9, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32579944

RESUMEN

Somatic cell nuclear transfer (SCNT) can reprogram a somatic nucleus to a totipotent state. However, the re-organization of 3D chromatin structure in this process remains poorly understood. Using low-input Hi-C, we revealed that, during SCNT, the transferred nucleus first enters a mitotic-like state (premature chromatin condensation). Unlike fertilized embryos, SCNT embryos show stronger topologically associating domains (TADs) at the 1-cell stage. TADs become weaker at the 2-cell stage, followed by gradual consolidation. Compartments A/B are markedly weak in 1-cell SCNT embryos and become increasingly strengthened afterward. By the 8-cell stage, somatic chromatin architecture is largely reset to embryonic patterns. Unexpectedly, we found cohesin represses minor zygotic genome activation (ZGA) genes (2-cell-specific genes) in pluripotent and differentiated cells, and pre-depleting cohesin in donor cells facilitates minor ZGA and SCNT. These data reveal multi-step reprogramming of 3D chromatin architecture during SCNT and support dual roles of cohesin in TAD formation and minor ZGA repression.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Cromatina/fisiología , Proteínas Cromosómicas no Histona/fisiología , Técnicas de Transferencia Nuclear , Cigoto/fisiología , Animales , Línea Celular , Núcleo Celular , Ensamble y Desensamble de Cromatina , Biología Computacional/métodos , Conjuntos de Datos como Asunto , Desarrollo Embrionario , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Cohesinas
5.
Mol Cell ; 77(4): 825-839.e7, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31837995

RESUMEN

In mammals, chromatin organization undergoes drastic reorganization during oocyte development. However, the dynamics of three-dimensional chromatin structure in this process is poorly characterized. Using low-input Hi-C (genome-wide chromatin conformation capture), we found that a unique chromatin organization gradually appears during mouse oocyte growth. Oocytes at late stages show self-interacting, cohesin-independent compartmental domains marked by H3K27me3, therefore termed Polycomb-associating domains (PADs). PADs and inter-PAD (iPAD) regions form compartment-like structures with strong inter-domain interactions among nearby PADs. PADs disassemble upon meiotic resumption from diplotene arrest but briefly reappear on the maternal genome after fertilization. Upon maternal depletion of Eed, PADs are largely intact in oocytes, but their reestablishment after fertilization is compromised. By contrast, depletion of Polycomb repressive complex 1 (PRC1) proteins attenuates PADs in oocytes, which is associated with substantial gene de-repression in PADs. These data reveal a critical role of Polycomb in regulating chromatin architecture during mammalian oocyte growth and early development.


Asunto(s)
Cromatina/química , Oocitos/crecimiento & desarrollo , Oogénesis/genética , Proteínas del Grupo Polycomb/fisiología , Animales , Blastocisto/química , Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Embrión de Mamíferos/química , Silenciador del Gen , Código de Histonas , Ratones , Oocitos/química , Transcripción Genética , Cohesinas
6.
Nat Genet ; 52(1): 95-105, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31844322

RESUMEN

Around implantation, the epiblast (Epi) transits from naïve to primed pluripotency, before giving rise to the three germ layers. How chromatin is reconfigured during this developmental window remains poorly understood. We performed a genome-wide investigation of chromatin landscapes during this period. We find that enhancers in ectoderm are already pre-accessible in embryonic day 6.5 (E6.5) Epi when cells enter a primed pluripotent state. Unexpectedly, strong trimethylation of histone H3 at lysine 4 (H3K4me3) emerges at developmental gene promoters in E6.5 Epi and positively correlates with H3K27me3, thus establishing bivalency. These genes also show enhanced spatial interactions. Both the strong bivalency and spatial clustering are virtually absent in preimplantation embryos and are markedly reduced in fate-committed lineages. Finally, we show that KMT2B is essential for establishing bivalent H3K4me3 at E6.5 but becomes partially dispensable later. Its deficiency leads to impaired activation of developmental genes and subsequent embryonic lethality. Thus, our data characterize lineage-specific chromatin reconfiguration and a unique chromatin state for primed pluripotency.


Asunto(s)
Cromatina/genética , Embrión de Mamíferos/citología , Células Madre Embrionarias/citología , Epigenómica/métodos , Gastrulación , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína de la Leucemia Mieloide-Linfoide/fisiología , Animales , Metilación de ADN , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Femenino , Histonas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Nat Genet ; 51(5): 844-856, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31040401

RESUMEN

The oocyte epigenome plays critical roles in mammalian gametogenesis and embryogenesis. Yet, how it is established remains elusive. Here, we report that histone-lysine N-methyltransferase SETD2, an H3K36me3 methyltransferase, is a crucial regulator of the mouse oocyte epigenome. Deficiency in Setd2 leads to extensive alterations of the oocyte epigenome, including the loss of H3K36me3, failure in establishing the correct DNA methylome, invasion of H3K4me3 and H3K27me3 into former H3K36me3 territories and aberrant acquisition of H3K4me3 at imprinting control regions instead of DNA methylation. Importantly, maternal depletion of SETD2 results in oocyte maturation defects and subsequent one-cell arrest after fertilization. The preimplantation arrest is mainly due to a maternal cytosolic defect, since it can be largely rescued by normal oocyte cytosol. However, chromatin defects, including aberrant imprinting, persist in these embryos, leading to embryonic lethality after implantation. Thus, these data identify SETD2 as a crucial player in establishing the maternal epigenome that in turn controls embryonic development.


Asunto(s)
Desarrollo Embrionario/genética , Epigénesis Genética , Impresión Genómica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , ADN (Citosina-5-)-Metiltransferasas/deficiencia , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Femenino , Código de Histonas/genética , N-Metiltransferasa de Histona-Lisina/deficiencia , Histonas/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Genéticos , Oocitos/metabolismo , Oogénesis/genética , Embarazo
8.
Elife ; 82019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30938678

RESUMEN

Imprinted X-inactivation silences genes exclusively on the paternally-inherited X-chromosome and is a paradigm of transgenerational epigenetic inheritance in mammals. Here, we test the role of maternal vs. zygotic Polycomb repressive complex 2 (PRC2) protein EED in orchestrating imprinted X-inactivation in mouse embryos. In maternal-null (Eedm-/-) but not zygotic-null (Eed-/-) early embryos, the maternal X-chromosome ectopically induced Xist and underwent inactivation. Eedm-/- females subsequently stochastically silenced Xist from one of the two X-chromosomes and displayed random X-inactivation. This effect was exacerbated in embryos lacking both maternal and zygotic EED (Eedmz-/-), suggesting that zygotic EED can also contribute to the onset of imprinted X-inactivation. Xist expression dynamics in Eedm-/- embryos resemble that of early human embryos, which lack oocyte-derived maternal PRC2 and only undergo random X-inactivation. Thus, expression of PRC2 in the oocyte and transmission of the gene products to the embryo may dictate the occurrence of imprinted X-inactivation in mammals.


Asunto(s)
Ratones/embriología , Complejo Represivo Polycomb 2/metabolismo , Inactivación del Cromosoma X , Animales , Ratones Noqueados , Complejo Represivo Polycomb 2/deficiencia , ARN Largo no Codificante/metabolismo
9.
Mol Cell ; 73(3): 547-561.e6, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30735655

RESUMEN

Chromatin organization undergoes drastic reconfiguration during gametogenesis. However, the molecular reprogramming of three-dimensional chromatin structure in this process remains poorly understood for mammals, including primates. Here, we examined three-dimensional chromatin architecture during spermatogenesis in rhesus monkey using low-input Hi-C. Interestingly, we found that topologically associating domains (TADs) undergo dissolution and reestablishment in spermatogenesis. Strikingly, pachytene spermatocytes, where synapsis occurs, are strongly depleted for TADs despite their active transcription state but uniquely show highly refined local compartments that alternate between transcribing and non-transcribing regions (refined-A/B). Importantly, such chromatin organization is conserved in mouse, where it remains largely intact upon transcription inhibition. Instead, it is attenuated in mutant spermatocytes, where the synaptonemal complex failed to be established. Intriguingly, this is accompanied by the restoration of TADs, suggesting that the synaptonemal complex may restrict TADs and promote local compartments. Thus, these data revealed extensive reprogramming of higher-order meiotic chromatin architecture during mammalian gametogenesis.


Asunto(s)
Reprogramación Celular , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Meiosis , Espermatogénesis , Espermatozoides/metabolismo , Animales , Cromatina/química , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Células HCT116 , Humanos , Macaca mulatta , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Conformación de Ácido Nucleico , Fase Paquiteno , Conformación Proteica , Relación Estructura-Actividad , Factores de Tiempo , Transcripción Genética , Inactivación del Cromosoma X
10.
Cell Stem Cell ; 23(4): 487-500.e6, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30122476

RESUMEN

Mesenchymal niche cells instruct activity of tissue-resident stem and progenitor cell populations. Epithelial stem cells in hair follicles (HFs) have region-specific activity, which may arise from intrinsic cellular heterogeneity within mesenchymal dermal papilla (DP) cells. Here we show that expression of Hoxc genes is sufficient to reprogram mesenchymal DP cells and alter the regenerative potential of epithelial stem cells. Hoxc gene expression in adult skin dermis closely correlates with regional HF regeneration patterns. Disrupting the region-specific expression patterns of Hoxc genes, by either decreasing their epigenetic repression via Bmi1 loss or inducing ectopic interactions of the Hoxc locus with an active epigenetic region, leads to precocious HF regeneration. We further show that a single Hoxc gene is sufficient to activate dormant DP niches and promote regional HF regeneration through canonical Wnt signaling. Altogether, these results reveal that Hoxc genes bestow mesenchymal niches with tissue-level heterogeneity and plasticity.


Asunto(s)
Autorrenovación de las Células , Genes Homeobox/genética , Folículo Piloso/citología , Folículo Piloso/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Plasticidad de la Célula , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Vía de Señalización Wnt
11.
Genome Biol ; 19(1): 18, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29422066

RESUMEN

BACKGROUND: Previous studies showed that the majority of developmental genes are devoid of DNA methylation at promoters even when they are repressed. Such hypomethylated regions at developmental genes are unusually large and extend well beyond proximal promoters, forming DNA methylation valleys (DMVs) or DNA methylation canyons. However, it remains elusive how most developmental genes can evade DNA methylation regardless of their transcriptional states. RESULTS: We show that DMVs are hypomethylated in development and are highly conserved across vertebrates. Importantly, DMVs are hotspots of regulatory regions for key developmental genes and show low levels of deamination mutation rates. By analyzing a panel of DNA methylomes from mouse tissues, we identify a subset of DMVs that are dynamically methylated. These DMVs are strongly enriched for Polycomb-deposited H3K27me3 when the associated genes are silenced, and surprisingly show elevated DNA methylation upon gene activation. 4C-seq analyses indicates that Polycomb-bound DMVs form insulated and self-interacting chromatin domains. Further investigations show that DNA hypomethylation is better correlated with the binding of Polycomb than with H3K27me3. In support of a role of Polycomb in DMV hypomethylation, we observe aberrant methylation in DMVs in mouse embryonic stem cells deficient in the EED protein. Finally, we show that Polycomb regulates hypomethylation of DMVs likely through ten-eleven translocation (TET) proteins. CONCLUSIONS: We show that Polycomb promotes the hypomethylation of DMVs near key developmental genes. These data reveal a delicate interplay between histone modifiers and DNA methylation, which contributes to their division at distinct gene targets, allowing lineage-specifying genes to largely maintain DNA methylation-free at regulatory elements.


Asunto(s)
Metilación de ADN , Proteínas del Grupo Polycomb/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Genoma , Crecimiento y Desarrollo/genética , Factores de Transcripción/metabolismo , Vertebrados/genética
12.
Nat Genet ; 50(1): 96-105, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29203909

RESUMEN

In mammals, all somatic development originates from lineage segregation in early embryos. However, the dynamics of transcriptomes and epigenomes acting in concert with initial cell fate commitment remains poorly characterized. Here we report a comprehensive investigation of transcriptomes and base-resolution methylomes for early lineages in peri- and postimplantation mouse embryos. We found allele-specific and lineage-specific de novo methylation at CG and CH sites that led to differential methylation between embryonic and extraembryonic lineages at promoters of lineage regulators, gene bodies, and DNA-methylation valleys. By using Hi-C experiments to define chromatin architecture across the same developmental period, we demonstrated that both global demethylation and remethylation in early development correlate with chromatin compartments. Dynamic local methylation was evident during gastrulation, which enabled the identification of putative regulatory elements. Finally, we found that de novo methylation patterning does not strictly require implantation. These data reveal dynamic transcriptomes, DNA methylomes, and 3D chromatin landscapes during the earliest stages of mammalian lineage specification.


Asunto(s)
Metilación de ADN , Embrión de Mamíferos/metabolismo , Epigénesis Genética , Alelos , Animales , Blastocisto/metabolismo , Linaje de la Célula/genética , Cromatina/química , Implantación del Embrión , Endodermo/metabolismo , Gastrulación/genética , Estratos Germinativos/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Transcriptoma
13.
Nature ; 547(7662): 232-235, 2017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28703188

RESUMEN

In mammals, chromatin organization undergoes drastic reprogramming after fertilization. However, the three-dimensional structure of chromatin and its reprogramming in preimplantation development remain poorly understood. Here, by developing a low-input Hi-C (genome-wide chromosome conformation capture) approach, we examined the reprogramming of chromatin organization during early development in mice. We found that oocytes in metaphase II show homogeneous chromatin folding that lacks detectable topologically associating domains (TADs) and chromatin compartments. Strikingly, chromatin shows greatly diminished higher-order structure after fertilization. Unexpectedly, the subsequent establishment of chromatin organization is a prolonged process that extends through preimplantation development, as characterized by slow consolidation of TADs and segregation of chromatin compartments. The two sets of parental chromosomes are spatially separated from each other and display distinct compartmentalization in zygotes. Such allele separation and allelic compartmentalization can be found as late as the 8-cell stage. Finally, we show that chromatin compaction in preimplantation embryos can partially proceed in the absence of zygotic transcription and is a multi-level hierarchical process. Taken together, our data suggest that chromatin may exist in a markedly relaxed state after fertilization, followed by progressive maturation of higher-order chromatin architecture during early development.


Asunto(s)
Alelos , Ensamble y Desensamble de Cromatina/genética , Cromatina/química , Cromatina/genética , Cromosomas de los Mamíferos/química , Cromosomas de los Mamíferos/genética , Desarrollo Embrionario/genética , Animales , Blastocisto/metabolismo , Cromatina/metabolismo , Cromosomas de los Mamíferos/metabolismo , Femenino , Fertilización , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones , Transcripción Genética , Cigoto/metabolismo
14.
Cell ; 170(2): 367-381.e20, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28709003

RESUMEN

High-order chromatin structure plays important roles in gene expression regulation. Knowledge of the dynamics of 3D chromatin structures during mammalian embryo development remains limited. We report the 3D chromatin architecture of mouse gametes and early embryos using an optimized Hi-C method with low-cell samples. We find that mature oocytes at the metaphase II stage do not have topologically associated domains (TADs). In sperm, extra-long-range interactions (>4 Mb) and interchromosomal interactions occur frequently. The high-order structures of both the paternal and maternal genomes in zygotes and two-cell embryos are obscure but are gradually re-established through development. The establishment of the TAD structure requires DNA replication but not zygotic genome activation. Furthermore, unmethylated CpGs are enriched in A compartment, and methylation levels are decreased to a greater extent in A compartment than in B compartment in embryos. In summary, the global reprogramming of chromatin architecture occurs during early mammalian development.


Asunto(s)
Cromatina/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Animales , Cromatina/química , Islas de CpG , Metilación de ADN , Replicación del ADN , Embrión de Mamíferos/química , Epigénesis Genética , Femenino , Células Germinativas/metabolismo , Masculino , Metafase , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Oocitos/citología , Espermatozoides/metabolismo , Cigoto/metabolismo
15.
Mol Cell ; 63(6): 1066-79, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27635762

RESUMEN

Polycomb group proteins and the related histone modification H3K27me3 can maintain the silencing of key developmental regulators and provide cellular memory. However, how such an epigenetic state is reprogrammed and inherited between generations is poorly understood. Using an ultra-sensitive approach, STAR ChIP-seq, we investigated H3K27me3 across 14 developmental stages along mouse gametogenesis and early development. Interestingly, highly pervasive H3K27me3 is found in regions depleted of transcription and DNA methylation in oocytes. Unexpectedly, we observed extensive loss of promoter H3K27me3 at Hox and other developmental genes upon fertilization. This is accompanied by global erasure of sperm H3K27me3 but inheritance of distal H3K27me3 from oocytes. The resulting allele-specific H3K27me3 patterns persist to blastocysts before being converted to canonical forms in postimplantation embryos, where both H3K4me3/H3K27me3 bivalent promoter marks are restored at developmental genes. Together, these data revealed widespread resetting of epigenetic memory and striking plasticity of epigenome during gametogenesis and early development.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Oocitos/metabolismo , Proteínas del Grupo Polycomb/genética , Espermatozoides/metabolismo , Animales , Reprogramación Celular , Embrión de Mamíferos , Desarrollo Embrionario/genética , Femenino , Fertilización , Gametogénesis/genética , Histonas/metabolismo , Patrón de Herencia , Masculino , Ratones , Ratones Endogámicos C57BL , Oocitos/citología , Oocitos/crecimiento & desarrollo , Proteínas del Grupo Polycomb/metabolismo , Regiones Promotoras Genéticas , Espermatozoides/citología , Espermatozoides/crecimiento & desarrollo , Cigoto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...