Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 23(5): 1356-1367, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28416808

RESUMEN

Synapse development and neuronal activity represent fundamental processes for the establishment of cognitive function. Structural organization as well as signalling pathways from receptor stimulation to gene expression regulation are mediated by synaptic activity and misregulated in neurodevelopmental disorders such as autism spectrum disorder (ASD) and intellectual disability (ID). Deleterious mutations in the PTCHD1 (Patched domain containing 1) gene have been described in male patients with X-linked ID and/or ASD. The structure of PTCHD1 protein is similar to the Patched (PTCH1) receptor; however, the cellular mechanisms and pathways associated with PTCHD1 in the developing brain are poorly determined. Here we show that PTCHD1 displays a C-terminal PDZ-binding motif that binds to the postsynaptic proteins PSD95 and SAP102. We also report that PTCHD1 is unable to rescue the canonical sonic hedgehog (SHH) pathway in cells depleted of PTCH1, suggesting that both proteins are involved in distinct cellular signalling pathways. We find that Ptchd1 deficiency in male mice (Ptchd1-/y) induces global changes in synaptic gene expression, affects the expression of the immediate-early expression genes Egr1 and Npas4 and finally impairs excitatory synaptic structure and neuronal excitatory activity in the hippocampus, leading to cognitive dysfunction, motor disabilities and hyperactivity. Thus our results support that PTCHD1 deficiency induces a neurodevelopmental disorder causing excitatory synaptic dysfunction.


Asunto(s)
Disfunción Cognitiva/metabolismo , Proteínas de la Membrana/deficiencia , Sinapsis/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cognición/fisiología , Disfunción Cognitiva/genética , Homólogo 4 de la Proteína Discs Large/genética , Homólogo 4 de la Proteína Discs Large/metabolismo , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Hipocampo/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Transducción de Señal , Sinapsis/genética , Transmisión Sináptica
2.
Br J Cancer ; 104(4): 554-8, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21245861

RESUMEN

PML-RAR (retinoic acid receptor)α is the hallmark protein of acute promyelocytic leukaemia, a highly malignant subtype of acute myeloid leukaemia that accounts for approximately 10% of all AML cases. Recently, several studies have been set out to obtain a comprehensive genome-wide view of the molecular actions of this chimeric protein. In this review, we highlight the new insights that arose from these studies, in particular focussing on newly identified PML-RARα target genes, its interplay with RXR and deregulation of epigenetic modifications.


Asunto(s)
Genoma Humano , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/fisiología , Animales , Epigénesis Genética/fisiología , Humanos , Modelos Biológicos , Proteínas de Fusión Oncogénica/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
3.
Arthritis Res Ther ; 23(1): 202, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321071

RESUMEN

OBJECTIVES: Hyperuricemia is a metabolic condition central to gout pathogenesis. Urate exposure primes human monocytes towards a higher capacity to produce and release IL-1ß. In this study, we assessed the epigenetic processes associated to urate-mediated hyper-responsiveness. METHODS: Freshly isolated human peripheral blood mononuclear cells or enriched monocytes were pre-treated with solubilized urate and stimulated with LPS with or without monosodium urate (MSU) crystals. Cytokine production was determined by ELISA. Histone epigenetic marks were assessed by sequencing immunoprecipitated chromatin. Mice were injected intraarticularly with MSU crystals and palmitate after inhibition of uricase and urate administration in the presence or absence of methylthioadenosine. DNA methylation was assessed by methylation array in whole blood of 76 participants with normouricemia or hyperuricemia. RESULTS: High concentrations of urate enhanced the inflammatory response in vitro in human cells and in vivo in mice, and broad-spectrum methylation inhibitors reversed this effect. Assessment of histone 3 lysine 4 trimethylation (H3K4me3) and histone 3 lysine 27 acetylation (H3K27ac) revealed differences in urate-primed monocytes compared to controls. Differentially methylated regions (e.g. HLA-G, IFITM3, PRKAB2) were found in people with hyperuricemia compared to normouricemia in genes relevant for inflammatory cytokine signaling. CONCLUSION: Urate alters the epigenetic landscape in selected human monocytes or whole blood of people with hyperuricemia compared to normouricemia. Both histone modifications and DNA methylation show differences depending on urate exposure. Subject to replication and validation, epigenetic changes in myeloid cells may be a therapeutic target in gout.


Asunto(s)
Gota , Ácido Úrico , Animales , Epigénesis Genética , Gota/genética , Humanos , Leucocitos Mononucleares , Proteínas de la Membrana , Ratones , Monocitos , Proteínas de Unión al ARN
4.
J Cell Biol ; 114(6): 1125-33, 1991 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1894691

RESUMEN

We have investigated the sorting and processing of the amphibian precursor prepro-dermorphin in mammalian cells. Dermorphin, a D-alanine-containing peptide with potent opioid activity, has been isolated from the skin of the frog Phyllomedusa sauvagei. The maturation of this peptide from the precursor involves several posttranslational steps. Recombinant vaccinia viruses were used to infect AtT-20, PC12, and HeLa cells to study the sorting and processing of prepro-dermorphin. While this precursor was not processed in any of the examined cell lines, AtT-20 cells were able to process approximately 40% of a chimeric precursor consisting of the first 241 amino acids of prepro-enkephalin fused to a carboxy-terminal part of pro-dermorphin. By immunogold-EM, we could show that the chimeric protein, but not pro-dermorphin, was sorted to dense-core secretion granules. The processing products could be released upon stimulation by 8-Br-cAMP. We conclude that the pro-enkephalin part of the fusion protein contains the information for targeting to the regulated pathway of secretion, while this sorting information is missing in pro-dermorphin. This indicates that sorting mechanisms may differ between amphibian and mammalian cells.


Asunto(s)
Encefalinas/genética , Oligopéptidos/genética , Precursores de Proteínas/genética , Procesamiento Proteico-Postraduccional , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Secuencia de Aminoácidos , Animales , Anuros , Línea Celular , Quimera , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Encefalinas/biosíntesis , Humanos , Cinética , Microscopía Inmunoelectrónica , Plásmidos , Precursores de Proteínas/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Transfección , Virus Vaccinia/genética
5.
Mucosal Immunol ; 11(5): 1512-1523, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30038215

RESUMEN

The tissue dendritic cell (DC) compartment is heterogeneous, and the ontogeny and functional specialization of human tissue conventional DC (cDC) subsets and their relationship with monocytes is unresolved. Here we identify monocyte-related CSF1R+Flt3- antigen presenting cells (APCs) that constitute about half of the cells classically defined as SIRPα+ DCs in the steady-state human small intestine. CSF1R+Flt3- APCs express calprotectin and very low levels of CD14, are transcriptionally related to monocyte-derived cells, and accumulate during inflammation. CSF1R+Flt3- APCs show typical macrophage characteristics functionally distinct from their Flt3+ cDC counterparts: under steady-state conditions they excel at antigen uptake, have a lower migratory potential, and are inefficient activators of naïve T cells. These results have important implications for the understanding of the ontogenetic and functional heterogeneity within human tissue DCs and their relation to the monocyte lineage.


Asunto(s)
Células Dendríticas/fisiología , Intestinos/fisiología , Macrófagos/fisiología , Monocitos/fisiología , Transcripción Genética/fisiología , Transcriptoma/fisiología , Anciano , Anciano de 80 o más Años , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/fisiología , Linaje de la Célula/fisiología , Células Dendríticas/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Linfocitos T/metabolismo , Linfocitos T/fisiología , Tirosina Quinasa 3 Similar a fms/metabolismo
6.
7.
Leukemia ; 32(3): 828-836, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28871137

RESUMEN

Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.


Asunto(s)
Reordenamiento Génico , Leucemia Mieloide Aguda/genética , Proteínas del Tejido Nervioso/genética , Dominios y Motivos de Interacción de Proteínas/genética , Activación Transcripcional , Translocación Genética , Línea Celular , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 16 , Epigénesis Genética , Exones , Regulación Leucémica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Intrones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas
8.
Mol Cell Biol ; 19(2): 967-78, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9891033

RESUMEN

Transmission of malaria depends on the successful development of the sexual stages of the parasite within the midgut of the mosquito vector. The differentiation process leading to the production of the sexual stages is delineated by several developmental switches. Arresting the progression through this sexual differentiation pathway would effectively block the spread of the disease. The successful development of such transmission-blocking agents is hampered by the lack of a detailed understanding of the program of gene expression that governs sexual differentiation of the parasite. Here we describe the isolation and functional characterization of the Plasmodium falciparum pfs16 and pfs25 promoters, whose activation marks the developmental switches executed during the sexual differentiation process. We have studied the differential activation of the pfs16 and pfs25 promoters during intraerythrocytic development by transfection of P. falciparum and during gametogenesis and early sporogonic development by transfection of the related malarial parasite P. gallinaceum. Our data indicate that the promoter of the pfs16 gene is activated at the onset of gametocytogenesis, while the activity of the pfs25 promoter is induced following the transition to the mosquito vector. Both promoters have unusual DNA compositions and are extremely A/T rich. We have identified the regions in the pfs16 and pfs25 promoters that are essential for high transcriptional activity. Furthermore, we have identified a DNA-binding protein, termed PAF-1, which activates pfs25 transcription in the mosquito midgut. The data presented here shed the first light on the details of processes of gene regulation in the important human pathogen P. falciparum.


Asunto(s)
ADN Protozoario/genética , ADN Protozoario/aislamiento & purificación , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/genética , Regiones Promotoras Genéticas , Animales , Secuencia de Bases , Sitios de Unión/genética , Mapeo Cromosómico , Culicidae/parasitología , Cartilla de ADN/genética , ADN Protozoario/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Protozoarios , Humanos , Insectos Vectores/parasitología , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Masculino , Datos de Secuencia Molecular , Plasmodium falciparum/patogenicidad , Diferenciación Sexual/genética , Factores de Transcripción/metabolismo , Transfección
9.
Leukemia ; 31(11): 2315-2325, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28216661

RESUMEN

Deregulation of epigenetic mechanisms, including microRNA, contributes to leukemogenesis and drug resistance by interfering with cancer-specific molecular pathways. Here, we show that the balance between miR-194-5p and its newly discovered target BCL2-associated transcription factor 1 (BCLAF1) regulates differentiation and survival of normal hematopoietic progenitors. In acute myeloid leukemias this balance is perturbed, locking cells into an immature, potentially 'immortal' state. Enhanced expression of miR-194-5p by treatment with the histone deacetylase inhibitor SAHA or by exogenous miR-194-5p expression re-sensitizes cells to differentiation and apoptosis by inducing BCLAF1 to shuttle between nucleus and cytosol. miR-194-5p/BCLAF1 balance was found commonly deregulated in 60 primary acute myeloid leukemia patients and was largely restored by ex vivo SAHA treatment. Our findings link treatment responsiveness to re-instatement of miR-194-5p/BCLAF1 balance.


Asunto(s)
Regulación de la Expresión Génica , Leucemia Mieloide Aguda/patología , MicroARNs/genética , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Apoptosis , Ciclo Celular , Diferenciación Celular , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Leucemia Mieloide Aguda/genética
10.
Oncogene ; 36(23): 3346-3356, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28114278

RESUMEN

In 11q23 leukemias, the N-terminal part of the mixed lineage leukemia (MLL) gene is fused to >60 different partner genes. In order to define a core set of MLL rearranged targets, we investigated the genome-wide binding of the MLL-AF9 and MLL-AF4 fusion proteins and associated epigenetic signatures in acute myeloid leukemia (AML) cell lines THP-1 and MV4-11. We uncovered both common as well as specific MLL-AF9 and MLL-AF4 target genes, which were all marked by H3K79me2, H3K27ac and H3K4me3. Apart from promoter binding, we also identified MLL-AF9 and MLL-AF4 binding at specific subsets of non-overlapping active distal regulatory elements. Despite this differential enhancer binding, MLL-AF9 and MLL-AF4 still direct a common gene program, which represents part of the RUNX1 gene program and constitutes of CD34+ and monocyte-specific genes. Comparing these data sets identified several zinc finger transcription factors (TFs) as potential MLL-AF9 co-regulators. Together, these results suggest that MLL fusions collaborate with specific subsets of TFs to deregulate the RUNX1 gene program in 11q23 AMLs.


Asunto(s)
Cromosomas Humanos Par 11/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Adulto , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Lactante , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Proteína de la Leucemia Mieloide-Linfoide/genética , Estadificación de Neoplasias , Proteínas de Fusión Oncogénica/genética , Pronóstico , Regiones Promotoras Genéticas
11.
Nucleic Acids Res ; 29(16): 3424-32, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11504880

RESUMEN

An important model system for studying the process leading to productive transcription is provided by the superfamily of nuclear receptors, which are for the most part ligand-controlled transcription factors. Over the past years several 'orphan' nuclear receptors have been isolated for which no ligand has yet been identified. Very little is known about how these 'orphan' receptors regulate transcription. In this study we have analysed the biochemical and transcriptional properties of the neuronally expressed orphan nuclear receptor RORbeta (NR1F2) and compared them with the retinoic acid receptor heterodimer RXRalpha-RARalpha (NR2B1-NR1B1) and Gal-VP16 in vitro. Although RORbeta binds to its DNA-binding sites with comparatively low affinity, it efficiently directs transcription in nuclear extracts derived from a neuronal cell line, Neuro2A, but not in nuclear extracts from non-neuronal HeLa cells. In contrast, RXRalpha-RARalpha and the acidic transcription factor Gal-VP16 support transcription in Neuro2A and HeLa nuclear extracts equally efficiently. These observations point to a different (co)factor requirement for transactivation by members of the NR1 subfamily of nuclear receptors.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Citoplasmáticos y Nucleares , Transcripción Genética/genética , Secuencia de Bases , Sitios de Unión , Extractos Celulares , ADN/genética , ADN/metabolismo , Dimerización , Vectores Genéticos/genética , Células HeLa , Proteína Vmw65 de Virus del Herpes Simple/metabolismo , Humanos , Neuronas/citología , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Unión Proteica , Receptores de Superficie Celular/genética , Receptores de Ácido Retinoico/metabolismo , Elementos de Respuesta/genética , Receptores X Retinoide , Termodinámica , Factores de Transcripción/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Virus Vaccinia/genética
12.
Oncogene ; 35(15): 1965-76, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26148230

RESUMEN

The ETS transcription factor ERG has been implicated as a major regulator of both normal and aberrant hematopoiesis. In acute myeloid leukemias harboring t(16;21), ERG function is deregulated due to a fusion with FUS/TLS resulting in the expression of a FUS-ERG oncofusion protein. How this oncofusion protein deregulates the normal ERG transcription program is unclear. Here, we show that FUS-ERG acts in the context of a heptad of proteins (ERG, FLI1, GATA2, LYL1, LMO2, RUNX1 and TAL1) central to proper expression of genes involved in maintaining a stem cell hematopoietic phenotype. Moreover, in t(16;21) FUS-ERG co-occupies genomic regions bound by the nuclear receptor heterodimer RXR:RARA inhibiting target gene expression and interfering with hematopoietic differentiation. All-trans retinoic acid treatment of t(16;21) cells as well as FUS-ERG knockdown alleviate the myeloid-differentiation block. Together, the results suggest that FUS-ERG acts as a transcriptional repressor of the retinoic acid signaling pathway.


Asunto(s)
Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 21/genética , Regulación Neoplásica de la Expresión Génica/genética , Hematopoyesis/fisiología , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Aguda/genética , Proteínas de Neoplasias/fisiología , Proteínas de Fusión Oncogénica/fisiología , Proteína FUS de Unión a ARN/fisiología , Transducción de Señal/fisiología , Translocación Genética , Tretinoina/fisiología , Secuencias de Aminoácidos , Línea Celular Tumoral , Cromosomas Humanos Par 16/ultraestructura , Cromosomas Humanos Par 21/ultraestructura , Dimerización , Elementos de Facilitación Genéticos , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/fisiopatología , Leucemia Mielomonocítica Aguda/patología , Leucemia Mielomonocítica Aguda/fisiopatología , Complejos Multiproteicos , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Regiones Promotoras Genéticas , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteína FUS de Unión a ARN/antagonistas & inhibidores , Proteína FUS de Unión a ARN/genética , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Receptores X Retinoide/metabolismo , Transducción de Señal/efectos de los fármacos , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Células U937
13.
Oncogene ; 20(24): 3100-9, 2001 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-11420726

RESUMEN

Transcriptional regulation at the level of chromatin plays crucial roles during eukaryotic development and differentiation. A plethora of studies revealed that the acetylation status of histones is controlled by multi-protein complexes containing (de)acetylase activities. In the current model, histone deacetylases and acetyltransferases are recruited to chromatin by DNA-bound repressors and activators, respectively. Shifting the balance between deacetylation, i.e. repressive chromatin and acetylation, i.e. active chromatin can lead to aberrant gene transcription and cancer. In human acute promyelocytic leukemia (APL) and avian erythroleukemia (AEL), chromosomal translocations and/or mutations in nuclear hormone receptors, RARalpha [NR1B1] and TRalpha [NR1A1], yielded oncoproteins that deregulate transcription and alter chromatin structure. The oncogenic receptors are locked in their 'off' mode thereby constitutively repressing transcription of genes that are critical for differentiation of hematopoietic cells. AEL involves an oncogenic version of the chicken TRalpha, v-ErbA. Apart from repression by v-ErbA via recruitment of corepressor complexes, other repressors and corepressors appear to be involved in repression of v-ErbA target genes, such as carbonic anhydrase II (CAII). Reactivation of repressed genes in APL and AEL by chromatin modifying agents such as inhibitors of histone deacetylase or of methylation provides new therapeutic strategies in the treatment of acute myeloid leukemia.


Asunto(s)
Leucosis Aviar/genética , Infecciones por Retroviridae/genética , Alpharetrovirus , Animales , Leucosis Aviar/virología , Pollos , Regulación de la Expresión Génica , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/virología , Modelos Biológicos , Neoplasias/genética , Proteínas Represoras/fisiología , Infecciones por Retroviridae/virología
14.
Oncogene ; 20(7): 775-87, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11314012

RESUMEN

v-ErbA is a mutated variant of thyroid hormone receptor (TRalpha/NR1A1) borne by the Avian Erythroblastosis virus causing erythroleukemia. TRalpha is known to activate transcription of specific genes in the presence of its cognate ligand, T3 hormone, while in its absence it represses it. v-ErbA is unable to bind ligand, and hence is thought to contribute to leukemogenesis by actively repressing erythroid-specific genes such as the carbonic anhydrase II gene (CA II). In the prevailing model, v-ErbA occludes liganded TR from binding to its cognate elements and constitutively interacts with the corepressors NCoR/SMRT. We previously identified a v-ErbA responsive element (VRE) within a DNase I hypersensitive region (HS2) located in the second intron of the CA II gene. We now show that HS2 fulfils all the requirements for a genuine enhancer that functions independent of its orientation and position with a profound erythroid-specific activity in normal erythroid progenitors (T2ECs) and in leukemic erythroid cell lines. We find that the HS2 enhancer activity is governed by two adjacent GATA-factor binding sites. v-ErbA as well as unliganded TR prevent HS2 activity by nullifying the positive function of factors bound to GATA-sites. However, v-ErbA, in contrast to TR, does not convey active repression to silence the transcriptional activity intrinsic to a heterologous tk promoter. We propose that depending on the sequence and context of the binding site, v-ErbA contributes to leukemogenesis by occluding liganded TR as well as unliganded TR thereby preventing activation or repression, respectively.


Asunto(s)
Anhidrasas Carbónicas/genética , Elementos de Facilitación Genéticos , Leucemia Eritroblástica Aguda/genética , Proteínas Oncogénicas v-erbA/genética , Receptores de Hormona Tiroidea/genética , Animales , Secuencia de Bases , Sitios de Unión , Pollos , Células Precursoras Eritroides , Regulación Leucémica de la Expresión Génica , Humanos , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Unión Proteica , Células Tumorales Cultivadas
15.
Biochim Biophys Acta ; 1494(3): 236-41, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11121580

RESUMEN

Nuclear receptors are ligand-inducible transcription factors that can be classified into two major groups according to their DNA-binding properties. Members of the first group bind to DNA as dimers, either homo- or heterodimers; members of the second group are also able to bind as monomers. While the first group has been extensively studied biochemically, very little is known about nuclear receptors that bind and act as monomers. In this study, we compared the binding and transcriptional behaviour of ROR alpha (NR1F1) and ROR beta (NR1F2), two representatives of the subgroup of monomer-binding receptors. We show that although they are highly related in their amino acid structures, they display remarkably different binding behaviours. Furthermore, we provide evidence that ROR beta can efficiently activate transcription in vitro as a monomer.


Asunto(s)
Receptores de Superficie Celular/genética , Receptores Citoplasmáticos y Nucleares , Sitios de Unión , Escherichia coli/genética , Expresión Génica , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Miembro 2 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Proteínas Tirosina Quinasas Receptoras , Receptores Huérfanos Similares al Receptor Tirosina Quinasa , Receptores de Superficie Celular/metabolismo , Transactivadores , Transcripción Genética , Virus Vaccinia/genética
16.
Biochim Biophys Acta ; 1350(3): 229-34, 1997 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-9061014

RESUMEN

Using crude in vitro systems, we have previously shown that RXR/RAR heterodimers are able to activate transcription from the RAR beta 2 promoter in a retinoid-dependent manner. Here we demonstrate that cofactors distinct from general transcription factors or receptors are required to mediate retinoic acid-dependent transcription in vitro.


Asunto(s)
Receptores de Ácido Retinoico/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología , Extractos Celulares , Sistema Libre de Células , Células HeLa , Humanos , Regiones Promotoras Genéticas/genética , Receptores de Ácido Retinoico/genética , Proteínas Recombinantes de Fusión , Receptor alfa de Ácido Retinoico , Receptores X Retinoide , Factor de Transcripción TFIID , Factores de Transcripción/genética , Factores de Transcripción TFII/aislamiento & purificación , Tretinoina/farmacología
17.
J Mol Biol ; 243(5): 840-5, 1994 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-7966304

RESUMEN

Basic mechanisms of transcription initiation are conserved from yeast to man. However, in contrast to genes transcribed by RNA polymerases II and III, ribosomal gene transcription by RNA polymerase I (Pol I) is species-specific. Promoter selectivity is mediated by SL1/TIF-IB, a multiprotein complex containing the TATA-binding protein (TBP) and TBP-associated factors (TAFs) which bind to DNA and nucleate the assembly of initiation complexes. Using a human cell line that expresses epitope-tagged yeast TBP, we have isolated a chimeric complex consisting of yeast TBP and human TAFs which faithfully promotes human rDNA transcription in vitro. This result argues that specific interactions between TBP and Pol I-specific TAFs have been evolutionarily conserved between distant species. In addition, this finding also underscores the importance of TAFs in determining promoter selectivity of Pol I.


Asunto(s)
Proteínas de Unión al ADN/genética , ARN Polimerasa I/genética , Factores de Transcripción/genética , Transcripción Genética/genética , Proteínas de Xenopus , Western Blotting , Candida/genética , Línea Celular , Humanos , Homología de Secuencia de Aminoácido , Proteína de Unión a TATA-Box
18.
Gene ; 132(2): 267-71, 1993 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-7693550

RESUMEN

A cDNA encoding the homologue of the rabbit immunophilin p59 was cloned from a mouse NIH-3T3 cell line library. Antibodies were generated against the N-terminal fragment of the protein produced in bacteria. Western blotting experiments suggest that homologous proteins are present in several other cell lines tested. Production of mouse p59 using recombinant vaccinia viruses resulted in a protein with the expected size of 59 kDa that can interact with the recombinant glucocorticoid receptor, as shown by co-immunoprecipitation experiments.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de Choque Térmico/genética , Receptores de Glucocorticoides/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Western Blotting , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Clonación Molecular , ADN , Proteínas de Choque Térmico/inmunología , Proteínas de Choque Térmico/metabolismo , Humanos , Sueros Inmunes/inmunología , Ratones , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Tacrolimus/metabolismo , Proteínas de Unión a Tacrolimus
19.
Biochem Soc Symp ; 62: 97-109, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8971343

RESUMEN

Retinoids play a fundamental role in regulating normal cell proliferation and differentiation. The most spectacular effects of retinoids in vitro can be observed with embryonal carcinoma (EC) cells that can be induced to differentiate into endodermal, mesodermal and neuroectodermal lineages. An early and essential step in the differentiation process is the activation of the retinoic acid receptor-beta 2 (RAR beta 2) promoter that requires a co-operation between RAR, the EC-cell specific adenovirus early gene product 1A (E1A)-like activity and the TATA-binding protein (TBP). In differentiated cells, this signalling pathway can be mimicked by ectopic expression of the adenoviral E1A protein. Here we show that E1A13S but not E1A12S augments the level of transcription. Analysis of the binding kinetics of E1A13S to TBP by the surface plasmon resonance (SPR) technique reveals that the affinity of TBP for a consensus TATA-box sequence is significantly and specifically increased by E1A13S only. Intriguingly, a specific interaction can only be obtained with crude TBP overexpressed in HeLa cells via vaccinia virus as opposed to bacterially expressed TBP, suggesting a cofactor requirement for the interaction. Co-immunoprecipitation experiments show that E1A13S is an integral component of the RNA polymerase II-specific TBP-containing complex in adenovirus transformed embryonal kidney 293 cells. Taken together the results suggest that E1A13S mediates transcriptional activation by providing a physical bridge between TBP/transcription factor IID (TFIID) and retinoic acid receptor.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Proteínas de Unión al ADN/metabolismo , Genes Inmediatos-Precoces , Receptores de Ácido Retinoico/fisiología , TATA Box , Factores de Transcripción/metabolismo , Transcripción Genética , Células HeLa , Humanos , Modelos Biológicos , Receptores de Ácido Retinoico/genética , Proteína de Unión a TATA-Box , Factor de Transcripción TFIID , Factores de Transcripción TFII/metabolismo , Transfección , Dedos de Zinc
20.
Hybridoma ; 15(1): 55-68, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9064287

RESUMEN

The TATA box-binding protein (TBP) is a key transcription factor required for transcription by all three eukaryotic RNA polymerases. It consists of a conserved carboxy-terminal DNA binding domain and a highly divergent amino terminal domain. TBP and different sets of TBP-associated factors (TAFs) constitute at least four multisubunit complexes referred to as SL1, TFIID, TFIIIB, and SNAPC. SL1, TFIID, and TFIIIB are required for transcription by RNA polymerases I, II, and III, respectively, while the SNAP complex is involved in transcription of the small nuclear RNA (snRNA) genes by RNA polymerases II and III. TBP also associates with a number of basal transcription factors such as TFIIA and TFIIB, and with several regulatory factors such as VP16, E1A, and p53. Here we describe the characterization of a panel of monoclonal antibodies (MAbs) directed against the amino-terminal domain of human TBP. These MAbs recognize different TBP epitopes, some of which have been precisely defined. Different MAbs recognize different TBP-containing complexes and several of them crossreact with TBP from other species. These antibodies can be used to purify TBP-containing complexes in a functional form and should be useful to identify new protein-protein interactions involving TBP.


Asunto(s)
Anticuerpos Monoclonales/química , Proteínas de Unión al ADN/inmunología , Mapeo Epitopo , Proteínas del Complejo de Iniciación de Transcripción Pol1 , Factores de Transcripción/inmunología , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Cricetinae , Reacciones Cruzadas , ADN/química , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/aislamiento & purificación , Drosophila , Electroforesis en Gel de Poliacrilamida , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Fragmentos de Péptidos/inmunología , Ratas , Erizos de Mar , Especificidad de la Especie , Proteína de Unión a TATA-Box , Factores de Transcripción/química , Factores de Transcripción/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA