Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 132(3): 422-33, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18237772

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas de los Mamíferos/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Factor de Unión a CCCTC , Diferenciación Celular , Línea Celular , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Citocinas/genética , Desoxirribonucleasa I/metabolismo , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Linfocitos T/citología , Linfocitos T/metabolismo , Cohesinas
2.
Cell ; 132(5): 860-74, 2008 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-18329371

RESUMEN

To explore the role of Dicer-dependent control mechanisms in B lymphocyte development, we ablated this enzyme in early B cell progenitors. This resulted in a developmental block at the pro- to pre-B cell transition. Gene-expression profiling revealed a miR-17 approximately 92 signature in the 3'UTRs of genes upregulated in Dicer-deficient pro-B cells; a top miR-17 approximately 92 target, the proapoptotic molecule Bim, was highly upregulated. Accordingly, B cell development could be partially rescued by ablation of Bim or transgenic expression of the prosurvival protein Bcl-2. This allowed us to assess the impact of Dicer deficiency on the V(D)J recombination program in developing B cells. We found intact Ig gene rearrangements in immunoglobulin heavy (IgH) and kappa chain loci, but increased sterile transcription and usage of D(H) elements of the DSP family in IgH, and increased N sequence addition in Igkappa due to deregulated transcription of the terminal deoxynucleotidyl transferase gene.


Asunto(s)
Diversidad de Anticuerpos , Linfocitos B/citología , Supervivencia Celular , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Regiones no Traducidas 3'/química , Regiones no Traducidas 3'/metabolismo , Animales , Northern Blotting , Perfilación de la Expresión Génica , Reordenamiento Génico de Linfocito B , Inmunoglobulinas/genética , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III , Organismos Libres de Patógenos Específicos
3.
J Immunol ; 195(12): 5667-77, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26538392

RESUMEN

CD4(+) regulatory T cells (Tregs) are essential for controlling immune responses and preventing autoimmunity. Their development requires regulation of gene expression by microRNAs (miRNAs). To understand miRNA function in Treg development, we searched for important miRNAs and their relevant target genes. Of the more abundantly expressed miRNAs in Tregs, only miR-15b/16, miR-24, and miR-29a impacted the production of in vitro-induced Tregs (iTregs) in overexpression and blocking experiments. miRNA mimics for these significantly enhanced the induction of iTregs in Dicer(-/-) CD4(+) T cells. Furthermore, the overexpression of miR-15b/16 in conventional CD4(+) T cells adoptively transferred into Rag2(-/-) mice increased the in vivo development of peripheral Tregs and diminished the severity of autoimmune colitis. In searching for targets of miR-15b/16, we observed that the mammalian target of rapamycin (mTOR) signaling pathway was enhanced in Dicer(-/-) CD4(+) T cells, and its pharmacological inhibition restored induction of iTregs. Suppression of mTOR signaling is essential for induction of iTregs from naive CD4(+) T cells, and the mTORC2 component, Rictor, contained a functional target site for miR-15b/16. Rictor was more abundantly expressed in Dicer(-/-) T cells as was mTOR, and their expression was downregulated by the overexpression of miR-15b/16. This led to a reduction in mTOR signaling, as measured by phosphorylation of the downstream target, ribosomal protein S6. Finally, knockdown of Rictor by small interfering RNAs enhanced Treg induction in Dicer(-/-) CD4(+) T cells. Therefore, an important mechanism of miRNA regulation of Treg development is through regulation of the mTOR signaling pathway.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Proteínas Portadoras/metabolismo , Colitis/inmunología , MicroARNs/metabolismo , Ribonucleasa III/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR/metabolismo , Traslado Adoptivo , Animales , Proteínas Portadoras/genética , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , ARN Interferente Pequeño/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Ribonucleasa III/genética , Transducción de Señal
4.
Immunology ; 149(1): 74-86, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27278750

RESUMEN

MicroRNAs (miRNAs) regulate many aspects of helper T cell (Th) development and function. Here we found that they are required for the suppression of interleukin-9 (IL-9) expression in Th9 cells and other Th subsets. Two highly related miRNAs (miR-15b and miR-16) that we previously found to play an important role in regulatory T (Treg) cell differentiation were capable of suppressing IL-9 expression when they were over-expressed in Th9 cells. We used these miRNAs as tools to identify novel regulators of IL-9 expression and found that they could regulate the expression of Epas1, which encodes hypoxia-inducible factor (HIF)-2α. HIF proteins regulate metabolic pathway usage that is important in determining appropriate Th differentiation. The related protein, HIF-1α enhances Th17 differentiation and inhibits Treg cell differentiation. Here we found that HIF-2α was required for IL-9 expression in Th9 cells, but its expression was not sufficient in other Th subsets. Furthermore, HIF-2α suppressed Treg cell differentiation like HIF-1α, demonstrating both similar and distinct roles of the HIF proteins in Th differentiation and adding a further dimension to their function. Ironically, even though miR-15b and miR-16 suppressed HIF-2α expression in Treg cells, inhibiting their function in Treg cells did not lead to an increase in IL-9 expression. Therefore, the physiologically relevant miRNAs that regulate IL-9 expression in Treg cells and other subsets remain unknown. Nevertheless, the analysis of miR-15b and miR-16 function led to the discovery of the importance of HIF-2α so this work demonstrated the utility of studying miRNA function to identify novel regulatory pathways in helper T-cell development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Interleucina-9/metabolismo , MicroARNs/genética , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Células HEK293 , Humanos , Interleucina-9/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ribonucleasa III/genética
5.
Cell Physiol Biochem ; 39(4): 1360-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27607422

RESUMEN

BACKGROUND/AIMS: Activation of T cell receptors (TCRs) in CD4+ T cells leads to a cascade of signalling reactions including increase of intracellular calcium (Ca2+) levels with subsequent Ca2+ dependent stimulation of gene expression, proliferation, cell motility and cytokine release. The increase of cytosolic Ca2+ results from intracellular Ca2+ release with subsequent activation of store-operated Ca2+ entry (SOCE). Previous studies suggested miRNAs are required for the development and functions of CD4+ T cells. An enzyme called Dicer is required during the process of manufacturing mature miRNAs from the precursor miRNAs. In this study, we explored whether loss of Dicer in CD4+ T cells affects SOCE and thus Ca2+ dependent regulation of cellular functions. METHODS: We tested the expression of Orai1 by q-RT-PCR and flow cytometry. Further, we measured SOCE by an inverted phase-contrast microscope with the Incident-light fluorescence illumination system using Fura-2. Intracellular Ca2+ was also measured by flow cytometry using Ca2+ sensitive dye Fluo-4. RESULTS: We found that in Dicer deficient (DicerΔ/Δ) mice Orai1 was downregulated at mRNA and protein level in CD4+ T cells. Further, SOCE was significantly smaller in DicerΔ/Δ CD4+ T cells than in CD4+ T cells isolated from wild-type (Dicerfl/fl) mice. CONCLUSION: Our data suggest that miRNAs are required for adequate Ca2+ entry into CD4+ T cells and thus triggering of Ca2+ sensitive immune functions.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Calcio/metabolismo , ARN Helicasas DEAD-box/genética , MicroARNs/genética , Proteína ORAI1/genética , ARN Mensajero/genética , Ribonucleasa III/genética , Compuestos de Anilina/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Quelantes del Calcio/metabolismo , ARN Helicasas DEAD-box/deficiencia , Colorantes Fluorescentes/metabolismo , Fura-2/metabolismo , Regulación de la Expresión Génica , Transporte Iónico/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , Proteína ORAI1/metabolismo , Cultivo Primario de Células , ARN Mensajero/metabolismo , Ribonucleasa III/deficiencia , Transducción de Señal , Tapsigargina/farmacología , Xantenos/metabolismo
6.
Blood ; 121(10): 1769-82, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23303821

RESUMEN

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.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular , Linaje de la Célula , Genoma , Factor de Transcripción Ikaros/metabolismo , Células Precursoras de Linfocitos B/citología , Factores de Transcripción/metabolismo , Animales , Linfocitos B/metabolismo , Sitios de Unión , Ciclo Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Factor de Transcripción Ikaros/genética , Activación de Linfocitos , Ratones , Células Precursoras de Linfocitos B/metabolismo , Transducción de Señal , Factores de Transcripción/genética
7.
Nature ; 460(7253): 410-3, 2009 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-19458616

RESUMEN

Cohesin-mediated sister chromatid cohesion is essential for chromosome segregation and post-replicative DNA repair. In addition, evidence from model organisms and from human genetics suggests that cohesin is involved in the control of gene expression. This non-canonical role has recently been rationalized by the findings that mammalian cohesin complexes are recruited to a subset of DNase I hypersensitive sites and to conserved noncoding sequences by the DNA-binding protein CTCF. CTCF functions at insulators (which control interactions between enhancers and promoters) and at boundary elements (which demarcate regions of distinct chromatin structure), and cohesin contributes to its enhancer-blocking activity. The underlying mechanisms remain unknown, and the full spectrum of cohesin functions remains to be determined. Here we show that cohesin forms the topological and mechanistic basis for cell-type-specific long-range chromosomal interactions in cis at the developmentally regulated cytokine locus IFNG. Hence, the ability of cohesin to constrain chromosome topology is used not only for the purpose of sister chromatid cohesion, but also to dynamically define the spatial conformation of specific loci. This new aspect of cohesin function is probably important for normal development and disease.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/genética , Cromosomas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Interferón gamma/genética , Animales , Factor de Unión a CCCTC , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Proteínas de Unión al ADN , Histonas/metabolismo , Humanos , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Cohesinas
8.
Cells ; 12(15)2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37566036

RESUMEN

MicroRNAs (miRNAs) are important regulators of embryonic stem cell (ESC) biology, and their study has identified key regulatory mechanisms. To find novel pathways regulated by miRNAs in ESCs, we undertook a bioinformatics analysis of gene pathways differently expressed in the absence of miRNAs due to the deletion of Dicer, which encodes an RNase that is essential for the synthesis of miRNAs. One pathway that stood out was Ca2+ signaling. Interestingly, we found that Dicer-/- ESCs had no difference in basal cytoplasmic Ca2+ levels but were hyperresponsive when Ca2+ import into the endoplasmic reticulum (ER) was blocked by thapsigargin. Remarkably, the increased Ca2+ response to thapsigargin in ESCs resulted in almost no increase in apoptosis and no differences in stress response pathways, despite the importance of miRNAs in the stress response of other cell types. The increased Ca2+ response in Dicer-/- ESCs was also observed during purinergic receptor activation, demonstrating a physiological role for the miRNA regulation of Ca2+ signaling pathways. In examining the mechanism of increased Ca2+ responsiveness to thapsigargin, neither store-operated Ca2+ entry nor Ca2+ clearance mechanisms from the cytoplasm appeared to be involved. Rather, it appeared to involve an increase in the expression of one isoform of the IP3 receptors (Itpr2). miRNA regulation of Itpr2 expression primarily appeared to be indirect, with transcriptional regulation playing a major role. Therefore, the miRNA regulation of Itpr2 expression offers a unique mechanism to regulate Ca2+ signaling pathways in the physiology of pluripotent stem cells.


Asunto(s)
MicroARNs , Animales , Ratones , MicroARNs/metabolismo , Tapsigargina/farmacología , Diferenciación Celular/genética , Células Madre Embrionarias , Homeostasis
9.
J Exp Med ; 203(11): 2519-27, 2006 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17060477

RESUMEN

Micro RNAs (miRNAs) regulate gene expression at the posttranscriptional level. Here we show that regulatory T (T reg) cells have a miRNA profile distinct from conventional CD4 T cells. A partial T reg cell-like miRNA profile is conferred by the enforced expression of Foxp3 and, surprisingly, by the activation of conventional CD4 T cells. Depleting miRNAs by eliminating Dicer, the RNAse III enzyme that generates functional miRNAs, reduces T reg cell numbers and results in immune pathology. Dicer facilitates, in a cell-autonomous fashion, the development of T reg cells in the thymus and the efficient induction of Foxp3 by transforming growth factor beta. These results suggest that T reg cell development involves Dicer-generated RNAs.


Asunto(s)
Ribonucleasa III/fisiología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Factores de Transcripción Forkhead/biosíntesis , Ratones , MicroARNs/biosíntesis , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Timo/citología , Timo/inmunología , Factor de Crecimiento Transformador beta/fisiología
10.
Proc Natl Acad Sci U S A ; 106(2): 516-21, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19116266

RESUMEN

B cell development requires the coordinated rearrangement of Ig heavy (IgH) and light chain loci (IgL). Most mature B cells express a single B cell receptor of unique specificity, and a central question in immunology concerns the mechanisms that prevent the productive rearrangement of >1 IgH and IgL allele per cell. Probabilistic models of allelic exclusion maintain that simultaneous rearrangement of both alleles is rare, because the likelihood of undergoing rearrangement is low for a given Ig allele. Strong support for this idea came from studies in which a GFP marker was inserted into the Igk locus. In this system, the probability of high-level germ-line transcription and subsequent locus rearrangement appeared to be low in pre-B cells. Readdressing the validity of GFP expression as a reporter for the level of germ-line transcription, we found a striking discordance between GFP transcript and protein levels at the pre-B cell stage, which is explained at least in part by the developmentally regulated usage of 2 alternative Igk-J germ-line promoters. These results question the validity of the kappa-GFP system as evidence for probabilistic models of allelic exclusion.


Asunto(s)
Alelos , Linfocitos B/inmunología , Reordenamiento Génico , Modelos Estadísticos , Receptores de Antígenos de Linfocitos B/genética , Animales , Linfocitos B/citología , Células de la Médula Ósea , Células Cultivadas , Proteínas Fluorescentes Verdes , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Inmunoglobulinas/genética , Ratones , Transcripción Genética
11.
J Exp Med ; 201(9): 1367-73, 2005 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-15867090

RESUMEN

The ribonuclease III enzyme Dicer is essential for the processing of micro-RNAs (miRNAs) and small interfering RNAs (siRNAs) from double-stranded RNA precursors. miRNAs and siRNAs regulate chromatin structure, gene transcription, mRNA stability, and translation in a wide range of organisms. To provide a model system to explore the role of Dicer-generated RNAs in the differentiation of mammalian cells in vivo, we have generated a conditional Dicer allele. Deletion of Dicer at an early stage of T cell development compromised the survival of alphabeta lineage cells, whereas the numbers of gammadelta-expressing thymocytes were not affected. In developing thymocytes, Dicer was not required for the maintenance of transcriptional silencing at pericentromeric satellite sequences (constitutive heterochromatin), the maintenance of DNA methylation and X chromosome inactivation in female cells (facultative heterochromatin), and the stable shutdown of a developmentally regulated gene (developmentally regulated gene silencing). Most remarkably, given that one third of mammalian mRNAs are putative miRNA targets, Dicer seems to be dispensable for CD4/8 lineage commitment, a process in which epigenetic regulation of lineage choice has been well documented. Thus, although Dicer seems to be critical for the development of the early embryo, it may have limited impact on the implementation of some lineage-specific gene expression programs.


Asunto(s)
Apoptosis/fisiología , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Ribonucleasa III/genética , Linfocitos T/fisiología , Animales , Apoptosis/genética , Southern Blotting , Diferenciación Celular/genética , Células Cultivadas , Islas de CpG/fisiología , Metilación de ADN , Heterocromatina/metabolismo , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Proc Natl Acad Sci U S A ; 105(6): 1949-54, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18238902

RESUMEN

Small noncoding RNAs, microRNAs (miRNAs), bind to messenger RNAs through base pairing to suppress gene expression. Despite accumulating evidence that miRNAs play critical roles in various biological processes across diverse organisms, their roles in mammalian skeletal development have not been demonstrated. Here, we show that Dicer, an essential component for biogenesis of miRNAs, is essential for normal skeletal development. Dicer-null growth plates show a progressive reduction in the proliferating pool of chondrocytes, leading to severe skeletal growth defects and premature death of mice. The reduction of proliferating chondrocytes in Dicer-null growth plates is caused by two distinct mechanisms: decreased chondrocyte proliferation and accelerated differentiation into postmitotic hypertrophic chondrocytes. These defects appear to be caused by mechanisms downstream or independent of the Ihh-PTHrP signaling pathway, a pivotal signaling system that regulates chondrocyte proliferation and differentiation. Microarray analysis of Dicer-null chondrocytes showed limited expression changes in miRNA-target genes, suggesting that, in the majority of cases, chondrocytic miRNAs do not directly regulate target RNA abundance. Our results demonstrate the critical role of the Dicer-dependent pathway in the regulation of chondrocyte proliferation and differentiation during skeletal development.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Condrocitos/citología , Ribonucleasa III/metabolismo , Animales , Secuencia de Bases , Desarrollo Óseo , Cartilla de ADN , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , MicroARNs/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III/fisiología
13.
Proc Natl Acad Sci U S A ; 105(22): 7797-802, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18509048

RESUMEN

Regulatory T (Treg) cells safeguard against autoimmunity and immune pathology. Because determinants of the Treg cell fate are not completely understood, we have delineated signaling events that control the de novo expression of Foxp3 in naive peripheral CD4 T cells and in thymocytes. We report that premature termination of TCR signaling and inibition of phosphatidyl inositol 3-kinase (PI3K) p110alpha, p110delta, protein kinase B (Akt), or mammalian target of rapamycin (mTOR) conferred Foxp3 expression and Treg-like gene expression profiles. Conversely, continued TCR signaling and constitutive PI3K/Akt/mTOR activity antagonised Foxp3 induction. At the chromatin level, di- and trimethylation of lysine 4 of histone H3 (H3K4me2 and -3) near the Foxp3 transcription start site (TSS) and within the 5' untranslated region (UTR) preceded active Foxp3 expression and, like Foxp3 inducibility, was lost upon continued TCR stimulation. These data demonstrate that the PI3K/Akt/mTOR signaling network regulates Foxp3 expression.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Regiones no Traducidas 5'/metabolismo , Animales , Factores de Transcripción Forkhead/genética , Histonas/metabolismo , Isoenzimas/metabolismo , Metilación , Ratones , Ratones Endogámicos , MicroARNs/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/agonistas , Transducción de Señal , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Serina-Treonina Quinasas TOR , Sitio de Iniciación de la Transcripción , Factor de Crecimiento Transformador beta/metabolismo
14.
J Vis Exp ; (117)2016 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-27842353

RESUMEN

Helper T cell development and function must be tightly regulated to induce an appropriate immune response that eliminates specific pathogens yet prevents autoimmunity. Many approaches involving different model organisms have been utilized to understand the mechanisms controlling helper T cell development and function. However, studies using mouse models have proven to be highly informative due to the availability of genetic, cellular, and biochemical systems. One genetic approach in mice used by many labs involves retroviral transduction of primary helper T cells. This is a powerful approach due to its relative ease, making it accessible to almost any laboratory with basic skills in molecular biology and immunology. Therefore, multiple genes in wild type or mutant forms can readily be tested for function in helper T cells to understand their importance and mechanisms of action. We have optimized this approach and describe here the protocols for production of high titer retroviruses, isolation of primary murine helper T cells, and their transduction by retroviruses and differentiation toward the different helper subsets. Finally, the use of this approach is described in uncovering mechanisms utilized by microRNAs (miRNAs) to regulate pathways controlling helper T cell development and function.


Asunto(s)
Diferenciación Celular , Linfocitos T Colaboradores-Inductores , Transducción Genética , Animales , Ratones , MicroARNs , Retroviridae
15.
Dev Cell ; 19(2): 207-19, 2010 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-20708584

RESUMEN

The two first cell fate decisions taken in the mammalian embryo generate three distinct cell lineages: one embryonic, the epiblast, and two extraembryonic, the trophoblast and primitive endoderm. miRNAs are essential for early development, but it is not known if they are utilized in the same way in these three lineages. We find that in the pluripotent epiblast they inhibit apoptosis by blocking the expression of the proapoptotic protein Bcl2l11 (Bim) but play little role in the initiation of gastrulation. In contrast, in the trophectoderm, miRNAs maintain the trophoblast stem cell compartment by directly inhibiting expression of Cdkn1a (p21) and Cdkn1c (p57), and in the primitive endoderm, they prevent differentiation by maintaining ERK1/2 phosphorylation through blocking the expression of Mapk inhibitors. Therefore, we show that there are fundamental differences in how stem cells maintain their developmental potential in embryonic and extraembryonic tissues through miRNAs.


Asunto(s)
Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , MicroARNs/metabolismo , Células Madre/fisiología , Animales , Apoptosis/fisiología , Tipificación del Cuerpo , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Linaje de la Célula , Proliferación Celular , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Estratos Germinativos/anatomía & histología , Estratos Germinativos/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , MicroARNs/genética , Ribonucleasa III , Células Madre/citología
16.
Immunol Lett ; 122(1): 37-43, 2009 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-19046990

RESUMEN

Naive CD4 T cells differentiate into functionally distinct T helper (Th) cells subsets or into regulatory T (Treg) cells in response to the cytokine milieu in which they encounter antigen. A recurring theme in post-thymic CD4 T cell differentiation is the cross-regulation of lineage choice by cytokines and transcription factors that are expressed in alternative lineages. For example, TGFbeta induces the de novo expression of the Treg cell signature transcription factor Foxp3, but iTreg differentiation is blocked by high concentrations of the Th2 cytokine IL4. However, whether IL4 can antagonise Foxp3 induction in more physiological settings remains to be addressed. Here we use a co-culture system to demonstrate that IL4 provided by Th2 cells in vitro is sufficient to block Foxp3 induction in naive CD4 T cells. In addition, we find that Foxp3 induction is efficiently blocked not only by the Th2 transcription factor Gata3, but also by PU.1, which is transiently induced during Th2 differentiation. These data suggest that iTreg differentiation may be affected by the polarity of immune responses.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Interleucina-4/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T Reguladores/metabolismo , Células Th2/metabolismo , Transactivadores/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Técnicas de Cocultivo , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Factor de Transcripción STAT6/deficiencia , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células Th2/citología , Células Th2/inmunología , Transactivadores/genética , Transactivadores/inmunología , Activación Transcripcional/inmunología
17.
Development ; 136(4): 525-30, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19144718

RESUMEN

Long noncoding RNAs are implicated in a number of regulatory functions in eukaryotic genomes. The paternally expressed long noncoding RNA (ncRNA) Kcnq1ot1 regulates epigenetic gene silencing in an imprinted gene cluster in cis over a distance of 400 kb in the mouse embryo, whereas the silenced region extends over 780 kb in the placenta. Gene silencing by the Kcnq1ot1 RNA involves repressive histone modifications, including H3K9me2 and H3K27me3, which are partly brought about by the G9a and Ezh2 histone methyltransferases. Here, we show that Kcnq1ot1 is transcribed by RNA polymerase II, is unspliced, is relatively stable and is localised in the nucleus. Analysis of conditional Dicer mutants reveals that the RNAi pathway is not involved in gene silencing in the Kcnq1ot1 cluster. Instead, using RNA/DNA FISH we show that the Kcnq1ot1 RNA establishes a nuclear domain within which the genes that are epigenetically inactivated in cis are frequently found, whereas nearby genes that are not regulated by Kcnq1ot1 are localised outside of the domain. The Kcnq1ot1 RNA domain is larger in the placenta than in the embryo, consistent with more genes in the cluster being silenced in the placenta. Our results show for the first time that autosomal long ncRNAs can establish nuclear domains, which might create a repressive environment for epigenetic silencing of adjacent genes. Long ncRNAs in imprinting clusters and the Xist RNA on the inactive X chromosome thus appear to regulate epigenetic gene silencing by similar mechanisms.


Asunto(s)
Linaje de la Célula , Núcleo Celular/genética , Silenciador del Gen , ARN no Traducido/metabolismo , Animales , Secuencia de Bases , Impresión Genómica , Ratones , Datos de Secuencia Molecular , Interferencia de ARN , Estabilidad del ARN , ARN Largo no Codificante , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN no Traducido/genética
18.
J Exp Med ; 206(11): 2329-37, 2009 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-19841090

RESUMEN

Runx proteins are essential for hematopoiesis and play an important role in T cell development by regulating key target genes, such as CD4 and CD8 as well as lymphokine genes, during the specialization of naive CD4 T cells into distinct T helper subsets. In regulatory T (T reg) cells, the signature transcription factor Foxp3 interacts with and modulates the function of several other DNA binding proteins, including Runx family members, at the protein level. We show that Runx proteins also regulate the initiation and the maintenance of Foxp3 gene expression in CD4 T cells. Full-length Runx promoted the de novo expression of Foxp3 during inducible T reg cell differentiation, whereas the isolated dominant-negative Runt DNA binding domain antagonized de novo Foxp3 expression. Foxp3 expression in natural T reg cells remained dependent on Runx proteins and correlated with the binding of Runx/core-binding factor beta to regulatory elements within the Foxp3 locus. Our data show that Runx and Foxp3 are components of a feed-forward loop in which Runx proteins contribute to the expression of Foxp3 and cooperate with Foxp3 proteins to regulate the expression of downstream target genes.


Asunto(s)
Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Factores de Transcripción Forkhead/metabolismo , Animales , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Subunidades alfa del Factor de Unión al Sitio Principal/química , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Retroalimentación Fisiológica , Genes Dominantes , Ratones , Estructura Terciaria de Proteína , Linfocitos T Reguladores/metabolismo
19.
Epigenetics Chromatin ; 1(1): 2, 2008 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19014663

RESUMEN

BACKGROUND: X chromosome inactivation is the mechanism used in mammals to achieve dosage compensation of X-linked genes in XX females relative to XY males. Chromosome silencing is triggered in cis by expression of the non-coding RNA Xist. As such, correct regulation of the Xist gene promoter is required to establish appropriate X chromosome activity both in males and females. Studies to date have demonstrated co-transcription of an antisense RNA Tsix and low-level sense transcription prior to onset of X inactivation. The balance of sense and antisense RNA is important in determining the probability that a given Xist allele will be expressed, termed the X inactivation choice, when X inactivation commences. RESULTS: Here we investigate further the mechanism of Xist promoter regulation. We demonstrate that both sense and antisense transcription modulate Xist promoter DNA methylation in undifferentiated embryonic stem (ES) cells, suggesting a possible mechanistic basis for influencing X chromosome choice. Given the involvement of sense and antisense RNAs in promoter methylation, we investigate a possible role for the RNA interference (RNAi) pathway. We show that the Xist promoter is hypomethylated in ES cells deficient for the essential RNAi enzyme Dicer, but that this effect is probably a secondary consequence of reduced levels of de novo DNA methyltransferases in these cells. Consistent with this we find that Dicer-deficient XY and XX embryos show appropriate Xist expression patterns, indicating that Xist gene regulation has not been perturbed. CONCLUSION: We conclude that Xist promoter methylation prior to the onset of random X chromosome inactivation is influenced by relative levels of sense and antisense transcription but that this probably occurs independent of the RNAi pathway. We discuss the implications for this data in terms of understanding Xist gene regulation and X chromosome choice in random X chromosome inactivation.

20.
Genome Biol ; 8(8): R169, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17705870

RESUMEN

BACKGROUND: The time of locus replication during S-phase is tightly regulated and correlates with chromatin state. Embryonic stem (ES) cells have an unusual chromatin profile where many developmental regulator genes that are not yet expressed are marked by both active and repressive histone modifications. This poised or bivalent state is also characterized by locus replication in early S-phase in ES cells, while replication timing is delayed in cells with restricted developmental options. RESULTS: Here we used a panel of mutant mouse ES cell lines lacking important chromatin modifiers to dissect the relationship between chromatin structure and replication timing. We show that temporal control of satellite DNA replication is sensitive to loss of a variety of chromatin modifiers, including Mll, Eed, Dnmt1, Suv39h1/h2 and Dicer. The replication times of many single copy loci, including a 5 Mb contiguous region surrounding the Rex1 gene, were retained in chromatin modifier mutant ES cells, although a subset of loci were affected. CONCLUSION: This analysis demonstrates the importance of chromatin modifiers for maintaining correct replication of satellite sequences in pluripotent ES cells and highlights the sensitivity of some single copy loci to the influence of chromatin modifiers. Abundant histone acetylation is shown to correlate well with early replication. Surprisingly, loss of DNA methylation or histone methylation was tolerated by many loci, suggesting that these modifications may be less influential for the timing of euchromatin replication.


Asunto(s)
Cromatina/metabolismo , Replicación del ADN , ADN Satélite/genética , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Acetilación , Animales , Línea Celular , Metilación de ADN , Histonas/metabolismo , Ratones , Mutación , Fase S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA