Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
J Cell Biol ; 223(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38625077

RESUMEN

The centromere is a fundamental higher-order structure in chromosomes ensuring their faithful segregation upon cell division. Centromeric transcripts have been described in several species and suggested to participate in centromere function. However, low sequence conservation of centromeric repeats appears inconsistent with a role in recruiting highly conserved centromeric proteins. Here, we hypothesized that centromeric transcripts may function through a secondary structure rather than sequence conservation. Using mouse embryonic stem cells (ESCs), we show that an imbalance in the levels of forward or reverse minor satellite (MinSat) transcripts leads to severe chromosome segregation defects. We further show that MinSat RNA adopts a stem-loop secondary structure, which is conserved in human α-satellite transcripts. We identify an RNA binding region in CENPC and demonstrate that MinSat transcripts function through the structured region of the RNA. Importantly, mutants that disrupt MinSat secondary structure do not cause segregation defects. We propose that the conserved role of centromeric transcripts relies on their secondary RNA structure.


Asunto(s)
Segregación Cromosómica , Satélite de ARN , Animales , Humanos , Ratones , División Celular , Células Madre Embrionarias de Ratones , Satélite de ARN/química , Satélite de ARN/metabolismo , Centrómero/metabolismo
2.
EMBO Rep ; 25(4): 1721-1733, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38528171

RESUMEN

Remnants of transposable elements (TEs) are widely expressed throughout mammalian embryo development. Originally infesting our genomes as selfish elements and acting as a source of genome instability, several of these elements have been co-opted as part of a complex system of genome regulation. Many TEs have lost transposition ability and their transcriptional potential has been tampered as a result of interactions with the host throughout evolutionary time. It has been proposed that TEs have been ultimately repurposed to function as gene regulatory hubs scattered throughout our genomes. In the early embryo in particular, TEs find a perfect environment of naïve chromatin to escape transcriptional repression by the host. As a consequence, it is thought that hosts found ways to co-opt TE sequences to regulate large-scale changes in chromatin and transcription state of their genomes. In this review, we discuss several examples of TEs expressed during embryo development, their potential for co-option in genome regulation and the evolutionary pressures on TEs and on our genomes.


Asunto(s)
Elementos Transponibles de ADN , Regulación de la Expresión Génica , Animales , Elementos Transponibles de ADN/genética , Evolución Biológica , Cromatina/genética , Embrión de Mamíferos , Evolución Molecular , Mamíferos/genética
3.
Nature ; 625(7994): 401-409, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38123678

RESUMEN

DNA replication enables genetic inheritance across the kingdoms of life. Replication occurs with a defined temporal order known as the replication timing (RT) programme, leading to organization of the genome into early- or late-replicating regions. RT is cell-type specific, is tightly linked to the three-dimensional nuclear organization of the genome1,2 and is considered an epigenetic fingerprint3. In spite of its importance in maintaining the epigenome4, the developmental regulation of RT in mammals in vivo has not been explored. Here, using single-cell Repli-seq5, we generated genome-wide RT maps of mouse embryos from the zygote to the blastocyst stage. Our data show that RT is initially not well defined but becomes defined progressively from the 4-cell stage, coinciding with strengthening of the A and B compartments. We show that transcription contributes to the precision of the RT programme and that the difference in RT between the A and B compartments depends on RNA polymerase II at zygotic genome activation. Our data indicate that the establishment of nuclear organization precedes the acquisition of defined RT features and primes the partitioning of the genome into early- and late-replicating domains. Our work sheds light on the establishment of the epigenome at the beginning of mammalian development and reveals the organizing principles of genome organization.


Asunto(s)
Momento de Replicación del ADN , Embrión de Mamíferos , Genoma , Animales , Ratones , Blastocisto/citología , Blastocisto/metabolismo , Cromatina/genética , Epigenoma/genética , Genoma/genética , ARN Polimerasa II/metabolismo , Cigoto/citología , Cigoto/crecimiento & desarrollo , Cigoto/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo
4.
Genes Dev ; 37(19-20): 901-912, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37914351

RESUMEN

Fertilization in mammals is accompanied by an intense period of chromatin remodeling and major changes in nuclear organization. How the earliest events in embryogenesis, including zygotic genome activation (ZGA) during maternal-to-zygotic transition, influence such remodeling remains unknown. Here, we have investigated the establishment of nuclear architecture, focusing on the remodeling of lamina-associated domains (LADs) during this transition. We report that LADs reorganize gradually in two-cell embryos and that blocking ZGA leads to major changes in nuclear organization, including altered chromatin and genomic features of LADs and redistribution of H3K4me3 toward the nuclear lamina. Our data indicate that the rearrangement of LADs is an integral component of the maternal-to-zygotic transition and that transcription contributes to shaping nuclear organization at the beginning of mammalian development.


Asunto(s)
ARN Polimerasa II , Transcripción Genética , Animales , Ratones , ARN Polimerasa II/genética , Desarrollo Embrionario/genética , Cigoto , Mamíferos/genética , Regulación del Desarrollo de la Expresión Génica , Cromatina
5.
EMBO Rep ; 24(9): e56194, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37432066

RESUMEN

Mouse embryonic stem cells (ESCs) display pluripotency features characteristic of the inner cell mass of the blastocyst. Mouse embryonic stem cell cultures are highly heterogeneous and include a rare population of cells, which recapitulate characteristics of the 2-cell embryo, referred to as 2-cell-like cells (2CLCs). Whether and how ESC and 2CLC respond to environmental cues has not been fully elucidated. Here, we investigate the impact of mechanical stress on the reprogramming of ESC to 2CLC. We show that hyperosmotic stress induces 2CLC and that this induction can occur even after a recovery time from hyperosmotic stress, suggesting a memory response. Hyperosmotic stress in ESCs leads to accumulation of reactive-oxygen species (ROS) and ATR checkpoint activation. Importantly, preventing either elevated ROS levels or ATR activation impairs hyperosmotic-mediated 2CLC induction. We further show that ROS generation and the ATR checkpoint act within the same molecular pathway in response to hyperosmotic stress to induce 2CLCs. Altogether, these results shed light on the response of ESC to mechanical stress and on our understanding of 2CLC reprogramming.


Asunto(s)
Células Madre Embrionarias , Transducción de Señal , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Blastocisto/metabolismo , Diferenciación Celular
6.
Curr Opin Genet Dev ; 81: 102083, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37421903

RESUMEN

In mammals, cells acquire totipotency at fertilization. Embryonic genome activation (EGA), which occurs at the 2-cell stage in the mouse and 4- to 8-cell stage in humans, occurs during the time window at which embryonic cells are totipotent and thus it is thought that EGA is mechanistically linked to the foundations of totipotency. The molecular mechanisms that lead to the establishment of totipotency and EGA had been elusive for a long time, however, recent advances have been achieved with the establishment of new cell lines with greater developmental potential and the application of novel low-input high-throughput techniques in embryos. These have unveiled several principles of totipotency related to its epigenetic makeup but also to characteristic features of totipotent cells. In this review, we summarize and discuss current views exploring some of the key drivers of totipotency from both in vitro cell culture models and embryogenesis in vivo.


Asunto(s)
Estructuras Embrionarias , Regulación del Desarrollo de la Expresión Génica , Humanos , Animales , Mamíferos , Genoma , Replicación del ADN
7.
Development ; 150(11)2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37294170

RESUMEN

A powerful feature of single-cell genomics is the possibility of identifying cell types from their molecular profiles. In particular, identifying novel rare cell types and their marker genes is a key potential of single-cell RNA sequencing. Standard clustering approaches perform well in identifying relatively abundant cell types, but tend to miss rarer cell types. Here, we have developed CIARA (Cluster Independent Algorithm for the identification of markers of RAre cell types), a cluster-independent computational tool designed to select genes that are likely to be markers of rare cell types. Genes selected by CIARA are subsequently integrated with common clustering algorithms to single out groups of rare cell types. CIARA outperforms existing methods for rare cell type detection, and we use it to find previously uncharacterized rare populations of cells in a human gastrula and among mouse embryonic stem cells treated with retinoic acid. Moreover, CIARA can be applied more generally to any type of single-cell omic data, thus allowing the identification of rare cells across multiple data modalities. We provide implementations of CIARA in user-friendly packages available in R and Python.


Asunto(s)
Algoritmos , Análisis de la Célula Individual , Animales , Humanos , Ratones , Análisis de Secuencia de ARN/métodos , Análisis por Conglomerados , Análisis de la Célula Individual/métodos , Perfilación de la Expresión Génica/métodos
8.
Cell Rep ; 42(4): 112380, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37061916

RESUMEN

Recent advances in synthetic embryology have opened new avenues for understanding the complex events controlling mammalian peri-implantation development. Here, we show that mouse embryonic stem cells (ESCs) solely exposed to chemical inhibition of SUMOylation generate embryo-like structures comprising anterior neural and trunk-associated regions. HypoSUMOylation-instructed ESCs give rise to spheroids that self-organize into gastrulating structures containing cell types spatially and functionally related to embryonic and extraembryonic compartments. Alternatively, spheroids cultured in a droplet microfluidic device form elongated structures that undergo axial organization reminiscent of natural embryo morphogenesis. Single-cell transcriptomics reveals various cellular lineages, including properly positioned anterior neuronal cell types and paraxial mesoderm segmented into somite-like structures. Transient SUMOylation suppression gradually increases DNA methylation genome wide and repressive mark deposition at Nanog. Interestingly, cell-to-cell variations in SUMOylation levels occur during early embryogenesis. Our approach provides a proof of principle for potentially powerful strategies to explore early embryogenesis by targeting chromatin roadblocks of cell fate change.


Asunto(s)
Embrión de Mamíferos , Sumoilación , Animales , Ratones , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Desarrollo Embrionario , Diferenciación Celular/fisiología , Mamíferos
9.
Genes Dev ; 37(1-2): 56-57, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37061962
10.
Genes Dev ; 37(7-8): 336-350, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-37072228

RESUMEN

The majority of our genome is composed of repeated DNA sequences that assemble into heterochromatin, a highly compacted structure that constrains their mutational potential. How heterochromatin forms during development and how its structure is maintained are not fully understood. Here, we show that mouse heterochromatin phase-separates after fertilization, during the earliest stages of mammalian embryogenesis. Using high-resolution quantitative imaging and molecular biology approaches, we show that pericentromeric heterochromatin displays properties consistent with a liquid-like state at the two-cell stage, which change at the four-cell stage, when chromocenters mature and heterochromatin becomes silent. Disrupting the condensates results in altered transcript levels of pericentromeric heterochromatin, suggesting a functional role for phase separation in heterochromatin function. Thus, our work shows that mouse heterochromatin forms membrane-less compartments with biophysical properties that change during development and provides new insights into the self-organization of chromatin domains during mammalian embryogenesis.


Asunto(s)
Cromatina , Heterocromatina , Animales , Ratones , Embrión de Mamíferos , Genoma , Mamíferos/genética
12.
Dev Cell ; 58(6): 435-449, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36977375

RESUMEN

Nuclear organization has emerged as a potential key regulator of genome function. During development, the deployment of transcriptional programs must be tightly coordinated with cell division and is often accompanied by major changes in the repertoire of expressed genes. These transcriptional and developmental events are paralleled by changes in the chromatin landscape. Numerous studies have revealed the dynamics of nuclear organization underlying them. In addition, advances in live-imaging-based methodologies enable the study of nuclear organization with high spatial and temporal resolution. In this Review, we summarize the current knowledge of the changes in nuclear architecture in the early embryogenesis of various model systems. Furthermore, to highlight the importance of integrating fixed-cell and live approaches, we discuss how different live-imaging techniques can be applied to examine nuclear processes and their contribution to our understanding of transcription and chromatin dynamics in early development. Finally, we provide future avenues for outstanding questions in this field.


Asunto(s)
Núcleo Celular , Cromatina , Desarrollo Embrionario/genética , Genoma
13.
Cell Rep ; 41(13): 111865, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36577375

RESUMEN

How transcription is regulated as development commences is fundamental to understand how the transcriptionally silent mature gametes are reprogrammed. The embryonic genome is activated for the first time during zygotic genome activation (ZGA). How RNA polymerase II (Pol II) and productive elongation are regulated during this process remains elusive. Here, we generate genome-wide maps of Serine 5 and Serine 2-phosphorylated Pol II during and after ZGA in mouse embryos. We find that both phosphorylated Pol II forms display similar distributions across genes during ZGA, with typical elongation enrichment of Pol II emerging after ZGA. Serine 2-phosphorylated Pol II occurs at genes prior to their activation, suggesting that Serine 2 phosphorylation may prime gene expression. Functional perturbations demonstrate that CDK9 and SPT5 are major ZGA regulators and that SPT5 prevents precocious activation of some genes. Overall, our work sheds molecular insights into transcriptional regulation at the beginning of mammalian development.


Asunto(s)
ARN Polimerasa II , Cigoto , Ratones , Animales , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Cigoto/metabolismo , Fosforilación , Genoma , Serina/metabolismo , Activación Transcripcional , Regulación del Desarrollo de la Expresión Génica , Mamíferos/metabolismo
14.
Nat Genet ; 54(3): 318-327, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35256805

RESUMEN

Totipotency emerges in early embryogenesis, but its molecular underpinnings remain poorly characterized. In the present study, we employed DNA fiber analysis to investigate how pluripotent stem cells are reprogrammed into totipotent-like 2-cell-like cells (2CLCs). We show that totipotent cells of the early mouse embryo have slow DNA replication fork speed and that 2CLCs recapitulate this feature, suggesting that fork speed underlies the transition to a totipotent-like state. 2CLCs emerge concomitant with DNA replication and display changes in replication timing (RT), particularly during the early S-phase. RT changes occur prior to 2CLC emergence, suggesting that RT may predispose to gene expression changes and consequent reprogramming of cell fate. Slowing down replication fork speed experimentally induces 2CLCs. In vivo, slowing fork speed improves the reprogramming efficiency of somatic cell nuclear transfer. Our data suggest that fork speed regulates cellular plasticity and that remodeling of replication features leads to changes in cell fate and reprogramming.


Asunto(s)
Embrión de Mamíferos , Células Madre Pluripotentes , Animales , Diferenciación Celular/genética , Reprogramación Celular/genética , Replicación del ADN/genética , Desarrollo Embrionario/genética , Ratones
17.
Nat Cell Biol ; 23(8): 814-816, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34354235
18.
Genes Dev ; 35(15-16): 1142-1160, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34244292

RESUMEN

The establishment of cell fates involves alterations of transcription factor repertoires and repurposing of transcription factors by post-translational modifications. In embryonic stem cells (ESCs), the chromatin organizers SATB2 and SATB1 balance pluripotency and differentiation by activating and repressing pluripotency genes, respectively. Here, we show that conditional Satb2 gene inactivation weakens ESC pluripotency, and we identify SUMO2 modification of SATB2 by the E3 ligase ZFP451 as a potential driver of ESC differentiation. Mutations of two SUMO-acceptor lysines of Satb2 (Satb2K →R ) or knockout of Zfp451 impair the ability of ESCs to silence pluripotency genes and activate differentiation-associated genes in response to retinoic acid (RA) treatment. Notably, the forced expression of a SUMO2-SATB2 fusion protein in either Satb2K →R or Zfp451-/- ESCs rescues, in part, their impaired differentiation potential and enhances the down-regulation of Nanog The differentiation defect of Satb2K →R ESCs correlates with altered higher-order chromatin interactions relative to Satb2wt ESCs. Upon RA treatment of Satb2wt ESCs, SATB2 interacts with ZFP451 and the LSD1/CoREST complex and gains binding at differentiation genes, which is not observed in RA-treated Satb2K →R cells. Thus, SATB2 SUMOylation may contribute to the rewiring of transcriptional networks and the chromatin interactome of ESCs in the transition of pluripotency to differentiation.


Asunto(s)
Células Madre Embrionarias , Sumoilación , Ubiquitina-Proteína Ligasas/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Nat Struct Mol Biol ; 28(6): 521-532, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34045724

RESUMEN

Totipotent cells hold enormous potential for regenerative medicine. Thus, the development of cellular models recapitulating totipotent-like features is of paramount importance. Cells resembling the totipotent cells of early embryos arise spontaneously in mouse embryonic stem (ES) cell cultures. Such '2-cell-like-cells' (2CLCs) recapitulate 2-cell-stage features and display expanded cell potential. Here, we used 2CLCs to perform a small-molecule screen to identify new pathways regulating the 2-cell-stage program. We identified retinoids as robust inducers of 2CLCs and the retinoic acid (RA)-signaling pathway as a key component of the regulatory circuitry of totipotent cells in embryos. Using single-cell RNA-seq, we reveal the transcriptional dynamics of 2CLC reprogramming and show that ES cells undergo distinct cellular trajectories in response to RA. Importantly, endogenous RA activity in early embryos is essential for zygotic genome activation and developmental progression. Overall, our data shed light on the gene regulatory networks controlling cellular plasticity and the totipotency program.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre Totipotentes/citología , Tretinoina/fisiología , Acitretina/farmacología , Animales , Masa Celular Interna del Blastocisto/citología , Diferenciación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Femenino , Redes Reguladoras de Genes/genética , Genes Reporteros , Isotretinoína/farmacología , Masculino , Ratones/embriología , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Piperazinas/farmacología , Pirazoles/farmacología , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , RNA-Seq , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/fisiología , Transducción de Señal/efectos de los fármacos , Células Madre Totipotentes/efectos de los fármacos , Transcripción Genética , Tretinoina/antagonistas & inhibidores , Tretinoina/farmacología , Receptor de Ácido Retinoico gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...