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.
Mol Cell Endocrinol ; 594: 112373, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39299378

RESUMEN

Thyroid hormone receptor α1 (TRα1) undergoes nucleocytoplasmic shuttling and mediates gene expression in response to thyroid hormone (T3). In Resistance to Thyroid Hormone Syndrome α (RTHα), certain TRα1 mutants have higher affinity for nuclear corepressor 1 (NCoR1) and may form stable complexes that are not released in the presence of T3. Here, we examined whether NCoR1 modulates intranuclear mobility and nuclear retention of TRα1 or RTHα-associated mutants in transfected human cells, as a way of analyzing critical structural components of TRα1 and to further explore the correlation between mutations in TRα1 and aberrant intracellular trafficking. We found no significant difference in intranuclear mobility, as measured by fluorescence recovery after photobleaching, between TRα1 and select RTHα mutants, irrespective of NCoR1 expression. Nuclear-to-cytoplasmic fluorescence ratios of RTHα mutants, however, varied from TRα1 when NCoR1 was overexpressed, with a significant increase in nuclear retention for A263V and a significant decrease for A263S and R384H. In NCoR1-knockout cells, nuclear retention of A263S, A263V, P389R, A382P, C392X, and F397fs406X was significantly decreased compared to control (wild-type) cells. Luciferase reporter gene transcription mediated by TRα1 was significantly repressed by both NCoR1 overexpression and NCoR1 knockout. Most RTHα mutants showed minimal induction regardless of NCoR1 levels, but T3-mediated transcriptional activity was decreased for R384C and F397fs406X when NCoR1 was overexpressed, and also decreased for N359Y in NCoR1-knockout cells. Our results suggest a complex interaction between NCoR1 and RTHα mutants characterized by aberrant intracellular localization patterns and transcriptional activity that potentially arise from variable repressor complex stability, and may provide insight into RTHα pathogenesis on a molecular and cellular level.

2.
Mol Cell Endocrinol ; 593: 112334, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39059458
3.
Mol Cell Endocrinol ; 559: 111781, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36191835

RESUMEN

Thyroid hormone receptor (TR) controls the expression of thyroid hormone (T3)-responsive genes, while undergoing rapid nucleocytoplasmic shuttling. In Resistance to Thyroid Hormone syndrome (RTH), mutant TR fails to activate T3-dependent transcription. Previously, we showed that Mediator subunit 1 (MED1) plays a role in TR nuclear retention. Here, we investigated MED1's effect on RTH mutants using nucleocytoplasmic scoring and fluorescence recovery after photobleaching in transfected cells. MED1 overexpression and knockout did not change the nucleocytoplasmic distribution or intranuclear mobility of C392X and P398R TRα1 at physiological T3 levels. At elevated T3 levels, however, overexpression increased P398R's nuclear retention and MED1 knockout decreased P398R's and A263V's intranuclear mobility, while not impacting C392X. Although A263V TRα1-transfected cells had a high percentage of aggregates, MED1 rescued A263V's impaired intranuclear mobility, suggesting that MED1 ameliorates nonfunctional aggregates. Results correlate with clinical severity, suggesting that altered interaction between MED1 and TRα1 mutants contributes to RTH pathology.


Asunto(s)
Receptores de Hormona Tiroidea , Síndrome de Resistencia a Hormonas Tiroideas , Humanos , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Mutación/genética , Síndrome de Resistencia a Hormonas Tiroideas/genética , Núcleo Celular/metabolismo , Hormonas Tiroideas/metabolismo , Triyodotironina/metabolismo , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/metabolismo
4.
J Biol Chem ; 296: 100677, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33887322

RESUMEN

A year ago, when I first contemplated writing this article, my intent was to provide a detailed review of the contributions of the diverse community of talented scientists in my lab to the nuclear receptor research field. In the throes of a deadly pandemic, political turmoil, and Black Lives Matter, however, I found myself compelled to tell a more personal story. While I will still cover milestones in our understanding of the intracellular trafficking of the thyroid hormone receptor, now these will be set against the backdrop of my path as a woman in STEM and on being intentionally inclusive. By sharing reflections on my journey, I hope to encourage young investigators to persist in their pursuit of a career in science.


Asunto(s)
Selección de Profesión , Narración , Receptores de Hormona Tiroidea/metabolismo , Femenino , Humanos , Transporte de Proteínas
5.
J Cell Biochem ; 121(4): 2909-2926, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31692077

RESUMEN

The thyroid hormone receptors (TRs) mediate thyroid hormone (T3 )-dependent gene expression. The nuclear import and export signals that direct TR shuttling are well characterized, but little is known about factors modulating nuclear retention. We used fluorescence-based nucleocytoplasmic scoring and fluorescence recovery after photobleaching in transfected cells to investigate whether Mediator subunits MED1 and MED13 play a role in nuclear retention of TR. When MED1 was overexpressed, there was a striking shift towards a greater nuclear localization of TRß1 and the oncoprotein v-ErbA, subtypes with cytosolic populations at steady-state, and TRß1 intranuclear mobility was reduced. For TRα1, there was no observable change in its predominantly nuclear distribution pattern or mobility. Consistent with a role for MED1 in nuclear retention, the cytosolic TRα1 and TRß1 population were significantly greater in MED1-/- cells, compared with MED1+/+ cells. Exposure to T3 and epidermal growth factor, which induces MED1 phosphorylation, also altered TR intranuclear dynamics. Overexpression of miR-208a, which downregulates MED13, led to a more cytosolic distribution of nuclear-localized TRα1; however, overexpression of MED13 had no effect on TRß1 localization. The known binding site of MED1 overlaps with a transactivation domain and nuclear export signal in helix 12 of TR's ligand-binding domain (LBD). Coimmunoprecipitation assays demonstrated that TR's LBD interacts directly with exportins 5 and 7, suggesting that binding of exportins and MED1 to TR may be mutually exclusive. Collectively, our data provide evidence that MED1 promotes nuclear retention of TR, and highlight the dual functionality of helix 12 in TR transactivation and nuclear export.


Asunto(s)
Subunidad 1 del Complejo Mediador/metabolismo , Proteínas Oncogénicas v-erbA/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Transporte Activo de Núcleo Celular , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , Fibroblastos/metabolismo , Expresión Génica , Genes erbA , Células HeLa , Humanos , Carioferinas/metabolismo , Complejo Mediador/metabolismo , Ratones , Fosforilación , Transporte de Proteínas , Receptores beta de Hormona Tiroidea/metabolismo , Hormonas Tiroideas/metabolismo , Transfección
6.
Artículo en Inglés | MEDLINE | ID: mdl-35280700

RESUMEN

The thyroid hormone receptor (TR) is essential for the proper regulation of metabolism and development, as it regulates gene expression in response to thyroid hormone. Nuclear localization signals (NLSs) and nuclear export signals (NESs) allow for TR transport into and out of the nucleus, respectively. Previous research suggests that nuclear import, nuclear retention, and nuclear export of TR are associated with modulation of gene expression, the alteration of which can contribute to various diseases. Here, we examined the impact of cancer-associated mutations on TR localization patterns as a way of analyzing key structural components of TR and to further explore the correlation between TR trafficking, misfolding, and disease. Through mammalian cell transfection of expression plasmids for green fluorescent protein (GFP) and mCherry-tagged TRα1 and quantitative fluorescence microscopy, we examined particular groups of TRα1 mutations that were observed in patients with hepatocellular carcinoma, renal cell carcinoma, and thyroid cancer, and are associated with NLSs and NESs of TRα1. We also investigated structural alterations of the mutants by in silico modeling. Our results show striking shifts towards a more cytoplasmic localization for many of the mutants and an increased tendency to form cytosolic and nuclear aggregates.

8.
Mol Cell Endocrinol ; 495: 110509, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31319097

RESUMEN

The thyroid hormone receptor (TR) undergoes nucleocytoplasmic shuttling, but is primarily nuclear-localized and mediates expression of genes involved in development and homeostasis. Given the proximity of TR acetylation and sumoylation sites to nuclear localization (NLS) and nuclear export signals, we investigated their role in regulating intracellular localization. The nuclear/cytosolic fluorescence ratio (N/C) of fluorescent protein-tagged acetylation mimic, nonacetylation mimic, and sumoylation-deficient TR was quantified in transfected mammalian cells. While nonacetylation mimic and sumoylation-deficient TRs displayed wild-type N/C, the acetylation mimic's N/C was significantly lower. Importins that interact with wild-type TR also interact with acetylation and nonacetylation mimics, suggesting factors other than reduced importin binding alter nuclear localization. FRAP analysis showed wild-type intranuclear dynamics of acetylation mimic and sumoylation-deficient TRs, whereas the nonacetylation mimic had significantly reduced mobility and transcriptional activity. Acetyltransferase CBP/p300 inhibition enhanced TR's nuclear localization, further suggesting that nonacetylation correlates with nuclear retention, while acetylation promotes cytosolic localization.


Asunto(s)
Núcleo Celular/metabolismo , Espacio Intracelular/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Acetilación , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Células Hep G2 , Humanos , Carioferinas/metabolismo , Ligandos , Mutación/genética , Unión Proteica , Transporte de Proteínas , Sumoilación , Transcripción Genética , Factores de Transcripción p300-CBP/metabolismo
9.
Vitam Horm ; 106: 45-66, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29407444

RESUMEN

The thyroid hormone receptors, TRα1 and TRß1, are members of the nuclear receptor superfamily that forms one of the most abundant classes of transcription factors in multicellular organisms. Although primarily localized to the nucleus, TRα1 and TRß1 shuttle rapidly between the nucleus and cytoplasm. The fine balance between nuclear import and export of TRs has emerged as a critical control point for modulating thyroid hormone-responsive gene expression. Mutagenesis studies have defined two nuclear localization signal (NLS) motifs that direct nuclear import of TRα1: NLS-1 in the hinge domain and NLS-2 in the N-terminal A/B domain. Three nuclear export signal (NES) motifs reside in the ligand-binding domain. A combined approach of shRNA-mediated knockdown and coimmunoprecipitation assays revealed that nuclear entry of TRα1 is facilitated by importin 7, likely through interactions with NLS-2, and importin ß1 and the adapter importin α1 interacting with both NLS-1 and NLS-2. Interestingly, TRß1 lacks NLS-2 and nuclear import depends solely on the importin α1/ß1 heterodimer. Heterokaryon and fluorescence recovery after photobleaching shuttling assays identified multiple exportins that play a role in nuclear export of TRα1, including CRM1 (exportin 1), and exportins 4, 5, and 7. Even single amino acid changes in TRs dramatically alter their intracellular distribution patterns. We conclude that mutations within NLS and NES motifs affect nuclear shuttling activity, and propose that TR mislocalization contributes to the development of some types of cancer and Resistance to Thyroid Hormone syndrome.


Asunto(s)
Transporte de Proteínas/fisiología , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética
10.
J Endocrinol ; 237(1): R19-R34, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29440347

RESUMEN

The thyroid hormone receptors, TRα1, TRß1 and other subtypes, are members of the nuclear receptor superfamily that mediate the action of thyroid hormone signaling in numerous tissues to regulate important physiological and developmental processes. Their most well-characterized role is as ligand-dependent transcription factors; TRs bind thyroid hormone response elements in the presence or absence of thyroid hormone to facilitate the expression of target genes. Although primarily residing in the nucleus, TRα1 and TRß1 shuttle rapidly between the nucleus and cytoplasm. We have identified multiple nuclear localization signals and nuclear export signals within TRα1 and TRß1 that interact with importins and exportins, respectively, to mediate translocation across the nuclear envelope. More recently, enigmatic cytoplasmic functions have been ascribed to other TR subtypes, expanding the diversity of the cellular response to thyroid hormone. By integrating data on localization signal motifs, this review provides an overview of the complex interplay between TR's dynamic transport pathways and thyroid hormone signaling activities. We examine the variation in TR subtype response to thyroid hormone signaling, and what is currently known about regulation of the variety of tissue-specific localization patterns, including targeting to the nucleus, the mitochondria and the inner surface of the plasma membrane.


Asunto(s)
Estructuras Animales/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Estructuras Animales/efectos de los fármacos , Animales , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Transporte de Proteínas , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/farmacología , Distribución Tisular
11.
Mol Cell Endocrinol ; 419: 185-97, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26525414

RESUMEN

The thyroid hormone receptor α1 (TRα1) is a nuclear receptor for thyroid hormone that shuttles rapidly between the nucleus and cytoplasm. Our prior studies showed that nuclear import of TRα1 is directed by two nuclear localization signals, one in the N-terminal A/B domain and the other in the hinge domain. Here, we showed using in vitro nuclear import assays that TRα1 nuclear localization is temperature and energy-dependent and can be reconstituted by the addition of cytosol. In HeLa cells expressing green fluorescent protein (GFP)-tagged TRα1, knockdown of importin 7, importin ß1 and importin α1 by RNA interference, or treatment with an importin ß1-specific inhibitor, significantly reduced nuclear localization of TRα1, while knockdown of other importins had no effect. Coimmunoprecipitation assays confirmed that TRα1 interacts with importin 7, as well as importin ß1 and the adapter importin α1, suggesting that TRα1 trafficking into the nucleus is mediated by two distinct pathways.


Asunto(s)
Núcleo Celular/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Células HeLa , Humanos , Transporte de Proteínas , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Temperatura
12.
Mol Cell Endocrinol ; 411: 86-96, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25911113

RESUMEN

The thyroid hormone receptor (TR) undergoes nucleocytoplasmic shuttling and regulates target genes involved in metabolism and development. Previously, we showed that TR follows a CRM1/calreticulin-mediated nuclear export pathway. However, two lines of evidence suggest TR also follows another pathway: export is only partially blocked by leptomycin B (LMB), a CRM1-specific inhibitor; and we identified nuclear export signals in TR that are LMB-resistant. To determine whether other exportins are involved in TR shuttling, we used RNA interference and fluorescence recovery after photobleaching shuttling assays in transfected cells. Knockdown of exportins 4, 5, and 7 altered TR shuttling dynamics, and when exportins 5 and 7 were overexpressed, TR distribution shifted toward the cytosol. To further assess the effects of exportin overexpression, we examined transactivation of a TR-responsive reporter gene. Our data indicate that multiple exportins influence TR localization, highlighting a fine balance of nuclear import, retention, and export that modulates TR function.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Carioferinas/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Carioferinas/genética
13.
J Biol Chem ; 287(37): 31280-97, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22815488

RESUMEN

Thyroid hormone receptor (TR) is a member of the nuclear receptor superfamily that shuttles between the cytosol and nucleus. The fine balance between nuclear import and export of TR has emerged as a critical control point for modulating thyroid hormone-responsive gene expression; however, sequence motifs of TR that mediate shuttling are not fully defined. Here, we characterized multiple signals that direct TR shuttling. Along with the known nuclear localization signal in the hinge domain, we identified a novel nuclear localization signal in the A/B domain of thyroid hormone receptor α1 that is absent in thyroid hormone receptor ß1 and inactive in the oncoprotein v-ErbA. Our prior studies showed that thyroid hormone receptor α1 exits the nucleus through two pathways, one dependent on the export factor CRM1 and the other CRM1-independent. Here, we identified three novel CRM1-independent nuclear export signal (NES) motifs in the ligand-binding domain as follows: a highly conserved NES in helix 12 (NES-H12) and two additional NES sequences spanning helix 3 and helix 6, respectively. Mutations predicted to disrupt the α-helical structure resulted in a significant decrease in NES-H12 activity. The high degree of conservation of helix 12 suggests that this region may function as a key NES in other nuclear receptors. Furthermore, our mutagenesis studies on NES-H12 suggest that altered shuttling of thyroid hormone receptor ß1 may be a contributing factor in resistance to thyroid hormone syndrome. Taken together, our findings provide a detailed mechanistic understanding of the multiple signals that work together to regulate TR shuttling and transcriptional activity, and they provide important insights into nuclear receptor function in general.


Asunto(s)
Señales de Localización Nuclear/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Secuencias de Aminoácidos , Animales , Células HeLa , Humanos , Mutación , Señales de Localización Nuclear/genética , Proteínas Oncogénicas v-erbA/genética , Proteínas Oncogénicas v-erbA/metabolismo , Estructura Terciaria de Proteína , Ratas , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/genética
14.
Sci Total Environ ; 416: 225-31, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22209374

RESUMEN

Bioaccumulation of contaminants can occur across ecosystem boundaries via transport by emergent aquatic insects. In the South River, Virginia, USA, aquatic mercury has contaminated songbirds nesting in adjacent riparian forests. Spiders contribute the majority of mercury to these songbirds' diets. We tested the hypothesis that massive annual mayfly emergences provide a vector for mercury from river sediments to the Lycosid spiders most frequently eaten by contaminated songbirds. We designed mayfly-specific PCR primers that amplified mtDNA from 76% of adult mayflies collected at this site. By combining this approach with an Agilent 2100 electrophoresis system, we created a highly sensitive test for mayfly predation by Lycosids, commonly known as wolf spiders. In laboratory spider feeding trials, mayfly DNA could be detected up to 192h post-ingestion; however, we detected no mayfly predation in a sample of 110 wolf spiders collected at the site during mayfly emergence. We suggest that mayfly predation is not an important mechanism for dietary transfer of mercury to wolf spiders and their avian predators at the South River. Instead, floodplain soil should be considered as a potential proximate source for mercury in the terrestrial food web.


Asunto(s)
Contaminantes Ambientales/efectos adversos , Insectos/efectos de los fármacos , Mercurio/efectos adversos , Reacción en Cadena de la Polimerasa/métodos , Arañas/efectos de los fármacos , Animales , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/análisis , Insectos/química , Insectos/genética , Datos de Secuencia Molecular , Conducta Predatoria/efectos de los fármacos , Alineación de Secuencia , Arañas/química , Arañas/genética , Arañas/fisiología
15.
Mol Cell Endocrinol ; 332(1-2): 196-212, 2011 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-21075170

RESUMEN

Aggresome formation, a cellular response to misfolded protein aggregates, is linked to cancer and neurodegenerative disorders. Previously we showed that Gag-v-ErbA (v-ErbA), a retroviral variant of the thyroid hormone receptor (TRα1), accumulates in and sequesters TRα1 into cytoplasmic foci. Here, we show that foci represent v-ErbA targeting to aggresomes. v-ErbA colocalizes with aggresomal markers, proteasomes, hsp70, HDAC6, and mitochondria. Foci have hallmark characteristics of aggresomes: formation is microtubule-dependent, accelerated by proteasome inhibitors, and they disrupt intermediate filaments. Proteasome-mediated degradation is critical for clearance of v-ErbA and T(3)-dependent TRα1 clearance. Our studies highlight v-ErbA's complex mode of action: the oncoprotein is highly mobile and trafficks between the nucleus, cytoplasm, and aggresome, carrying out distinct activities within each compartment. Dynamic trafficking to aggresomes contributes to the dominant negative activity of v-ErbA and may be enhanced by the viral Gag sequence. These studies provide insight into novel modes of oncogenesis across multiple cellular compartments.


Asunto(s)
Cuerpos de Inclusión/metabolismo , Proteínas Oncogénicas v-erbA/metabolismo , Alpharetrovirus/genética , Alpharetrovirus/metabolismo , Transporte Biológico , Biomarcadores/metabolismo , Dineínas/metabolismo , Eritroblastos/citología , Eritroblastos/metabolismo , Eritroblastos/virología , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Células HeLa , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Proteínas Oncogénicas v-erbA/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Vimentina/metabolismo
16.
Biochem J ; 427(3): 349-57, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20180778

RESUMEN

MK-STYX [MAPK (mitogen-activated protein kinase) phospho-serine/threonine/tyrosine-binding protein] is a pseudophosphatase member of the dual-specificity phosphatase subfamily of the PTPs (protein tyrosine phosphatases). MK-STYX is catalytically inactive due to the absence of two amino acids from the signature motif that are essential for phosphatase activity. The nucleophilic cysteine residue and the adjacent histidine residue, which are conserved in all active dual-specificity phosphatases, are replaced by serine and phenylalanine residues respectively in MK-STYX. Mutations to introduce histidine and cysteine residues into the active site of MK-STYX generated an active phosphatase. Using MS, we identified G3BP1 [Ras-GAP (GTPase-activating protein) SH3 (Src homology 3) domain-binding protein-1], a regulator of Ras signalling, as a binding partner of MK-STYX. We observed that G3BP1 bound to native MK-STYX; however, binding to the mutant catalytically active form of MK-STYX was dramatically reduced. G3BP1 is also an RNA-binding protein with endoribonuclease activity that is recruited to 'stress granules' after stress stimuli. Stress granules are large subcellular structures that serve as sites of mRNA sorting, in which untranslated mRNAs accumulate. We have shown that expression of MK-STYX inhibited stress granule formation induced either by aresenite or expression of G3BP itself; however, the catalytically active mutant MK-STYX was impaired in its ability to inhibit G3BP-induced stress granule assembly. These results reveal a novel facet of the function of a member of the PTP family, illustrating a role for MK-STYX in regulating the ability of G3BP1 to integrate changes in growth-factor stimulation and environmental stress with the regulation of protein synthesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Gránulos Citoplasmáticos/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Animales , Células COS , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , ADN Helicasas , Fosfatasas de Especificidad Dual/genética , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica/genética , Unión Proteica/fisiología , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Transfección
17.
J Biol Chem ; 283(37): 25576-25588, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18641393

RESUMEN

The thyroid hormone receptor alpha1 (TRalpha) exhibits a dual role as an activator or repressor of its target genes in response to thyroid hormone (T(3)). Previously, we have shown that TRalpha, formerly thought to reside solely in the nucleus bound to DNA, actually shuttles rapidly between the nucleus and cytoplasm. An important aspect of the shuttling activity of TRalpha is its ability to exit the nucleus through the nuclear pore complex. TRalpha export is not sensitive to treatment with the CRM1-specific inhibitor leptomycin B (LMB) in heterokaryon assays, suggesting a role for an export receptor other than CRM1. Here, we have used a combined approach of in vivo fluorescence recovery after photobleaching experiments, in vitro permeabilized cell nuclear export assays, and glutathione S-transferase pull-down assays to investigate the export pathway used by TRalpha. We show that, in addition to shuttling in heterokaryons, TRalpha shuttles rapidly in an unfused monokaryon system as well. Furthermore, our data show that TRalpha directly interacts with calreticulin, and point to the intriguing possibility that TRalpha follows a cooperative export pathway in which both calreticulin and CRM1 play a role in facilitating efficient translocation of TRalpha from the nucleus to cytoplasm.


Asunto(s)
Calreticulina/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , Transporte Activo de Núcleo Celular , Animales , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Ácidos Grasos Insaturados/farmacología , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Ratones , Modelos Biológicos , Proteína Exportina 1
18.
J Biol Chem ; 283(16): 10568-80, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18270197

RESUMEN

The androgen receptor undergoes nuclear import in response to ligand, but the mechanism by which it undergoes nuclear export is poorly understood. We developed a permeabilized cell assay to characterize nuclear export of the androgen receptor in LNCaP prostate cancer cells. We found that nuclear export of endogenous androgen receptor can be stimulated by short double-stranded DNA oligonucleotides. This androgen receptor export pathway is dependent on ATP hydrolysis and is enhanced by phosphatase inhibition with okadaic acid. Fluorescence recovery after photobleaching in permeabilized cells, under the conditions that stimulate androgen receptor export, suggested that double-stranded DNA-dependent export does not simply reflect the relief of a nuclear retention mechanism. A radiolabeled androgen was used to show that the androgen receptor remains ligand-bound during translocation through the nuclear pore complex. A specific inhibitor to the DNA-dependent protein kinase, NU7026, inhibits androgen receptor export and phosphorylation. In living cells, NU7026 treatment increases androgen-dependent transcription from endogenous genes that are regulated by androgen receptor. We suggest that DNA-dependent protein kinase phosphorylation of the androgen receptor, or an interacting component, helps target the androgen receptor for export from the nucleus.


Asunto(s)
Cromonas/farmacología , Proteína Quinasa Activada por ADN/metabolismo , Morfolinas/farmacología , Receptores Androgénicos/metabolismo , Transporte Activo de Núcleo Celular , Adenosina Trifosfato/química , Línea Celular Tumoral , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Proteínas Fluorescentes Verdes/química , Humanos , Hidrólisis , Técnicas In Vitro , Ligandos , Modelos Biológicos , Oligonucleótidos/química , Fosforilación , Unión Proteica
19.
Nucl Recept Signal ; 4: e008, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16741566

RESUMEN

Nuclear receptors (NRs) are transcription factors whose activity is modulated by ligand binding. These receptors are at the core of complex signaling pathways and act as integrators of many cellular signals. In the last decade our understanding of NRs has greatly evolved. In particular, regulation of NR subcellular dynamics has emerged as central to their activity. Research on the subcellular distribution of the thyroid hormone receptor (TR) has revealed new dimensions in the complexity of NR regulation, and points to the possibility that NR mislocalization plays a key role in oncogenesis. For many years, TR was thought to reside exclusively in the nucleus. It is now known that TR is a dynamic protein that shuttles between the nucleus and cytoplasm. TR is localized to the nucleus in a phosphorylated form, suggesting that compartment-specific phosphorylation mediates cross-talk between multiple cell signaling pathways. The oncoprotein v-ErbA, a viral-derived dominant negative variant of TR is actively exported to the cytoplasm by the CRM1 export receptor. Strikingly, the oncoprotein causes mislocalization of cellular TR and some of its coactivators by direct interaction. Here, we offer some perspectives on the role of subcellular trafficking in the oncogenic conversion of TR, and propose a new model for oncoprotein dominant negative activity.

20.
Mol Endocrinol ; 19(5): 1213-30, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15650025

RESUMEN

The retroviral v-ErbA oncoprotein is a highly mutated variant of the thyroid hormone receptor alpha (TRalpha), which is unable to bind T(3) and interferes with the action of TRalpha in mammalian and avian cancer cells. v-ErbA dominant-negative activity is attributed to competition with TRalpha for T(3)-responsive DNA elements and/or auxiliary factors involved in the transcriptional regulation of T(3)-responsive genes. However, competition models do not address the altered subcellular localization of v-ErbA and its possible implications in oncogenesis. Here, we report that v-ErbA dimerizes with TRalpha and the retinoid X receptor and sequesters a significant fraction of the two nuclear receptors in the cytoplasm. Recruitment of TRalpha to the cytoplasm by v-ErbA can be partially reversed in the presence of ligand and when chromatin is disrupted by the histone deacetylase inhibitor trichostatin A. These results define a new mode of action of v-ErbA and illustrate the importance of cellular compartmentalization in transcriptional regulation and oncogenesis.


Asunto(s)
Neoplasias/metabolismo , Proteínas Oncogénicas v-erbA/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Citoplasma/metabolismo , Dimerización , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Carioferinas/metabolismo , Ligandos , Ratones , Células 3T3 NIH , Proteínas Oncogénicas v-erbA/genética , Transporte de Proteínas/fisiología , Receptor beta X Retinoide/metabolismo , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA