Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 41(12): e109457, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35603814

RESUMO

The mammalian germline is characterized by extensive epigenetic reprogramming during its development into functional eggs and sperm. Specifically, the epigenome requires resetting before parental marks can be established and transmitted to the next generation. In the female germline, X-chromosome inactivation and reactivation are among the most prominent epigenetic reprogramming events, yet very little is known about their kinetics and biological function. Here, we investigate X-inactivation and reactivation dynamics using a tailor-made in vitro system of primordial germ cell-like cell (PGCLC) differentiation from mouse embryonic stem cells. We find that X-inactivation in PGCLCs in vitro and in germ cell-competent epiblast cells in vivo is moderate compared to somatic cells, and frequently characterized by escaping genes. X-inactivation is followed by step-wise X-reactivation, which is mostly completed during meiotic prophase I. Furthermore, we find that PGCLCs which fail to undergo X-inactivation or reactivate too rapidly display impaired meiotic potential. Thus, our data reveal fine-tuned X-chromosome remodelling as a critical feature of female germ cell development towards meiosis and oogenesis.


Assuntos
Células Germinativas , Meiose , Animais , Diferenciação Celular , Cromossomos , Mamíferos/genética , Meiose/genética , Camundongos , Inativação do Cromossomo X/genética
2.
Proc Natl Acad Sci U S A ; 120(4): e2213810120, 2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36669113

RESUMO

Reactivation of the inactive X chromosome is a hallmark epigenetic event during reprogramming of mouse female somatic cells to induced pluripotent stem cells (iPSCs). This involves global structural remodeling from a condensed, heterochromatic into an open, euchromatic state, thereby changing a transcriptionally inactive into an active chromosome. Despite recent advances, very little is currently known about the molecular players mediating this process and how this relates to iPSC-reprogramming in general. To gain more insight, here we perform a RNAi-based knockdown screen during iPSC-reprogramming of mouse fibroblasts. We discover factors important for X chromosome reactivation (XCR) and iPSC-reprogramming. Among those, we identify the cohesin complex member SMC1a as a key molecule with a specific function in XCR, as its knockdown greatly affects XCR without interfering with iPSC-reprogramming. Using super-resolution microscopy, we find SMC1a to be preferentially enriched on the active compared with the inactive X chromosome and that SMC1a is critical for the decompacted state of the active X. Specifically, depletion of SMC1a leads to contraction of the active X both in differentiated and in pluripotent cells, where it normally is in its most open state. In summary, we reveal cohesin as a key factor for remodeling of the X chromosome from an inactive to an active structure and that this is a critical step for XCR during iPSC-reprogramming.


Assuntos
Células-Tronco Pluripotentes Induzidas , Feminino , Animais , Camundongos , Reprogramação Celular , Inativação do Cromossomo X/genética , Cromossomo X/genética , Estruturas Cromossômicas , Coesinas
3.
Mol Cell ; 64(4): 645-658, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863225

RESUMO

The cellular plasticity of pluripotent stem cells is thought to be sustained by genomic regions that display both active and repressive chromatin properties. These regions exhibit low levels of gene expression, yet the mechanisms controlling these levels remain unknown. Here, we describe Elongin BC as a binding factor at the promoters of bivalent sites. Biochemical and genome-wide analyses show that Elongin BC is associated with Polycomb Repressive Complex 2 (PRC2) in pluripotent stem cells. Elongin BC is recruited to chromatin by the PRC2-associated factor EPOP (Elongin BC and Polycomb Repressive Complex 2 Associated Protein, also termed C17orf96, esPRC2p48, E130012A19Rik), a protein expressed in the inner cell mass of the mouse blastocyst. Both EPOP and Elongin BC are required to maintain low levels of expression at PRC2 genomic targets. Our results indicate that keeping the balance between activating and repressive cues is a more general feature of chromatin in pluripotent stem cells than previously appreciated.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 2/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Cromatina/química , Cromatina/metabolismo , Proteínas Cromossômicas não Histona , DNA Polimerase II/genética , DNA Polimerase II/metabolismo , Elonguina , Implantação do Embrião , Embrião de Mamíferos , Histonas/genética , Histonas/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Pluripotentes/citologia , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/metabolismo , Transcrição Gênica
4.
Bioessays ; 44(10): e2200105, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36028473

RESUMO

The spatial organization of genomes is becoming increasingly understood. In mammals, where it is most investigated, this organization ties in with transcription, so an important research objective is to understand whether gene activity is a cause or a consequence of genome folding in space. In this regard, the phenomena of X-chromosome inactivation and reactivation open a unique window of investigation because of the singularities of the inactive X chromosome. Here we focus on the cause-consequence nexus between genome conformation and transcription and explain how recent results about the structural changes associated with inactivation and reactivation of the X chromosome shed light on this problem.


Assuntos
Inativação do Cromossomo X , Cromossomo X , Animais , Genoma/genética , Mamíferos/genética , Inativação do Cromossomo X/genética
5.
Biochem J ; 480(8): 521-537, 2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37096944

RESUMO

In female mammals, the two X chromosomes are subject to epigenetic gene regulation in order to balance X-linked gene dosage with autosomes and in relation to males, which have one X and one Y chromosome. This is achieved by an intricate interplay of several processes; X-chromosome inactivation and reactivation elicit global epigenetic regulation of expression from one X chromosome in a stage-specific manner, whilst the process of X-chromosome upregulation responds to this by fine-tuning transcription levels of the second X. The germline is unique in its function of transmitting both the genetic and epigenetic information from one generation to the next, and remodelling of the X chromosome is one of the key steps in setting the stage for successful development. Here, we provide an overview of the complex dynamics of X-chromosome dosage control during embryonic and germ cell development, and aim to decipher its potential role for normal germline competency.


Assuntos
Mecanismo Genético de Compensação de Dose , Epigênese Genética , Masculino , Animais , Feminino , Cromossomo X , Células Germinativas/metabolismo , Mamíferos/genética
6.
PLoS Comput Biol ; 17(11): e1009582, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34762642

RESUMO

Isogenic cells cultured together show heterogeneity in their proliferation rate. To determine the differences between fast and slow-proliferating cells, we developed a method to sort cells by proliferation rate, and performed RNA-seq on slow and fast proliferating subpopulations of pluripotent mouse embryonic stem cells (mESCs) and mouse fibroblasts. We found that slowly proliferating mESCs have a more naïve pluripotent character. We identified an evolutionarily conserved proliferation-correlated transcriptomic signature that is common to all eukaryotes: fast cells have higher expression of genes for protein synthesis and protein degradation. This signature accurately predicted growth rate in yeast and cancer cells, and identified lineage-specific proliferation dynamics during development, using C. elegans scRNA-seq data. In contrast, sorting by mitochondria membrane potential revealed a highly cell-type specific mitochondria-state related transcriptome. mESCs with hyperpolarized mitochondria are fast proliferating, while the opposite is true for fibroblasts. The mitochondrial electron transport chain inhibitor antimycin affected slow and fast subpopulations differently. While a major transcriptional-signature associated with cell-to-cell heterogeneity in proliferation is conserved, the metabolic and energetic dependency of cell proliferation is cell-type specific.


Assuntos
Linhagem da Célula , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Proliferação de Células , Regulação da Expressão Gênica no Desenvolvimento , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Análise de Sequência de RNA/métodos , Transcriptoma
7.
Mol Cell ; 52(6): 805-18, 2013 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-24268575

RESUMO

Transitions between pluripotent and differentiated states are marked by dramatic epigenetic changes. Cellular differentiation is tightly linked to X chromosome inactivation (XCI), whereas reprogramming to induced pluripotent stem cells (iPSCs) is associated with X chromosome reactivation (XCR). XCR reverses the silent state of the inactive X, occurring in mouse blastocysts and germ cells. In spite of its importance, little is known about underlying mechanisms. Here, we examine the role of the long noncoding Tsix RNA and the germline factor, PRDM14. In blastocysts, XCR is perturbed by mutation of either Tsix or Prdm14. In iPSCs, XCR is disrupted only by PRDM14 deficiency, which also affects iPSC derivation and maintenance. We show that Tsix and PRDM14 directly link XCR to pluripotency: first, PRDM14 represses Rnf12 by recruiting polycomb repressive complex 2; second, Tsix enables PRDM14 to bind Xist. Thus, our study provides functional and mechanistic links between cellular and X chromosome reprogramming.


Assuntos
Blastocisto/metabolismo , Reprogramação Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo , Inativação do Cromossomo X , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Proteínas de Ligação a DNA , Implantação do Embrião , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transfecção , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(7): 1619-1624, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28143937

RESUMO

Rett syndrome (RS) is a debilitating neurological disorder affecting mostly girls with heterozygous mutations in the gene encoding the methyl-CpG-binding protein MeCP2 on the X chromosome. Because restoration of MeCP2 expression in a mouse model reverses neurologic deficits in adult animals, reactivation of the wild-type copy of MeCP2 on the inactive X chromosome (Xi) presents a therapeutic opportunity in RS. To identify genes involved in MeCP2 silencing, we screened a library of 60,000 shRNAs using a cell line with a MeCP2 reporter on the Xi and found 30 genes clustered in seven functional groups. More than half encoded proteins with known enzymatic activity, and six were members of the bone morphogenetic protein (BMP)/TGF-ß pathway. shRNAs directed against each of these six genes down-regulated X-inactive specific transcript (XIST), a key player in X-chromosome inactivation that encodes an RNA that coats the silent X chromosome, and modulation of regulators of this pathway both in cell culture and in mice demonstrated robust regulation of XIST. Moreover, we show that Rnf12, an X-encoded ubiquitin ligase important for initiation of X-chromosome inactivation and XIST transcription in ES cells, also plays a role in maintenance of the inactive state through regulation of BMP/TGF-ß signaling. Our results identify pharmacologically suitable targets for reactivation of MeCP2 on the Xi and a genetic circuitry that maintains XIST expression and X-chromosome inactivation in differentiated cells.


Assuntos
Proteína Morfogenética Óssea 2/genética , Proteína 2 de Ligação a Metil-CpG/genética , RNA Longo não Codificante/genética , Fator de Crescimento Transformador beta/genética , Inativação do Cromossomo X , Animais , Linhagem Celular , Feminino , Perfilação da Expressão Gênica , Biblioteca Gênica , Humanos , Camundongos , RNA Interferente Pequeno/genética , Síndrome de Rett/genética , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases/genética
9.
Proc Natl Acad Sci U S A ; 113(50): 14366-14371, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-28182563

RESUMO

X-chromosome inactivation is a mechanism of dosage compensation in which one of the two X chromosomes in female mammals is transcriptionally silenced. Once established, silencing of the inactive X (Xi) is robust and difficult to reverse pharmacologically. However, the Xi is a reservoir of >1,000 functional genes that could be potentially tapped to treat X-linked disease. To identify compounds that could reactivate the Xi, here we screened ∼367,000 small molecules in an automated high-content screen using an Xi-linked GFP reporter in mouse fibroblasts. Given the robust nature of silencing, we sensitized the screen by "priming" cells with the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5azadC). Compounds that elicited GFP activity include VX680, MLN8237, and 5azadC, which are known to target the Aurora kinase and DNA methylation pathways. We demonstrate that the combinations of VX680 and 5azadC, as well as MLN8237 and 5azadC, synergistically up-regulate genes on the Xi. Thus, our work identifies a synergism between the DNA methylation and Aurora kinase pathways as being one of interest for possible pharmacological reactivation of the Xi.


Assuntos
Aurora Quinases/antagonistas & inibidores , Metilação de DNA/efeitos dos fármacos , Inativação do Cromossomo X/efeitos dos fármacos , Animais , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase B/genética , Aurora Quinases/genética , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Azepinas/administração & dosagem , Linhagem Celular , Decitabina , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Técnicas de Silenciamento de Genes , Genes Ligados ao Cromossomo X , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Ensaios de Triagem em Larga Escala , Camundongos , Camundongos Transgênicos , Piperazinas/administração & dosagem , Pirimidinas/administração & dosagem , Cromossomo X/efeitos dos fármacos , Cromossomo X/genética
10.
Semin Cell Dev Biol ; 56: 88-99, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27112543

RESUMO

With the emergence of sex-determination by sex chromosomes, which differ in composition and number between males and females, appeared the need to equalize X-chromosomal gene dosage between the sexes. Mammals have devised the strategy of X-chromosome inactivation (XCI), in which one of the two X-chromosomes is rendered transcriptionally silent in females. In the mouse, the best-studied model organism with respect to XCI, this inactivation process occurs in different forms, imprinted and random, interspersed by periods of X-chromosome reactivation (XCR), which is needed to switch between the different modes of XCI. In this review, I describe the recent advances with respect to the developmental control of XCI and XCR and in particular their link to differentiation and pluripotency. Furthermore, I review the mechanisms, which influence the timing and choice, with which one of the two X-chromosomes is chosen for inactivation during random XCI. This has an impact on how females are mosaics with regard to which X-chromosome is active in different cells, which has implications on the severity of diseases caused by X-linked mutations.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Inativação do Cromossomo X/genética , Animais , Diferenciação Celular/genética , Impressão Genômica , Humanos , Mosaicismo
11.
Proc Natl Acad Sci U S A ; 112(47): 14415-22, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26489649

RESUMO

The long noncoding X-inactivation-specific transcript (Xist gene) is responsible for mammalian X-chromosome dosage compensation between the sexes, the process by which one of the two X chromosomes is inactivated in the female soma. Xist is essential for both the random and imprinted forms of X-chromosome inactivation. In the imprinted form, Xist is paternally marked to be expressed in female embryos. To investigate the mechanism of Xist imprinting, we introduce Xist transgenes (Tg) into the male germ line. Although ectopic high-level Xist expression on autosomes can be compatible with viability, transgenic animals demonstrate reduced fitness, subfertility, defective meiotic pairing, and other germ-cell abnormalities. In the progeny, paternal-specific expression is recapitulated by the 200-kb Xist Tg. However, Xist imprinting occurs efficiently only when it is in an unpaired or unpartnered state during male meiosis. When transmitted from a hemizygous father (+/Tg), the Xist Tg demonstrates paternal-specific expression in the early embryo. When transmitted by a homozygous father (Tg/Tg), the Tg fails to show imprinted expression. Thus, Xist imprinting is directed by sequences within a 200-kb X-linked region, and the hemizygous (unpaired) state of the Xist region promotes its imprinting in the male germ line.


Assuntos
Impressão Genômica , Células Germinativas/metabolismo , RNA Longo não Codificante/genética , Animais , Blastocisto/metabolismo , Epigênese Genética , Feminino , Hemizigoto , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Masculino , Camundongos Transgênicos , Fenótipo , RNA Longo não Codificante/síntese química , RNA Longo não Codificante/metabolismo , Transgenes
12.
RNA Biol ; 11(7): 798-807, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25137047

RESUMO

X-chromosome inactivation (XCI) in female mammals is a dramatic example of epigenetic gene regulation, which entails the silencing of an entire chromosome through a wide range of mechanisms involving noncoding RNAs, chromatin-modifications, and DNA-methylation. While XCI is associated with the differentiated cell state, it is reversed by X-chromosome reactivation (XCR) ex vivo in pluripotent stem cells and in vivo in the early mouse embryo and the germline. Critical in the regulation of XCI vs. XCR is the X-inactivation center, a multigene locus on the X-chromosome harboring several long noncoding RNA genes including, most prominently, Xist and Tsix. These genes, which sit at the top of the XCI hierarchy, are by themselves controlled by pluripotency factors, coupling XCR with the naïve pluripotent stem cell state. In this point-of-view article we review the latest findings regarding this intricate relationship between cell differentiation state and epigenetic control of the X-chromosome. In particular, we discuss the emerging picture of complex multifactorial regulatory mechanisms, ensuring both a fine-tuned and robust X-reactivation process.


Assuntos
Células-Tronco Pluripotentes/metabolismo , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo , Inativação do Cromossomo X , Animais , Diferenciação Celular , Feminino , Humanos , Camundongos , Cromossomo X/genética
13.
Hum Genet ; 130(2): 265-80, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21667284

RESUMO

X-chromosome inactivation is an epigenetic hallmark of mammalian development. Chromosome-wide regulation of the X-chromosome is essential in embryonic and germ cell development. In the male germline, the X-chromosome goes through meiotic sex chromosome inactivation, and the chromosome-wide silencing is maintained from meiosis into spermatids before the transmission to female embryos. In early female mouse embryos, X-inactivation is imprinted to occur on the paternal X-chromosome, representing the epigenetic programs acquired in both parental germlines. Recent advances revealed that the inactive X-chromosome in both females and males can be dissected into two elements: repeat elements versus unique coding genes. The inactive paternal X in female preimplantation embryos is reactivated in the inner cell mass of blastocysts in order to subsequently allow the random form of X-inactivation in the female embryo, by which both Xs have an equal chance of being inactivated. X-chromosome reactivation is regulated by pluripotency factors and also occurs in early female germ cells and in pluripotent stem cells, where X-reactivation is a stringent marker of naive ground state pluripotency. Here we summarize recent progress in the study of X-inactivation and X-reactivation during mammalian reproduction and development as well as in pluripotent stem cells.


Assuntos
Epigênese Genética/fisiologia , Impressão Genômica/fisiologia , Células Germinativas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Inativação do Cromossomo X/fisiologia , Cromossomo X/fisiologia , Animais , Epigênese Genética/genética , Feminino , Humanos , Masculino , Mamíferos , Modelos Biológicos , RNA Longo não Codificante , RNA não Traduzido/fisiologia , Especificidade da Espécie , Cromossomo X/genética , Inativação do Cromossomo X/genética
14.
Nature ; 436(7048): 207-13, 2005 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-15937476

RESUMO

Germ cell fate in mice is induced in pluripotent epiblast cells in response to signals from extraembryonic tissues. The specification of approximately 40 founder primordial germ cells and their segregation from somatic neighbours are important events in early development. We have proposed that a critical event during this specification includes repression of a somatic programme that is adopted by neighbouring cells. Here we show that Blimp1 (also known as Prdm1), a known transcriptional repressor, has a critical role in the foundation of the mouse germ cell lineage, as its disruption causes a block early in the process of primordial germ cell formation. Blimp1-deficient mutant embryos form a tight cluster of about 20 primordial germ cell-like cells, which fail to show the characteristic migration, proliferation and consistent repression of homeobox genes that normally accompany specification of primordial germ cells. Furthermore, our genetic lineage-tracing experiments indicate that the Blimp1-positive cells originating from the proximal posterior epiblast cells are indeed the lineage-restricted primordial germ cell precursors.


Assuntos
Linhagem da Célula , Células Germinativas/citologia , Células Germinativas/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Gástrula/citologia , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Camundongos , Mutação/genética , Fenótipo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Repressoras/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
15.
Nat Commun ; 12(1): 3499, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34108480

RESUMO

A hallmark of chromosome organization is the partition into transcriptionally active A and repressed B compartments, and into topologically associating domains (TADs). Both structures were regarded to be absent from the inactive mouse X chromosome, but to be re-established with transcriptional reactivation and chromatin opening during X-reactivation. Here, we combine a tailor-made mouse iPSC reprogramming system and high-resolution Hi-C to produce a time course combining gene reactivation, chromatin opening and chromosome topology during X-reactivation. Contrary to previous observations, we observe A/B-like compartments on the inactive X harbouring multiple subcompartments. While partial X-reactivation initiates within a compartment rich in X-inactivation escapees, it then occurs rapidly along the chromosome, concomitant with downregulation of Xist. Importantly, we find that TAD formation precedes transcription and initiates from Xist-poor compartments. Here, we show that TAD formation and transcriptional reactivation are causally independent during X-reactivation while establishing Xist as a common denominator.


Assuntos
Transcrição Gênica , Inativação do Cromossomo X/genética , Cromossomo X/metabolismo , Animais , Reprogramação Celular/genética , Montagem e Desmontagem da Cromatina , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Cromatina Sexual/genética , Cromatina Sexual/metabolismo , Cromossomo X/genética
16.
Genome Biol ; 22(1): 171, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34082786

RESUMO

BACKGROUND: Somatic cell reprogramming is the process that allows differentiated cells to revert to a pluripotent state. In contrast to the extensively studied rewiring of epigenetic and transcriptional programs required for reprogramming, the dynamics of post-transcriptional changes and their associated regulatory mechanisms remain poorly understood. Here we study the dynamics of alternative splicing changes occurring during efficient reprogramming of mouse B cells into induced pluripotent stem (iPS) cells and compare them to those occurring during reprogramming of mouse embryonic fibroblasts. RESULTS: We observe a significant overlap between alternative splicing changes detected in the two reprogramming systems, which are generally uncoupled from changes in transcriptional levels. Correlation between gene expression of potential regulators and specific clusters of alternative splicing changes enables the identification and subsequent validation of CPSF3 and hnRNP UL1 as facilitators, and TIA1 as repressor of mouse embryonic fibroblasts reprogramming. We further find that these RNA-binding proteins control partially overlapping programs of splicing regulation, involving genes relevant for developmental and morphogenetic processes. CONCLUSIONS: Our results reveal common programs of splicing regulation during reprogramming of different cell types and identify three novel regulators of this process and their targets.


Assuntos
Processamento Alternativo/genética , Reprogramação Celular/genética , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Antígeno-1 Intracelular de Células T/metabolismo , Animais , Linfócitos B/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos
17.
Aging Cell ; 20(5): e13360, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33908703

RESUMO

Female fertility is inversely correlated with maternal age due to a depletion of the oocyte pool and a reduction in oocyte developmental competence. Few studies have addressed the effect of maternal age on the human mature oocyte (MII) transcriptome, which is established during oocyte growth and maturation, however, the pathways involved remain unclear. Here, we characterize and compare the transcriptomes of a large cohort of fully grown germinal vesicle stage (GV) and in vitro matured (IVM-MII) oocytes from women of varying reproductive age. First, we identified two clusters of cells reflecting the oocyte maturation stage (GV and IVM-MII) with 4445 and 324 putative marker genes, respectively. Furthermore, we identified genes for which transcript representation either progressively increased or decreased with age. Our results indicate that the transcriptome is more affected by age in IVM-MII oocytes (1219 genes) than in GV oocytes (596 genes). In particular, we found that transcripts of genes involved in chromosome segregation and RNA splicing significantly increased representation with age, while genes related to mitochondrial activity showed a lower representation. Gene regulatory network analysis facilitated the identification of potential upstream master regulators of the genes involved in those biological functions. Our analysis suggests that advanced maternal age does not globally affect the oocyte transcriptome at GV or IVM-MII stages. Nonetheless, hundreds of genes displayed altered transcript representation, particularly in IVM-MII oocytes, which might contribute to the age-related quality decline in human oocytes.


Assuntos
Envelhecimento/genética , Oócitos/metabolismo , Transcriptoma , Adolescente , Adulto , Índice de Massa Corporal , Feminino , Regulação da Expressão Gênica , Humanos , Oócitos/crescimento & desenvolvimento , RNA-Seq , Análise de Célula Única , Adulto Jovem
18.
Epigenetics Chromatin ; 12(1): 38, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31221220

RESUMO

BACKGROUND: In order to prepare the genome for gametogenesis, primordial germ cells (PGCs) undergo extensive epigenetic reprogramming during migration toward the gonads in mammalian embryos. This includes changes on a genome-wide scale and additionally in females the remodeling of the inactive X-chromosome to enable X-chromosome reactivation (XCR). However, if global remodeling and X-chromosomal remodeling are related, how they occur in PGCs in vivo in relation to their migration progress and which factors are important are unknown. RESULTS: Here we identify the germ cell determinant PR-domain containing protein 14 (PRDM14) as the first known factor that is instrumental for both global reprogramming and X-chromosomal reprogramming in migrating mouse PGCs. We find that global upregulation of the repressive histone H3 lysine 27 trimethylation (H3K27me3) mark is PRDM14 dosage dependent in PGCs of both sexes. When focusing on XCR, we observed that PRDM14 is required for removal of H3K27me3 from the inactive X-chromosome, which, in contrast to global upregulation, takes place progressively along the PGC migration path. Furthermore, we show that global and X-chromosomal reprogramming of H3K27me3 are functionally separable, despite their common regulation by PRDM14. CONCLUSIONS: In summary, here we provide new insight and spatiotemporal resolution to the progression and regulation of epigenome remodeling along mouse PGC migration in vivo and link epigenetic reprogramming to its developmental context.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Germinativas Embrionárias/metabolismo , Histonas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Cromossomo X/metabolismo , Animais , Movimento Celular/fisiologia , Reprogramação Celular , Metilação de DNA , Proteínas de Ligação a DNA/genética , Células Germinativas Embrionárias/citologia , Epigênese Genética , Feminino , Histonas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Cromossomo X/genética , Inativação do Cromossomo X
19.
Neuron ; 37(4): 597-609, 2003 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-12597858

RESUMO

Two-photon excitation microscopy was used to reconstruct cell divisions in living zebrafish embryonic retinas. Contrary to proposed models for vertebrate asymmetric divisions, no apico-basal cell divisions take place in the zebrafish retina during the generation of postmitotic neurons. However, a surprising shift in the orientation of cell division from central-peripheral to circumferential occurs within the plane of the ventricular surface. In the sonic you (syu) and lakritz (lak) mutants, the shift from central-peripheral to circumferential divisions is absent or delayed, correlating with the delay in neuronal differentiation and neurogenesis in these mutants. The reconstructions here show that mitotic cells always remain in contact with the opposite basal surface by means of a thin basal process that can be inherited asymmetrically.


Assuntos
Neurônios/citologia , Retina/citologia , Retina/embriologia , Animais , Divisão Celular/fisiologia , Corantes Fluorescentes , Proteínas Luminescentes/genética , Microscopia de Vídeo , Mitose , Mutação , Ratos , Proteínas Recombinantes de Fusão/genética , Células-Tronco/citologia , Fatores de Tempo , Peixe-Zebra
20.
Curr Biol ; 13(23): 2110-7, 2003 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-14654002

RESUMO

stella is a novel gene specifically expressed in primordial germ cells, oocytes, preimplantation embryos, and pluripotent cells. It encodes a protein with a SAP-like domain and a splicing factor motif-like structure, suggesting possible roles in chromosomal organization or RNA processing. Here, we have investigated the effects of a targeted mutation of stella in mice. We show that while matings between heterozygous animals resulted in the birth of apparently normal stella null offspring, stella-deficient females displayed severely reduced fertility due to a lack of maternally inherited Stella-protein in their oocytes. Indeed, we demonstrate that embryos without Stella are compromised in preimplantation development and rarely reach the blastocyst stage. stella is thus one of few known mammalian maternal effect genes, as the phenotypic effect on embryonic development is mainly a consequence of the maternal stella mutant genotype. Furthermore, we show that STELLA that is expressed in human oocytes is also expressed in human pluripotent cells and in germ cell tumors. Interestingly, human chromosome 12p, which harbours STELLA, is consistently overrepresented in these tumors. These findings suggest a similar role for STELLA during early human development as in mice and a potential involvement in germ cell tumors.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Camundongos/embriologia , Camundongos/genética , Proteínas Repressoras/genética , Sequência de Aminoácidos , Animais , Proteínas Cromossômicas não Histona , Mapeamento Cromossômico , Cromossomos Humanos Par 12 , Feminino , Fertilidade/genética , Fertilidade/fisiologia , Humanos , Masculino , Camundongos Mutantes , Dados de Sequência Molecular , Mutação , Oócitos/citologia , Oócitos/fisiologia , Homologia de Sequência , Testículo/citologia , Testículo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA