Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nat Protoc ; 18(1): 157-187, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36280749

RESUMEN

The ability to visualize RNA in its native subcellular environment by using single-molecule fluorescence in situ hybridization (smFISH) has reshaped our understanding of gene expression and cellular functions. A major hindrance of smFISH is the difficulty to perform systematic experiments in medium- or high-throughput formats, principally because of the high cost of generating the individual fluorescent probe sets. Here, we present high-throughput smFISH (HT-smFISH), a simple and cost-efficient method for imaging hundreds to thousands of single endogenous RNA molecules in 96-well plates. HT-smFISH uses RNA probes transcribed in vitro from a large pool of unlabeled oligonucleotides. This allows the generation of individual probes for many RNA species, replacing commercial DNA probe sets. HT-smFISH thus reduces costs per targeted RNA compared with many smFISH methods and is easily scalable and flexible in design. We provide a protocol that combines oligo pool design, probe set generation, optimized hybridization conditions and guidelines for image acquisition and analysis. The pipeline requires knowledge of standard molecular biology tools, cell culture and fluorescence microscopy. It is achievable in ~20 d. In brief, HT-smFISH is tailored for medium- to high-throughput screens that image RNAs at single-molecule sensitivity.


Asunto(s)
Diagnóstico por Imagen , ARN , ARN/genética , Hibridación Fluorescente in Situ/métodos , Análisis Costo-Beneficio , Flujo de Trabajo
2.
J Cell Sci ; 135(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35343565

RESUMEN

Senescence is an irreversible withdrawal from cell proliferation that can be initiated after DNA damage-induced cell cycle arrest in G2 phase to prevent genomic instability. Senescence onset in G2 requires p53 (also known as TP53) and retinoblastoma protein (RB, also known as RB1) family tumour suppressors, but how they are regulated to convert a temporary cell cycle arrest into a permanent one remains unknown. Here, we show that a previously unrecognised balance between the cyclin-dependent kinase (CDK) inhibitor p21 and the checkpoint kinase Chk1 controls cyclin D-CDK activity during G2 arrest. In non-transformed cells, p21 activates RB in G2 by inhibiting cyclin D1 complexed with CDK2 or CDK4. The resulting G2 exit, which precedes the appearance of senescence markers, is associated with a mitotic bypass, Chk1 downregulation and reduction in the number of DNA damage foci. In p53/RB-proficient cancer cells, a compromised G2 exit correlates with sustained Chk1 activity, delayed p21 induction, untimely cyclin E1 re-expression and genome reduplication. Conversely, Chk1 depletion promotes senescence by inducing p21 binding to cyclin D1- and cyclin E1-CDK complexes and downregulating CDK6, whereas knockdown of the checkpoint kinase Chk2 enables RB phosphorylation and delays G2 exit. In conclusion, p21 and Chk2 oppose Chk1 to maintain RB activity, thus promoting the onset of senescence induced by DNA damage in G2.


Asunto(s)
Ciclina D1 , Proteína p53 Supresora de Tumor , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Fosforilación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Nat Commun ; 12(1): 1352, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33649340

RESUMEN

Local translation allows for a spatial control of gene expression. Here, we use high-throughput smFISH to screen centrosomal protein-coding genes, and we describe 8 human mRNAs accumulating at centrosomes. These mRNAs localize at different stages during cell cycle with a remarkable choreography, indicating a finely regulated translational program at centrosomes. Interestingly, drug treatments and reporter analyses reveal a common translation-dependent localization mechanism requiring the nascent protein. Using ASPM and NUMA1 as models, single mRNA and polysome imaging reveals active movements of endogenous polysomes towards the centrosome at the onset of mitosis, when these mRNAs start localizing. ASPM polysomes associate with microtubules and localize by either motor-driven transport or microtubule pulling. Remarkably, the Drosophila orthologs of the human centrosomal mRNAs also localize to centrosomes and also require translation. These data identify a conserved family of centrosomal mRNAs that localize by active polysome transport mediated by nascent proteins.


Asunto(s)
Centrosoma/metabolismo , Polirribosomas/metabolismo , Transporte de ARN , Animales , Proteínas de Ciclo Celular/metabolismo , Centrosoma/efectos de los fármacos , Cicloheximida/farmacología , Drosophila/genética , Células HeLa , Humanos , Mitosis/efectos de los fármacos , Sistemas de Lectura Abierta/genética , Polirribosomas/efectos de los fármacos , Puromicina/farmacología , Transporte de ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33402530

RESUMEN

The recent emergence and reemergence of viruses in the human population has highlighted the need to develop broader panels of therapeutic molecules. High-throughput screening assays opening access to untargeted steps of the viral replication cycle will provide powerful leverage to identify innovative antiviral molecules. We report here the development of an innovative protein complementation assay, termed αCentauri, to measure viral translocation between subcellular compartments. As a proof of concept, the Centauri fragment was either tethered to the nuclear pore complex or sequestered in the nucleus, while the complementary α fragment (<16 amino acids) was attached to the integrase proteins of infectious HIV-1. The translocation of viral ribonucleoproteins from the cytoplasm to the nuclear envelope or to the nucleoplasm efficiently reconstituted superfolder green fluorescent protein or NanoLuc αCentauri reporters. These fluorescence- or bioluminescence-based assays offer a robust readout of specific steps of viral infection in a multiwell format that is compatible for high-throughput screening and is validated by a short hairpin RNA-based prototype screen.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Virosis/metabolismo , Replicación Viral/fisiología , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Infecciones por VIH/metabolismo , Células HeLa , Humanos , Membrana Nuclear/metabolismo , Poro Nuclear/metabolismo , Ribonucleoproteínas/metabolismo , Replicación Viral/efectos de los fármacos
5.
Sci Rep ; 9(1): 10311, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31312011

RESUMEN

To build and maintain mitotic spindle architecture, molecular motors exert spatially regulated forces on microtubules (MT) minus-ends. This spatial regulation is required to allow proper chromosomes alignment through the organization of kinetochore fibers (k-fibers). NuMA was recently shown to target dynactin to MT minus-ends and thus to spatially regulate dynein activity. However, given that k-fibers are embedded in the spindle, our understanding of the machinery involved in the targeting of proteins to their minus-ends remains limited. Intraflagellar transport (IFT) proteins were primarily studied for their ciliary roles but they also emerged as key regulators of cell division. Taking advantage of MT laser ablation, we show here that IFT88 concentrates at k-fibers minus-ends and is required for their re-anchoring into spindles by controlling NuMA accumulation. Indeed, IFT88 interacts with NuMA and is required for its enrichment at newly generated k-fibers minus-ends. Combining nocodazole washout experiments and IFT88 depletion, we further show that IFT88 is required for the reorganization of k-fibers into spindles and thus for efficient chromosomes alignment in mitosis. Overall, we propose that IFT88 could serve as a mitotic MT minus-end adaptor to concentrate NuMA at minus-ends thus facilitating k-fibers incorporation into the main spindle.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Huso Acromático/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular , Células HCT116 , Humanos , Terapia por Láser , Nocodazol/farmacología , Huso Acromático/efectos de los fármacos , Sus scrofa
6.
Sci Rep ; 7(1): 13429, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-29044141

RESUMEN

Although cyclin-dependent kinase 2 (Cdk2) controls the G1/S transition and promotes DNA replication, it is dispensable for cell cycle progression due to redundancy with Cdk1. Yet Cdk2 also has non-redundant functions that can be revealed in certain genetic backgrounds and it was reported to promote the G2/M DNA damage response checkpoint in TP53 (p53)-deficient cancer cells. However, in p53-proficient cells subjected to DNA damage, Cdk2 is inactivated by the CDK inhibitor p21. We therefore investigated whether Cdk2 differentially affects checkpoint responses in p53-proficient and deficient cell lines. We show that, independently of p53 status, Cdk2 stimulates the ATR/Chk1 pathway and is required for an efficient DNA replication checkpoint response. In contrast, Cdk2 is not required for a sustained DNA damage response and G2 arrest. Rather, eliminating Cdk2 delays S/G2 progression after DNA damage and accelerates appearance of early markers of cell cycle exit. Notably, Cdk2 knockdown leads to down-regulation of Cdk6, which we show is a non-redundant pRb kinase whose elimination compromises cell cycle progression. Our data reinforce the notion that Cdk2 is a key p21 target in the DNA damage response whose inactivation promotes exit from the cell cycle in G2.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Daño del ADN , Puntos de Control de la Fase S del Ciclo Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células HCT116 , Humanos , Proteína p53 Supresora de Tumor/metabolismo
7.
Biol Cell ; 109(5): 210-221, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28225561

RESUMEN

Zebrafish gastrulation and particularly epiboly that involves coordinated movements of several cell layers is a dynamic process for which regulators remain to be identified. We show here that Flotillin 1 and 2, ubiquitous and highly conserved proteins, are required for epiboly. Flotillins knockdown compromised embryo survival, strongly delayed epiboly and impaired deep cell radial intercalation and directed collective migration without affecting enveloping layer cell movement. At the molecular level, we identified that Flotillins are required for the formation of E-cadherin-mediated cell-cell junctions. These results provide the first in vivo evidence that Flotillins regulate E-cadherin-mediated cell-cell junctions to allow epiboly progression.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Proteínas de la Membrana/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Adhesión Celular , Comunicación Celular , Técnicas de Silenciamiento del Gen , beta Catenina/metabolismo
8.
Nat Commun ; 7: 10765, 2016 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-26899482

RESUMEN

ERBB2 overexpression in human breast cancer leads to invasive carcinoma but the mechanism is not clearly understood. Here we report that TOM1L1 is co-amplified with ERBB2 and defines a subgroup of HER2(+)/ER(+) tumours with early metastatic relapse. TOM1L1 encodes a GAT domain-containing trafficking protein and is a SRC substrate that negatively regulates tyrosine kinase signalling. We demonstrate that TOM1L1 upregulation enhances the invasiveness of ERBB2-transformed cells. This pro-tumoural function does not involve SRC, but implicates membrane-bound membrane-type 1 MMP (MT1-MMP)-dependent activation of invadopodia, membrane protrusions specialized in extracellular matrix degradation. Mechanistically, ERBB2 elicits the indirect phosphorylation of TOM1L1 on Ser321. The phosphorylation event promotes GAT-dependent association of TOM1L1 with the sorting protein TOLLIP and trafficking of the metalloprotease MT1-MMP from endocytic compartments to invadopodia for tumour cell invasion. Collectively, these results show that TOM1L1 is an important element of an ERBB2-driven proteolytic invasive programme and that TOM1L1 amplification potentially enhances the metastatic progression of ERBB2-positive breast cancers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Receptor ErbB-2/metabolismo , Células 3T3 , Animales , Línea Celular Tumoral , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Invasividad Neoplásica
9.
PLoS One ; 6(7): e22296, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21811582

RESUMEN

Ilf3 and NF90, two proteins containing double-stranded RNA-binding domains, are generated by alternative splicing and involved in several functions. Their heterogeneity results from posttranscriptional and posttranslational modifications. Alternative splicing of exon 3, coding for a 13 aa N-terminal motif, generates for each protein a long and short isoforms. Subcellular fractionation and localization of recombinant proteins showed that this motif acts as a nucleolar localization signal. Deletion and substitution mutants identified four arginines, essential for nucleolar targeting, and three histidines to stabilize the proteins within the nucleolus. The short isoforms are never found in the nucleoli, whereas the long isoforms are present in the nucleoplasm and the nucleoli. For Ilf3, only the posttranslationally-unmodified long isoform is nucleolar, suggesting that this nucleolar targeting is abrogated by posttranslational modifications. Confocal microscopy and FRAP experiments have shown that the long Ilf3 isoform localizes to the granular component of the nucleolus, and that L-Ilf3 and L-NF90 exchange rapidly between nucleoli. The presence of this 13 aminoacid motif, combined with posttranslational modifications, is responsible for the differences in Ilf3 and NF90 isoforms subcellular localizations. The protein polymorphism of Ilf3/NF90 and the various subcellular localizations of their isoforms may partially explain the various functions previously reported for these proteins.


Asunto(s)
Empalme Alternativo/genética , Nucléolo Celular/metabolismo , Exones/genética , Proteínas del Factor Nuclear 90/genética , Proteínas del Factor Nuclear 90/metabolismo , Señales de Clasificación de Proteína , Isoformas de ARN/genética , Empalme Alternativo/efectos de los fármacos , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Nucléolo Celular/efectos de los fármacos , Diclororribofuranosil Benzoimidazol/farmacología , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas del Factor Nuclear 90/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Señales de Clasificación de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Eliminación de Secuencia/genética , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
10.
Development ; 138(14): 2903-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21653611

RESUMEN

Female meiotic divisions are extremely asymmetric, giving rise to a large oocyte and small degenerating polar bodies, keeping the maternal stores for further embryo development. This asymmetry is achieved via off-center positioning of the division spindle. Mouse oocytes have developed a formin-2-dependent actin-based spindle positioning mechanism that allows the meiotic spindle to migrate towards the closest cortex. Using spinning disk microscopy and FRAP analysis, we studied the changes in the organization of the cytoplasmic F-actin meshwork during the first meiotic division. It is very dense in prophase I, undergoes a significant density drop upon meiosis resumption and reforms progressively later on. This meshwork remodeling correlates with endogenous formin 2 regulation. High formin 2 levels at meiosis I entry induce meshwork maintenance, leading to equal forces being exerted on the chromosomes, preventing spindle migration. Hence, the meshwork density drop at meiosis resumption is germane to the symmetry-breaking event required for successful asymmetric meiotic divisions.


Asunto(s)
Actinas/metabolismo , Meiosis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Oocitos/citología , Huso Acromático/fisiología , Animales , Femenino , Recuperación de Fluorescencia tras Fotoblanqueo , Técnica del Anticuerpo Fluorescente , Immunoblotting , Ratones , Microscopía Confocal , Oocitos/fisiología , Plásmidos/genética
11.
Curr Biol ; 18(19): 1514-9, 2008 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-18848445

RESUMEN

Female meiosis in higher organisms consists of highly asymmetric divisions, which retain most maternal stores in the oocyte for embryo development. Asymmetric partitioning of the cytoplasm results from the spindle's "off-center" positioning, which, in mouse oocytes, depends mainly on actin filaments [1, 2]. This is a unique situation compared to most systems, in which spindle positioning requires interactions between astral microtubules and cortical actin filaments [3]. Formin 2, a straight-actin-filament nucleator, is required for the first meiotic spindle migration to the cortex and cytokinesis in mouse oocytes [4, 5]. Although the requirement for actin filaments in the control of spindle positioning is well established in this model, no one has been able to detect them in the cytoplasm [6]. Through the expression of an F-actin-specific probe and live confocal microscopy, we show the presence of a cytoplasmic actin meshwork, organized by Formin 2, that controls spindle migration. In late meiosis I, these filaments organize into a spindle-like F-actin structure, which is connected to the cortex. At anaphase, global reorganization of this meshwork allows polar-body extrusion. In addition, using actin-YFP, our FRAP analysis confirms the presence of a highly dynamic cytoplasmic actin meshwork that is tightly regulated in time and space.


Asunto(s)
Citoesqueleto de Actina/fisiología , Oocitos/fisiología , Huso Acromático/fisiología , Actinas/metabolismo , Animales , Femenino , Ratones , Proteínas de Microfilamentos/metabolismo
12.
Nat Neurosci ; 11(4): 440-9, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18327254

RESUMEN

During their migration, cerebellar granule cells switch from a tangential to a radial mode of migration. We have previously demonstrated that this involves the transmembrane semaphorin Sema6A. We show here that plexin-A2 is the receptor that controls Sema6A function in migrating granule cells. In plexin-A2-deficient (Plxna2(-/-)) mice, which were generated by homologous recombination, many granule cells remained in the molecular layer, as we saw in Sema6a mutants. A similar phenotype was observed in mutant mice that were generated by mutagenesis with N-ethyl-N-nitrosourea and had a single amino-acid substitution in the semaphorin domain of plexin-A2. We found that this mutation abolished the ability of Sema6A to bind to plexin-A2. Mouse chimera studies further suggested that plexin-A2 acts in a cell-autonomous manner. We also provide genetic evidence for a ligand-receptor relationship between Sema6A and plexin-A2 in this system. Using time-lapse video microscopy, we found that centrosome-nucleus coupling and coordinated motility were strongly perturbed in Sema6a(-/-) and Plxna2(-/-) granule cells. This suggests that semaphorin-plexin signaling modulates cell migration by controlling centrosome positioning.


Asunto(s)
Movimiento Celular/fisiología , Núcleo Celular/metabolismo , Centrosoma/metabolismo , Cerebelo/crecimiento & desarrollo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Animales , Células Cultivadas , Cerebelo/citología , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuronas/citología , Neuronas/metabolismo , Receptores de Superficie Celular/genética , Semaforinas/genética
13.
J Cell Biol ; 176(3): 295-305, 2007 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-17261848

RESUMEN

Spindle formation is essential for stable inheritance of genetic material. Experiments in various systems indicate that Ran GTPase is crucial for meiotic and mitotic spindle assembly. Such an important role for Ran in chromatin-induced spindle assembly was initially demonstrated in Xenopus laevis egg extracts. However, the requirement of RanGTP in living meiotic cells has not been shown. In this study, we used a fluorescence resonance energy transfer probe to measure RanGTP-regulated release of importin beta. A RanGTP-regulated gradient was established during meiosis I and was centered on chromosomes throughout mouse meiotic maturation. Manipulating levels of RanGTP in mice and X. laevis oocytes did not inhibit assembly of functional meiosis I spindles. However, meiosis II spindle assembly did not tolerate changes in the level of RanGTP in both species. These findings suggest that a mechanism common to vertebrates promotes meiosis I spindle formation in the absence of chromatin-induced microtubule production and centriole-based microtubule organizing centers.


Asunto(s)
Centriolos/metabolismo , Meiosis/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Oocitos/citología , Huso Acromático/metabolismo , Proteína de Unión al GTP ran/metabolismo , Animales , Cromosomas de los Mamíferos/metabolismo , Femenino , Transferencia Resonante de Energía de Fluorescencia , Guanosina Trifosfato/metabolismo , Ratones , Ratones Endogámicos , Proteínas de Unión al GTP Monoméricas/genética , Oligonucleótidos Antisentido , Oocitos/metabolismo , Vertebrados , Xenopus laevis , beta Carioferinas/metabolismo , Proteína de Unión al GTP ran/genética
14.
Eur Biophys J ; 36(2): 153-61, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17021805

RESUMEN

Two-photon, two-color fluorescence cross-correlation spectroscopy (TPTCFCCS) was used to directly detect ligand-dependent interaction between an eCFP-fusion of the androgen receptor (eCFP-AR) and an eYFP fusion of the nuclear receptor co-activator, Tif2 (eYFP-Tif2) in live cells. As expected, these two proteins were co-localized in the nucleus in the presence of ligand. Analysis of the cross-correlation amplitude revealed that AR was on average 81% bound to Tif2 in the presence of agonist, whereas the fractional complex formation decreased to 56% in the presence of antagonist. Residual AR-Tif2 interaction in presence of antagonist is likely mediated by its ligand-independent activation function. These studies demonstrate that using TPTCFCCS it is possible to quantify ligand-dependent interaction of nuclear receptors with co-regulator partners in live cells, making possible a vast array of structure-function studies for these important transcriptional regulators.


Asunto(s)
Coactivador 2 del Receptor Nuclear/metabolismo , Mapeo de Interacción de Proteínas/métodos , Receptores Androgénicos/metabolismo , Espectrometría de Fluorescencia/métodos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Ligandos , Microscopía de Fluorescencia por Excitación Multifotónica , Unión Proteica
15.
Mol Endocrinol ; 20(7): 1506-18, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16527872

RESUMEN

The androgen receptor (AR) is a ligand-activated transcription factor that controls growth and survival of prostate cancer cells. In the present study, we investigated the regulation of AR activity by the receptor-interacting protein 140 (RIP140). We first showed that RIP140 could be coimmunoprecipitated with the receptor when coexpressed in 293T cells. This interaction appeared physiologically relevant because chromatin immunoprecipitation assays revealed that, under R1881 treatment, RIP140 could be recruited to the prostate-specific antigen encoding gene in LNCaP cells. In vitro glutathione S-transferase pull-down assays provided evidence that the carboxy-terminal domain of AR could interact with different regions of RIP140. By means of fluorescent proteins, we demonstrated that ligand-activated AR was not only able to translocate to the nucleus but also to relocate RIP140 from very structured nuclear foci to a diffuse pattern. Overexpression of RIP140 strongly repressed AR-dependent transactivation by preferentially targeting the ligand binding domain-dependent activity. Moreover, disruption of RIP140 expression induced AR overactivation, thus revealing RIP140 as a strong AR repressor. We analyzed its mechanism of transrepression and first demonstrated that different regions of RIP140 could mediate AR-dependent repression. We then showed that the carboxy-terminal end of RIP140 could reverse transcriptional intermediary factor 2-dependent overactivation of AR. The use of mutants of RIP140 allowed us to suggest that C-terminal binding protein played no role in RIP140-dependent inhibition of AR activity, whereas histone deacetylases partly regulated that transrepression. Finally, we provided evidence for a stimulation of RIP140 mRNA expression in LNCaP cells under androgen treatment, further emphasizing the role of RIP140 in androgen signaling.


Asunto(s)
Antagonistas de Receptores Androgénicos , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Oxidorreductasas de Alcohol , Animales , Células COS , Compartimento Celular , Chlorocebus aethiops , Cricetinae , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Metribolona/farmacología , Proteína de Interacción con Receptores Nucleares 1 , Fosfoproteínas/metabolismo , Unión Proteica , ARN Mensajero/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba/efectos de los fármacos
16.
Mol Endocrinol ; 20(4): 724-34, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16373394

RESUMEN

The mutation of a single amino acid in the ligand binding domain of the human androgen receptor (AR) can induce functional abnormalities; for example, in androgen binding or interactions with coregulators. We report here on the structure/function analysis of the ARE709K substitution that is associated with partial androgen insensitivity syndrome. We introduced several mutations at position 709 and tested the consequences of these changes on AR structure and activity in the presence of androgen and antiandrogens. Our results demonstrate that a strong interaction between helix H12 and residue 709 in H3 is required to obtain a fully functional AR. We show that glutamic acid 709 can be replaced by a bulky tyrosine residue without significant effect on the activation by agonists. In contrast, smaller or linear residues that are unable to maintain a tight interaction with H12 induce a substantial loss of androgen-induced AR activity. We also show that the agonist activity of partial antiandrogens is dependent on the side-chain residue at position 709. Strikingly, the ARE709Y substitution causes the conversion of cyproterone acetate into a pure antiandrogen and bicalutamide into a partial agonist. Together, our structural and functional data reveal the key role of glutamic acid 709 in androgenic and antiandrogenic activities.


Asunto(s)
Síndrome de Resistencia Androgénica/genética , Síndrome de Resistencia Androgénica/fisiopatología , Receptores Androgénicos/química , Receptores Androgénicos/genética , Sustitución de Aminoácidos , Antagonistas de Andrógenos/farmacología , Andrógenos/farmacología , Animales , Secuencia de Bases , Línea Celular , Acetato de Ciproterona/farmacología , ADN Complementario/genética , Humanos , Técnicas In Vitro , Lactante , Masculino , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Conformación Proteica , Estructura Secundaria de Proteína , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
FEBS Lett ; 579(20): 4278-86, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16051232

RESUMEN

Resistance to 4-hydroxy-tamoxifen (OHT), which appears in breast cancer cells after long-term antiestrogen treatment, may involve irreversible changes of gene expression. We previously developed a MCF-7 derived cell line (MVLN), in which OHT rapidly and irreversibly inactivates the expression of an estrogen-regulated luciferase transgene (Vit-tk-luciferase). In chromatin immunoprecipitation experiments, heterochromatin protein 1 (HP1alpha) was found to be associated with the Vit-tk-luciferase transgene, only when it was inactivated by OHT treatment. Chimeras composed of either HP1alpha or the Krupple-associated box (KRAB) module of KOX-1 protein (known to repress gene expression by recruitment of HP1 proteins), fused to the estrogen receptor (ER)-DNA binding domain (DBD) and the androgen receptor (AR)-ligand binding domain (LBD) were generated and appeared as potent transcriptional repressors. In stably transfected MVLN cells, irreversible inactivation of the luciferase transgene expression obtained with HP1alpha-ER(DBD)-AR(LBD) was partial, whereas inactivation obtained with KRAB-ER(DBD)-AR(LBD) was comparable to that obtained with OHT, although with a slower kinetics. Altogether, these data suggest that HP1alpha is involved in the silencing effects associated with long-term OHT treatments.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Cromosómicas no Histona/metabolismo , Antagonistas de Estrógenos/farmacología , Silenciador del Gen , Tamoxifeno/análogos & derivados , Transcripción Genética/efectos de los fármacos , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Inmunoprecipitación de Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/genética , Humanos , Factores de Transcripción de Tipo Kruppel , Luciferasas/análisis , Luciferasas/genética , Isoformas de Proteínas/metabolismo , Receptores Androgénicos/análisis , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/análisis , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/análisis , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Tamoxifeno/farmacología , Transgenes , Células Tumorales Cultivadas
18.
J Biol Chem ; 279(35): 36972-81, 2004 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-15215236

RESUMEN

Epidermal growth factor receptor (EGFR) signaling is initiated by ligand binding followed by homodimerization and rapid receptor autophosphorylation. Monitoring EGFR phosphorylation was achieved by measuring translocation and binding of an enhanced yellow fluorescent protein (EYFP)-labeled phosphotyrosine-binding domain (PTB) to enhanced cyan fluorescent protein (ECFP)-tagged EGFR using fluorescence lifetime imaging microscopy or sensitized emission measurements. To simplify dynamic phosphorylation pattern measurements in cells, FLAME, a ratiometric sensor containing both EGFR-ECFP and PTB-EYFP in one molecule, was designed and examined in COS7 cells. Epidermal growth factor (EGF) treatment demonstrated rapid and reversible changes in the EYFP/ECFP fluorescence emission ratios, due to binding of the PTB domain to its consensus binding sites upon phosphorylation at the cell periphery, whereas perinuclear regions failed to respond to EGF but were responsive to tyrosine kinase inhibition. Long-term EGF treatment resulted in accumulation of dephosphorylated receptor in the perinuclear region due to active dephosphorylation occurring at intracellular sites. This indicates that the sensor closely approaches the true dynamics of tyrosine kinase autophosphorylation and dephosphorylation. Phosphatase inhibition by pervanadate resulted in an irreversible response in all cellular compartments. These data show that EGFR is under tonic phosphatase suppression maintaining the receptor in an unphosphorylated (silent) state and is dephosphorylated at endomembranes after ligand-mediated endocytosis.


Asunto(s)
Receptores ErbB/química , Animales , Células COS , Línea Celular Tumoral , Citoesqueleto/metabolismo , Dimerización , Endocitosis , Receptores ErbB/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Immunoblotting , Ligandos , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Fosforilación , Fosfotirosina/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factores de Tiempo , Transfección
19.
FEBS Lett ; 531(2): 245-9, 2002 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-12417320

RESUMEN

Spectral variants of the green fluorescent protein (GFP) have been extensively used as reporters to image molecular interactions in living cells by fluorescence resonance energy transfer (FRET). However, those GFP variants which are the most efficient donor acceptor pairs for FRET measurements show a high degree of spectral overlap which has hampered in the past their use in FRET applications. Here we use spectral imaging and subsequent un-mixing to quantitatively separate highly overlapping donor and acceptor emissions in FRET measurements. We demonstrate the method in fixed and living cells using a novel GFP based FRET pair (GFP2-YFP (yellow)), which has an increased FRET efficiency compared to the most commonly used FRET pair consisting of cyan fluorescent protein and YFP. Moreover, GFP2 has its excitation maximum at 396 nm at which the YFP acceptor is excited only below the detection level and thus this FRET pair is ideal for applications involving sensitized emission.


Asunto(s)
Proteínas Bacterianas/química , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Luminiscentes/química , Microscopía Fluorescente , Proteínas Bacterianas/genética , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Indicadores y Reactivos , Proteínas Luminiscentes/genética , Proteínas Recombinantes de Fusión/química
20.
Biochemistry ; 41(39): 11824-31, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12269826

RESUMEN

Activation of the androgen receptor (AR) is induced by ligand binding through conformational changes leading to control of gene expression. Antiandrogens compete with androgens for AR occupancy and subsequently block at least one step in AR action. Analysis of nuclear transfer kinetics using the GFP-AR fusion protein and partial proteolysis analysis provided evidence that the ligand-bound receptor was in equilibrium between at least two distinct conformations, leading to the production of 35 and 29 kDa trypsin-resistant fragments. It also indicated that this equilibrium may regulate the rate of nuclear transfer. The slowing of nuclear transfer by antiandrogens was correlated with the amount of receptor in conformation leading to the 35 kDa trypsin-resistant fragment. To establish the role of heat shock protein (hsp) 90 activity in antiandrogenic action, the effect of geldanamycin (GA) was evaluated in both in vitro assays and live cells. We demonstrated that in vitro hsp90s are required to stabilize the receptor in the inactive conformation and that hsp90 activity is involved in the integrity and nuclear transfer of agonist- and antagonist-bound AR. Furthermore, nuclear transfer is not the only step affected by GA since this compound was also active on a constitutively nuclear AR (GFP-NLS-AR). Hsp90 inactivation impedes interaction of androgen-bound GFP-NLS-AR with nuclear components and inhibits transcriptional activity. We conclude that hsp90s are required for the acquisition of active conformation in agonist-bound AR to regulate nuclear transfer, nuclear matrix binding, and transcriptional activity. Pure antiandrogens block the transconformational change of AR in an intermediary complex unable to acquire the active conformation and to dissociate the hsp90.


Asunto(s)
Antagonistas de Andrógenos/química , Antagonistas de Andrógenos/farmacología , Proteínas HSP90 de Choque Térmico/fisiología , Receptores Androgénicos/química , Receptores Androgénicos/fisiología , Transactivadores/química , Transactivadores/fisiología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Animales , Benzoquinonas , Células COS , Línea Celular , Núcleo Celular/química , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Endopeptidasas , Proteínas Fluorescentes Verdes , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Hidrólisis , Cinética , Lactamas Macrocíclicas , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Plásmidos , Conformación Proteica , Quinonas/farmacología , Receptores Androgénicos/genética , Transactivadores/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA