Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 84(3): 413-414, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38307002

RESUMO

In a recent study in Cell, Malachowski et al.1 show that the trinucleotide expansion in the FMR1 gene underlying fragile X syndrome triggers formation of large heterochromatin domains across the genome, resulting in the repression of synaptic genes housed within these domains.


Assuntos
Síndrome do Cromossomo X Frágil , Humanos , Síndrome do Cromossomo X Frágil/genética , Expansão das Repetições de Trinucleotídeos/genética , Heterocromatina/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Regiões Promotoras Genéticas , Repetições de Trinucleotídeos/genética
2.
Mol Cell ; 82(1): 190-208.e17, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34932975

RESUMO

Developmental genes such as Xist, which initiates X chromosome inactivation, are controlled by complex cis-regulatory landscapes, which decode multiple signals to establish specific spatiotemporal expression patterns. Xist integrates information on X chromosome dosage and developmental stage to trigger X inactivation in the epiblast specifically in female embryos. Through a pooled CRISPR screen in differentiating mouse embryonic stem cells, we identify functional enhancer elements of Xist at the onset of random X inactivation. Chromatin profiling reveals that X-dosage controls the promoter-proximal region, while differentiation cues activate several distal enhancers. The strongest distal element lies in an enhancer cluster associated with a previously unannotated Xist-enhancing regulatory transcript, which we named Xert. Developmental cues and X-dosage are thus decoded by distinct regulatory regions, which cooperate to ensure female-specific Xist upregulation at the correct developmental time. With this study, we start to disentangle how multiple, functionally distinct regulatory elements interact to generate complex expression patterns in mammals.


Assuntos
Elementos Facilitadores Genéticos , Loci Gênicos , Células-Tronco Embrionárias Murinas/metabolismo , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Inativação do Cromossomo X , Cromossomo X , Animais , Diferenciação Celular , Linhagem Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regulação para Cima
3.
Development ; 151(10)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38813842

RESUMO

This Review elucidates the regulatory principles of random monoallelic expression by focusing on two well-studied examples: the X-chromosome inactivation regulator Xist and the olfactory receptor gene family. Although the choice of a single X chromosome or olfactory receptor occurs in different developmental contexts, common gene regulatory principles guide monoallelic expression in both systems. In both cases, an event breaks the symmetry between genetically and epigenetically identical copies of the gene, leading to the expression of one single random allele, stabilized through negative feedback control. Although many regulatory steps that govern the establishment and maintenance of monoallelic expression have been identified, key pieces of the puzzle are still missing. We provide an overview of the current knowledge and models for the monoallelic expression of Xist and olfactory receptors. We discuss their similarities and differences, and highlight open questions and approaches that could guide the study of other monoallelically expressed genes.


Assuntos
Alelos , RNA Longo não Codificante , Receptores Odorantes , Inativação do Cromossomo X , Animais , Humanos , Inativação do Cromossomo X/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento
4.
Mol Syst Biol ; 19(11): e11510, 2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-37735975

RESUMO

For a short period during early development of mammalian embryos, both X chromosomes in females are active, before dosage compensation is ensured through X-chromosome inactivation. In female mouse embryonic stem cells (mESCs), which carry two active X chromosomes, increased X-dosage affects cell signaling and impairs differentiation. The underlying mechanisms, however, remain poorly understood. To dissect X-dosage effects on the signaling network in mESCs, we combine systematic perturbation experiments with mathematical modeling. We quantify the response to a variety of inhibitors and growth factors for cells with one (XO) or two X chromosomes (XX). We then build models of the signaling networks in XX and XO cells through a semi-quantitative modeling approach based on modular response analysis. We identify a novel negative feedback in the PI3K/AKT pathway through GSK3. Moreover, the presence of a single active X makes mESCs more sensitive to the differentiation-promoting Activin A signal and leads to a stronger RAF1-mediated negative feedback in the FGF-triggered MAPK pathway. The differential response to these differentiation-promoting pathways can explain the impaired differentiation propensity of female mESCs.


Assuntos
Células-Tronco Embrionárias , Células-Tronco Embrionárias Murinas , Feminino , Animais , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias/metabolismo , Caracteres Sexuais , Quinase 3 da Glicogênio Sintase , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Diferenciação Celular/genética , Mamíferos
5.
Genome Res ; 29(7): 1087-1099, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31175153

RESUMO

To initiate X-Chromosome inactivation (XCI), the long noncoding RNA Xist mediates chromosome-wide gene silencing of one X Chromosome in female mammals to equalize gene dosage between the sexes. The efficiency of gene silencing is highly variable across genes, with some genes even escaping XCI in somatic cells. A gene's susceptibility to Xist-mediated silencing appears to be determined by a complex interplay of epigenetic and genomic features; however, the underlying rules remain poorly understood. We have quantified chromosome-wide gene silencing kinetics at the level of the nascent transcriptome using allele-specific Precision nuclear Run-On sequencing (PRO-seq). We have developed a Random Forest machine-learning model that can predict the measured silencing dynamics based on a large set of epigenetic and genomic features and tested its predictive power experimentally. The genomic distance to the Xist locus, followed by gene density and distance to LINE elements, are the prime determinants of the speed of gene silencing. Moreover, we find two distinct gene classes associated with different silencing pathways: a class that requires Xist-repeat A for silencing, which is known to activate the SPEN pathway, and a second class in which genes are premarked by Polycomb complexes and tend to rely on the B repeat in Xist for silencing, known to recruit Polycomb complexes during XCI. Moreover, a series of features associated with active transcriptional elongation and chromatin 3D structure are enriched at rapidly silenced genes. Our machine-learning approach can thus uncover the complex combinatorial rules underlying gene silencing during X inactivation.


Assuntos
Epigênese Genética , Inativação Gênica , Aprendizado de Máquina , RNA Longo não Codificante/fisiologia , Inativação do Cromossomo X/genética , Animais , Linhagem Celular , Células-Tronco Embrionárias , Feminino , Genes Ligados ao Cromossomo X , Genoma , Cinética , Camundongos , Modelos Genéticos
6.
Bioessays ; 42(4): e1900163, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32189388

RESUMO

X-chromosome inactivation ensures dosage compensation between the sexes in mammals by randomly choosing one out of the two X chromosomes in females for inactivation. This process imposes a plethora of questions: How do cells count their X chromosome number and ensure that exactly one stays active? How do they randomly choose one of two identical X chromosomes for inactivation? And how do they stably maintain this state of monoallelic expression? Here, different regulatory concepts and their plausibility are evaluated in the context of theoretical studies that have investigated threshold behavior, ultrasensitivity, and bistability through mathematical modeling. It is discussed how a twofold difference between a single and a double dose of X-linked genes might be converted to an all-or-nothing response and how mutually exclusive expression can be initiated and maintained. Finally, candidate factors that might mediate the proposed regulatory principles are reviewed.


Assuntos
Mamíferos/genética , Biologia de Sistemas/métodos , Inativação do Cromossomo X , Cromossomo X/genética , Alelos , Animais , Desenvolvimento Embrionário/genética , Epigenômica/métodos , Retroalimentação Fisiológica/fisiologia , Feminino , Humanos , Masculino , Mamíferos/embriologia , Ploidias , RNA Longo não Codificante/genética
7.
Genome Res ; 27(3): 479-490, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28057745

RESUMO

Understanding how regulatory sequences interact in the context of chromosomal architecture is a central challenge in biology. Chromosome conformation capture revealed that mammalian chromosomes possess a rich hierarchy of structural layers, from multi-megabase compartments to sub-megabase topologically associating domains (TADs) and sub-TAD contact domains. TADs appear to act as regulatory microenvironments by constraining and segregating regulatory interactions across discrete chromosomal regions. However, it is unclear whether other (or all) folding layers share similar properties, or rather TADs constitute a privileged folding scale with maximal impact on the organization of regulatory interactions. Here, we present a novel algorithm named CaTCH that identifies hierarchical trees of chromosomal domains in Hi-C maps, stratified through their reciprocal physical insulation, which is a single and biologically relevant parameter. By applying CaTCH to published Hi-C data sets, we show that previously reported folding layers appear at different insulation levels. We demonstrate that although no structurally privileged folding level exists, TADs emerge as a functionally privileged scale defined by maximal boundary enrichment in CTCF and maximal cell-type conservation. By measuring transcriptional output in embryonic stem cells and neural precursor cells, we show that the likelihood that genes in a domain are coregulated during differentiation is also maximized at the scale of TADs. Finally, we observe that regulatory sequences occur at genomic locations corresponding to optimized mutual interactions at the same scale. Our analysis suggests that the architectural functionality of TADs arises from the interplay between their ability to partition interactions and the specific genomic position of regulatory sequences.


Assuntos
Algoritmos , Montagem e Desmontagem da Cromatina , Cromossomos/química , Elementos Isolantes , Animais , Células Cultivadas , Cromossomos/genética , Cromossomos/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Modelos Teóricos , Células-Tronco Neurais/metabolismo
8.
Immunity ; 30(5): 673-83, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19409816

RESUMO

Differentiation of naive T lymphocytes into type I T helper (Th1) cells requires interferon-gamma and interleukin-12. It is puzzling that interferon-gamma induces the Th1 transcription factor T-bet, whereas interleukin-12 mediates Th1 cell lineage differentiation. We use mathematical modeling to analyze the expression kinetics of T-bet, interferon-gamma, and the IL-12 receptor beta2 chain (IL-12Rbeta2) during Th1 cell differentiation, in the presence or absence of interleukin-12 or interferon-gamma signaling. We show that interferon-gamma induced initial T-bet expression, whereas IL-12Rbeta2 was repressed by T cell receptor (TCR) signaling. The termination of TCR signaling permitted upregulation of IL-12Rbeta2 by T-bet and interleukin-12 signaling that maintained T-bet expression. This late expression of T-bet, accompanied by the upregulation of the transcription factors Runx3 and Hlx, was required to imprint the Th cell for interferon-gamma re-expression. Thus initial polarization and subsequent imprinting of Th1 cells are mediated by interlinked, sequentially acting positive feedback loops of TCR-interferon-gamma-Stat1-T-bet and interleukin-12-Stat4-T-bet signaling.


Assuntos
Polaridade Celular/genética , Impressão Genômica , Proteínas de Homeodomínio/metabolismo , Interferon gama/genética , Interleucina-12/genética , Células Th1/imunologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Polaridade Celular/imunologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Proteínas de Homeodomínio/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Receptores de Interleucina-12/imunologia , Receptores de Interleucina-12/metabolismo , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/imunologia
9.
Nature ; 485(7398): 381-5, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22495304

RESUMO

In eukaryotes transcriptional regulation often involves multiple long-range elements and is influenced by the genomic environment. A prime example of this concerns the mouse X-inactivation centre (Xic), which orchestrates the initiation of X-chromosome inactivation (XCI) by controlling the expression of the non-protein-coding Xist transcript. The extent of Xic sequences required for the proper regulation of Xist remains unknown. Here we use chromosome conformation capture carbon-copy (5C) and super-resolution microscopy to analyse the spatial organization of a 4.5-megabases (Mb) region including Xist. We discover a series of discrete 200-kilobase to 1 Mb topologically associating domains (TADs), present both before and after cell differentiation and on the active and inactive X. TADs align with, but do not rely on, several domain-wide features of the epigenome, such as H3K27me3 or H3K9me2 blocks and lamina-associated domains. TADs also align with coordinately regulated gene clusters. Disruption of a TAD boundary causes ectopic chromosomal contacts and long-range transcriptional misregulation. The Xist/Tsix sense/antisense unit illustrates how TADs enable the spatial segregation of oppositely regulated chromosomal neighbourhoods, with the respective promoters of Xist and Tsix lying in adjacent TADs, each containing their known positive regulators. We identify a novel distal regulatory region of Tsix within its TAD, which produces a long intervening RNA, Linx. In addition to uncovering a new principle of cis-regulatory architecture of mammalian chromosomes, our study sets the stage for the full genetic dissection of the X-inactivation centre.


Assuntos
RNA não Traduzido/genética , Inativação do Cromossomo X/genética , Cromossomo X/genética , Animais , Diferenciação Celular , DNA Intergênico/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Epigenômica , Feminino , Fibroblastos , Regulação da Expressão Gênica , Histonas/metabolismo , Hibridização in Situ Fluorescente , Masculino , Metilação , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , RNA Longo não Codificante , Transcriptoma , Cromossomo X/química
10.
Curr Opin Genet Dev ; 86: 102198, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663040

RESUMO

Genomes are organised through hierarchical structures, ranging from local kilobase-scale cis-regulatory contacts to large chromosome territories. Most notably, (sub)-compartments partition chromosomes according to transcriptional activity, while topologically associating domains (TADs) define cis-regulatory landscapes. The inactive X chromosome in mammals has provided unique insights into the regulation and function of the three-dimensional (3D) genome. Concurrent with silencing of the majority of genes and major alterations of its chromatin state, the X chromosome undergoes profound spatial rearrangements at multiple scales. These include the emergence of megadomains, alterations of the compartment structure and loss of the majority of TADs. Moreover, the Xist locus, which orchestrates X-chromosome inactivation, has provided key insights into regulation and function of regulatory domains. This review provides an overview of recent insights into the control of these structural rearrangements and contextualises them within a broader understanding of 3D genome organisation.


Assuntos
Cromatina , Inativação do Cromossomo X , Inativação do Cromossomo X/genética , Animais , Humanos , Cromatina/genética , Cromossomo X/genética , Genoma/genética , RNA Longo não Codificante/genética
11.
Curr Opin Genet Dev ; 81: 102063, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37356341

RESUMO

X-chromosome inactivation (XCI) has evolved in mammals to compensate for the difference in X-chromosomal dosage between the sexes. In placental mammals, XCI is initiated during early embryonic development through upregulation of the long noncoding RNA Xist from one randomly chosen X chromosome in each female cell. The Xist locus must thus integrate both X-linked and developmental trans-regulatory factors in a dosage-dependent manner. Furthermore, the two alleles must coordinate to ensure inactivation of exactly one X chromosome per cell. In this review, we summarize the regulatory principles that govern the onset of XCI. We go on to provide an overview over the factors that have been implicated in Xist regulation and discuss recent advances in our understanding of how Xist's cis-regulatory landscape integrates information in a precise fashion.


Assuntos
Placenta , RNA Longo não Codificante , Gravidez , Animais , Feminino , Inativação do Cromossomo X/genética , Cromossomo X/genética , RNA Longo não Codificante/genética , Mamíferos/genética
12.
Nat Commun ; 14(1): 3225, 2023 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-37270532

RESUMO

Certain cellular processes are dose-dependent, requiring specific quantities or stoichiometries of gene products, as exemplified by haploinsufficiency and sex-chromosome dosage compensation. Understanding dosage-sensitive processes requires tools to quantitatively modulate protein abundance. Here we present CasTuner, a CRISPR-based toolkit for analog tuning of endogenous gene expression. The system exploits Cas-derived repressors that are quantitatively tuned by ligand titration through a FKBP12F36V degron domain. CasTuner can be applied at the transcriptional or post-transcriptional level using a histone deacetylase (hHDAC4) fused to dCas9, or the RNA-targeting CasRx, respectively. We demonstrate analog tuning of gene expression homogeneously across cells in mouse and human cells, as opposed to KRAB-dependent CRISPR-interference systems, which exhibit digital repression. Finally, we quantify the system's dynamics and use it to measure dose-response relationships of NANOG and OCT4 with their target genes and with the cellular phenotype. CasTuner thus provides an easy-to-implement tool to study dose-responsive processes in their physiological context.


Assuntos
Sistemas CRISPR-Cas , Fatores de Transcrição , Camundongos , Humanos , Animais , Sistemas CRISPR-Cas/genética , Fatores de Transcrição/metabolismo , Genes Homeobox , Expressão Gênica
13.
Nat Cell Biol ; 25(11): 1704-1715, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37932452

RESUMO

X-chromosome inactivation (XCI) balances gene expression between the sexes in female mammals. Shortly after fertilization, upregulation of Xist RNA from one X chromosome initiates XCI, leading to chromosome-wide gene silencing. XCI is maintained in all cell types, except the germ line and the pluripotent state where XCI is reversed. The mechanisms triggering Xist upregulation have remained elusive. Here we identify GATA transcription factors as potent activators of Xist. Through a pooled CRISPR activation screen in murine embryonic stem cells, we demonstrate that GATA1, as well as other GATA transcription factors can drive ectopic Xist expression. Moreover, we describe GATA-responsive regulatory elements in the Xist locus bound by different GATA factors. Finally, we show that GATA factors are essential for XCI induction in mouse preimplantation embryos. Deletion of GATA1/4/6 or GATA-responsive Xist enhancers in mouse zygotes effectively prevents Xist upregulation. We propose that the activity or complete absence of various GATA family members controls initial Xist upregulation, XCI maintenance in extra-embryonic lineages and XCI reversal in the epiblast.


Assuntos
Fatores de Transcrição GATA , RNA Longo não Codificante , Animais , Feminino , Camundongos , Fertilização/genética , Fatores de Transcrição GATA/genética , Mamíferos , RNA Longo não Codificante/genética , Regulação para Cima , Cromossomo X , Inativação do Cromossomo X/genética
14.
Sci Adv ; 9(39): eadg1936, 2023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37774033

RESUMO

Human pluripotent stem cells (hPSCs) are of fundamental relevance in regenerative medicine. Naïve hPSCs hold promise to overcome some of the limitations of conventional (primed) hPSCs, including recurrent epigenetic anomalies. Naïve-to-primed transition (capacitation) follows transcriptional dynamics of human embryonic epiblast and is necessary for somatic differentiation from naïve hPSCs. We found that capacitated hPSCs are transcriptionally closer to postimplantation epiblast than conventional hPSCs. This prompted us to comprehensively study epigenetic and related transcriptional changes during capacitation. Our results show that CpG islands, gene regulatory elements, and retrotransposons are hotspots of epigenetic dynamics during capacitation and indicate possible distinct roles of specific epigenetic modifications in gene expression control between naïve and primed hPSCs. Unexpectedly, PRC2 activity appeared to be dispensable for the capacitation. We find that capacitated hPSCs acquire an epigenetic state similar to conventional hPSCs. Significantly, however, the X chromosome erosion frequently observed in conventional female hPSCs is reversed by resetting and subsequent capacitation.


Assuntos
Células-Tronco Pluripotentes , Humanos , Feminino , Diferenciação Celular/genética , Embrião de Mamíferos , Epigênese Genética
15.
Cell Syst ; 13(11): 857-858, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36395723

RESUMO

One snapshot of the peer-review process for "Somatic XIST activation and features of X chromosome inactivation in male human cancers" (Sadagopan et al., 2022).


Assuntos
Neoplasias , RNA Longo não Codificante , Masculino , Humanos , Inativação do Cromossomo X/genética , RNA não Traduzido/genética , RNA Longo não Codificante/genética , Fatores de Transcrição/genética , Neoplasias/genética
16.
Methods Mol Biol ; 2520: 1-24, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35218528

RESUMO

Pooled CRISPR screens are emerging as a powerful tool to dissect regulatory networks, by assessing how a protein responds to genetic perturbations in a highly multiplexed manner. A large number of genes are perturbed in a cell population through genomic integration of one single-guide RNA (sgRNA) per cell. A subset of cells with the phenotype of interest can then be enriched through fluorescence-activated cell sorting (FACS). SgRNAs with altered abundance after phenotypic enrichment allow identification of genes that either promote or attenuate the investigated phenotype. Here we provide detailed guidelines on how to design and execute a pooled CRISPR screen to investigate molecular phenotypes. We describe how to generate a custom sgRNA library and how to perform a FACS-based screen using readouts such as intracellular antibody staining or Flow-FISH to assess phosphorylation levels or RNA abundance. Through the variety of available perturbation systems and readout options many different molecular and cellular phenotypes can now be tackled with pooled CRISPR screens.


Assuntos
Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Citometria de Fluxo , Fenótipo , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo
17.
Mol Syst Biol ; 6: 359, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20393579

RESUMO

Although cell-to-cell variability has been recognized as an unavoidable consequence of stochasticity in gene expression, it may also serve a functional role for tuning physiological responses within a cell population. In the immune system, remarkably large variability in the expression of cytokine genes has been observed in homogeneous populations of lymphocytes, but the underlying molecular mechanisms are incompletely understood. Here, we study the interleukin-4 gene (il4) in T-helper lymphocytes, combining mathematical modeling with the experimental quantification of expression variability and critical parameters. We show that a stochastic rate-limiting step upstream of transcription initiation, but acting at the level of an individual allele, controls il4 expression. Only a fraction of cells reaches an active, transcription-competent state in the transient time window determined by antigen stimulation. We support this finding by experimental evidence of a previously unknown short-term memory that was predicted by the model to arise from the long lifetime of the active state. Our analysis shows how a stochastic mechanism acting at the chromatin level can be integrated with transcriptional regulation to quantitatively control cell-to-cell variability.


Assuntos
Regulação da Expressão Gênica , Interleucina-4/genética , Ativação Transcricional , Animais , Cromatina/metabolismo , Citometria de Fluxo , Fator de Transcrição GATA3/metabolismo , Interleucina-4/biossíntese , Camundongos , Modelos Genéticos , Fatores de Transcrição NFATC/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Processos Estocásticos , Células Th2/metabolismo
18.
Genome Biol ; 22(1): 110, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863351

RESUMO

BACKGROUND: X-chromosomal genes contribute to sex differences, in particular during early development, when both X chromosomes are active in females. Double X-dosage shifts female pluripotent cells towards the naive stem cell state by increasing pluripotency factor expression, inhibiting the differentiation-promoting MAP kinase (MAPK) signaling pathway, and delaying differentiation. RESULTS: To identify the genetic basis of these sex differences, we use a two-step CRISPR screening approach to comprehensively identify X-linked genes that cause the female pluripotency phenotype in murine embryonic stem cells. A primary chromosome-wide CRISPR knockout screen and three secondary screens assaying for different aspects of the female pluripotency phenotype allow us to uncover multiple genes that act in concert and to disentangle their relative roles. Among them, we identify Dusp9 and Klhl13 as two central players. While Dusp9 mainly affects MAPK pathway intermediates, Klhl13 promotes pluripotency factor expression and delays differentiation, with both factors jointly repressing MAPK target gene expression. CONCLUSIONS: Here, we elucidate the mechanisms that drive sex-induced differences in pluripotent cells and our approach serves as a blueprint to discover the genetic basis of the phenotypic consequences of other chromosomal effects.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células-Tronco Embrionárias/metabolismo , Genes Ligados ao Cromossomo X , Estudos de Associação Genética/métodos , Caracteres Sexuais , Animais , Biomarcadores , Proteínas de Transporte , Diferenciação Celular/genética , Variações do Número de Cópias de DNA , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Mutação , Fosforilação , Ligação Proteica , Fatores Sexuais , Cromossomo X
19.
Nat Commun ; 12(1): 3638, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131144

RESUMO

To ensure dosage compensation between the sexes, one randomly chosen X chromosome is silenced in each female cell in the process of X-chromosome inactivation (XCI). XCI is initiated during early development through upregulation of the long non-coding RNA Xist, which mediates chromosome-wide gene silencing. Cell differentiation, Xist upregulation and gene silencing are thought to be coupled at multiple levels to ensure inactivation of exactly one out of two X chromosomes. Here we perform an integrated analysis of all three processes through allele-specific single-cell RNA-sequencing. Specifically, we assess the onset of random XCI in differentiating mouse embryonic stem cells, and develop dedicated analysis approaches. By exploiting the inter-cellular heterogeneity of XCI onset, we identify putative Xist regulators. Moreover, we show that transient Xist upregulation from both X chromosomes results in biallelic gene silencing right before transitioning to the monoallelic state, confirming a prediction of the stochastic model of XCI. Finally, we show that genetic variation modulates the XCI process at multiple levels, providing a potential explanation for the long-known X-controlling element (Xce) effect, which leads to preferential inactivation of a specific X chromosome in inter-strain crosses. We thus draw a detailed picture of the different levels of regulation that govern the initiation of XCI. The experimental and computational strategies we have developed here will allow us to profile random XCI in more physiological contexts, including primary human cells in vivo.


Assuntos
RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Regulação para Cima , Inativação do Cromossomo X , Alelos , Animais , Mecanismo Genético de Compensação de Dose , Feminino , Inativação Gênica , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas , Análise de Sequência de RNA , Cromossomo X , Inativação do Cromossomo X/genética , Inativação do Cromossomo X/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA