Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 185(17): 3169-3185.e20, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35908548

RESUMO

Mice deficient for all ten-eleven translocation (TET) genes exhibit early gastrulation lethality. However, separating cause and effect in such embryonic failure is challenging. To isolate cell-autonomous effects of TET loss, we used temporal single-cell atlases from embryos with partial or complete mutant contributions. Strikingly, when developing within a wild-type embryo, Tet-mutant cells retain near-complete differentiation potential, whereas embryos solely comprising mutant cells are defective in epiblast to ectoderm transition with degenerated mesoderm potential. We map de-repressions of early epiblast factors (e.g., Dppa4 and Gdf3) and failure to activate multiple signaling from nascent mesoderm (Lefty, FGF, and Notch) as likely cell-intrinsic drivers of TET loss phenotypes. We further suggest loss of enhancer demethylation as the underlying mechanism. Collectively, our work demonstrates an unbiased approach for defining intrinsic and extrinsic embryonic gene function based on temporal differentiation atlases and disentangles the intracellular effects of the demethylation machinery from its broader tissue-level ramifications.


Assuntos
Gastrulação , Mesoderma , Animais , Diferenciação Celular/genética , Embrião de Mamíferos/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteínas Nucleares/metabolismo , Transdução de Sinais
2.
Cell ; 184(11): 2825-2842.e22, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33932341

RESUMO

Mouse embryonic development is a canonical model system for studying mammalian cell fate acquisition. Recently, single-cell atlases comprehensively charted embryonic transcriptional landscapes, yet inference of the coordinated dynamics of cells over such atlases remains challenging. Here, we introduce a temporal model for mouse gastrulation, consisting of data from 153 individually sampled embryos spanning 36 h of molecular diversification. Using algorithms and precise timing, we infer differentiation flows and lineage specification dynamics over the embryonic transcriptional manifold. Rapid transcriptional bifurcations characterize the commitment of early specialized node and blood cells. However, for most lineages, we observe combinatorial multi-furcation dynamics rather than hierarchical transcriptional transitions. In the mesoderm, dozens of transcription factors combinatorially regulate multifurcations, as we exemplify using time-matched chimeric embryos of Foxc1/Foxc2 mutants. Our study rejects the notion of differentiation being governed by a series of binary choices, providing an alternative quantitative model for cell fate acquisition.


Assuntos
Desenvolvimento Embrionário/fisiologia , Gastrulação/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/genética , Feminino , Expressão Gênica , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas , Gravidez , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos
3.
Cell ; 173(6): 1520-1534.e20, 2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29856957

RESUMO

The emergence and diversification of cell types is a leading factor in animal evolution. So far, systematic characterization of the gene regulatory programs associated with cell type specificity was limited to few cell types and few species. Here, we perform whole-organism single-cell transcriptomics to map adult and larval cell types in the cnidarian Nematostella vectensis, a non-bilaterian animal with complex tissue-level body-plan organization. We uncover eight broad cell classes in Nematostella, including neurons, cnidocytes, and digestive cells. Each class comprises different subtypes defined by the expression of multiple specific markers. In particular, we characterize a surprisingly diverse repertoire of neurons, which comparative analysis suggests are the result of lineage-specific diversification. By integrating transcription factor expression, chromatin profiling, and sequence motif analysis, we identify the regulatory codes that underlie Nematostella cell-specific expression. Our study reveals cnidarian cell type complexity and provides insights into the evolution of animal cell-specific genomic regulation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Neurônios/fisiologia , RNA , Anêmonas-do-Mar/fisiologia , Actinas/química , Motivos de Aminoácidos , Animais , Cromatina/metabolismo , Análise por Conglomerados , Perfilação da Expressão Gênica , Genoma , Genômica , Filogenia , Anêmonas-do-Mar/genética , Análise de Sequência de RNA , Transcriptoma , Tubulina (Proteína)/química
4.
Nature ; 540(7632): 296-300, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27919068

RESUMO

Chromosomes are folded into highly compacted structures to accommodate physical constraints within nuclei and to regulate access to genomic information. Recently, global mapping of pairwise contacts showed that loops anchoring topological domains (TADs) are highly conserved between cell types and species. Whether pairwise loops synergize to form higher-order structures is still unclear. Here we develop a conformation capture assay to study higher-order organization using chromosomal walks (C-walks) that link multiple genomic loci together into proximity chains in human and mouse cells. This approach captures chromosomal structure at varying scales. Inter-chromosomal contacts constitute only 7-10% of the pairs and are restricted by interfacing TADs. About half of the C-walks stay within one chromosome, and almost half of those are restricted to intra-TAD spaces. C-walks that couple 2-4 TADs indicate stochastic associations between transcriptionally active, early replicating loci. Targeted analysis of thousands of 3-walks anchored at highly expressed genes support pairwise, rather than hub-like, chromosomal topology at active loci. Polycomb-repressed Hox domains are shown by the same approach to enrich for synergistic hubs. Together, the data indicate that chromosomal territories, TADs, and intra-TAD loops are primarily driven by nested, possibly dynamic, pairwise contacts.


Assuntos
Passeio de Cromossomo , Cromossomos/química , Cromossomos/genética , Loci Gênicos , Conformação de Ácido Nucleico , Animais , Cromatina/química , Cromatina/genética , Regulação da Expressão Gênica , Genes Homeobox , Loci Gênicos/genética , Humanos , Imageamento Tridimensional , Camundongos , Proteínas do Grupo Polycomb/metabolismo , Processos Estocásticos , Transcrição Gênica
5.
Nature ; 513(7516): 115-9, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25043040

RESUMO

Stable maintenance of gene regulatory programs is essential for normal function in multicellular organisms. Epigenetic mechanisms, and DNA methylation in particular, are hypothesized to facilitate such maintenance by creating cellular memory that can be written during embryonic development and then guide cell-type-specific gene expression. Here we develop new methods for quantitative inference of DNA methylation turnover rates, and show that human embryonic stem cells preserve their epigenetic state by balancing antagonistic processes that add and remove methylation marks rather than by copying epigenetic information from mother to daughter cells. In contrast, somatic cells transmit considerable epigenetic information to progenies. Paradoxically, the persistence of the somatic epigenome makes it more vulnerable to noise, since random epimutations can accumulate to massively perturb the epigenomic ground state. The rate of epigenetic perturbation depends on the genomic context, and, in particular, DNA methylation loss is coupled to late DNA replication dynamics. Epigenetic perturbation is not observed in the pluripotent state, because the rapid turnover-based equilibrium continuously reinforces the canonical state. This dynamic epigenetic equilibrium also explains how the epigenome can be reprogrammed quickly and to near perfection after induced pluripotency.


Assuntos
Metilação de DNA , Epigênese Genética , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Alelos , Linhagem Celular , Linhagem Celular Tumoral , Células Clonais/citologia , Células Clonais/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/citologia , Genoma Humano/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
6.
Proc Natl Acad Sci U S A ; 114(20): E4030-E4039, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461505

RESUMO

Children with Down syndrome (DS) are prone to development of high-risk B-cell precursor ALL (DS-ALL), which differs genetically from most sporadic pediatric ALLs. Increased expression of cytokine receptor-like factor 2 (CRLF2), the receptor to thymic stromal lymphopoietin (TSLP), characterizes about half of DS-ALLs and also a subgroup of sporadic "Philadelphia-like" ALLs. To understand the pathogenesis of relapsed DS-ALL, we performed integrative genomic analysis of 25 matched diagnosis-remission and -relapse DS-ALLs. We found that the CRLF2 rearrangements are early events during DS-ALL evolution and generally stable between diagnoses and relapse. Secondary activating signaling events in the JAK-STAT/RAS pathway were ubiquitous but highly redundant between diagnosis and relapse, suggesting that signaling is essential but that no specific mutations are "relapse driving." We further found that activated JAK2 may be naturally suppressed in 25% of CRLF2pos DS-ALLs by loss-of-function aberrations in USP9X, a deubiquitinase previously shown to stabilize the activated phosphorylated JAK2. Interrogation of large ALL genomic databases extended our findings up to 25% of CRLF2pos, Philadelphia-like ALLs. Pharmacological or genetic inhibition of USP9X, as well as treatment with low-dose ruxolitinib, enhanced the survival of pre-B ALL cells overexpressing mutated JAK2. Thus, somehow counterintuitive, we found that suppression of JAK-STAT "hypersignaling" may be beneficial to leukemic B-cell precursors. This finding and the reduction of JAK mutated clones at relapse suggest that the therapeutic effect of JAK specific inhibitors may be limited. Rather, combined signaling inhibitors or direct targeting of the TSLP receptor may be a useful therapeutic strategy for DS-ALL.


Assuntos
Síndrome de Down/complicações , Janus Quinases/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Fatores de Transcrição STAT/metabolismo , Adolescente , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Citocinas/genética , Recidiva , Transdução de Sinais , Ubiquitina Tiolesterase/genética , Adulto Jovem
7.
Nat Methods ; 13(8): 685-91, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27376768

RESUMO

We developed a targeted chromosome conformation capture (4C) approach that uses unique molecular identifiers (UMIs) to derive high-complexity quantitative chromosome contact profiles with controlled signal-to-noise ratios. UMI-4C detects chromosomal interactions with improved sensitivity and specificity, and it can easily be multiplexed to allow robust comparison of contact distributions between loci and conditions. This approach may open the way to the incorporation of contact distributions into quantitative models of gene regulation.


Assuntos
Mapeamento Cromossômico/métodos , Cromossomos Humanos/química , Cromossomos Humanos/metabolismo , Regulação da Expressão Gênica , Genômica/métodos , Leucemia/genética , Cromatina/genética , Interpretação Estatística de Dados , Loci Gênicos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Conformação de Ácido Nucleico , Razão Sinal-Ruído , Software , Células Tumorais Cultivadas , Globinas beta/genética
8.
Nature ; 504(7479): 282-6, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24172903

RESUMO

Mouse embryonic stem (ES) cells are isolated from the inner cell mass of blastocysts, and can be preserved in vitro in a naive inner-cell-mass-like configuration by providing exogenous stimulation with leukaemia inhibitory factor (LIF) and small molecule inhibition of ERK1/ERK2 and GSK3ß signalling (termed 2i/LIF conditions). Hallmarks of naive pluripotency include driving Oct4 (also known as Pou5f1) transcription by its distal enhancer, retaining a pre-inactivation X chromosome state, and global reduction in DNA methylation and in H3K27me3 repressive chromatin mark deposition on developmental regulatory gene promoters. Upon withdrawal of 2i/LIF, naive mouse ES cells can drift towards a primed pluripotent state resembling that of the post-implantation epiblast. Although human ES cells share several molecular features with naive mouse ES cells, they also share a variety of epigenetic properties with primed murine epiblast stem cells (EpiSCs). These include predominant use of the proximal enhancer element to maintain OCT4 expression, pronounced tendency for X chromosome inactivation in most female human ES cells, increase in DNA methylation and prominent deposition of H3K27me3 and bivalent domain acquisition on lineage regulatory genes. The feasibility of establishing human ground state naive pluripotency in vitro with equivalent molecular and functional features to those characterized in mouse ES cells remains to be defined. Here we establish defined conditions that facilitate the derivation of genetically unmodified human naive pluripotent stem cells from already established primed human ES cells, from somatic cells through induced pluripotent stem (iPS) cell reprogramming or directly from blastocysts. The novel naive pluripotent cells validated herein retain molecular characteristics and functional properties that are highly similar to mouse naive ES cells, and distinct from conventional primed human pluripotent cells. This includes competence in the generation of cross-species chimaeric mouse embryos that underwent organogenesis following microinjection of human naive iPS cells into mouse morulas. Collectively, our findings establish new avenues for regenerative medicine, patient-specific iPS cell disease modelling and the study of early human development in vitro and in vivo.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Animais , Blastocisto/citologia , Reprogramação Celular , Quimera/embriologia , Cromatina/metabolismo , Metilação de DNA , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Feminino , Camadas Germinativas/citologia , Histonas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Masculino , Camundongos , Mórula/citologia , Organogênese , Regiões Promotoras Genéticas/genética , Medicina Regenerativa , Reprodutibilidade dos Testes , Transdução de Sinais , Inativação do Cromossomo X
9.
Nature ; 502(7469): 65-70, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24048479

RESUMO

Somatic cells can be inefficiently and stochastically reprogrammed into induced pluripotent stem (iPS) cells by exogenous expression of Oct4 (also called Pou5f1), Sox2, Klf4 and Myc (hereafter referred to as OSKM). The nature of the predominant rate-limiting barrier(s) preventing the majority of cells to successfully and synchronously reprogram remains to be defined. Here we show that depleting Mbd3, a core member of the Mbd3/NuRD (nucleosome remodelling and deacetylation) repressor complex, together with OSKM transduction and reprogramming in naive pluripotency promoting conditions, result in deterministic and synchronized iPS cell reprogramming (near 100% efficiency within seven days from mouse and human cells). Our findings uncover a dichotomous molecular function for the reprogramming factors, serving to reactivate endogenous pluripotency networks while simultaneously directly recruiting the Mbd3/NuRD repressor complex that potently restrains the reactivation of OSKM downstream target genes. Subsequently, the latter interactions, which are largely depleted during early pre-implantation development in vivo, lead to a stochastic and protracted reprogramming trajectory towards pluripotency in vitro. The deterministic reprogramming approach devised here offers a novel platform for the dissection of molecular dynamics leading to establishing pluripotency at unprecedented flexibility and resolution.


Assuntos
Reprogramação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Modelos Biológicos , Animais , Linhagem Celular , Células Cultivadas , Reprogramação Celular/genética , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Fatores de Transcrição/genética
10.
PLoS Genet ; 12(11): e1006330, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27846223

RESUMO

The development of niches for tissue-specific stem cells is an important aspect of stem cell biology. Determination of niche size and niche numbers during organogenesis involves precise control of gene expression. How this is achieved in the context of a complex chromatin landscape is largely unknown. Here we show that the nuclear protein Combgap (Cg) supports correct ovarian niche formation in Drosophila by controlling ecdysone-Receptor (EcR)- mediated transcription and long-range chromatin contacts in the broad locus (BR-C). Both cg and BR-C promote ovarian growth and the development of niches for germ line stem cells. BR-C levels were lower when Combgap was either reduced or over-expressed, indicating an intricate regulation of the BR-C locus by Combgap. Polytene chromosome stains showed that Cg co-localizes with EcR, the major regulator of BR-C, at the BR-C locus and that EcR binding to chromatin was sensitive to changes in Cg levels. Proximity ligation assay indicated that the two proteins could reside in the same complex. Finally, chromatin conformation analysis revealed that EcR-bound regions within BR-C, which span ~30 KBs, contacted each other. Significantly, these contacts were stabilized in an ecdysone- and Combgap-dependent manner. Together, these results highlight Combgap as a novel regulator of chromatin structure that promotes transcription of ecdysone target genes and ovarian niche formation.


Assuntos
Cromatina/genética , Proteínas de Drosophila/genética , Receptores de Esteroides/genética , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Ecdisona/biossíntese , Ecdisona/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Especificidade de Órgãos , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Cromossomos Politênicos/genética , Nicho de Células-Tronco/genética
13.
Cell Rep ; 42(6): 112569, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37256750

RESUMO

Long non-coding RNAs (lncRNAs) are implicated in a plethora of cellular processes, but an in-depth understanding of their functional features or their mechanisms of action is currently lacking. Here we study Meteor, a lncRNA transcribed near the gene encoding EOMES, a pleiotropic transcription factor implicated in various processes throughout development and in adult tissues. Using a wide array of perturbation techniques, we show that transcription elongation through the Meteor locus is required for Eomes activation in mouse embryonic stem cells, with Meteor repression linked to a change in the subpopulation primed to differentiate to the mesoderm lineage. We further demonstrate that a distinct functional feature of the locus-namely, the underlying DNA element-is required for suppressing Eomes expression following neuronal differentiation. Our results demonstrate the complex regulation that can be conferred by a single locus and emphasize the importance of careful selection of perturbation techniques when studying lncRNA loci.


Assuntos
RNA Longo não Codificante , Proteínas com Domínio T , Animais , Camundongos , Diferenciação Celular/genética , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo
14.
Nat Commun ; 14(1): 3844, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37386027

RESUMO

Embryonic development involves massive proliferation and differentiation of cell lineages. This must be supported by chromosome replication and epigenetic reprogramming, but how proliferation and cell fate acquisition are balanced in this process is not well understood. Here we use single cell Hi-C to map chromosomal conformations in post-gastrulation mouse embryo cells and study their distributions and correlations with matching embryonic transcriptional atlases. We find that embryonic chromosomes show a remarkably strong cell cycle signature. Despite that, replication timing, chromosome compartment structure, topological associated domains (TADs) and promoter-enhancer contacts are shown to be variable between distinct epigenetic states. About 10% of the nuclei are identified as primitive erythrocytes, showing exceptionally compact and organized compartment structure. The remaining cells are broadly associated with ectoderm and mesoderm identities, showing only mild differentiation of TADs and compartment structures, but more specific localized contacts in hundreds of ectoderm and mesoderm promoter-enhancer pairs. The data suggest that while fully committed embryonic lineages can rapidly acquire specific chromosomal conformations, most embryonic cells are showing plastic signatures driven by complex and intermixed enhancer landscapes.


Assuntos
Gastrulação , Sequências Reguladoras de Ácido Nucleico , Feminino , Gravidez , Animais , Camundongos , Conformação Molecular , Regiões Promotoras Genéticas/genética , Cromossomos
15.
J Neurosci ; 31(32): 11437-42, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21832174

RESUMO

Autism spectrum disorder (ASD) is a highly heritable, behaviorally defined, heterogeneous disorder of unknown pathogenesis. Several genetic risk genes have been identified, including the gene encoding the receptor tyrosine kinase MET, which regulates neuronal differentiation and growth. An ASD-associated polymorphism disrupts MET gene transcription, and there are reduced levels of MET protein expression in the mature temporal cortex of subjects with ASD. To address the possible neurodevelopmental contribution of MET to ASD pathogenesis, we examined the expression and transcriptional regulation of MET by a transcription factor, FOXP2, which is implicated in regulation of cognition and language, two functions altered in ASD. MET mRNA expression in the midgestation human fetal cerebral cortex is strikingly restricted, localized to portions of the temporal and occipital lobes. Within the cortical plate of the temporal lobe, the pattern of MET expression is highly complementary to the expression pattern of FOXP2, suggesting the latter may play a role in repression of gene expression. Consistent with this, MET and FOXP2 also are reciprocally expressed by differentiating normal human neuronal progenitor cells (NHNPs) in vitro, leading us to assess whether FOXP2 transcriptionally regulates MET. Indeed, FOXP2 binds directly to the 5' regulatory region of MET, and overexpression of FOXP2 results in transcriptional repression of MET. The expression of MET in restricted human neocortical regions, and its regulation in part by FOXP2, is consistent with genetic evidence for MET contributing to ASD risk.


Assuntos
Transtorno Autístico/genética , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Proto-Oncogênicas c-met/biossíntese , Receptores de Fatores de Crescimento/biossíntese , Regiões 5' não Traduzidas/genética , Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/metabolismo , Transtornos Globais do Desenvolvimento Infantil/patologia , Transtornos Cognitivos/patologia , Feminino , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/metabolismo , Humanos , Recém-Nascido , Masculino , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Receptores de Fatores de Crescimento/antagonistas & inibidores , Receptores de Fatores de Crescimento/genética , Fatores de Risco
16.
Nat Struct Mol Biol ; 29(12): 1252-1265, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36510023

RESUMO

In mammalian embryos, DNA methylation is initialized to maximum levels in the epiblast by the de novo DNA methyltransferases DNMT3A and DNMT3B before gastrulation diversifies it across regulatory regions. Here we show that DNMT3A and DNMT3B are differentially regulated during endoderm and mesoderm bifurcation and study the implications in vivo and in meso-endoderm embryoid bodies. Loss of both Dnmt3a and Dnmt3b impairs exit from the epiblast state. More subtly, independent loss of Dnmt3a or Dnmt3b leads to small biases in mesoderm-endoderm bifurcation and transcriptional deregulation. Epigenetically, DNMT3A and DNMT3B drive distinct methylation kinetics in the epiblast, as can be predicted from their strand-specific sequence preferences. The enzymes compensate for each other in the epiblast, but can later facilitate lineage-specific methylation kinetics as their expression diverges. Single-cell analysis shows that differential activity of DNMT3A and DNMT3B combines with replication-linked methylation turnover to increase epigenetic plasticity in gastrulation. Together, these findings outline a dynamic model for the use of DNMT3A and DNMT3B sequence specificity during gastrulation.


Assuntos
DNA Metiltransferase 3A , Gastrulação , Animais , Camundongos , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Embrião de Mamíferos/metabolismo , DNA/metabolismo , Mamíferos/genética
17.
Nat Plants ; 7(6): 800-813, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34135484

RESUMO

The vegetative-to-floral transition is a dramatic developmental change of the shoot apical meristem, promoted by the systemic florigen signal. However, poor molecular temporal resolution of this dynamic process has precluded characterization of how meristems respond to florigen induction. Here, we develop a technology that allows sensitive transcriptional profiling of individual shoot apical meristems. Computational ordering of hundreds of tomato samples reconstructed the floral transition process at fine temporal resolution and uncovered novel short-lived gene expression programs that are activated before flowering. These programs are annulled only when both florigen and a parallel signalling pathway are eliminated. Functional screening identified genes acting at the onset of pre-flowering programs that are involved in the regulation of meristem morphogenetic changes but dispensable for the timing of floral transition. Induced expression of these short-lived transition-state genes allowed us to determine their genetic hierarchies and to bypass the need for the main flowering pathways. Our findings illuminate how systemic and autonomous pathways are integrated to control a critical developmental switch.


Assuntos
Flores/genética , Perfilação da Expressão Gênica/métodos , Meristema/genética , Proteínas de Plantas/genética , Solanum lycopersicum/genética , Simulação por Computador , Florígeno/metabolismo , Flores/crescimento & desenvolvimento , Regulação da Expressão Gênica de Plantas , Solanum lycopersicum/citologia , Solanum lycopersicum/crescimento & desenvolvimento , Meristema/citologia , Meristema/crescimento & desenvolvimento , Meristema/metabolismo , Microscopia Eletrônica de Varredura , Mutação , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas
18.
Nat Genet ; 52(7): 709-718, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32601473

RESUMO

Propagation of clonal regulatory programs contributes to cancer development. It is poorly understood how epigenetic mechanisms interact with genetic drivers to shape this process. Here, we combine single-cell analysis of transcription and DNA methylation with a Luria-Delbrück experimental design to demonstrate the existence of clonally stable epigenetic memory in multiple types of cancer cells. Longitudinal transcriptional and genetic analysis of clonal colon cancer cell populations reveals a slowly drifting spectrum of epithelial-to-mesenchymal transcriptional identities that is seemingly independent of genetic variation. DNA methylation landscapes correlate with these identities but also reflect an independent clock-like methylation loss process. Methylation variation can be explained as an effect of global trans-acting factors in most cases. However, for a specific class of promoters-in particular, cancer-testis antigens-de-repression is correlated with and probably driven by loss of methylation in cis. This study indicates how genetic sub-clonal structure in cancer cells can be diversified by epigenetic memory.


Assuntos
Evolução Clonal , Epigênese Genética , Neoplasias/genética , Neoplasias/patologia , Transcrição Gênica , Linhagem Celular , Proliferação de Células/genética , Metilação de DNA , DNA de Neoplasias/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , RNA Neoplásico/metabolismo , Análise de Célula Única
19.
Nat Microbiol ; 3(1): 90-98, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29085076

RESUMO

The evolutionary pressure imposed by phage predation on bacteria and archaea has resulted in the development of effective anti-phage defence mechanisms, including restriction-modification and CRISPR-Cas systems. Here, we report on a new defence system, DISARM (defence island system associated with restriction-modification), which is widespread in bacteria and archaea. DISARM is composed of five genes, including a DNA methylase and four other genes annotated as a helicase domain, a phospholipase D (PLD) domain, a DUF1998 domain and a gene of unknown function. Engineering the Bacillus paralicheniformis 9945a DISARM system into Bacillus subtilis has rendered the engineered bacteria protected against phages from all three major families of tailed double-stranded DNA phages. Using a series of gene deletions, we show that four of the five genes are essential for DISARM-mediated defence, with the fifth (PLD) being redundant for defence against some of the phages. We further show that DISARM restricts incoming phage DNA and that the B. paralicheniformis DISARM methylase modifies host CCWGG motifs as a marker of self DNA akin to restriction-modification systems. Our results suggest that DISARM is a new type of multi-gene restriction-modification module, expanding the arsenal of defence systems known to be at the disposal of prokaryotes against their viruses.


Assuntos
Bacillus subtilis/genética , Bacillus subtilis/virologia , Proteínas de Bactérias/metabolismo , Bacteriófagos/fisiologia , Enzimas de Restrição-Modificação do DNA/genética , Família Multigênica/genética , Proteínas de Bactérias/genética , Bacteriófagos/classificação , Bacteriófagos/crescimento & desenvolvimento , Clonagem Molecular , Biologia Computacional , Genoma Bacteriano/genética , Ilhas Genômicas , Metiltransferases/genética , Modelos Genéticos , Deleção de Sequência , Replicação Viral
20.
Nat Ecol Evol ; 2(7): 1176-1188, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29942020

RESUMO

A hallmark of metazoan evolution is the emergence of genomic mechanisms that implement cell-type-specific functions. However, the evolution of metazoan cell types and their underlying gene regulatory programmes remains largely uncharacterized. Here, we use whole-organism single-cell RNA sequencing to map cell-type-specific transcription in Porifera (sponges), Ctenophora (comb jellies) and Placozoa species. We describe the repertoires of cell types in these non-bilaterian animals, uncovering diverse instances of previously unknown molecular signatures, such as multiple types of peptidergic cells in Placozoa. Analysis of the regulatory programmes of these cell types reveals variable levels of complexity. In placozoans and poriferans, sequence motifs in the promoters are predictive of cell-type-specific programmes. By contrast, the generation of a higher diversity of cell types in ctenophores is associated with lower specificity of promoter sequences and the existence of distal regulatory elements. Our findings demonstrate that metazoan cell types can be defined by networks of transcription factors and proximal promoters, and indicate that further genome regulatory complexity may be required for more diverse cell type repertoires.


Assuntos
Evolução Biológica , Ctenóforos/citologia , Placozoa/citologia , Poríferos/citologia , Transcrição Gênica/fisiologia , Animais , Ctenóforos/genética , Placozoa/genética , Poríferos/genética , Análise de Sequência de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA