Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(36): E7526-E7535, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827334

RESUMEN

The human genome contains ∼30,000 CpG islands (CGIs). While CGIs associated with promoters nearly always remain unmethylated, many of the ∼9,000 CGIs lying within gene bodies become methylated during development and differentiation. Both promoter and intragenic CGIs may also become abnormally methylated as a result of genome rearrangements and in malignancy. The epigenetic mechanisms by which some CGIs become methylated but others, in the same cell, remain unmethylated in these situations are poorly understood. Analyzing specific loci and using a genome-wide analysis, we show that transcription running across CGIs, associated with specific chromatin modifications, is required for DNA methyltransferase 3B (DNMT3B)-mediated DNA methylation of many naturally occurring intragenic CGIs. Importantly, we also show that a subgroup of intragenic CGIs is not sensitive to this process of transcription-mediated methylation and that this correlates with their individual intrinsic capacity to initiate transcription in vivo. We propose a general model of how transcription could act as a primary determinant of the patterns of CGI methylation in normal development and differentiation, and in human disease.


Asunto(s)
Diferenciación Celular/genética , Islas de CpG/genética , Metilación de ADN/genética , Transcripción Genética/genética , Animales , Línea Celular , Epigénesis Genética/genética , Genoma Humano/genética , Humanos , Ratones , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN/métodos
2.
Genes Dev ; 25(15): 1583-8, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21828268

RESUMEN

Remote distal enhancers may be located tens or thousands of kilobases away from their promoters. How they control gene expression is still poorly understood. Here, we analyze the influence of a remote enhancer on the balance between repression (Polycomb-PcG) and activation (Trithorax-TrxG) of a developmentally regulated gene associated with a CpG island. We reveal its essential, nonredundant role in clearing the PcG complex and H3K27me3 from the CpG island. In the absence of the enhancer, the H3K27me3 demethylase (JMJD3) is not recruited to the CpG island. We propose a new role of long-range regulatory elements in removing repressive PcG complexes.


Asunto(s)
Elementos de Facilitación Genéticos/fisiología , Proteínas Represoras/metabolismo , Animales , Línea Celular , Células Cultivadas , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Islas de CpG , Eritropoyesis/fisiología , Regulación de la Expresión Génica , Humanos , Proteínas del Grupo Polycomb , Unión Proteica , Proteína Metiltransferasas/metabolismo
3.
Nat Rev Genet ; 13(1): 14-20, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-22179716

RESUMEN

Mouse models have become an invaluable tool for understanding human health and disease owing to our ability to manipulate the mouse genome exquisitely. Recent progress in genomic analysis has led to an increase in the number and type of disease-causing mutations detected and has also highlighted the importance of non-coding regions. As a result, there is increasing interest in creating 'genomically' humanized mouse models, in which entire human genomic loci are transferred into the mouse genome. The technical challenges towards achieving this aim are large but are starting to be tackled with success.


Asunto(s)
Cromosomas Artificiales de los Mamíferos/genética , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Ratones Transgénicos/genética , Transgenes/genética , Animales , Modelos Animales de Enfermedad , Enfermedades Genéticas Congénitas/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Ratones , Regiones Promotoras Genéticas , Recombinación Genética , Elementos Reguladores de la Transcripción
4.
EMBO J ; 31(2): 317-29, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22056776

RESUMEN

The role of DNA sequence in determining chromatin state is incompletely understood. We have previously demonstrated that large chromosomal segments from human cells recapitulate their native chromatin state in mouse cells, but the relative contribution of local sequences versus their genomic context remains unknown. In this study, we compare orthologous chromosomal regions for which the human locus establishes prominent sites of Polycomb complex recruitment in pluripotent stem cells, whereas the corresponding mouse locus does not. Using recombination-mediated cassette exchange at the mouse locus, we establish the primacy of local sequences in the encoding of chromatin state. We show that the signal for chromatin bivalency is redundantly encoded across a bivalent domain and that this reflects competition between Polycomb complex recruitment and transcriptional activation. Furthermore, our results suggest that a high density of unmethylated CpG dinucleotides is sufficient for vertebrate Polycomb recruitment. This model is supported by analysis of DNA methyltransferase-deficient embryonic stem cells.


Asunto(s)
Islas de CpG/fisiología , Regulación de la Expresión Génica/genética , Proteínas Represoras/metabolismo , Globinas alfa/genética , Animales , Células Cultivadas/metabolismo , Cromatina/genética , Mapeo Cromosómico , Cromosomas Humanos Par 16 , Metilación de ADN , ADN Recombinante/genética , Células Madre Embrionarias/metabolismo , Humanos , Ratones , Ratones Transgénicos , Células Madre Pluripotentes/metabolismo , Proteínas del Grupo Polycomb , Recombinación Genética , Secuencias Reguladoras de Ácidos Nucleicos , Especificidad de la Especie , Transcripción Genética
5.
BMC Dev Biol ; 15: 35, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26453549

RESUMEN

BACKGROUND: Pluripotent cells are present in early embryos until the levels of the pluripotency regulator Oct4 drop at the beginning of somitogenesis. Elevating Oct4 levels in explanted post-pluripotent cells in vitro restores their pluripotency. Cultured pluripotent cells can participate in normal development when introduced into host embryos up to the end of gastrulation. In contrast, pluripotent cells efficiently seed malignant teratocarcinomas in adult animals. In humans, extragonadal teratomas and teratocarcinomas are most frequently found in the sacrococcygeal region of neonates, suggesting that these tumours originate from cells in the posterior of the embryo that either reactivate or fail to switch off their pluripotent status. However, experimental models for the persistence or reactivation of pluripotency during embryonic development are lacking. METHODS: We manually injected embryonic stem cells into conceptuses at E9.5 to test whether the presence of pluripotent cells at this stage correlates with teratocarcinoma formation. We then examined the effects of reactivating embryonic Oct4 expression ubiquitously or in combination with Nanog within the primitive streak (PS)/tail bud (TB) using a transgenic mouse line and embryo chimeras carrying a PS/TB-specific heterologous gene expression cassette respectively. RESULTS: Here, we show that pluripotent cells seed teratomas in post-gastrulation embryos. However, at these stages, induced ubiquitous expression of Oct4 does not lead to restoration of pluripotency (indicated by Nanog expression) and tumour formation in utero, but instead causes a severe phenotype in the extending anteroposterior axis. Use of a more restricted T(Bra) promoter transgenic system enabling inducible ectopic expression of Oct4 and Nanog specifically in the posteriorly-located primitive streak (PS) and tail bud (TB) led to similar axial malformations to those induced by Oct4 alone. These cells underwent induction of pluripotency marker expression in Epiblast Stem Cell (EpiSC) explants derived from somitogenesis-stage embryos, but no teratocarcinoma formation was observed in vivo. CONCLUSIONS: Our findings show that although pluripotent cells with teratocarcinogenic potential can be produced in vitro by the overexpression of pluripotency regulators in explanted somitogenesis-stage somatic cells, the in vivo induction of these genes does not yield tumours. This suggests a restrictive regulatory role of the embryonic microenvironment in the induction of pluripotency.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/metabolismo , Teratoma/metabolismo , Teratoma/patología , Animales , Embrión de Mamíferos/patología , Proteínas Fetales/metabolismo , Proteínas de Homeodominio/genética , Humanos , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas de Dominio T Box/metabolismo , Cola (estructura animal)/embriología
6.
Stem Cells ; 31(8): 1511-22, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23649667

RESUMEN

Robust development of the early embryo may benefit from mechanisms that ensure that not all pluripotent cells differentiate at exactly the same time: such mechanisms would build flexibility into the process of lineage allocation. This idea is supported by the observation that pluripotent stem cells differentiate at different rates in vitro. We use a clonal commitment assay to confirm that pluripotent cells commit to differentiate asynchronously even under uniform differentiation conditions. Stochastic variability in expression of the Notch target gene Hes1 has previously been reported to influence neural versus mesodermal differentiation through modulation of Notch activity. Here we report that Hes1 also has an earlier role to delay exit from the pluripotent state into all lineages. The early function of Hes1 to delay differentiation can be explained by an ability of Hes1 to amplify STAT3 responsiveness in a cell-autonomous manner. Variability in Hes1 expression therefore helps to explain why STAT3 responsiveness varies between individual ES cells, and this in turn helps to explain why pluripotent cells commit to differentiate asynchronously.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Receptores Notch/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Diferenciación Celular/fisiología , Regulación hacia Abajo , Humanos , Ratones , Proteína Homeótica Nanog , Transducción de Señal , Factor de Transcripción HES-1 , Transfección
7.
Nature ; 453(7195): 662-6, 2008 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-18449193

RESUMEN

Phosphoinositide 3-kinases (PI3Ks) signal downstream of multiple cell-surface receptor types. Class IA PI3K isoforms couple to tyrosine kinases and consist of a p110 catalytic subunit (p110alpha, p110beta or p110delta), constitutively bound to one of five distinct p85 regulatory subunits. PI3Ks have been implicated in angiogenesis, but little is known about potential selectivity among the PI3K isoforms and their mechanism of action in endothelial cells during angiogenesis in vivo. Here we show that only p110alpha activity is essential for vascular development. Ubiquitous or endothelial cell-specific inactivation of p110alpha led to embryonic lethality at mid-gestation because of severe defects in angiogenic sprouting and vascular remodelling. p110alpha exerts this critical endothelial cell-autonomous function by regulating endothelial cell migration through the small GTPase RhoA. p110alpha activity is particularly high in endothelial cells and preferentially induced by tyrosine kinase ligands (such as vascular endothelial growth factor (VEGF)-A). In contrast, p110beta in endothelial cells signals downstream of G-protein-coupled receptor (GPCR) ligands such as SDF-1alpha, whereas p110delta is expressed at low level and contributes only minimally to PI3K activity in endothelial cells. These results provide the first in vivo evidence for p110-isoform selectivity in endothelial PI3K signalling during angiogenesis.


Asunto(s)
Movimiento Celular , Células Endoteliales/citología , Células Endoteliales/enzimología , Neovascularización Fisiológica , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/genética , Interferencia de ARN , Ratas , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Heridas y Lesiones , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
8.
Nucleic Acids Res ; 40(19): e150, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22753106

RESUMEN

Transgenesis is a cornerstone of molecular biology. The ability to integrate a specifically engineered piece of DNA into the genome of a living system is fundamental to our efforts to understand life and exploit its implications for medicine, nanotechnology and bioprospecting. However, transgenesis has been hampered by position effects and multi-copy integration problems, which are mainly due to the use of small, plasmid-based transgenes. Large transgenes based on native genomic regions cloned into bacterial artificial chromosomes (BACs) circumvent these problems but are prone to fragmentation. Herein, we report that contrary to widely held notions, large BAC-sized constructs do not prohibit transposition. We also report the first reliable method for BAC transgenesis in human embryonic stem cells (hESCs). The PiggyBac or Sleeping Beauty transposon inverted repeats were integrated into BAC vectors by recombineering, followed by co-lipofection with the corresponding transposase in hESCs to generate robust fluorescent protein reporter lines for OCT4, NANOG, GATA4 and PAX6. BAC transposition delivers several advantages, including increased frequencies of single-copy, full-length integration, which will be useful in all transgenic systems but especially in difficult venues like hESCs.


Asunto(s)
Cromosomas Artificiales Bacterianos , Elementos Transponibles de ADN , Técnicas de Transferencia de Gen , Transgenes , Animales , Línea Celular , Células Madre Embrionarias , Dosificación de Gen , Genes Reporteros , Proteínas de Homeodominio/genética , Humanos , Proteínas Luminiscentes/genética , Ratones , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética
9.
PLoS Genet ; 7(11): e1002348, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22072979

RESUMEN

The forkhead transcription factor Foxn1 is indispensable for thymus development, but the mechanisms by which it mediates thymic epithelial cell (TEC) development are poorly understood. To examine the cellular and molecular basis of Foxn1 function, we generated a novel and revertible hypomorphic allele of Foxn1. By varying levels of its expression, we identified a number of features of the Foxn1 system. Here we show that Foxn1 is a powerful regulator of TEC differentiation that is required at multiple intermediate stages of TE lineage development in the fetal and adult thymus. We find no evidence for a role for Foxn1 in TEC fate-choice. Rather, we show it is required for stable entry into both the cortical and medullary TEC differentiation programmes and subsequently is needed at increasing dosage for progression through successive differentiation states in both cortical and medullary TEC. We further demonstrate regulation by Foxn1 of a suite of genes with diverse roles in thymus development and/or function, suggesting it acts as a master regulator of the core thymic epithelial programme rather than regulating a particular aspect of TEC biology. Overall, our data establish a genetics-based model of cellular hierarchies in the TE lineage and provide mechanistic insight relating titration of a single transcription factor to control of lineage progression. Our novel revertible hypomorph system may be similarly applied to analyzing other regulators of development.


Asunto(s)
Médula Suprarrenal/citología , Diferenciación Celular/genética , Linaje de la Célula/genética , Desarrollo Embrionario/genética , Factores de Transcripción Forkhead/metabolismo , Timo/crecimiento & desarrollo , Médula Suprarrenal/metabolismo , Alelos , Animales , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Integrasas/química , Integrasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamoxifeno/química
10.
Nature ; 441(7091): 366-70, 2006 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-16625210

RESUMEN

The eight catalytic subunits of the mammalian phosphoinositide-3-OH kinase (PI(3)K) family form the backbone of an evolutionarily conserved signalling pathway; however, the roles of most PI(3)K isoforms in organismal physiology and disease are unknown. To delineate the role of p110alpha, a ubiquitously expressed PI(3)K involved in tyrosine kinase and Ras signalling, here we generated mice carrying a knockin mutation (D933A) that abrogates p110alpha kinase activity. Homozygosity for this kinase-dead p110alpha led to embryonic lethality. Mice heterozygous for this mutation were viable and fertile, but displayed severely blunted signalling via insulin-receptor substrate (IRS) proteins, key mediators of insulin, insulin-like growth factor-1 and leptin action. Defective responsiveness to these hormones led to reduced somatic growth, hyperinsulinaemia, glucose intolerance, hyperphagia and increased adiposity in mice heterozygous for the D933A mutation. This signalling function of p110alpha derives from its highly selective recruitment and activation to IRS signalling complexes compared to p110beta, the other broadly expressed PI(3)K isoform, which did not contribute to IRS-associated PI(3)K activity. p110alpha was the principal IRS-associated PI(3)K in cancer cell lines. These findings demonstrate a critical role for p110alpha in growth factor and metabolic signalling and also suggest an explanation for selective mutation or overexpression of p110alpha in a variety of cancers.


Asunto(s)
Crecimiento/fisiología , Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Adiposidad , Animales , Peso Corporal , Dominio Catalítico , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I , Ingestión de Alimentos , Pérdida del Embrión/enzimología , Pérdida del Embrión/genética , Pérdida del Embrión/metabolismo , Activación Enzimática , Glucosa/metabolismo , Heterocigoto , Homocigoto , Hiperinsulinismo/metabolismo , Proteínas Sustrato del Receptor de Insulina , Leptina/metabolismo , Ratones , Mutación/genética , Neoplasias/metabolismo , Neoplasias/patología , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Fosfoproteínas/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal
11.
Blood ; 114(19): 4253-60, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19696202

RESUMEN

Previous studies in the mouse have shown that high levels of alpha-globin gene expression in late erythropoiesis depend on long-range, physical interactions between remote upstream regulatory elements and the globin promoters. Using quantitative chromosome conformation capture (q3C), we have now analyzed all interactions between 4 such elements lying 10 to 50 kb upstream of the human alpha cluster and their interactions with the alpha-globin promoter. All of these elements interact with the alpha-globin gene in an erythroid-specific manner. These results were confirmed in a mouse model of human alpha globin expression in which the human cluster replaces the mouse cluster in situ (humanized mouse). We have also shown that expression and all of the long-range interactions depend largely on just one of these elements; removal of the previously characterized major regulatory element (called HS -40) results in loss of all the interactions and alpha-globin expression. Reinsertion of this element at an ectopic location restores both expression and the intralocus interactions. In contrast to other more complex systems involving multiple upstream elements and promoters, analysis of the human alpha-globin cluster during erythropoiesis provides a simple and tractable model to understand the mechanisms underlying long-range gene regulation.


Asunto(s)
Cromosomas Humanos/genética , Globinas alfa/genética , Animales , Secuencia de Bases , Línea Celular , Células Cultivadas , Sondas de ADN/genética , Eritropoyesis/genética , Femenino , Redes Reguladoras de Genes , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Familia de Multigenes , Regiones Promotoras Genéticas , Elementos Reguladores de la Transcripción
12.
Proc Natl Acad Sci U S A ; 105(24): 8292-7, 2008 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-18544649

RESUMEN

The p110 isoforms of phosphoinositide 3-kinase (PI3K) are acutely regulated by extracellular stimuli. The class IA PI3K catalytic subunits (p110alpha, p110beta, and p110delta) occur in complex with a Src homology 2 (SH2) domain-containing p85 regulatory subunit, which has been shown to link p110alpha and p110delta to Tyr kinase signaling pathways. The p84/p101 regulatory subunits of the p110gamma class IB PI3K lack SH2 domains and instead couple p110gamma to G protein-coupled receptors (GPCRs). Here, we show, using small-molecule inhibitors with selectivity for p110beta and cells derived from a p110beta-deficient mouse line, that p110beta is not a major effector of Tyr kinase signaling but couples to GPCRs. In macrophages, both p110beta and p110gamma contributed to Akt activation induced by the GPCR agonist complement 5a, but not by the Tyr kinase ligand colony-stimulating factor-1. In fibroblasts, which express p110beta but not p110gamma, p110beta mediated Akt activation by the GPCR ligands stromal cell-derived factor, sphingosine-1-phosphate, and lysophosphatidic acid but not by the Tyr kinase ligands PDGF, insulin, and insulin-like growth factor 1. Introduction of p110gamma in these cells reduced the contribution of p110beta to GPCR signaling. Taken together, these data show that p110beta and p110gamma can couple redundantly to the same GPCR agonists. p110beta, which shows a much broader tissue distribution than the leukocyte-restricted p110gamma, could thus provide a conduit for GPCR-linked PI3K signaling in the many cell types where p110gamma expression is low or absent.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase I , Complemento C5a/farmacología , Fibroblastos/enzimología , Prueba de Complementación Genética , Isoenzimas/genética , Isoenzimas/metabolismo , Ligandos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/enzimología , Ratones , Ratones Mutantes , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal
13.
J Vis Exp ; (159)2020 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-32449724

RESUMEN

Custom designed endonucleases, such as RNA-guided Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9, enable efficient genome editing in mammalian cells. Here we describe detailed procedures to seamlessly genome edit the hepatocyte nuclear factor 4 alpha (HNF4α) locus as an example in human pluripotent stem cells. Combining a piggyBac-based donor plasmid and the CRISPR-Cas9 nickase mutant in a two-step genetic selection, we demonstrate correct and efficient targeting of the HNF4α locus.


Asunto(s)
Edición Génica/métodos , Células Madre Pluripotentes/metabolismo , Mutación Puntual/genética , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Elementos Transponibles de ADN/genética , Vectores Genéticos/genética , Genoma Humano , Humanos
14.
Neuron ; 48(5): 737-42, 2005 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-16337912

RESUMEN

Voltage-gated sodium channels are concentrated in myelinated nerves at the nodes of Ranvier flanked by paranodal axoglial junctions. Establishment of these essential nodal and paranodal domains is determined by myelin-forming glia, but the mechanisms are not clear. Here, we show that two isoforms of Neurofascin, Nfasc155 in glia and Nfasc186 in neurons, are required for the assembly of these specialized domains. In Neurofascin-null mice, neither paranodal adhesion junctions nor nodal complexes are formed. Transgenic expression of Nfasc155 in the myelinating glia of Nfasc-/- nerves rescues the axoglial adhesion complex by recruiting the axonal proteins Caspr and Contactin to the paranodes. However, in the absence of Nfasc186, sodium channels remain diffusely distributed along the axon. Our study shows that the two major Neurofascins play essential roles in assembling the nodal and paranodal domains of myelinated axons; therefore, they are essential for the transition to saltatory conduction in developing vertebrate nerves.


Asunto(s)
Axones/fisiología , Moléculas de Adhesión Celular/fisiología , Factores de Crecimiento Nervioso/fisiología , Conducción Nerviosa/fisiología , Canales de Sodio/fisiología , Animales , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Espacio Extracelular , Uniones Intercelulares/fisiología , Ratones , Ratones Noqueados/genética , Ratones Transgénicos , Vaina de Mielina/metabolismo , Fibras Nerviosas Mielínicas/fisiología , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Neuroglía/metabolismo , Neuroglía/fisiología , Fenotipo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína/fisiología , Nódulos de Ranvier/fisiología
15.
PLoS Genet ; 2(4): e58, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16628246

RESUMEN

ATRX is an X-encoded member of the SNF2 family of ATPase/helicase proteins thought to regulate gene expression by modifying chromatin at target loci. Mutations in ATRX provided the first example of a human genetic disease associated with defects in such proteins. To better understand the role of ATRX in development and the associated abnormalities in the ATR-X (alpha thalassemia mental retardation, X-linked) syndrome, we conditionally inactivated the homolog in mice, Atrx, at the 8- to 16-cell stage of development. The protein, Atrx, was ubiquitously expressed, and male embryos null for Atrx implanted and gastrulated normally but did not survive beyond 9.5 days postcoitus due to a defect in formation of the extraembryonic trophoblast, one of the first terminally differentiated lineages in the developing embryo. Carrier female mice that inherit a maternal null allele should be affected, since the paternal X chromosome is normally inactivated in extraembryonic tissues. Surprisingly, however, some carrier females established a normal placenta and appeared to escape the usual pattern of imprinted X-inactivation in these tissues. Together these findings demonstrate an unexpected, specific, and essential role for Atrx in the development of the murine trophoblast and present an example of escape from imprinted X chromosome inactivation.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Inactivación del Cromosoma X , Alelos , Animales , Linaje de la Célula , Metilación de ADN , Compensación de Dosificación (Genética) , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Trofoblastos/metabolismo , Proteína Nuclear Ligada al Cromosoma X
16.
iScience ; 16: 206-217, 2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31185456

RESUMEN

During mammalian development, liver differentiation is driven by signals that converge on multiple transcription factor networks. The hepatocyte nuclear factor signaling network is known to be essential for hepatocyte specification and maintenance. In this study, we have generated deletion and point mutants of hepatocyte nuclear factor-4alpha (HNF4α) to precisely evaluate the function of protein domains during hepatocyte specification from human pluripotent stem cells. We demonstrate that nuclear HNF4α is essential for hepatic progenitor specification, and the introduction of point mutations in HNF4α's Small Ubiquitin-like Modifier (SUMO) consensus motif leads to disrupted hepatocyte differentiation. Taking a multiomics approach, we identified key deficiencies in cell biology, which included dysfunctional metabolism, substrate adhesion, tricarboxylic acid cycle flux, microRNA transport, and mRNA processing. In summary, the combination of genome editing and multiomics analyses has provided valuable insight into the diverse functions of HNF4α during pluripotent stem cell entry into the hepatic lineage and during hepatocellular differentiation.

17.
Dev Cell ; 44(3): 362-377.e7, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29290585

RESUMEN

ETV6-RUNX1 is associated with childhood acute B-lymphoblastic leukemia (cALL) functioning as a first-hit mutation that initiates a clinically silent pre-leukemia in utero. Because lineage commitment hierarchies differ between embryo and adult, and the impact of oncogenes is cell-context dependent, we hypothesized that the childhood affiliation of ETV6-RUNX1 cALL reflects its origins in a progenitor unique to embryonic life. We characterize the first emerging B cells in first-trimester human embryos, identifying a developmentally restricted CD19-IL-7R+ progenitor compartment, which transitions from a myeloid to lymphoid program during ontogeny. This developmental series is recapitulated in differentiating human pluripotent stem cells (hPSCs), thereby providing a model for the initiation of cALL. Genome-engineered hPSCs expressing ETV6-RUNX1 from the endogenous ETV6 locus show expansion of the CD19-IL-7R+ compartment, show a partial block in B lineage commitment, and produce proB cells with aberrant myeloid gene expression signatures and potential: features (collectively) consistent with a pre-leukemic state.


Asunto(s)
Linfocitos B/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Desarrollo Embrionario , Regulación Leucémica de la Expresión Génica , Células Madre Pluripotentes Inducidas/patología , Células Mieloides/patología , Proteínas de Fusión Oncogénica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Enfermedad Aguda , Linfocitos B/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Células Mieloides/metabolismo , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Embarazo , Primer Trimestre del Embarazo , Receptores de Interleucina-7 , Transcriptoma
18.
Stem Cell Reports ; 10(6): 1895-1907, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29779895

RESUMEN

Human embryonic stem cells (hESCs) display substantial heterogeneity in gene expression, implying the existence of discrete substates within the stem cell compartment. To determine whether these substates impact fate decisions of hESCs we used a GFP reporter line to investigate the properties of fractions of putative undifferentiated cells defined by their differential expression of the endoderm transcription factor, GATA6, together with the hESC surface marker, SSEA3. By single-cell cloning, we confirmed that substates characterized by expression of GATA6 and SSEA3 include pluripotent stem cells capable of long-term self-renewal. When clonal stem cell colonies were formed from GATA6-positive and GATA6-negative cells, more of those derived from GATA6-positive cells contained spontaneously differentiated endoderm cells than similar colonies derived from the GATA6-negative cells. We characterized these discrete cellular states using single-cell transcriptomic analysis, identifying a potential role for SOX17 in the establishment of the endoderm-biased stem cell state.


Asunto(s)
Autorrenovación de las Células , Endodermo/citología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Biomarcadores , Diferenciación Celular/genética , Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/metabolismo , Perfilación de la Expresión Génica , Genes Reporteros , Humanos , Inmunofenotipificación , Análisis de la Célula Individual/métodos
19.
J Clin Invest ; 111(1): 35-41, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12511586

RESUMEN

Using physiological, pharmacological, and gene disruption approaches, we demonstrate that proteinase-activated receptor-2 (PAR-2) plays a pivotal role in mediating chronic inflammation. Using an adjuvant monoarthritis model of chronic inflammation, joint swelling was substantially inhibited in PAR-2-deficient mice, being reduced by more than fourfold compared with wild-type mice, with virtually no histological evidence of joint damage. Mice heterozygous for PAR-2 gene disruption showed an intermediate phenotype. PAR-2 expression, normally limited to endothelial cells in small arterioles, was substantially upregulated 2 weeks after induction of inflammation, both in synovium and in other periarticular tissues. PAR-2 agonists showed potent proinflammatory effects as intra-articular injection of ASKH95, a novel synthetic PAR-2 agonist, induced prolonged joint swelling and synovial hyperemia. Given the absence of the chronic inflammatory response in the PAR-2-deficient mice, our findings demonstrate a key role for PAR-2 in mediating chronic inflammation, thereby identifying a novel and important therapeutic target for the management of chronic inflammatory diseases such as rheumatoid arthritis.


Asunto(s)
Artritis/metabolismo , Receptores de Trombina/biosíntesis , Receptores de Trombina/fisiología , Regulación hacia Arriba , Alelos , Animales , Cartílago/lesiones , Endotelio/metabolismo , Exones , Fémur/lesiones , Vectores Genéticos , Heterocigoto , Hibridación in Situ , Inflamación , Ratones , Modelos Químicos , Modelos Genéticos , Oligopéptidos/farmacología , Péptidos/farmacología , Fenotipo , Receptor PAR-2 , Receptores de Trombina/agonistas , Recombinación Genética , Factores de Tiempo
20.
Nat Biotechnol ; 21(4): 443-7, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12627172

RESUMEN

The mouse is the leading vertebrate model because its genome can be altered by both random transgenesis and homologous recombination with targeting constructs. Both approaches have been hindered by the size and site limitations implicit in conventional Escherichia coli DNA-engineering methods. Homologous recombination in E. coli, or 'recombineering', has overcome these limitations for bacterial artificial chromosome (BAC) transgenesis. Here we applied Red/ET recombineering (using the lambda Redalpha/Redbeta recombinase pair) to generate a 64 kilobase targeting construct that carried two selectable cassettes permitting the simultaneous mutation of the target gene, Mll, at sites 43 kb apart in one round of mouse embryonic stem (ES) cell targeting. The targeting frequency after dual selection was 6%. Because the two selectable cassettes were flanked by FRT or loxP sites, three more alleles can be generated by site-specific recombination. Our approach represents a simple way to introduce changes at two or more sites in a genetic locus, and thereafter generate allele combinations. The size of BAC templates offers new freedom for the design of targeting constructs. Combined with the use of two selectable cassettes placed far apart, BAC-based targeting constructs may be applicable to tasks such as regional exchanges, deletions, and insertions.


Asunto(s)
Alelos , Cromosomas Artificiales Bacterianos/genética , Marcación de Gen/métodos , Ingeniería Genética/métodos , Genoma , Animales , Células Cultivadas , Escherichia coli/genética , Ratones , Mutagénesis Sitio-Dirigida , Recombinación Genética , Células Madre/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA