Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 147(3): 653-65, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-22036571

RESUMEN

Sensory-motor circuits in the spinal cord are constructed with a fine specificity that coordinates motor behavior, but the mechanisms that direct sensory connections with their motor neuron partners remain unclear. The dorsoventral settling position of motor pools in the spinal cord is known to match the distal-to-proximal position of their muscle targets in the limb, but the significance of invariant motor neuron positioning is unknown. An analysis of sensory-motor connectivity patterns in FoxP1 mutant mice, where motor neuron position has been scrambled, shows that the final pattern of sensory-motor connections is initiated by the projection of sensory axons to discrete dorsoventral domains of the spinal cord without regard for motor neuron subtype or, indeed, the presence of motor neurons. By implication, the clustering and dorsoventral settling position of motor neuron pools serve as a determinant of the pattern of sensory input specificity and thus motor coordination.


Asunto(s)
Tipificación del Cuerpo , Neuronas Motoras/metabolismo , Médula Espinal/embriología , Médula Espinal/metabolismo , Animales , Electromiografía , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Miembro Posterior/inervación , Ratones , Mutación , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Células Receptoras Sensoriales/metabolismo
2.
Cell ; 134(2): 304-16, 2008 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-18662545

RESUMEN

The precision with which motor neurons innervate target muscles depends on a regulatory network of Hox transcription factors that translates neuronal identity into patterns of connectivity. We show that a single transcription factor, FoxP1, coordinates motor neuron subtype identity and connectivity through its activity as a Hox accessory factor. FoxP1 is expressed in Hox-sensitive motor columns and acts as a dose-dependent determinant of columnar fate. Inactivation of Foxp1 abolishes the output of the motor neuron Hox network, reverting the spinal motor system to an ancestral state. The loss of FoxP1 also changes the pattern of motor neuron connectivity, and in the limb motor axons appear to select their trajectories and muscle targets at random. Our findings show that FoxP1 is a crucial determinant of motor neuron diversification and connectivity, and clarify how this Hox regulatory network controls the formation of a topographic neural map.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/metabolismo , Neuronas Motoras/metabolismo , Proteínas Represoras/metabolismo , Médula Espinal/metabolismo , Animales , Embrión de Pollo , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Transgénicos , Neuronas Motoras/citología , Proteínas Represoras/genética , Médula Espinal/citología
3.
Development ; 139(14): 2500-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22675208

RESUMEN

The molecular pathways regulating cell lineage determination and regeneration in epithelial tissues are poorly understood. The secretory epithelium of the lung is required for production of mucus to help protect the lung against environmental insults, including pathogens and pollution, that can lead to debilitating diseases such as asthma and chronic obstructive pulmonary disease. We show that the transcription factors Foxp1 and Foxp4 act cooperatively to regulate lung secretory epithelial cell fate and regeneration by directly restricting the goblet cell lineage program. Loss of Foxp1/4 in the developing lung and in postnatal secretory epithelium leads to ectopic activation of the goblet cell fate program, in part, through de-repression of the protein disulfide isomerase anterior gradient 2 (Agr2). Forced expression of Agr2 is sufficient to promote the goblet cell fate in the developing airway epithelium. Finally, in a model of lung secretory cell injury and regeneration, we show that loss of Foxp1/4 leads to catastrophic loss of airway epithelial regeneration due to default differentiation of secretory cells into the goblet cell lineage. These data demonstrate the importance of Foxp1/4 in restricting cell fate choices during development and regeneration, thereby providing the proper balance of functional epithelial lineages in the lung.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Pulmón/metabolismo , Mucoproteínas/metabolismo , Proteínas Represoras/metabolismo , Animales , Southern Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Inmunoprecipitación de Cromatina , Factores de Transcripción Forkhead/genética , Células Caliciformes/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucoproteínas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas , Reacción en Cadena de la Polimerasa , Regeneración/fisiología , Proteínas Represoras/genética
4.
Nature ; 460(7259): 1093-7, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19657335

RESUMEN

The contribution of changes in cis-regulatory elements or trans-acting factors to interspecies differences in gene expression is not well understood. The mammalian beta-globin loci have served as a model for gene regulation during development. Transgenic mice containing the human beta-globin locus, consisting of the linked embryonic (epsilon), fetal (gamma) and adult (beta) genes, have been used as a system to investigate the temporal switch from fetal to adult haemoglobin, as occurs in humans. Here we show that the human gamma-globin (HBG) genes in these mice behave as murine embryonic globin genes, revealing a limitation of the model and demonstrating that critical differences in the trans-acting milieu have arisen during mammalian evolution. We show that the expression of BCL11A, a repressor of human gamma-globin expression identified by genome-wide association studies, differs between mouse and human. Developmental silencing of the mouse embryonic globin and human gamma-globin genes fails to occur in mice in the absence of BCL11A. Thus, BCL11A is a critical mediator of species-divergent globin switching. By comparing the ontogeny of beta-globin gene regulation in mice and humans, we have shown that alterations in the expression of a trans-acting factor constitute a critical driver of gene expression changes during evolution.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación del Desarrollo de la Expresión Génica , Globinas/genética , Proteínas Nucleares/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Unión al ADN , Embrión de Mamíferos/metabolismo , Evolución Molecular , Feto/metabolismo , Silenciador del Gen , Hematopoyesis , Humanos , Ratones , Proteínas Nucleares/genética , Proteínas Represoras , Especificidad de la Especie , Globinas beta/genética , gamma-Globinas/genética
5.
EMBO J ; 28(6): 711-24, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19214191

RESUMEN

Regulation of BCR signalling strength is crucial for B-cell development and function. Bright is a B-cell-restricted factor that complexes with Bruton's tyrosine kinase (Btk) and its substrate, transcription initiation factor-I (TFII-I), to activate immunoglobulin heavy chain gene transcription in the nucleus. Here we show that a palmitoylated pool of Bright is diverted to lipid rafts of resting B cells where it associates with signalosome components. After BCR ligation, Bright transiently interacts with sumoylation enzymes, blocks calcium flux and phosphorylation of Btk and TFII-I and is then discharged from lipid rafts as a Sumo-I-modified form. The resulting lipid raft concentration of Bright contributes to the signalling threshold of B cells, as their sensitivity to BCR stimulation decreases as the levels of Bright increase. Bright regulates signalling independent of its role in IgH transcription, as shown by specific dominant-negative titration of rafts-specific forms. This study identifies a BCR tuning mechanism in lipid rafts that is regulated by differential post-translational modification of a transcription factor with implications for B-cell tolerance and autoimmunity.


Asunto(s)
Microdominios de Membrana/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Antígenos/metabolismo , Linfocitos B/enzimología , Proteínas de Unión al ADN , Humanos , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Lipoilación , Activación de Linfocitos , Microdominios de Membrana/enzimología , Ratones , Mutación/genética , Oncogenes , Fosforilación , Unión Proteica , Transporte de Proteínas , Proteínas Tirosina Quinasas/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción TFII/metabolismo , Transcripción Genética
6.
Nat Cell Biol ; 8(7): 756-63, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16767080

RESUMEN

The presence of actin in the nucleus has been well established, and several studies have implicated nuclear actin in transcriptional regulation. Neuronal Wiskott-Aldrich syndrome protein (N-WASP) is a member of the WASP family of proteins; these proteins function in the cytoplasm as key regulators of cortical actin filament. Interestingly, N-WASP has also been observed in the nucleus. However, a potential nuclear function for N-WASP has not been established. Here, we report the identification of nuclear N-WASP within a large nuclear-protein complex containing PSF-NonO (polypyrimidine-tract-binding-protein-associated splicing factor-non-Pou-domain octamer-binding protein/p54(nrb)), nuclear actin and RNA polymerase II. The PSF-NonO complex is involved in the regulation of many cellular processes, such as transcription, RNA processing, DNA unwinding and repair. We demonstrate that the interaction of N-WASP with the PSF-NonO complex can couple N-WASP with RNA polymerase II to regulate transcription. We also provide evidence that the potential function of N-WASP in promoting polymerization of nuclear actins has an important role in this process. Based on these results, we propose a nuclear function for N-WASP in transcriptional regulation.


Asunto(s)
Actinas/biosíntesis , Núcleo Celular/metabolismo , ARN Polimerasa II/metabolismo , Elementos Reguladores de la Transcripción/genética , Transcripción Genética/genética , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Núcleo Celular/genética , Proteínas de Unión al ADN , Humanos , Sustancias Macromoleculares/metabolismo , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Factor de Empalme Asociado a PTB , Polímeros/metabolismo , ARN Polimerasa II/genética , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética
7.
Proc Natl Acad Sci U S A ; 107(2): 604-9, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20080725

RESUMEN

The N-linked glycan of immunoglobulin G (IgG) is indispensable for the interaction of the Fc domain with Fcgamma receptors on effector cells and the clearance of target cells via antibody dependent cell-mediated cytotoxicity (ADCC). Escherichia coli expressed, aglycosylated Fc domains bind effector FcgammaRs poorly and cannot elicit ADCC. Using a novel bacterial display/flow cytometric library screening system we isolated Fc variants that bind to FcgammaRI (CD64) with nanomolar affinity. Binding was critically dependent on amino acid substitutions (E382V, and to a lesser extent, M428I) distal to the putative FcgammaRI binding epitope within the CH3 domain. These mutations did not adversely affect its pH-dependent interaction with FcRn in vitro nor its serum persistence in vivo. Remarkably, the anti-Her2 IgG trastuzumab containing the E382V, M428I substitutions and expressed in E. coli exhibited highly selective binding to FcgammaRI but not to the other activating receptors (FcgammaRIIa, FcgammaRIIIa) nor to the inhibitory receptor, FcgammaRIIb. In contrast, the glycosylated version of trastuzumab (E382V, M428I) purified from HEK293T cells bound to all Fcgamma receptors in a manner similar to that of clinical grade trastuzumab. E. coli-purified trastuzumab (E382V, M428I), but not glycosylated trastuzumab (E382V, M428I) or clinical grade trastuzumab, was capable of potentiating the killing of Her2 overexpressing tumor cells with dendritic cells (DCs) as effectors. These results indicate that aglycosylated IgGs can be engineered to display unique FcgammaR selectivity profiles that, in turn, mediate ADCC via mechanisms that are not normally displayed by glycosylated monoclonal antibodies.


Asunto(s)
Células Dendríticas/inmunología , Inmunoglobulina G/genética , Monocitos/inmunología , Receptores de IgG/inmunología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Escherichia coli/inmunología , Citometría de Flujo , Ingeniería Genética , Variación Genética , Glicosilación , Humanos , Modelos Moleculares , Conformación Proteica , Receptores de IgG/química , Proteínas Recombinantes/inmunología , Trastuzumab
8.
Blood ; 115(3): 510-8, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965654

RESUMEN

Proper thymocyte development is required to establish T-cell central tolerance and to generate naive T cells, both of which are essential for T-cell homeostasis and a functional immune system. Here we demonstrate that the loss of transcription factor Foxp1 results in the abnormal development of T cells. Instead of generating naive T cells, Foxp1-deficient single-positive thymocytes acquire an activated phenotype prematurely in the thymus and lead to the generation of peripheral CD4(+) T and CD8(+) T cells that exhibit an activated phenotype and increased apoptosis and readily produce cytokines upon T-cell receptor engagement. These results identify Foxp1 as an essential transcriptional regulator for thymocyte development and the generation of quiescent naive T cells.


Asunto(s)
Diferenciación Celular/genética , Factores de Transcripción Forkhead/fisiología , Proteínas Represoras/fisiología , Linfocitos T/fisiología , Timo/fisiología , Animales , Apoptosis/genética , Apoptosis/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Factores de Transcripción Forkhead/genética , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Proteínas Represoras/genética , Linfocitos T/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología
9.
J Biol Chem ; 285(17): 13304-13, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20185820

RESUMEN

To determine the underlying mechanism of Foxp1/2/4-mediated transcriptional repression, a yeast two-hybrid screen was performed that identified p66beta, a transcriptional repressor and component of the NuRD chromatin-remodeling complex. We show that direct interactions between Foxp1/4 and p66beta are mediated by the CR2 domain within p66beta and the zinc finger/leucine zipper repression domain found in Foxp1/2/4. These direct interactions are functionally relevant as overexpression of p66beta in combination with Foxp factors cooperatively represses Foxp target gene expression, whereas loss of p66 and Foxp factors results in de-repression of endogenous Foxp target genes in lung epithelial cells. Moreover, the NuRD components HDAC1/2 associate in a macromolecular complex with Foxp proteins, and loss of expression or inhibition of HDAC1/2 activity leads to de-repression of Foxp target gene expression. Importantly, we show in vivo that Foxp1 and HDAC2 act cooperatively to regulate expression of the cytoprotective cytokine interleukin-6, which results in increased resistance to hyperoxic lung injury in Foxp1/HDAC2 compound mutant animals. These data reveal an important interaction between the Foxp transcription factors and the NuRD chromatin-remodeling complex that modulates transcriptional repression critical for the lung epithelial injury response.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Hiperoxia/metabolismo , Interleucina-6/biosíntesis , Pulmón/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas Represoras/metabolismo , Mucosa Respiratoria/lesiones , Mucosa Respiratoria/metabolismo , Animales , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Hiperoxia/genética , Interleucina-6/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Ratones Mutantes , Células 3T3 NIH , Proteínas Represoras/genética , Técnicas del Sistema de Dos Híbridos , Dedos de Zinc
10.
Stem Cells ; 28(9): 1560-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20680960

RESUMEN

B-cell regulator of immunoglobulin heavy chain transcription (Bright)/ARID3a, an A+T-rich interaction domain protein, was originally discovered in B lymphocyte lineage cells. However, expression patterns and high lethality levels in knockout mice suggested that it had additional functions. Three independent lines of evidence show that functional inhibition of Bright results in increased developmental plasticity. Bright-deficient cells from two mouse models expressed a number of pluripotency-associated gene products, expanded indefinitely, and spontaneously differentiated into cells of multiple lineages. Furthermore, direct knockdown of human Bright resulted in colonies capable of expressing multiple lineage markers. These data suggest that repression of this single molecule confers adult somatic cells with new developmental options.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/deficiencia , Animales , Biomarcadores/metabolismo , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Dominantes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Interferencia de ARN , Teratoma/genética , Teratoma/metabolismo , Factores de Transcripción/genética
11.
Mol Cell Biol ; 26(6): 2187-201, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16507996

RESUMEN

Bright/ARID3a has been implicated in mitogen- and growth factor-induced up-regulation of immunoglobulin heavy-chain (IgH) genes and in E2F1-dependent G1/S cell cycle progression. For IgH transactivation, Bright binds to nuclear matrix association regions upstream of certain variable region promoters and flanking the IgH intronic enhancer. While Bright protein was previously shown to reside within the nuclear matrix, we show here that a significant amount of Bright resides in the cytoplasm of normal and transformed B cells. Leptomycin B, chromosome region maintenance 1 (CRM1) overexpression, and heterokaryon experiments indicate that Bright actively shuttles between the nucleus and the cytoplasm in a CRM1-dependent manner. We mapped the functional nuclear localization signal to the N-terminal region of REKLES, a domain conserved within ARID3 paralogues. Residues within the C terminus of REKLES contain its nuclear export signal, whose regulation is primarily responsible for Bright shuttling. Growth factor depletion and cell synchronization experiments indicated that Bright shuttling during S phase of the cell cycle leads to an increase in its nuclear abundance. Finally, we show that shuttle-incompetent Bright point mutants, even if sequestered within the nucleus, are incapable of transactivating an IgH reporter gene. Therefore, regulation of Bright's cellular localization appears to be required for its function.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Unión al ADN/metabolismo , Inmunoglobulinas/genética , Transactivadores/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Medio de Cultivo Libre de Suero/farmacología , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Ácidos Grasos Insaturados/farmacología , Regulación de la Expresión Génica , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Regiones de Fijación a la Matriz , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Señales de Localización Nuclear , Oncogenes/efectos de los fármacos , Oncogenes/genética , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Fase S/genética , Transactivadores/efectos de los fármacos , Transactivadores/genética , Factores de Transcripción , Células Tumorales Cultivadas , Proteína Exportina 1
12.
Mol Immunol ; 46(1): 207-11, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18722016

RESUMEN

Intrathymic signals induce the differentiation of immature CD4(+)CD8(+) double positive (DP) thymocytes into mature CD4(+) or CD8(+) single positive (SP) T cells. The transcriptional mechanism by which CD8 lineage is determined is not fully understood. The best evidence, which favors the kinetic signaling/coreceptor reversal model, indicates that signaled DP thymocytes terminate CD8 transcription prior to their subsequent re-initiation of CD8 transcription and ultimate differentiation into CD8SP T cells. We and others have shown that CD8 lineage commitment is severely perturbed in mice in which expression of the transcription factor SATB1 is either conventionally knocked out or T cell-specifically knocked down. Here, we demonstrate that, as with normal thymocytes, cultured SATB1-deficient DP thymocytes inactivate CD8 coreceptor transcription following receipt of signals (PMA plus ionomycin) that mimic TCR-mediated positive selection. However, this terminated CD8 transcription is not re-initiated by signals (IL-7) conducive to CD8 differentiation in SATB1-deficient DP. We show that SATB1 specifically binds to a cis-regulatory element within the CD8 enhancer (E8(III)) known to be required for coreceptor reversal. A requirement in CD8 coreceptor reversal identifies SATB1 as an essential trans-regulator of CD8 lineage fate, whose action may be mediated via recruitment to the E8(III) DP enhancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/inmunología , Receptores de Superficie Celular/inmunología , Animales , Sitios de Unión , Linfocitos T CD8-positivos/efectos de los fármacos , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-7/farmacología , Proteínas de Unión a la Región de Fijación a la Matriz/deficiencia , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Unión Proteica/efectos de los fármacos
13.
Biotechnol Bioeng ; 99(1): 146-54, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17546686

RESUMEN

The direct correlation between levels of heat shock protein expression and efficiency of its tissue protection function motivates this study of how thermal doses can be used for an optimal stress protocol design. Heat shock protein 70 (HSP70) expression kinetics were visualized continuously in cultured bovine aortic endothelial cells (BAECs) on a microscope heating stage using green fluorescent protein (GFP) as a reporter. BAECs were transfected with a DNA vector, HSP(p)-HSP70-GFP which expresses an HSP70-GFP fusion protein under control of the HSP70 promoter. Expression levels were validated by western blot analysis. Transfected cells were heated on a controlled temperature microscope stage at 42 degrees C for a defined period, then shifted to 37 degrees C for varied post-heating times. The expression of HSP70-GFP and its sub-cellular localization were visualized via fluorescence microscopy. The progressive expression kinetics were measured by quantitative analysis of serial fluorescence images captured during heating protocols from 1 to 2 h and post-heating times from 0 to 20 h. The results show two sequential peaks in HSP70 expression at approximately 3 and 12 h post-heat shock. A progressive translocation of HSP70 from the cytoplasm to the nucleus was observed from 6 to 16 h. We conclude that we have successfully combined molecular cloning and optical imaging to study HSP70 expression kinetics. The kinetic profile for HSP70-GFP fusion protein is consistent with the endogenous HSP70. Furthermore, information on dynamic intracellular translocation of HSP70 was extracted from the same experimental data.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Microscopía Fluorescente/métodos , Fracciones Subcelulares/metabolismo , Animales , Bovinos , Células Cultivadas , Células Endoteliales/ultraestructura , Proteínas Fluorescentes Verdes/metabolismo , Calefacción/métodos , Cinética , Tasa de Depuración Metabólica , Perfusión , Proteínas Recombinantes de Fusión/metabolismo , Fracciones Subcelulares/ultraestructura , Temperatura
14.
FEBS Lett ; 581(2): 223-32, 2007 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-17188683

RESUMEN

PSF (PTB-associated splicing factor) is a multi-functional protein that participates in transcription and RNA processing. While phosphorylation of PSF has been shown to be important for some functions, the sites and the kinases involved are not well understood. Although PSF does not contain a typical RS domain, we report here that PSF is phosphorylated in vivo to generate an epitope(s) that can be recognized by a monoclonal antibody specific for phosphorylated RS motifs within SR proteins. PSF can be phosphorylated by human and yeast SR kinases in vivo and in vitro at an isolated RS motif within its N terminus. A functional consequence of SR phosphorylation of PSF is to inhibit its binding to the 3' polypyrimidine tract of pre-mRNA. These results indicate that PSF is a substrate of SR kinases whose phosphorylation regulates its RNA binding capacity and ultimate biological function.


Asunto(s)
Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Precursores del ARN/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Fosfo-Específicos/inmunología , Humanos , Datos de Secuencia Molecular , Fosforilación , Proteína de Unión al Tracto de Polipirimidina/inmunología , Pirimidinas/metabolismo , Especificidad por Sustrato
15.
Mol Cancer ; 5: 35, 2006 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-16956402

RESUMEN

Uncensored exchange of scientific results hastens progress. Open Access does not stop at the removal of price and permission barriers; still, censorship and reading disabilities, to name a few, hamper access to information. Here, we invite the scientific community and the public to discuss new methods to distribute, store and manage literature in order to achieve unfettered access to literature.


Asunto(s)
Acceso a la Información , Internet , Bases de Datos Bibliográficas , Humanos , Publicaciones Periódicas como Asunto/tendencias , PubMed , Edición/tendencias
16.
Mol Cancer ; 5: 26, 2006 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-16805913

RESUMEN

BACKGROUND: Disrupting the balance of histone lysine methylation alters the expression of genes involved in tumorigenesis including proto-oncogenes and cell cycle regulators. Methylation of lysine residues is commonly catalyzed by a family of proteins that contain the SET domain. Here, we report the identification and characterization of the SET domain-containing protein, Smyd2. RESULTS: Smyd2 mRNA is most highly expressed in heart and brain tissue, as demonstrated by northern analysis and in situ hybridization. Over-expressed Smyd2 localizes to the cytoplasm and the nucleus in 293T cells. Although accumulating evidence suggests that methylation of histone 3, lysine 36 (H3K36) is associated with actively transcribed genes, we show that the SET domain of Smyd2 mediates H3K36 dimethylation and that Smyd2 represses transcription from an SV40-luciferase reporter. Smyd2 associates specifically with the Sin3A histone deacetylase complex, which was recently linked to H3K36 methylation within the coding regions of active genes in yeast. Finally, we report that exogenous expression of Smyd2 suppresses cell proliferation. CONCLUSION: We propose that Sin3A-mediated deacetylation within the coding regions of active genes is directly linked to the histone methyltransferase activity of Smyd2. Moreover, Smyd2 appears to restrain cell proliferation, likely through direct modulation of chromatin structure.


Asunto(s)
Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/química , Metiltransferasas/química , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatina/química , N-Metiltransferasa de Histona-Lisina/química , Humanos , Lisina/química , Metiltransferasas/biosíntesis , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Estructura Terciaria de Proteína , Proteínas Represoras/química , Homología de Secuencia de Aminoácido , Complejo Correpresor Histona Desacetilasa y Sin3
17.
Mol Cancer ; 5: 18, 2006 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-16704730

RESUMEN

BACKGROUND: Chromosomal aberrations of BCL11A at 2p16.1 have been reported in a variety of B-cell malignancies and its deficiency in mice leads to a profound block in B-cell development. RESULTS: Alternative pre-mRNA splicing of BCL11A produces multiple isoforms sharing a common N-terminus. The most abundant isoform we have identified in human lymphoid samples is BCL11A-XL, the longest transcript produced at this locus, and here we report the conservation of this major isoform and its functional characterization. We show that BCL11A-XL is a DNA-sequence-specific transcriptional repressor that associates with itself and with other BCL11A isoforms, as well as with the BCL6 proto-oncogene. Western blot data for BCL11A-XL expression coupled with data previously published for BCL6 indicates that these genes are expressed abundantly in germinal-center-derived B cells but that expression is extinguished upon terminal differentiation to the plasma cell stage. Although BCL11A-XL/BCL6 interaction can modulate BCL6 DNA binding in vitro, their heteromeric association does not alter the homomeric transcriptional properties of either on model reporter activity. BCL11A-XL partitions into the nuclear matrix and colocalizes with BCL6 in nuclear paraspeckles. CONCLUSION: We propose that the conserved N-terminus of BCL11A defines a superfamily of C2HC zinc-finger transcription factors involved in hematopoietic malignancies.


Asunto(s)
Proteínas Portadoras/metabolismo , Centro Germinal/metabolismo , Linfoma de Células B/metabolismo , Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Empalme Alternativo/genética , Animales , Western Blotting , Células COS , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Perfilación de la Expresión Génica , Centro Germinal/patología , Células HeLa , Humanos , Inmunoprecipitación , Linfoma de Células B/genética , Linfoma de Células B/patología , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/análisis , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Represoras
18.
Endocrinology ; 143(4): 1280-90, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11897684

RESUMEN

The first 57 bp upstream of the transcription initiation site of the human CYP17 (hCYP17) gene are essential for both basal and cAMP-dependent transcription. EMSA carried out by incubating H295R adrenocortical cell nuclear extracts with radiolabeled -57/-38 probe from the hCYP17 promoter showed the formation of three DNA-protein complexes. The fastest complex contained steroidogenic factor (SF-1) and p54(nrb)/NonO, the intermediate complex contained p54(nrb)/NonO and polypyrimidine tract-binding protein-associated splicing factor (PSF), and the slowest complex contained an SF-1/PSF/p54(nrb)/NonO complex. (Bu)(2)cAMP treatment resulted in a cAMP-inducible increase in the binding intensity of only the upper complex and also activated hCYP17 gene transcription. SF-1 coimmunoprecipitated with p54(nrb)/NonO, indicating direct interaction between these proteins. Functional assays revealed that PSF represses basal transcription. Further, the repression of hCYP17 promoter-reporter construct luciferase activity resulted from PSF interacting with the corepressor mSin3A. Trichostatin A attenuated the inhibition of basal transcription, suggesting that a histone deacetylase interacts with the SF-1/PSF/p54(nrb)/NonO/mSin3A complex. Our studies lend support to the idea that the balance between transcriptional activation and repression is essential in the control of adrenocortical steroid hormone biosynthesis.


Asunto(s)
Corteza Suprarrenal/enzimología , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/genética , Esteroide 17-alfa-Hidroxilasa/genética , Esteroide 17-alfa-Hidroxilasa/metabolismo , Factores de Transcripción/fisiología , Activación Transcripcional/genética , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Western Blotting , Núcleo Celular/genética , Células Cultivadas , Cicloheximida/farmacología , Factores de Transcripción Fushi Tarazu , Regulación Enzimológica de la Expresión Génica/genética , Histona Desacetilasas/genética , Proteínas de Homeodominio , Humanos , Técnicas para Inmunoenzimas , Datos de Secuencia Molecular , Plásmidos/genética , Regiones Promotoras Genéticas/genética , Inhibidores de la Síntesis de la Proteína/farmacología , Receptores Citoplasmáticos y Nucleares , Factores de Empalme Serina-Arginina , Factor Esteroidogénico 1 , Transcripción Genética/genética
19.
Mol Cell Biol ; 33(9): 1768-81, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23438597

RESUMEN

Recombination-activating gene 1 protein (RAG1) and RAG2 are critical enzymes for initiating variable-diversity-joining (VDJ) segment recombination, an essential process for antigen receptor expression and lymphocyte development. The transcription factor BCL11A is required for B cell development, but its molecular function(s) in B cell fate specification and commitment is unknown. We show here that the major B cell isoform, BCL11A-XL, binds the RAG1 promoter and Erag enhancer to activate RAG1 and RAG2 transcription in pre-B cells. We employed BCL11A overexpression with recombination substrates in a cultured pre-B cell line as well as Cre recombinase-mediated Bcl11a(lox/lox) deletion in explanted murine pre-B cells to demonstrate direct consequences of BCL11A/RAG modulation on V(D)J recombination. We conclude that BCL11A is a critical component of a transcriptional network that regulates B cell fate by controlling V(D)J recombination.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Proteínas Nucleares/metabolismo , Activación Transcripcional , Recombinación V(D)J , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Línea Celular , Células Cultivadas , Eliminación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Células Precursoras de Linfocitos B/metabolismo , Regiones Promotoras Genéticas , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Represoras , Regulación hacia Arriba
20.
PLoS One ; 7(12): e51944, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300581

RESUMEN

In vitro bioreactor-based cultures are being extensively investigated for large-scale production of differentiated cells from embryonic stem cells (ESCs). However, it is unclear whether in vitro ESC-derived progenitors have similar gene expression profiles and functionalities as their in vivo counterparts. This is crucial in establishing the validity of ESC-derived cells as replacements for adult-isolated cells for clinical therapies. In this study, we compared the gene expression profiles of Lin-ckit+Sca-1+ (LKS) cells generated in vitro from mouse ESCs using either static or bioreactor-based cultures, with that of native LKS cells isolated from mouse fetal liver (FL) or bone marrow (BM). We found that in vitro-generated LKS cells were more similar to FL- than to BM LKS cells in gene expression. Further, when compared to cells derived from bioreactor cultures, static culture-derived LKS cells showed fewer differentially expressed genes relative to both in vivo LKS populations. Overall, the expression of hematopoietic genes was lower in ESC-derived LKS cells compared to cells from BM and FL, while the levels of non-hematopoietic genes were up-regulated. In order to determine if these molecular profiles correlated with functionality, we evaluated ESC-derived LKS cells for in vitro hematopoietic-differentiation and colony formation (CFU assay). Although static culture-generated cells failed to form any colonies, they did differentiate into CD11c+ and B220+ cells indicating some hematopoietic potential. In contrast, bioreactor-derived LKS cells, when differentiated under the same conditions failed to produce any B220+ or CD11c+ cells and did not form colonies, indicating that these cells are not hematopoietic progenitors. We conclude that in vitro culture conditions significantly affect the transcriptome and functionality of ESC-derived LKS cells and although in vitro differentiated LKS cells were lineage negative and expressed both ckit and Sca-1, these cells, especially those obtained from dynamic cultures, are significantly different from native cells of the same phenotype.


Asunto(s)
Biomarcadores/metabolismo , Médula Ósea/metabolismo , Células Madre Embrionarias/metabolismo , Feto/metabolismo , Perfilación de la Expresión Génica , Hígado/metabolismo , Animales , Antígenos Ly/metabolismo , Médula Ósea/crecimiento & desarrollo , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Feto/citología , Hígado/citología , Proteínas de la Membrana/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA