Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
Nat Immunol ; 20(10): 1372-1380, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31451789

RESUMO

In multicellular organisms, duplicated genes can diverge through tissue-specific gene expression patterns, as exemplified by highly regulated expression of RUNX transcription factor paralogs with apparent functional redundancy. Here we asked what cell-type-specific biologies might be supported by the selective expression of RUNX paralogs during Langerhans cell and inducible regulatory T cell differentiation. We uncovered functional nonequivalence between RUNX paralogs. Selective expression of native paralogs allowed integration of transcription factor activity with extrinsic signals, while non-native paralogs enforced differentiation even in the absence of exogenous inducers. DNA binding affinity was controlled by divergent amino acids within the otherwise highly conserved RUNT domain and evolutionary reconstruction suggested convergence of RUNT domain residues toward submaximal strength. Hence, the selective expression of gene duplicates in specialized cell types can synergize with the acquisition of functional differences to enable appropriate gene expression, lineage choice and differentiation in the mammalian immune system.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/genética , Sistema Imunitário/fisiologia , Células de Langerhans/fisiologia , Especificidade de Órgãos/genética , Linfócitos T Reguladores/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Sequência Conservada , Evolução Molecular , Duplicação Gênica , Humanos , Mamíferos , Transdução de Sinais , Transcriptoma
2.
Nat Immunol ; 24(4): 559, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36879068
3.
Nat Immunol ; 19(9): 932-941, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30127433

RESUMO

Cohesin is important for 3D genome organization. Nevertheless, even the complete removal of cohesin has surprisingly little impact on steady-state gene transcription and enhancer activity. Here we show that cohesin is required for the core transcriptional response of primary macrophages to microbial signals, and for inducible enhancer activity that underpins inflammatory gene expression. Consistent with a role for inflammatory signals in promoting myeloid differentiation of hematopoietic stem and progenitor cells (HPSCs), cohesin mutations in HSPCs led to reduced inflammatory gene expression and increased resistance to differentiation-inducing inflammatory stimuli. These findings uncover an unexpected dependence of inducible gene expression on cohesin, link cohesin with myeloid differentiation, and may help explain the prevalence of cohesin mutations in human acute myeloid leukemia.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Autorrenovação Celular/genética , Proteínas Cromossômicas não Histona/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Leucemia Mieloide Aguda/genética , Macrófagos/fisiologia , Proteínas Nucleares/genética , Fosfoproteínas/genética , Animais , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Inflamação/genética , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Knockout , Mutação/genética , Coesinas
4.
Mol Cell ; 82(20): 3769-3780.e5, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36182691

RESUMO

Complex genomes show intricate organization in three-dimensional (3D) nuclear space. Current models posit that cohesin extrudes loops to form self-interacting domains delimited by the DNA binding protein CTCF. Here, we describe and quantitatively characterize cohesin-propelled, jet-like chromatin contacts as landmarks of loop extrusion in quiescent mammalian lymphocytes. Experimental observations and polymer simulations indicate that narrow origins of loop extrusion favor jet formation. Unless constrained by CTCF, jets propagate symmetrically for 1-2 Mb, providing an estimate for the range of in vivo loop extrusion. Asymmetric CTCF binding deflects the angle of jet propagation as experimental evidence that cohesin-mediated loop extrusion can switch from bi- to unidirectional and is controlled independently in both directions. These data offer new insights into the physiological behavior of in vivo cohesin-mediated loop extrusion and further our understanding of the principles that underlie genome organization.


Assuntos
Cromatina , Proteínas Cromossômicas não Histona , Animais , Cromatina/genética , Fator de Ligação a CCCTC/genética , Fator de Ligação a CCCTC/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Polímeros/metabolismo , Mamíferos/metabolismo , Coesinas
5.
Cell ; 152(4): 873-83, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23415233

RESUMO

Embryonic stem cells (ESCs) can instruct the conversion of differentiated cells toward pluripotency following cell-to-cell fusion by a mechanism that is rapid but poorly understood. Here, we used centrifugal elutriation to enrich for mouse ESCs at sequential stages of the cell cycle and showed that ESCs in S/G2 phases have an enhanced capacity to dominantly reprogram lymphocytes and fibroblasts in heterokaryon and hybrid assays. Reprogramming success was associated with an ability to induce precocious nucleotide incorporation within the somatic partner nuclei in heterokaryons. BrdU pulse-labeling experiments revealed that virtually all successfully reprogrammed somatic nuclei, identified on the basis of Oct4 re-expression, had undergone DNA synthesis within 24 hr of fusion with ESCs. This was essential for successful reprogramming because drugs that inhibited DNA polymerase activity effectively blocked pluripotent conversion. These data indicate that nucleotide incorporation is an early and critical event in the epigenetic reprogramming of somatic cells in experimental ESC-heterokaryons.


Assuntos
Replicação do DNA , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Linfócitos B/citologia , Fusão Celular , Núcleo Celular/metabolismo , Reprogramação Celular , Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Humanos , Camundongos , Nucleotídeos/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo
6.
Trends Immunol ; 41(11): 994-1005, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33036908

RESUMO

The remarkable process of light emission by living organisms has fascinated mankind for thousands of years. A recent expansion in the repertoire of catalytic luciferase enzymes, coupled with the discovery of the genes and pathways that encode different luciferin substrates, means that bioluminescence imaging (BLI) is set to revolutionize longitudinal and dynamic studies of gene control within biomedicine, including the regulation of immune responses. In this review article, we summarize recent advances in bioluminescence-based imaging approaches that promise to enlighten our understanding of in vivo gene and epigenetic control within the immune system.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica , Sistema Imunitário , Padrões de Herança , Medições Luminescentes , Animais , Humanos , Luciferases/genética , Luciferases/metabolismo
7.
Cell ; 132(3): 422-33, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18237772

RESUMO

Cohesins mediate sister chromatid cohesion, which is essential for chromosome segregation and postreplicative DNA repair. In addition, cohesins appear to regulate gene expression and enhancer-promoter interactions. These noncanonical functions remained unexplained because knowledge of cohesin-binding sites and functional interactors in metazoans was lacking. We show that the distribution of cohesins on mammalian chromosome arms is not driven by transcriptional activity, in contrast to S. cerevisiae. Instead, mammalian cohesins occupy a subset of DNase I hypersensitive sites, many of which contain sequence motifs resembling the consensus for CTCF, a DNA-binding protein with enhancer blocking function and boundary-element activity. We find cohesins at most CTCF sites and show that CTCF is required for cohesin localization to these sites. Recruitment by CTCF suggests a rationale for noncanonical cohesin functions and, because CTCF binding is sensitive to DNA methylation, allows cohesin positioning to integrate DNA sequence and epigenetic state.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos de Mamíferos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Animais , Sequência de Bases , Fator de Ligação a CCCTC , Diferenciação Celular , Linhagem Celular , Montagem e Desmontagem da Cromatina , Imunoprecipitação da Cromatina , Citocinas/genética , Desoxirribonuclease I/metabolismo , Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Linfócitos T/citologia , Linfócitos T/metabolismo , Coesinas
8.
Genes Dev ; 29(1): 23-38, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25561493

RESUMO

Cohesin is implicated in establishing and maintaining pluripotency. Whether this is because of essential cohesin functions in the cell cycle or in gene regulation is unknown. Here we tested cohesin's contribution to reprogramming in systems that reactivate the expression of pluripotency genes in the absence of proliferation (embryonic stem [ES] cell heterokaryons) or DNA replication (nuclear transfer). Contrary to expectations, cohesin depletion enhanced the ability of ES cells to initiate somatic cell reprogramming in heterokaryons. This was explained by increased c-Myc (Myc) expression in cohesin-depleted ES cells, which promoted DNA replication-dependent reprogramming of somatic fusion partners. In contrast, cohesin-depleted somatic cells were poorly reprogrammed in heterokaryons, due in part to defective DNA replication. Pluripotency gene induction was rescued by Myc, which restored DNA replication, and by nuclear transfer, where reprogramming does not require DNA replication. These results redefine cohesin's role in pluripotency and reveal a novel function for Myc in promoting the replication-dependent reprogramming of somatic nuclei.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Reprogramação Celular/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Replicação do DNA , Humanos , Camundongos , Dados de Sequência Molecular , Oócitos/metabolismo , Células-Tronco Pluripotentes/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Xenopus , Coesinas
9.
PLoS Biol ; 17(4): e2006506, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30978178

RESUMO

The differentiation of self-renewing progenitor cells requires not only the regulation of lineage- and developmental stage-specific genes but also the coordinated adaptation of housekeeping functions from a metabolically active, proliferative state toward quiescence. How metabolic and cell-cycle states are coordinated with the regulation of cell type-specific genes is an important question, because dissociation between differentiation, cell cycle, and metabolic states is a hallmark of cancer. Here, we use a model system to systematically identify key transcriptional regulators of Ikaros-dependent B cell-progenitor differentiation. We find that the coordinated regulation of housekeeping functions and tissue-specific gene expression requires a feedforward circuit whereby Ikaros down-regulates the expression of Myc. Our findings show how coordination between differentiation and housekeeping states can be achieved by interconnected regulators. Similar principles likely coordinate differentiation and housekeeping functions during progenitor cell differentiation in other cell lineages.


Assuntos
Linfócitos B/citologia , Genes myc , Células Precursoras de Linfócitos B/citologia , Animais , Linfócitos B/metabolismo , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Linhagem da Célula , Bases de Dados Genéticas , Regulação para Baixo , Regulação da Expressão Gênica , Genes Essenciais , Humanos , Fator de Transcrição Ikaros/metabolismo , Ativação Linfocitária , Camundongos , Células Precursoras de Linfócitos B/metabolismo , Fatores de Transcrição/metabolismo
10.
Mol Cell ; 49(6): 1023-33, 2013 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-23453809

RESUMO

Genomic imprinting directs the allele-specific marking and expression of loci according to their parental origin. Differential DNA methylation at imprinted control regions (ICRs) is established in gametes and, although largely preserved through development, can be experimentally reset by fusing somatic cells with embryonic germ cell (EGC) lines. Here, we show that the Ten-Eleven Translocation proteins Tet1 and Tet2 participate in the efficient erasure of imprints in this model system. The fusion of B cells with EGCs initiates pluripotent reprogramming, in which rapid re-expression of Oct4 is accompanied by an accumulation of 5-hydroxymethylcytosine (5hmC) at several ICRs. Tet2 was required for the efficient reprogramming capacity of EGCs, whereas Tet1 was necessary to induce 5-methylcytosine oxidation specifically at ICRs. These data show that the Tet1 and Tet2 proteins have discrete roles in cell-fusion-mediated pluripotent reprogramming and imprint erasure in somatic cells.


Assuntos
Fusão Celular , Proteínas de Ligação a DNA/fisiologia , Impressão Genômica , Proteínas Proto-Oncogênicas/fisiologia , 5-Metilcitosina/análogos & derivados , Animais , Linfócitos B/citologia , Sequência de Bases , Linhagem Celular , Citosina/análogos & derivados , Citosina/metabolismo , Metilação de DNA , Dioxigenases , Células-Tronco Embrionárias/citologia , Expressão Gênica , Células Germinativas/citologia , Proteínas de Fluorescência Verde/biossíntese , Humanos , Fator de Crescimento Insulin-Like II/genética , Camundongos , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Proteínas/metabolismo , RNA Longo não Codificante/genética , Análise de Sequência de DNA
11.
Nucleic Acids Res ; 45(21): e174, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28981838

RESUMO

Mouse embryonic stem (ES) cells are a popular model system to study biological processes, though uncovering recessive phenotypes requires inactivating both alleles. Building upon resources from the International Knockout Mouse Consortium (IKMC), we developed a targeting vector for second allele inactivation in conditional-ready IKMC 'knockout-first' ES cell lines. We applied our technology to several epigenetic regulators, recovering bi-allelic targeted clones with a high efficiency of 60% and used Flp recombinase to restore expression in two null cell lines to demonstrate how our system confirms causality through mutant phenotype reversion. We designed our strategy to select against re-targeting the 'knockout-first' allele and identify essential genes in ES cells, including the histone methyltransferase Setdb1. For confirmation, we exploited the flexibility of our system, enabling tamoxifen inducible conditional gene ablation while controlling for genetic background and tamoxifen effects. Setdb1 ablated ES cells exhibit severe growth inhibition, which is not rescued by exogenous Nanog expression or culturing in naive pluripotency '2i' media, suggesting that the self-renewal defect is mediated through pluripotency network independent pathways. Our strategy to generate null mutant mouse ES cells is applicable to thousands of genes and repurposes existing IKMC Intermediate Vectors.


Assuntos
Alelos , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Técnicas de Inativação de Genes/métodos , Animais , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Expressão Gênica , Vetores Genéticos , Histona-Lisina N-Metiltransferase/genética , Camundongos , Complexo Repressor Polycomb 2/genética
12.
Genome Res ; 25(4): 504-13, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25677180

RESUMO

In addition to mediating sister chromatid cohesion during the cell cycle, the cohesin complex associates with CTCF and with active gene regulatory elements to form long-range interactions between its binding sites. Genome-wide chromosome conformation capture had shown that cohesin's main role in interphase genome organization is in mediating interactions within architectural chromosome compartments, rather than specifying compartments per se. However, it remains unclear how cohesin-mediated interactions contribute to the regulation of gene expression. We have found that the binding of CTCF and cohesin is highly enriched at enhancers and in particular at enhancer arrays or "super-enhancers" in mouse thymocytes. Using local and global chromosome conformation capture, we demonstrate that enhancer elements associate not just in linear sequence, but also in 3D, and that spatial enhancer clustering is facilitated by cohesin. The conditional deletion of cohesin from noncycling thymocytes preserved enhancer position, H3K27ac, H4K4me1, and enhancer transcription, but weakened interactions between enhancers. Interestingly, ∼ 50% of deregulated genes reside in the vicinity of enhancer elements, suggesting that cohesin regulates gene expression through spatial clustering of enhancer elements. We propose a model for cohesin-dependent gene regulation in which spatial clustering of enhancer elements acts as a unified mechanism for both enhancer-promoter "connections" and "insulation."


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Família Multigênica/genética , Proteínas Repressoras/metabolismo , Timócitos/citologia , Animais , Sítios de Ligação/genética , Fator de Ligação a CCCTC , Células Cultivadas , Histonas/genética , Camundongos , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Coesinas
13.
Mol Cell ; 38(5): 675-88, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20542000

RESUMO

Polycomb proteins maintain cell identity by repressing the expression of developmental regulators specific for other cell types. Polycomb repressive complex-2 (PRC2) catalyzes trimethylation of histone H3 lysine-27 (H3K27me3). Although repressed, PRC2 targets are generally associated with the transcriptional initiation marker H3K4me3, but the significance of this remains unclear. Here, we identify a class of short RNAs, approximately 50-200 nucleotides in length, transcribed from the 5' end of polycomb target genes in primary T cells and embryonic stem cells. Short RNA transcription is associated with RNA polymerase II and H3K4me3, occurs in the absence of mRNA transcription, and is independent of polycomb activity. Short RNAs form stem-loop structures resembling PRC2 binding sites in Xist, interact with PRC2 through SUZ12, cause gene repression in cis, and are depleted from polycomb target genes activated during cell differentiation. We propose that short RNAs play a role in the association of PRC2 with its target genes.


Assuntos
RNA/metabolismo , Proteínas Repressoras/metabolismo , Transcrição Gênica , Animais , Sequência de Bases , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Histonas/genética , Histonas/metabolismo , Lisina/metabolismo , Camundongos , Dados de Sequência Molecular , Neurônios/citologia , Neurônios/fisiologia , Conformação de Ácido Nucleico , Proteínas do Grupo Polycomb , Regiões Promotoras Genéticas , RNA/química , RNA/genética , Proteínas Repressoras/genética , Linfócitos T/citologia , Linfócitos T/fisiologia
14.
PLoS Genet ; 11(2): e1005020, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25714103

RESUMO

The development and homeostasis of multicellular organisms relies on gene regulation within individual constituent cells. Gene regulatory circuits that increase the robustness of gene expression frequently incorporate microRNAs as post-transcriptional regulators. Computational approaches, synthetic gene circuits and observations in model organisms predict that the co-regulation of microRNAs and their target mRNAs can reduce cell-to-cell variability in the expression of target genes. However, whether microRNAs directly regulate variability of endogenous gene expression remains to be tested in mammalian cells. Here we use quantitative flow cytometry to show that microRNAs impact on cell-to-cell variability of protein expression in developing mouse thymocytes. We find two distinct mechanisms that control variation in the activation-induced expression of the microRNA target CD69. First, the expression of miR-17 and miR-20a, two members of the miR-17-92 cluster, is co-regulated with the target mRNA Cd69 to form an activation-induced incoherent feed-forward loop. Another microRNA, miR-181a, acts at least in part upstream of the target mRNA Cd69 to modulate cellular responses to activation. The ability of microRNAs to render gene expression more uniform across mammalian cell populations may be important for normal development and for disease.


Assuntos
Sobrevivência Celular/genética , MicroRNAs/genética , Biossíntese de Proteínas/genética , Timócitos/metabolismo , Animais , Linhagem Celular Tumoral , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Camundongos , RNA Mensageiro/biossíntese
15.
Nature ; 476(7361): 467-71, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21832993

RESUMO

Cohesin enables post-replicative DNA repair and chromosome segregation by holding sister chromatids together from the time of DNA replication in S phase until mitosis. There is growing evidence that cohesin also forms long-range chromosomal cis-interactions and may regulate gene expression in association with CTCF, mediator or tissue-specific transcription factors. Human cohesinopathies such as Cornelia de Lange syndrome are thought to result from impaired non-canonical cohesin functions, but a clear distinction between the cell-division-related and cell-division-independent functions of cohesion--as exemplified in Drosophila--has not been demonstrated in vertebrate systems. To address this, here we deleted the cohesin locus Rad21 in mouse thymocytes at a time in development when these cells stop cycling and rearrange their T-cell receptor (TCR) α locus (Tcra). Rad21-deficient thymocytes had a normal lifespan and retained the ability to differentiate, albeit with reduced efficiency. Loss of Rad21 led to defective chromatin architecture at the Tcra locus, where cohesion-binding sites flank the TEA promoter and the Eα enhancer, and demarcate Tcra from interspersed Tcrd elements and neighbouring housekeeping genes. Cohesin was required for long-range promoter-enhancer interactions, Tcra transcription, H3K4me3 histone modifications that recruit the recombination machinery and Tcra rearrangement. Provision of pre-rearranged TCR transgenes largely rescued thymocyte differentiation, demonstrating that among thousands of potential target genes across the genome, defective Tcra rearrangement was limiting for the differentiation of cohesin-deficient thymocytes. These findings firmly establish a cell-division-independent role for cohesin in Tcra locus rearrangement and provide a comprehensive account of the mechanisms by which cohesin enables cellular differentiation in a well-characterized mammalian system.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Proteínas Cromossômicas não Histona/metabolismo , Rearranjo Gênico do Linfócito T , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Timo/citologia , Animais , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/deficiência , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Rearranjo Gênico do Linfócito T/genética , Genes RAG-1/genética , Camundongos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Recombinases/metabolismo , Timo/metabolismo , Transcrição Gênica , Coesinas
16.
Genes Dev ; 23(22): 2625-38, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19933152

RESUMO

Satb1 and the closely related Satb2 proteins regulate gene expression and higher-order chromatin structure of multigene clusters in vivo. In examining the role of Satb proteins in murine embryonic stem (ES) cells, we find that Satb1(-/-) cells display an impaired differentiation potential and augmented expression of the pluripotency determinants Nanog, Klf4, and Tbx3. Metastable states of self-renewal and differentiation competence have been attributed to heterogeneity of ES cells in the expression of Nanog. Satb1(-/-) cultures have a higher proportion of Nanog(high) cells, and an increased potential to reprogram human B lymphocytes in cell fusion experiments. Moreover, Satb1-deficient ES cells show an increased expression of Satb2, and we find that forced Satb2 expression in wild-type ES cells antagonizes differentiation-associated silencing of Nanog and enhances the induction of NANOG in cell fusions with human B lymphocytes. An antagonistic function of Satb1 and Satb2 is also supported by the almost normal differentiation potential of Satb1(-/-)Satb2(-/-) ES cells. Taken together with the finding that both Satb1 and Satb2 bind the Nanog locus in vivo, our data suggest that the balance of Satb1 and Satb2 contributes to the plasticity of Nanog expression and ES cell pluripotency.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Proteína Homeobox Nanog
17.
Genome Res ; 23(12): 2066-77, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24002784

RESUMO

Chromosome conformation capture approaches have shown that interphase chromatin is partitioned into spatially segregated Mb-sized compartments and sub-Mb-sized topological domains. This compartmentalization is thought to facilitate the matching of genes and regulatory elements, but its precise function and mechanistic basis remain unknown. Cohesin controls chromosome topology to enable DNA repair and chromosome segregation in cycling cells. In addition, cohesin associates with active enhancers and promoters and with CTCF to form long-range interactions important for gene regulation. Although these findings suggest an important role for cohesin in genome organization, this role has not been assessed on a global scale. Unexpectedly, we find that architectural compartments are maintained in noncycling mouse thymocytes after genetic depletion of cohesin in vivo. Cohesin was, however, required for specific long-range interactions within compartments where cohesin-regulated genes reside. Cohesin depletion diminished interactions between cohesin-bound sites, whereas alternative interactions between chromatin features associated with transcriptional activation and repression became more prominent, with corresponding changes in gene expression. Our findings indicate that cohesin-mediated long-range interactions facilitate discrete gene expression states within preexisting chromosomal compartments.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/fisiologia , Regulação da Expressão Gênica , Proteínas Repressoras/metabolismo , Timócitos/metabolismo , Animais , Fator de Ligação a CCCTC , Ciclo Celular/genética , Cromossomos de Mamíferos , Proteínas de Ligação a DNA , Dosagem de Genes , Genoma , Modelos Lineares , Camundongos , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/metabolismo , Coesinas
18.
Nature ; 467(7311): E3-4; discussion E5, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20811409

RESUMO

The contribution of REST to embryonic stem (ES) cell pluripotency has been uncertain. Two years ago, Singh et al. claimed that Rest(+/-) and REST knock-down ES cells expressed reduced levels of pluripotency markers, in contrast to a prior and subsequent reports. To understand the basis of this difference, we analysed the YHC334 (YHC) and RRC160 (RRC) gene-trap ES cell lines used by Singh et al., obtained directly from BayGenomics. Both REST mutant lines generated REST-betaGeo fusion proteins, but expressed pluripotency genes at levels similar to appropriately matched parental wild ES cells, consistent with expression being REST-independent.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Repressoras/genética , Animais , Linhagem Celular , Camundongos , Mutagênese Insercional , Proteínas Recombinantes de Fusão/genética
19.
Development ; 139(21): 3891-6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23048179

RESUMO

Understanding the basis of epigenetic memory is a fast-moving challenge in modern biology. At a recent Company of Biologists Workshop held at Steyning's historic Wiston House, thirty researchers led by John Gurdon interrogated three central questions: how are cell type-specific programs generated, what mechanisms duplicate this programmatic information as cells divide, and how does epigenetics contribute to trans-generational inheritance? We report some of the emerging themes arising from this debate.


Assuntos
Epigênese Genética/genética , Hereditariedade/genética , Animais , Centrômero/genética , Cromatina/genética , Congressos como Assunto , Humanos , Mitose/genética , Modelos Biológicos
20.
Blood ; 121(10): 1769-82, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23303821

RESUMO

Ikaros family DNA-binding proteins are critical regulators of B-cell development. Because the current knowledge of Ikaros targets in B-cell progenitors is limited, we have identified genes that are bound and regulated by Ikaros in pre-B cells. To elucidate the role of Ikaros in B-cell lineage specification and differentiation, we analyzed the differential expression of Ikaros targets during the progression of multipotent to lymphoid-restricted progenitors, B- and T-cell lineage specification, and progression along the B-cell lineage. Ikaros targets accounted for one-half of all genes up-regulated during B-cell lineage specification in vivo, explaining the essential role of Ikaros in this process. Expression of the Ikaros paralogs Ikzf1 and Ikzf3 increases incrementally during B-cell progenitor differentiation, and, remarkably, inducible Ikaros expression in cycling pre-B cells was sufficient to drive transcriptional changes resembling the differentiation of cycling to resting pre-Bcells in vivo. The data suggest that Ikaros transcription factor dosage drives the progression of progenitors along a predetermined lineage by regulating multiple targets in key pathways, including pre-B­cell receptor signaling, cell cycle progression, and lymphocyte receptor rearrangement.Our approachmay be of general use to map the contribution of transcription factors to cell lineage commitment and differentiation.


Assuntos
Linfócitos B/citologia , Diferenciação Celular , Linhagem da Célula , Genoma , Fator de Transcrição Ikaros/metabolismo , Células Precursoras de Linfócitos B/citologia , Fatores de Transcrição/metabolismo , Animais , Linfócitos B/metabolismo , Sítios de Ligação , Ciclo Celular , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica , Fator de Transcrição Ikaros/genética , Ativação Linfocitária , Camundongos , Células Precursoras de Linfócitos B/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA